[go: up one dir, main page]

WO2025087287A1 - Use of cholesin in modulating cholesterol homeostasis - Google Patents

Use of cholesin in modulating cholesterol homeostasis Download PDF

Info

Publication number
WO2025087287A1
WO2025087287A1 PCT/CN2024/126779 CN2024126779W WO2025087287A1 WO 2025087287 A1 WO2025087287 A1 WO 2025087287A1 CN 2024126779 W CN2024126779 W CN 2024126779W WO 2025087287 A1 WO2025087287 A1 WO 2025087287A1
Authority
WO
WIPO (PCT)
Prior art keywords
cholesin
cholesterol
protein
subject
plasma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2024/126779
Other languages
French (fr)
Chinese (zh)
Inventor
王一国
胡晓丽
陈丰艺
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tsinghua University
Original Assignee
Tsinghua University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tsinghua University filed Critical Tsinghua University
Publication of WO2025087287A1 publication Critical patent/WO2025087287A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics

Definitions

  • the present invention belongs to the field of biomedicine, and particularly relates to the use of the intestinal secretory hormone Cholesin in regulating cholesterol homeostasis, and the use of the intestinal secretory hormone Cholesin in regulating cholesterol homeostasis after being used in combination with other cholesterol-lowering drugs.
  • Cholesterol homeostasis is coordinated by various tissues to maintain a balance between dietary cholesterol absorption, new synthesis, and bile clearance and excretion. Elevated blood cholesterol levels, especially low-density lipoprotein cholesterol (LDL-C), contribute to the development of atherosclerosis and are considered an important risk factor for cardiovascular disease (CVD). CVD is a pressing public health problem, accounting for more than 30% of deaths worldwide. Both dietary cholesterol and bile cholesterol are taken up by Niemann-Pick type C1 like 1 (NPC1L1) proteins on the apical surface of enterocytes in the intestine. After absorption, dietary cholesterol is released in the form of chylomicrons, which are then taken up by the liver, the major site of cholesterol synthesis. Cholesterol molecules from new synthesis and intestinal absorption are transported to cells for use via the circulation. To maintain cholesterol homeostasis, excess cholesterol is excreted from the body via bile secretion into the feces.
  • NPC1L1 Niemann-P
  • New cholesterol synthesis is an energy-consuming process in which cholesterol is generated from acetyl CoA under the control of a series of enzymes.
  • HMGCR 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase
  • SREBP2 sterol regulatory element binding protein 2
  • Cholesterol biosynthesis and uptake are tightly regulated by a negative feedback mechanism that senses the cholesterol level of the cell.
  • SREBP2 When cells are cholesterol-deficient, SREBP2, together with its escort protein SCAP (SREBP cleavage-activating protein), is transported from the endoplasmic reticulum (ER) to the Golgi apparatus in COPII vesicles.
  • SREBP2 is cleaved by site 1 and site 2 proteases in sequence.
  • the N-terminal structure of SREBP2 released by this cleavage travels to the nucleus, where it acts as a transcription factor, enhancing the expression of genes involved in cholesterol synthesis and uptake.
  • INSIG insulin-induced gene
  • VLDL very low-density lipoproteins
  • LDLR LDL receptor
  • statins reduce cholesterol synthesis by inhibiting HMGCR and increase LDL uptake by upregulating LDLR.
  • PCSK9 inhibitors stabilize LDLR and promote liver absorption of LDL.
  • statins and PCSK9 inhibitors are both LDLR-dependent, which poses a challenge for patients with defective or insufficient LDLR.
  • compensatory increases in HMGCR levels worsen the outcomes of some patients who stop taking statins after long-term treatment.
  • statins can greatly increase the expression of Hmgcr and other cholesterol-causing genes, and their efficacy has been questioned.
  • the search for new cholesterol-lowering drugs is of great biological significance for the treatment of diseases associated with elevated plasma cholesterol levels, including hypercholesterolemia and atherosclerosis.
  • the present invention aims to provide a method for treating diseases associated with elevated plasma cholesterol levels, including hypercholesterolemia and atherosclerosis.
  • Cholesin is a hormone secreted by the intestine in response to cholesterol absorption, which has not been reported before, and can inhibit the synthesis of cholesterol and the secretion of VLDL in the liver, thereby reducing the level of plasma cholesterol to prevent diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • Mechanistic studies have shown that Cholesin inhibits cholesterol synthesis controlled by PKA signaling and SREBP2 by binding to its receptor GPR146 (Figure 30).
  • the Cholesin-GPR146 axis is a molecular link between intestinal absorption of cholesterol and inhibition of cholesterol synthesis in the liver. Therefore, exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin protein in a subject can reduce the level of plasma cholesterol and/or triglycerides, thereby preventing or treating diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • Cholesin has an LDLR-independent downregulation effect on the expression of genes involved in cholesterol synthesis and reduces plasma cholesterol levels, which means that Cholesin treatment is an effective strategy to combat diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • Cholesin can reduce Combining Cholesin with existing cholesterol-lowering drugs such as statins is also a promising strategy for treating diseases associated with elevated plasma cholesterol levels, including hyperlipidemia and atherosclerosis.
  • the present invention provides a method for lowering plasma cholesterol level and/or triglyceride level in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the plasma cholesterol is selected from plasma total cholesterol and plasma LDL-cholesterol.
  • the present invention provides a method for inhibiting hepatic cholesterol synthesis in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the method inhibits the expression of a gene associated with cholesterol synthesis, and the gene is selected from Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle. In some embodiments, the method inhibits the expression of a gene associated with cholesterol uptake, and the gene is Ldlr.
  • the method inhibits the expression of a transcription factor that promotes the expression of genes involved in cholesterol synthesis or uptake, and the transcription factor is SREBP2.
  • the present invention provides a method for inhibiting hepatic VLDL-cholesterol secretion in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the present invention provides a method for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the Cholesin protein is a human Cholesin protein.
  • the nucleic acid is DNA or RNA.
  • the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • a lentiviral vector an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • AAV adeno-associated virus
  • the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.
  • hyperlipidemia e.g., hypercholesterolemia, hypertriglyceridemia
  • cardiovascular disease e.g., myocardial infarction, coronary heart disease
  • cerebrovascular disease e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction
  • hyperglycemia e.g., hypercholesterolemia, hypertriglyceridemia
  • atherosclerosis e.g., myocardial infarction, coronary heart disease
  • the disease associated with elevated plasma cholesterol levels is hypercholesterolemia.
  • the disease associated with elevated plasma cholesterol levels is atherosclerosis.
  • the method further comprises the step of administering a second therapeutic agent, preferably, the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.
  • the second therapeutic agent is a cholesterol-lowering drug selected from the group consisting of an HMGCR inhibitor, a PCSK9 inhibitor, and a cholesterol absorption inhibitor.
  • the HMGCR inhibitor is a statin.
  • the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin.
  • the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.
  • the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.
  • the second therapeutic agent is administered before, after, or simultaneously with the Cholesin protein, the nucleic acid, or the expression vector.
  • the subject is a human.
  • the subject is unresponsive or intolerant to one or more of a cholesterol absorption inhibitor, an HMGCR inhibitor, and a PCSK9 inhibitor, for example, the subject has an LDL receptor (LDLR) defect.
  • LDLR LDL receptor
  • the present invention provides a composition comprising a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of human endogenous Cholesin protein, and a pharmaceutically acceptable carrier or excipient.
  • composition is used for one or more of the following:
  • the Cholesin protein is a human Cholesin protein.
  • the composition further comprises a second therapeutic agent, preferably, the second therapeutic agent is selected from protein Proteins or peptides, nucleic acids and small molecule drugs.
  • the second therapeutic agent is selected from a cholesterol-lowering drug.
  • the cholesterol-lowering drug is selected from the group consisting of an HMGCR inhibitor, a PCSK9 inhibitor, and a cholesterol absorption inhibitor.
  • the HMGCR inhibitor is a statin.
  • the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin.
  • the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.
  • the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.
  • the composition is formulated as an injection preparation or an oral preparation, such as a tablet, a capsule, a granule, a suspension.
  • Figure 1 Silver staining and immunoblotting results of serum proteins from mice fed a normal diet (RD) and a Western diet (WD).
  • RD normal diet
  • WD Western diet
  • FIG. 1A Localization of human Cholesin (hCholesin) and mouse Cholesin (mCholesin) in cells detected by immunofluorescence experiments.
  • Figure 2B Secretion of human and mouse Cholesin in the culture medium detected by immunoblotting.
  • FIG. 3A Plasma Cholesin levels of fasting, RD, and WD mice measured by ELISA.
  • FIG. 3B Changes in plasma Cholesin levels in human clinical samples after fasting and 1 h of feeding.
  • FIG. 4 Cholesin protein levels in different mouse tissues. Cholesin whole body knockout (KO) mice were used as controls.
  • Fig. 5A Plasma Cholesin levels and small intestinal total cholesterol levels in fasted and WD-fed WT and IKO mice.
  • FIG. 5B Plasma total cholesterol levels and plasma triglyceride levels in fasted and WD-fed WT and IKO mice.
  • FIG. 5C Plasma Cholesin levels and small intestinal total cholesterol levels in WT mice and IKO mice gavaged with corn oil (chol-) and cholesterol (chol+).
  • FIG. 5D Plasma total cholesterol levels and plasma triglyceride levels in WT mice and IKO mice gavaged with corn oil (chol-) and cholesterol (chol+).
  • Fig. 6 Cellular localization of Cholesin observed by immunofluorescence experiments.
  • Figure 7 Cholesin levels secreted in the culture medium and total cholesterol levels in cells after cholesterol stimulation of HCT116 cells overexpressing Cholesin.
  • Fig. 8A Cholesin secretion from Cholesin-overexpressing HCT116 cells with NPC1L1 knockdown (NPC1L1 KD) detected by immunoblotting. Cholesin-overexpressing HCT116 cells without NPC1L1 knockdown (KD) served as a control.
  • Fig. 8B The secreted Cholesin level in the culture medium and the total cholesterol level in the cells after co-culture with cholesterol in NPC1L1 knockdown (NPC1L1 KD) Cholesin-overexpressing HCT116 cells. NPC1L1 non-knockdown (NT) Cholesin-overexpressing HCT116 cells served as a control.
  • Figure 9A Cholesin levels in plasma and total cholesterol levels in the small intestine of mice treated with ezetimibe (Ezetimlbe+) after oral administration of cholesterol (chol+). Mice not treated with ezetimibe (Ezetimlbe-) served as controls.
  • Figure 9B Total cholesterol levels in plasma of mice treated with ezetimibe after oral administration of cholesterol. Mice not treated with ezetimibe served as controls.
  • Fig. 9C Cholesin secreted by Npc1l1 knockout (Npc1l1 -/- ) mice detected by immunoblotting. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout served as controls.
  • Fig. 9D shows the level of Cholesin in plasma and the level of total cholesterol in small intestine of Npc1l1 knockout (Npc1l1 -/- ) mice after oral administration of cholesterol. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout were used as controls.
  • Fig. 9E shows the total cholesterol level in plasma of Npc1l1 knockout (Npc1l1 -/- ) mice after oral administration of cholesterol. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout were used as controls.
  • FIG. 10A Correlation between plasma Cholesin level and plasma total cholesterol (TC) level in humans.
  • FIG. 10B Correlation between plasma Cholesin level and plasma low-density lipoprotein cholesterol (LDL-C) level.
  • FIG. 11A Plasma total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.
  • FIG. 11B Levels of total cholesterol and triglycerides in the liver of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.
  • FIG. 11C Plasma total cholesterol levels and levels of HDL, IDL/LDL, and VLDL in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD as measured by fast protein liquid chromatography.
  • FIG. 12A Body weight of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.
  • FIG 12B Wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD After body fat percentage.
  • FIG. 12C Food intake of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.
  • FIG. 12D Water intake of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.
  • FIG. 12E Small intestinal total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.
  • FIG. 12F Chylomicron breakdown rates in the small intestine of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD, as assessed by lipid tolerance assay.
  • FIG. 13A Gallbladder volume, bile total cholesterol concentration, and bile acid concentration in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.
  • FIG. 13B Fecal total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.
  • FIG. 14A mRNA levels of cholesterol synthesis-related genes in the liver of WT and IKO mice fed RD and WD as determined by qPCR.
  • FIG. 14B Protein expression levels of SREBP2, HMGCR, HMGCS1, and LDLR in the livers of WT and IKO mice fed RD and WD as determined by immunoblotting.
  • FIG. 15 Poloxamer 407-injected plasma VLDL secretion rate in WT and IKO mice fed RD and WD.
  • FIG. 16A Immunofluorescence staining of Cholesin binding to various tissues of wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 ⁇ / ⁇ ) mice.
  • FIG. 16B Immunofluorescence staining of Cholesin binding in primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 ⁇ / ⁇ ) mice.
  • FIG. 16C Saturation curves of Cholesin binding to wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 ⁇ / ⁇ ) mouse primary hepatocytes.
  • FIG. 17 Binding affinity curves of Cholesin to wild-type (WT) and mutant (Mut) GPR146.
  • FIG. 18 Interaction of GPR146 with GNAI1 detected by co-immunoprecipitation in the presence and absence of Cholesin.
  • FIG. 19 cAMP levels in primary hepatocytes of wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 ⁇ / ⁇ ) mice after Cholesin treatment.
  • FIG. 20 Hmgcr mRNA levels in mouse liver after intraperitoneal injection of Cholesin.
  • FIG. 21A Experimental scheme for intraperitoneal injection of Cholesin into mice.
  • Figure 21B Hmgcr mRNA levels in the liver of mice injected intraperitoneally with Cholesin.
  • FIG. 21C Total cholesterol levels in plasma of mice injected intraperitoneally with Cholesin.
  • FIG. 21D Total cholesterol levels in the liver of mice injected intraperitoneally with Cholesin.
  • FIG. 22A Experimental scheme for injection of adenovirus and Cholesin into mice.
  • Figure 22B mRNA levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, and Pmvk in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut), with or without Cholesin injection.
  • Vec adenovirus control
  • WT wild-type GPR146
  • Mot adenovirus expressing mutant GPR146
  • FIG22C Protein levels of SREBP2, HMGCR, HMGCS1, LDLR, and pKA Sub in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut) with or without Cholesin injection.
  • the expression level of HSP90 was used as an internal control.
  • FIG. 22D Total cholesterol levels in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut), with or without Cholesin injection.
  • Gpr146 fl/fl wild-type mice and Gpr146 knockout mice injected with adenovirus control
  • Vec adenovirus control
  • WT adenovirus expressing wild-type GPR146
  • Mot adenovirus expressing mutant GPR146
  • FIG. 23 Schematic diagram of the dosing regimen for Ldlr knockout (Ldlr ⁇ / ⁇ ) mice.
  • FIG. 24A Changes in plasma total cholesterol levels in Ldlr ⁇ / ⁇ mice under different dosing regimens and statistics of the area under the curve (AUC).
  • FIG. 24B Total cholesterol levels in the liver of Ldlr ⁇ / ⁇ mice after 8 weeks of drug administration under different dosing regimens.
  • FIG. 24C Changes in plasma triglyceride levels in Ldlr ⁇ / ⁇ mice under different dosing regimens and statistics of the area under the curve (AUC).
  • FIG. 24D Hepatic triglyceride levels in Ldlr ⁇ / ⁇ mice after 8 weeks of drug administration under different dosing regimens.
  • FIG. 25A mRNA levels of genes related to cholesterol synthesis in the liver of Ldlr ⁇ / ⁇ mice after 8 weeks of drug administration under different drug administration regimens.
  • FIG. 25B Expression levels of liver cholesterol synthesis-related genes Hmgcs1, Hmgcr and transcription factor SREBP2 in Ldlr -/- mice after 8 weeks of drug administration under different dosing regimens.
  • Figure 26 Oil red staining of the whole aorta of Ldlr -/- mice after 8 weeks of drug administration under different drug administration regimens and their statistical results.
  • FIG. 27A Body weight changes of Ldlr ⁇ / ⁇ mice under different dosing regimens.
  • FIG. 27B Changes in food intake of Ldlr ⁇ / ⁇ mice under different dosing regimens.
  • Figure 28 HE staining of liver tissue of Ldlr -/- mice after 8 weeks of drug administration under different dosing regimens.
  • FIG. 29A Plasma alanine aminotransferase (ALT) levels in Ldlr ⁇ / ⁇ mice after 8 weeks of drug administration under different dosing regimens.
  • FIG. 29B Plasma aspartate aminotransferase (AST) levels in Ldlr ⁇ / ⁇ mice after 8 weeks of administration under different dosing regimens.
  • Figure 30 Schematic diagram of the mechanism by which Cholesin regulates cholesterol metabolism through its receptor GPR146.
  • the term “and/or” is considered a specific disclosure of each of the two specified features or components with or without the other.
  • the term “and/or” as used in phrases such as "A and/or B” is intended to include A and B; A or B; A (alone); and B (alone).
  • the term “and/or” as used in phrases such as "A, B, and/or C” is intended to cover each of the following: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • Cholesin or “Cholesin protein” is a homolog of the unknown human protein C7orf50.
  • the human Cholesin gene is located in the p22 region of human chromosome 7.
  • the gene encoding human Cholesin has a gene ID of 84310 in NCBI, but its specific function has not yet been reported.
  • the inventors discovered its inhibitory effect on cholesterol synthesis through characterization of the protein, so it was named "Cholesin”.
  • the amino acid sequence of Cholesin protein is highly conserved in various species.
  • the amino acid sequences of Cholesin proteins in different species are as follows:
  • Brown rat (Rattus norvegicus) (Gene ID 498154)
  • Cholesin protein should be understood in its broadest sense, including wild-type, isolated or purified, or recombinant Cholesin protein, its variants or derivatives, as long as the desired biological activity (e.g., activity of reducing plasma cholesterol level and/or triglyceride level in a subject) is retained.
  • Variants or derivatives of Cholesin protein encompass, for example, variants or derivatives of Cholesin protein described herein that retain the ability to reduce plasma cholesterol level and/or triglyceride level in a subject to a degree similar to that of wild-type Cholesin protein, to the same degree as that of wild-type Cholesin protein, or to a degree higher than that of wild-type Cholesin protein.
  • the variant or derivative of the Cholesin protein retains at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8%, at least 99.9%, or 100% of the biological activity of the wild-type Cholesin protein, or has a higher biological activity than the wild-type Cholesin protein, particularly the activity of reducing the plasma cholesterol level and/or triglyceride level in a subject.
  • Variants of Cholesin protein include polypeptides that have significant sequence identity to wild-type Cholesin protein and retain the biological activity of Cholesin protein (e.g., activity that reduces plasma cholesterol levels and/or triglyceride levels in a subject). Variants may have one or more amino acid mutations compared to wild-type Cholesin protein. Mutations may include, for example, amino acid insertions, deletions, or substitutions.
  • the amino acid sequence of the variant may, for example, have at least about 65%, 70%, 75%, 80%, 90%, 95%, 96%, 97%, 98%, 98.2%, 98.4%, 98.6%, 98.8%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or higher identity to the amino acid sequence of wild-type Cholesin protein.
  • Derivatives of cholesin proteins generally refer to those that have been chemically modified, especially by one or more substituents.
  • Typical derivatives can be, for example, amidated, glycosylated, alkylated, acylated, esterified, pegylated Cholesin protein or its variants.
  • a step of enhancing the expression or activity of an endogenous Cholesin protein in a subject refers to any method or substance that can enhance the stability or expression of a nucleic acid encoding a Cholesin protein, enhance the transcription and translation of Cholesin, increase the level of Cholesin protein, enhance the activity of Cholesin protein, enhance the stability of Cholesin protein, and increase the effective action time of Cholesin protein.
  • an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject can be a small molecule, a protein, a polypeptide, a nucleic acid, etc.
  • GPR146 is a G protein-coupled receptor (GPCR) that is involved in the regulation of cholesterol metabolism and inhibits cholesterol synthesis controlled by PKA signaling and SREBP2.
  • GPCR G protein-coupled receptor
  • Previous studies have found that GPR146 regulates the secretion of very low-density lipoprotein (VLDL) in the liver by activating the extracellular signal-regulated kinase (ERK) signaling pathway and promoting the activity of liver sterol regulatory element binding protein 2 (SREBP2), thereby regulating the levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) in the circulation.
  • ERK extracellular signal-regulated kinase
  • SREBP2 liver sterol regulatory element binding protein 2
  • the GPR146/ERK axis has a regulatory role in systemic cholesterol metabolism, and inhibiting GPR146 may be an effective strategy to reduce plasma cholesterol levels and atherosclerosis.
  • the human GPR146 gene ID is 115330, and the amino acid sequence is: MWSCSWFNGTGLVEELPACQDLQLGLSLLSLLGLVVGVPVGLCYNALLVLANLHSKASMTMPDVYFVNMAVAGLVLSALAPVHLLGPPSSRWALWSVGGEVHVALQIPFNVSSLVAMYSTALLSLDHYIERALPRTYMASVYNTRHVCGFVWGGALLTS FSSLLFYICSHVSTRALECAKMQNAEAADATLVFIGYVVPALATLYALVLLSRVRREDTPLDRDTGRLEPSAHRLLVATVCTQFGLWTPHYLILLGHTVIISRGKPVDAHYLGLLHFVKDFSKLLAFSSSFVTPLLYRYMNQSFPSKLQRLMKKLPCGDRHCSPDHMGVQQVLA(SEQ ID NO:7)
  • the mouse GPR146 gene ID is 80290, and the amino acid sequence is: MWSCGPLNSTAWAEEPLCRNLRLGLWVLSLLYLGAGVPVSLGYNALLVLANLASKNTMTMPDVYFVNMAVAGLVLTALAPAYLLGPAHSRWALWSLSSEAHVTLLILFNVASLVTMYSTALLSLDYYIERALPRTYMASVYNTRHVCGFVWGGAVLTSFS SLLFYICSHVSSRIAECARMQNTEAADAILVLIGYVVPGLAVLYALALISRIGKEDTPLDQDTSRLDPSVHRLLVATVCTQFGLWTPYYLSLGHTVLTSRGRTVEGHYLGILQVAKDLAKFLAFSSSSVTPLLYRYINKAFPGKLRRLMKKMHCGRRHCSPDPSGIQQVMAQA(SEQ ID NO: 8).
  • Cholesterol is the main component of cell membranes and is also the main component of the synthesis of adrenal cortex hormones, sex hormones, bile acids and vitamins.
  • concentration of cholesterol in human blood can be used as an indicator of lipid metabolism to assess the risk of cardiovascular and cerebrovascular diseases, hyperlipidemia, atherosclerosis and other diseases.
  • Cholesterol often exists in the form of lipoproteins in the blood. Lipoproteins are divided into five types according to particle size and density, namely chylomicrons, very low-density lipoproteins, intermediate-density lipoproteins, low-density lipoproteins and high-density lipoproteins.
  • the very low-density lipoproteins synthesized by the liver transport triglycerides into the blood, where they are broken down by lipase to produce very low-density lipoprotein remnants, also called intermediate-density lipoproteins.
  • Low-density lipoproteins act on the whole body and are used by binding to low-density lipoprotein receptors of cells and tissues.
  • Low-density lipoprotein (LDL) in plasma is the main carrier for transporting endogenous cholesterol, which is degraded and converted by binding to the low-density lipoprotein receptor (LDLR) on its cell membrane. LDLR functional defects will reduce the clearance ability of plasma LDL, ultimately leading to the formation of atherosclerotic plaques in the arterial intima.
  • Low-density lipoprotein cholesterol (LDL-C) is the main lipoprotein in fasting plasma, accounting for about 2/3 of plasma lipoproteins, and is the main carrier for transporting cholesterol to extrahepatic tissues. Therefore, the content of LDL-C is related to the incidence and degree of cardiovascular disease, hyperlipidemia, atherosclerosis and other diseases.
  • LDL low-density lipoprotein
  • VLDL Very low-density lipoprotein
  • VLDL is synthesized in the liver and transports triglycerides and cholesterol to tissues through the blood. Since this type of lipoprotein carries a relatively small amount of cholesterol and their particles are relatively large, it is not easy to penetrate the vascular endothelium. Therefore, normal very low-density lipoprotein generally does not cause atherosclerosis.
  • VLDL very low-density lipoprotein
  • VLDL-C very low-density lipoprotein
  • Intermediate density lipoprotein is mainly an intermediate metabolite of very low density lipoprotein (VLDL), so it is also called residual VLDL. IDL can also be directly secreted by the liver, but its amount is very small. It is the last lipoprotein to be decomposed by lipoprotein lipase (LPL) among lipoproteins containing apolipoprotein B.
  • VLDL very low density lipoprotein
  • LPL lipoprotein lipase
  • High-density lipoprotein is a group of plasma lipoproteins with the smallest diameter but the highest density. In normal plasma, most HDL particles are spherical and consist of a hydrophobic core surrounded by phospholipids, unacylated cholesterol, and apolipoproteins. High-density lipoprotein cholesterol (HDL-C) can transport cholesterol from extrahepatic tissues to the liver for metabolism and excretion from the body by bile. In this article, “HDL” and “HDL-C” can be used interchangeably.
  • Chylomicrons are the largest lipoprotein particles in plasma and are the main lipoproteins for transporting exogenous triglycerides. Cholesterol and triglycerides contained in food are absorbed in the small intestine, through the lymphatic vessels, and absorbed through chylomicrons. After entering the blood, chylomicrons will become mature chylomicrons and, under the action of lipase, will be converted into free fatty acids. Free fatty acids are converted into energy by the human body on the one hand, and are also further synthesized and stored in fat.
  • Cholesterol synthesis occurs mainly in the liver. Cholesterol is generated from acetyl CoA under the control of a series of enzymes. Among them, HMGCR (3-hydroxy-3-methylglutaryl CoA reductase) is the rate-limiting enzyme, and its activity is controlled at the transcriptional and post-transcriptional levels. SREBP2 (sterol regulatory element binding protein 2) acts as a master transcription factor, dominating the expression of HMGCR and other genes involved in cholesterol synthesis and uptake. Cholesterol biosynthesis and uptake are strictly regulated by a negative feedback mechanism that senses the cholesterol level of the cell.
  • HMGCR 3-hydroxy-3-methylglutaryl CoA reductase
  • SREBP2 sterol regulatory element binding protein 2
  • Cholesterol biosynthesis and uptake are strictly regulated by a negative feedback mechanism that senses the cholesterol level of the cell.
  • SREBP2 When cells are cholesterol-deficient, SREBP2, together with its escort protein SCAP (SREBP cleavage-activating protein), is transported from the endoplasmic reticulum (ER) to the Golgi apparatus in COPII vesicles.
  • SREBP2 is cleaved by site 1 and site 2 proteases in sequence.
  • the N-terminal structure of SREBP2 released by this cleavage travels to the nucleus, where it acts as a transcription factor, enhancing the expression of genes involved in cholesterol synthesis (e.g., Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk, and Sqle).
  • VLDL very low-density lipoprotein
  • LDLR LDL receptor
  • expression vector is a nucleic acid molecule used as a medium for transferring (exogenous) genetic material into a host cell, where the nucleic acid molecule as a vector can, for example, be replicated and/or expressed.
  • expression vector encompasses, but is not limited to, plasmids, viral vectors (including, for example, retroviral vectors, lentiviral vectors, adenoviral vectors, vaccinia virus vectors, polyoma virus vectors, and adenovirus-associated vectors (AAV)), phages, phagemids, cosmids, and artificial chromosomes (including, for example, BACs and YACs).
  • viral vectors including, for example, retroviral vectors, lentiviral vectors, adenoviral vectors, vaccinia virus vectors, polyoma virus vectors, and adenovirus-associated vectors (AAV)
  • phages phagemids
  • cosmids
  • treatment includes therapeutic or prophylactic treatment in a subject in need thereof.
  • “Therapeutic or prophylactic treatment” includes prophylactic treatment aimed at completely preventing clinical and/or pathological manifestations or therapeutic treatment aimed at improving or alleviating clinical and/or pathological manifestations. Therefore, the term “treatment” also includes ameliorating or preventing a disease.
  • the term “effective amount” means an amount of a therapeutic agent that, when administered to a subject for the treatment or prevention of a disease, is sufficient to achieve such treatment or prevention.
  • the “effective amount” may vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • a “therapeutically effective amount” refers to an effective amount for therapeutic treatment.
  • a “prophylactically effective amount” refers to an effective amount for prophylactic treatment.
  • the terms "subject” or “individual” or “animal” or “patient” are used interchangeably herein and refer to any subject in need of treatment, particularly a mammalian subject.
  • mammalian subjects include humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cows, dairy cows, etc.
  • Dyslipidemia is an abnormal metabolism of lipoproteins in the human body, mainly including elevated total cholesterol, low-density lipoprotein cholesterol, triglycerides and/or reduced high-density lipoprotein cholesterol, etc.
  • Dyslipidemia can lead to hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease, angina pectoris, malignant arrhythmia), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, liver failure, nephrotic syndrome, gallstones, pancreatitis and other diseases or symptoms.
  • hyperlipidemia e.g., hypercholesterolemia, hypertriglyceridemia
  • atherosclerosis e.g., myocardial infarction, coronary heart disease,
  • Diseases related to fat metabolism disorder are a general term for related diseases caused by fat metabolism disorders, including, for example: hyperlipidemia, hypertension, atherosclerosis, obesity, fatty liver, cirrhosis, coronary heart disease, angina pectoris, myocardial infarction, inflammatory bowel disease, indigestion and gastrointestinal ulcers.
  • Hyperlipidemia usually refers to elevated levels of plasma cholesterol and/or triglycerides. Hyperlipidemia is a manifestation of abnormal fat metabolism in the human body and is mainly divided into three categories: hypercholesterolemia, hypertriglyceridemia, and mixed hyperlipidemia. Among them, hypercholesterolemia refers to elevated levels of total plasma cholesterol or low-density lipoprotein cholesterol. Hypercholesterolemia can lead to various life-threatening cardiovascular diseases, atherosclerosis and other complications.
  • Atherosclerosis is the most common and important type of vascular disease of arteriosclerosis.
  • Lipid metabolism disorder is the pathological basis of atherosclerosis, and its characteristic is that the lesions of the affected arteries start from the intima.
  • the lesions often involve elastic and large and medium muscular arteries. Once they develop to the point of blocking the arterial lumen, the tissues or organs supplied by the artery will be ischemic or necrotic. Because the lipids accumulated in the intima of the artery appear yellow and porridge-like, it is called atherosclerosis.
  • Atherosclerosis is often accompanied by diseases such as hypertension, diabetes, and obesity.
  • HMGCR inhibitor is also referred to as an HMG-CoA reductase inhibitor, and refers to a compound capable of inhibiting HMG-CoA reductase. It inhibits the biosynthesis of isoprenoids by inhibiting HMG-CoA reductase, blocks the isoprenylation of proteins, and thereby reduces plasma cholesterol levels.
  • PCSK9 inhibitor refers to a class of compounds that inhibit PCSK9 (the ninth member of the Kexin-like proconvertase subtilisin family).
  • PCSK9 is a liver-derived secretory protein that can reduce the number of LDLRs on hepatocytes, affect LDL internalization, and prevent LDL from being cleared from the blood, thereby leading to hypercholesterolemia. Therefore, inhibiting PCSK9 can stabilize LDLR and promote the liver's absorption of LDL, thereby reducing the level of LDL in the blood.
  • cholesterol absorption inhibitor refers to a compound that is capable of inhibiting the absorption of cholesterol, for example, by inhibiting NPC1L1-mediated endocytosis, thereby blocking the uptake of cholesterol.
  • composition refers in particular to a composition suitable for administration to humans. However, the term also generally encompasses compositions suitable for administration to non-human animals.
  • the composition and its components i.e., active agent and optional carrier or excipient
  • pharmaceutically acceptable means that the carrier or excipient is compatible with the other ingredients of the composition and not substantially toxic to the recipient thereof, and/or such carrier or excipient is approved or available for inclusion in the composition.
  • Cholesin is a hormone secreted by the intestine in response to cholesterol absorption, which can inhibit cholesterol synthesis and VLDL secretion in the liver, thereby reducing the levels of plasma cholesterol and/or triglycerides to prevent diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • Mechanistic studies have shown that Cholesin inhibits cholesterol synthesis controlled by PKA signaling and SREBP2 by binding to its receptor GPR146.
  • Cholesin has an LDLR-independent downregulation effect on the expression of genes involved in cholesterol synthesis and reduces plasma cholesterol levels, which means that Cholesin treatment is an effective strategy to combat diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin proteins in subjects can reduce the levels of plasma cholesterol and/or triglycerides, thereby preventing or treating diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis.
  • exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin protein in a subject with existing cholesterol-lowering drugs such as statins is also a promising strategy for treating diseases associated with elevated plasma cholesterol levels, including hyperlipidemia and atherosclerosis.
  • the present invention provides a method for lowering plasma cholesterol level and/or triglyceride level in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • plasma cholesterol is selected from plasma total cholesterol and plasma LDL-cholesterol. In some embodiments, plasma cholesterol is plasma total cholesterol. In some embodiments, plasma cholesterol is plasma LDL-cholesterol. In some embodiments, plasma cholesterol is plasma total cholesterol and plasma LDL-cholesterol.
  • the present invention provides a method for inhibiting liver cholesterol synthesis in a subject, comprising administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or a nucleic acid comprising the Cholesin protein.
  • the method inhibits the expression of genes related to cholesterol synthesis.
  • the genes related to cholesterol synthesis are selected from one or more of Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle.
  • the gene related to cholesterol synthesis is Hmgcr.
  • the gene related to cholesterol synthesis is Hmgcs1.
  • the gene related to cholesterol synthesis is Mvd.
  • the gene related to cholesterol synthesis is Mvk.
  • the gene related to cholesterol synthesis is Pmvk.
  • the gene related to cholesterol synthesis is Sqle.
  • the gene related to cholesterol synthesis is Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle.
  • the method inhibits the expression of a gene associated with cholesterol uptake.
  • the gene associated with cholesterol synthesis and uptake is Ldlr.
  • the method inhibits the expression of genes associated with cholesterol synthesis and uptake.
  • the genes associated with cholesterol synthesis are Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle; and the gene associated with cholesterol synthesis and uptake is Ldlr.
  • the method inhibits the expression of transcription factors that promote the expression of cholesterol synthesis-related genes. In some embodiments, the method inhibits the expression of transcription factors that promote the expression of cholesterol uptake-related genes. In some embodiments, the transcription factor that promotes the expression of cholesterol synthesis and/or uptake-related genes is SREBP2.
  • the present invention provides a method for inhibiting hepatic VLDL-cholesterol secretion in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the present invention provides a method for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.
  • the Cholesin protein is isolated or purified.
  • the Cholesin protein is a mammalian Cholesin protein.
  • mammalian Cholesin proteins include but are not limited to Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cows.
  • the Cholesin protein is a non-human primate Cholesin protein.
  • the Cholesin protein is a human Cholesin protein.
  • the human Cholesin protein comprises an amino acid sequence as shown in SEQ ID NO:1.
  • the Cholesin protein is a mouse Cholesin protein.
  • the mouse Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2.
  • the Cholesin protein is a macaque Cholesin protein.
  • the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3.
  • the Cholesin protein is a bovine Cholesin protein.
  • the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4.
  • the Cholesin protein is a Rattus norvegicus Cholesin protein.
  • the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5.
  • the Cholesin protein is a Gallus gallus Cholesin protein.
  • the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.
  • the nucleic acid is DNA or RNA. In some embodiments, the nucleic acid is DNA. In some embodiments, the nucleic acid encodes human Cholesin protein.
  • the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • a lentiviral vector an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • AAV adeno-associated virus
  • the expression vector itself is usually a nucleotide sequence, usually a DNA sequence containing an insert (transgene) and a larger sequence as a vector "backbone".
  • Engineered vectors usually include an origin of autonomous replication in a host cell (if stable expression of the polynucleotide is desired), a selection marker, and a restriction enzyme cleavage site (such as a multiple cloning site, MCS).
  • the vector may additionally include a promoter, a genetic marker, a reporter gene, a targeting sequence, and/or a protein purification tag. As known to those skilled in the art, a large number of suitable expression vectors are known to those skilled in the art, and many are commercially available.
  • the Cholesin protein disclosed herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent for enhancing the expression or activity of the endogenous Cholesin protein of a subject can be used for treating and/or preventing diseases associated with elevated plasma cholesterol levels, i.e., diseases, conditions or disorders induced by elevated plasma cholesterol levels, for example, for treating and/or preventing:
  • Dyslipidemia and its sequelae such as atherosclerosis, cardiovascular and cerebrovascular diseases, etc., especially those diseases characterized by one or more of the following factors (but not limited to):
  • metabolic syndrome A variety of other conditions that may be associated with metabolic syndrome, such as:
  • Heart failure such as (but not limited to) heart failure following myocardial infarction, hypertensive heart disease or cardiomyopathy;
  • diabetic vascular disease such as damage to large and small blood vessels leading to lesions of the heart, brain, kidneys, peripheral nerves, eyes, feet and other tissues and organs, including diabetic eye disease, diabetic heart disease, diabetic nephropathy, diabetic neuropathy and distal limb necrosis of the lower limbs, etc.
  • Fat metabolism disorders and related diseases that are common in diabetes, occur with diabetes, or are aggravated by diabetes, including hyperlipidemia, hypertension, atherosclerosis, obesity, fatty liver, and cirrhosis;
  • - atherosclerosis such as (but not limited to) coronary artery atherosclerosis, including angina pectoris or myocardial infarction, stroke,
  • the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.
  • hyperlipidemia e.g., hypercholesterolemia, hypertriglyceridemia
  • cardiovascular disease e.g., myocardial infarction, coronary heart disease
  • cerebrovascular disease e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction
  • hyperglycemia e.g., hypercholesterolemia, hypertriglyceridemia
  • atherosclerosis e.g., myocardial infarction, coronary heart disease
  • the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In other preferred embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.
  • the method further comprises the step of administering a second therapeutic agent.
  • the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.
  • the second therapeutic agent is a cholesterol-lowering drug.
  • the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.
  • the cholesterol-lowering drug is an HMGCR inhibitor.
  • the HMGCR inhibitor is a statin.
  • the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin.
  • the second therapeutic agent is rosuvastatin.
  • the cholesterol-lowering drug is a PCSK9 inhibitor.
  • the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.
  • the second therapeutic agent is Evolocumab.
  • the second therapeutic agent is Alirocumab.
  • the second therapeutic agent is Inclisiran.
  • the second therapeutic agent is Tafolecimab.
  • Evolocumab (Evolocumab, trade name ) is a fully human IgG2 monoclonal antibody. Evolocumab can bind to PCSK9 and inhibit the binding of circulating PCSK9 to the low-density lipoprotein receptor (LDLR), thereby preventing PCSK9-mediated low-density lipoprotein receptor degradation. Its approved indications are hypercholesterolemia and mixed dyslipidemia.
  • Alirocumab (Alirocumab, trade name ) is a fully human IgG1 monoclonal antibody that binds to PCSK9 and inhibits the binding of circulating PCSK9 to the low-density lipoprotein receptor (LDLR), thereby preventing PCSK9-mediated degradation of the low-density lipoprotein receptor.
  • the drug is used to treat adults with heterozygous familial hypercholesterolemia and clinical atherosclerotic cardiovascular disease (e.g., heart disease or stroke requiring low-density cholesterol reduction).
  • Inclisiran (Inclisiran, trade name ) is a small interfering nucleic acid (siRNA) drug targeting PCSK9 for lowering low-density lipoprotein cholesterol. It has been approved for the treatment of adult patients with primary hypercholesterolemia (heterozygous familial and non-familial) or mixed dyslipidemia.
  • siRNA small interfering nucleic acid
  • Tafolecimab (Tafolecimab, trade name ) is a fully human IgG2 monoclonal antibody. It can specifically bind to the PCSK9 molecule, increase the low-density lipoprotein receptor (LDLR) level by reducing PCSK9-mediated endocytosis of the LDLR, and then increase the clearance of low-density lipoprotein cholesterol (LDL-C) and reduce the LDL-C level.
  • LDLR low-density lipoprotein receptor
  • LDL-C low-density lipoprotein cholesterol
  • the cholesterol-lowering drug is a cholesterol absorption inhibitor.
  • the cholesterol absorption inhibitor is an NPC1L1 inhibitor.
  • NPC1L1 inhibitors target the cholesterol transporter NPC1L1, and inhibit the intracellular transport of cholesterol by acting on the key transporter NPC1L1 for cholesterol uptake and absorption on the brush border of the small intestinal villi, thereby reducing the absorption of cholesterol in food and bile in the small intestine, hindering the transport pathway of cholesterol from the small intestine to the liver, reducing the cholesterol storage in the liver, and accelerating the clearance of cholesterol.
  • the NPC1L1 inhibitor is selected from Ezetimibe and Hybutimibe.
  • the second therapeutic agent is Ezetimibe. In some embodiments, the second therapeutic agent is Hybutimibe.
  • the second therapeutic agent is administered before, after, or simultaneously with the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered before the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered after the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered simultaneously with the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein.
  • treatment may require a single administration of a therapeutically effective dose or multiple administrations of a therapeutically effective dose of an active agent of the invention.
  • the active agent may be administered one to three times a day, once every two days, once every 3 to 4 days, once a week, or once every two weeks, or once in a month.
  • the active agents disclosed herein can be applied to administration by various routes. Typically, administration is completed parenterally. Parenteral delivery methods include topical, intravenous, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.
  • the subject is a mammal. In a preferred embodiment, the subject is a human. In some embodiments, the subject is resistant to cholesterol absorption inhibitors, HMGCR inhibitors, and PCSK9 inhibitors. The subject may be unresponsive or intolerant to one or more of the agents, for example, the subject has an LDL receptor (LDLR) defect.
  • LDLR LDL receptor
  • the effective amount of the Cholesin protein disclosed herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the endogenous Cholesin protein of the subject can be determined by those skilled in the art using known techniques.
  • the appropriate dosage provides a sufficient amount of the active agent of the present invention, and is preferably therapeutically effective, that is, sufficient to cause, for example, a therapeutic or preventive response in the subject or animal within a reasonable time frame.
  • the dosage of the Cholesin protein of the present invention, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the endogenous Cholesin protein of the subject should be sufficient to produce a therapeutic or preventive response within a period of about 20 minutes, 30 minutes, 1 hour, 2 hours or longer, such as 12 hours to 24 hours or longer (e.g., 1 month, 2 months, 3 months, 6 months, 12 months, 24 months, etc.) from the time of administration. In certain embodiments, the time period can be even longer.
  • the attending physician determines the dosage of the disclosed active agent to be administered to each individual patient, taking into account a variety of factors such as age, weight, general health, diet, sex, the active agent to be administered, the route of administration, and the severity of the condition being treated.
  • the present invention provides a composition comprising a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of human endogenous Cholesin protein, and a pharmaceutically acceptable carrier or excipient.
  • composition is used for one or more of the following:
  • the Cholesin protein is isolated or purified.
  • the Cholesin protein is a mammalian Cholesin protein.
  • mammalian Cholesin proteins include, but are not limited to, Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cattle.
  • the Cholesin protein is a non-human primate Cholesin protein.
  • the Cholesin protein is a human Cholesin protein.
  • the human Cholesin protein comprises the amino acid sequence shown in SEQ ID NO: 1.
  • the Cholesin protein is a mouse Cholesin protein. In some embodiments, the mouse Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2. In other embodiments, the Cholesin protein is a macaque Cholesin protein. In some embodiments, the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3. In other embodiments, the Cholesin protein is a bovine Cholesin protein. In some embodiments, the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4. In other embodiments, the Cholesin protein is a Rattus norvegicus Cholesin protein. In some embodiments, the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5. In other embodiments, the Cholesin protein is a Gallus gallus Cholesin protein. In some embodiments, the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.
  • the second therapeutic agent is a cholesterol-lowering drug.
  • the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.
  • the HMGCR inhibitor is a statin.
  • the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin.
  • the second therapeutic agent is rosuvastatin.
  • the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.
  • the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.
  • the second therapeutic agent is ezetimibe.
  • compositions comprising, for example, the Cholesin protein described herein, an expression vector encoding the Cholesin protein, or comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein of a subject are generally provided in liquid form, and preferably contain a pharmaceutically acceptable buffer.
  • the liquid preparation may be a solution or a suspension.
  • the composition of the present invention is formulated as an injection preparation or an oral preparation.
  • oral preparations include, but are not limited to, tablets, capsules, granules, suspensions.
  • the composition of the present invention It can be used for various routes of administration, such as parenteral administration.
  • the composition of the present invention can be administered intravenously, intramuscularly, subcutaneously, intraperitoneally, etc.
  • the composition of the present invention can be administered orally.
  • examples of carriers and excipients include, but are not limited to, fillers, adhesives, disintegrants, coating agents, adsorbents, antiadhesives, glidants, preservatives, antioxidants, flavoring agents, coloring agents, sweeteners, solvents, cosolvents, buffers, chelating agents, viscosity imparting agents, surfactants, diluents, wetting agents, diluents, preservatives, emulsifiers, stabilizers and tension regulators. It is known to those skilled in the art that suitable excipients are selected to prepare the compositions of the present invention.
  • Exemplary carriers used in the compositions of the present invention include saline, buffered saline, glucose and water.
  • suitable excipients depends especially on the desired dosage form of the active agent used, the disease to be treated and the compositions.
  • the present invention provides the use of a composition as described herein in the preparation of a medicament for one or more of:
  • the present invention provides a method for treating a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of the composition of the present invention.
  • the present invention provides a method for diagnosing a disease associated with elevated plasma cholesterol levels in a subject, comprising the steps of:
  • an increased level of Cholesin protein in the sample compared to a healthy control indicates that the subject is at risk for a disease associated with increased plasma cholesterol levels.
  • the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.
  • hyperlipidemia e.g., hypercholesterolemia, hypertriglyceridemia
  • cardiovascular disease e.g., myocardial infarction, coronary heart disease
  • cerebrovascular disease e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction
  • hyperglycemia e.g., diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.
  • the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In some embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.
  • the blood sample is selected from blood, serum and plasma.
  • the method is performed in vitro.
  • the method further comprises the step of isolating Cholesin protein from the sample. Methods for isolating Cholesin protein are known in the art.
  • the level of Cholesin protein in the sample can be determined by anti-Cholesin protein antibodies.
  • the antibody is preferably a monoclonal antibody, but can also be Fab, Fab', (Fab') 2 , Fv, scFv, bi-scFv, bispecific antibody, trispecific antibody, tetraspecific antibody, etc.
  • the present invention provides use of a reagent for determining the level of Cholesin protein in a blood sample from a subject in the preparation of a kit for diagnosing a disease associated with elevated plasma cholesterol levels in the subject.
  • the present invention provides a kit for diagnosing a disease associated with elevated plasma cholesterol levels in a subject, comprising reagents for determining the level of Cholesin protein in a blood sample from the subject.
  • the reagent is used to determine the level of Cholesin protein in the sample in a method selected from the following: mass spectrometry, fluorescence detection, chemiluminescence, ELISA, Western blot, radioimmunoassay, immunohistochemical staining, multiple immunoassay and dot blot assay.
  • the reagent is an anti-Cholesin protein antibody.
  • the antibody is preferably a monoclonal antibody, but can also be Fab, Fab', (Fab') 2 , Fv, scFv, bi-scFv, bispecific antibody, trispecific antibody, tetraspecific antibody, etc.
  • the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.
  • the disease associated with elevated plasma cholesterol levels is hypercholesterolemia.
  • the disease associated with elevated plasma cholesterol levels is atherosclerosis.
  • the blood sample is selected from the group consisting of blood, serum, and plasma.
  • the present disclosure provides a method for preparing a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject.
  • a composition for reducing plasma cholesterol and/or triglyceride levels in a subject is plasma total cholesterol or plasma LDL-cholesterol.
  • the present disclosure provides use of a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject in the preparation of a composition for inhibiting liver cholesterol synthesis in a subject.
  • the present disclosure provides use of a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject in the preparation of a composition for inhibiting hepatic VLDL-cholesterol secretion in a subject.
  • the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject, for use in reducing plasma cholesterol and/or triglyceride levels in a subject.
  • plasma cholesterol is plasma total cholesterol or plasma LDL-cholesterol.
  • the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject, for use in inhibiting liver cholesterol synthesis in a subject.
  • the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject, for use in inhibiting hepatic VLDL-cholesterol secretion in a subject.
  • the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject, for use in treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.
  • the Cholesin protein is isolated or purified.
  • the Cholesin protein is a mammalian Cholesin protein.
  • mammalian Cholesin proteins include but are not limited to Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cows.
  • the Cholesin protein is a non-human primate Cholesin protein.
  • the Cholesin protein is a human Cholesin protein.
  • the human Cholesin protein comprises an amino acid sequence as shown in SEQ ID NO:1.
  • the Cholesin protein is a mouse Cholesin protein.
  • the Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2.
  • the Cholesin protein is a macaque Cholesin protein.
  • the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3.
  • the Cholesin protein is a bovine Cholesin protein.
  • the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4.
  • the Cholesin protein is a Rattus norvegicus Cholesin protein.
  • the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5.
  • the Cholesin protein is a Gallus gallus Cholesin protein.
  • the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.
  • the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • a lentiviral vector an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.
  • AAV adeno-associated virus
  • the Cholesin protein described herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein is combined with a second therapeutic agent.
  • the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.
  • the second therapeutic agent is a cholesterol-lowering drug.
  • the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.
  • the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.
  • the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.
  • the second therapeutic agent is ezetimibe.
  • the subject is a mammal. In a preferred embodiment, the subject is In some embodiments, the subject is unresponsive or intolerant to one or more of a cholesterol absorption inhibitor, an HMGCR inhibitor, and a PCSK9 inhibitor, for example, the subject has an LDL receptor (LDLR) defect.
  • LDLR LDL receptor
  • Example 1 Cholesin is a cholesterol-induced intestinal secretory hormone
  • mice 10-12 week old wild-type male mice with a genetic background of C57BL/6J were selected.
  • serum proteins of mice fed with a normal diet (RD, containing 0.02% cholesterol) and a Western diet (WD, containing 1.25% cholesterol) for 1 hour after fasting overnight were analyzed.
  • the mice were randomly divided into 2 groups, 6 in each group. Among them, the first group of mice were fed a normal diet after starvation overnight (normal diet group (RD group)); the second group of mice were fed a Western diet after starvation overnight (Western diet group (WD group)).
  • RD normal diet
  • WD group Western diet after starvation overnight
  • mice The blood of the two groups of mice was collected in anticoagulant tubes treated with lithium heparin, and centrifuged at 4°C and 4000rpm for 15 minutes. The supernatant was plasma, which was transferred to a new centrifuge tube. The plasma was treated with a column albumin/immunoglobulin removal kit (Bio-Rad) to remove IgG and albumin. Subsequently, the ProteoMiner Protein Enrichment Kit (BIO-RAD) was used to dilute high-abundance proteins and concentrate low-abundance proteins.
  • Bio-Rad column albumin/immunoglobulin removal kit
  • BIO-RAD ProteoMiner Protein Enrichment Kit
  • the treated plasma was mixed with 5 ⁇ SDS loading buffer (250mM Tris-HCl, pH 6.8, 10% SDS, 0.5% bromophenol blue, 50% glycerol, 5% ⁇ -ME), boiled at 100°C for 10 minutes, and subjected to SDS-PAGE protein electrophoresis. Silver staining was performed after electrophoresis. The results showed that compared with the normal diet group, an enhanced band was observed at around 23kDa in the Western diet group (Figure 1).
  • 5 ⁇ SDS loading buffer 250mM Tris-HCl, pH 6.8, 10% SDS, 0.5% bromophenol blue, 50% glycerol, 5% ⁇ -ME
  • differential bands were proteins encoded by 3110082I17Rik (mouse homologous gene of C7ORF50), which had not been reported in previous literature. Based on subsequent work, the inventors found that it had an inhibitory effect on cholesterol synthesis and named it Cholesin.
  • the cells were sonicated on ice according to the program of 6 seconds on, 6 seconds off, 18 seconds in total, and 40% power.
  • the cells were centrifuged at the maximum speed of 4°C for 15 minutes, and the supernatant obtained by centrifugation was added with 5 ⁇ SDS loading buffer.
  • the samples were boiled at 100°C for 10 minutes to obtain the total protein extract. Cholesin in the culture medium and lysis supernatant was detected by immunoblotting. The results showed that the secretion of Cholesin could be detected in the culture medium of cells overexpressing human or mouse Cholesin (Figure 2B), indicating that Cholesin also has the ability to be secreted under in vitro conditions.
  • a baculovirus overexpressing mouse Cholesin with a C-terminal Flag tag was constructed and used to infect insect Hi5 cells.
  • the specific method is as follows: 1) culture 1L Hi5 cells to a density of 2-3 ⁇ 10 6 cells/ml, add baculovirus at a ratio of 1:50, culture for 48-72 hours, collect the culture supernatant by centrifugation, and filter the culture supernatant with a 0.22 ⁇ m filter to remove cell debris; 2) use an equilibration buffer (20 mM NaH 2 PO 4 , 0.5 M NaCl, pH 7.4) to equilibrate a HisTrap excel (Cytiva) chromatography column for at least 5 column volumes; 3) use a pump head to load the filtered culture medium onto the column; 4) rinse with a wash buffer (20 mM NaH 2 PO 4 , 0.5 M NaCl, 30 mM imidazole, pH 7.4) for at least 20 column volumes until no protein flows out
  • Cholesin is a hormone that responds to eating
  • mice 10-12 week old wild-type male mice with a genetic background of C57BL/6J were randomly divided into 9 groups, 8 mice in each group.
  • Group 1 was starved overnight (fasting group);
  • Groups 2 to 5 were starved overnight and then fed with a normal diet (RD) for 1, 2, 4, and 12 hours (normal diet group containing 0.02% cholesterol);
  • Groups 6 to 9 were starved overnight and then fed with a Western diet (WD) for 1, 2, 4, and 12 hours (Western diet group containing 1.25% cholesterol).
  • the 9 groups of mice were killed, and then plasma was collected and the Cholesin level in plasma was detected by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • the specific operation is as follows: 3 ⁇ l mouse plasma and 100 ⁇ l coating solution (0.05 mol/L Na2CO3/NaHCO3, pH 8.0) were added to the polystyrene microplate.
  • the Cholesin whole body knockout mouse plasma was set as the control group for calibration in the 96-well microplate.
  • the standard curve was set with the purified and quantified Cholesin protein standard. The concentration of the standard curve was 0, 12.5, 25, 50, 100, and 200 pmol. After the addition of the sample, it was incubated at 4°C overnight to allow the protein to adsorb to the bottom of the well.
  • the microplate After overnight coating on a 4°C shaker, the microplate was removed, the liquid in the well was shaken dry, and 100 ⁇ l of 1 ⁇ gelatin was added to each well to block the microplate for 2 hours at room temperature. The blocking solution was discarded, and 200 ⁇ l of TBST solution was added to each well to wash 4 times. The liquid was shaken dry as much as possible for the last time.
  • the primary antibody for detection was prepared in TBST, 100 ⁇ l of primary antibody was added to each well, and incubated at 4°C shaker overnight. Discard the primary antibody, add 200 ⁇ l TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time.
  • mice Five wild-type (WT) and Cholesin knockout (KO) male mice of 10-12 weeks old with a genetic background of C57BL/6J were selected, and the brain, stomach, small intestine, colon, liver, kidney, and brown fat tissues of the mice were collected.
  • RIPA lysis buffer 25mM Tris, 150mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, pH 7.4 was prepared, and protease inhibitors and phosphatase inhibitors were added before use according to the volume of the lysis buffer to prepare tissue protein extract. Weigh an appropriate amount of tissue, add 500 ⁇ l RIPA lysis buffer and magnetic beads, and grind at 60Hz for 1 minute in a tissue grinder.
  • the results are shown in FIG4 .
  • the size of the endogenous Cholesin protein is slightly less than 25 kDa, and its expression level is highest in the small intestine, followed by the colon and stomach, and its expression level in other tissues is low, showing strong tissue specificity.
  • Cholesin is an intestinal secretory hormone that responds to cholesterol absorption
  • Cholesin intestinal-specific knockout mice were constructed as a research tool.
  • WT and IKO mice were randomly divided into 2 groups, each with 8 mice. The first group of mice were starved overnight, and the second group of mice were starved overnight and then fed with a Western diet (WD) containing 1.25% cholesterol for 1 hour. Then the plasma and small intestine of the mice were collected respectively, and the content of total cholesterol (TC) in the small intestine and the content of cholesin, TC and triglyceride (TG) in plasma were measured. The results are shown in Figures 5A and 5B.
  • mice were randomly divided into 2 groups, 7 mice in each group, and the mice were starved overnight.
  • the first group of mice were gavaged with 150 ⁇ l corn oil containing 20% anhydrous ethanol (chol-), and the second group of mice were gavaged with 150 ⁇ l 200 mg kg -1 body weight of cholesterol (chol+, where cholesterol is dissolved in corn oil containing 20% anhydrous ethanol).
  • Plasma and small intestinal tissues were collected from mice 1 hour after gavage. The content of TC in the small intestine and the content of Cholesin, TC and TG in plasma were measured. The results are shown in Figures 5C and 5D.
  • the results of Figure 5A show that the Cholesin content in the plasma of WT mice increased significantly after feeding WD, while the Cholesin content in the plasma of IKO mice hardly increased, and the Cholesin content in the plasma of IKO mice was significantly lower than that of WT mice under starvation.
  • the results of Figure 5A also show that the total cholesterol content in the small intestine increased after feeding WD, but there was no significant difference between the WT and IKO groups in both the starving and fed states. This result suggests that there is no difference in the cholesterol absorption capacity of the small intestine of WT and IKO group mice, and their response to the Western diet is not due to different absorption capacities of the small intestine.
  • the plasma TG of IKO mice was slightly higher than that of the WT group, indicating that eating would reduce the total triglycerides circulating in the blood, and the difference in plasma TG between the WT and IKO groups also suggests that Cholesin may have an effect on plasma TG.
  • the results of Figure 5C show that the Cholesin content in the plasma of WT mice increased significantly after oral administration of cholesterol, while IKO mice basically did not respond to the stimulation of cholesterol, and the absorption of cholesterol by IKO mice was also unaffected.
  • the results of Figure 5D show that there was no significant change in the plasma TC content of mice after oral administration of cholesterol, but the plasma TC of IKO mice was higher than that of the WT group in both the control group and the cholesterol-treated group.
  • the total TG content of mouse plasma increased after oral administration of cholesterol, and there was no significant difference in the plasma TG of WT and IKO mice in the experimental group and the control group. This is because cholesterol is dissolved in corn oil, so the TG in plasma will increase after oral administration.
  • the total cholesterol level in the cells was also detected, and the specific method was as follows: 1) After the cells absorbed the culture medium to detect the amount of Cholesin secretion, the remaining cells were digested with Trypsin, and the cells were resuspended in PBS and counted; 2) The supernatant was discarded after high-speed centrifugation, 150 ⁇ l of isopropanol was added to each sample, and the cells were ultrasonically disrupted on ice (40% intensity, ultrasonic for 2 seconds, interval of 3 seconds, total time of 3 minutes); 3) After the end of ultrasonication, centrifuge at 10000g and 4°C for 10 minutes, and the supernatant was the total cholesterol extract of the cells; 4) The total cholesterol concentration in the extract was determined using the CHOD-PAP cholesterol determination kit (Sino-Biotech Holdings), and the total cholesterol amount was calculated based on the volume of the extract, and finally the total cholesterol amount was divided by the total number of cells to convert the cholesterol content per 106 cells.
  • NPC1L1 knockdown in HCT116 cells disrupts cholesterol absorption and Cholesin secretion
  • NPC1L1 was knocked down (Npc1l1 KD) based on the HCT116 stable cell line overexpressing Cholesin. Subsequently, Western blotting confirmed that NPC1L1 knockdown had no effect on Cholesin expression at the protein level (Figure 8A).
  • non-target knockdown control cells NT
  • NPC1L1 knockdown cells Npc1l1 KD
  • serum-free RPMI1640 containing 0 ⁇ M and 10 ⁇ M cholesterol for 1 hour.
  • the culture medium was collected and the secretion of Cholesin was detected by ELISA.
  • Total cholesterol was extracted by ultrasonication with propanol, and the total cholesterol concentration in the extract was determined using the CHOD-PAP cholesterol assay kit (Zhongsheng Beikong). The total cholesterol amount was calculated based on the volume of the extract, and finally the total cholesterol amount was divided by the total number of cells to convert the cholesterol content per 10 6 cells.
  • the results showed that knocking down NPC1L1 in HCT116 cells impaired cholesterol absorption and Cholesin secretion (Figure 8B).
  • NPC1L1 inhibitor treatment and Npc1l1 gene knockout in mice effectively blocked small intestinal cholesterol absorption and cholesin secretion
  • mice Male mice aged 10-12 weeks were fasted overnight, and then gavaged with ezetimibe (Ezetimlbe+) at a dose of 10 mg kg -1 , and gavage with corn oil solvent as a control (Ezetimlbe-). One hour after gavage, the mice were gavaged with 150 ⁇ l corn oil solvent control (chol-) or cholesterol (chol+, 200 mg kg -1 body weight). One hour after gavage, plasma was collected to detect total cholesterol (TC) and Cholesin levels in plasma, and small intestinal tissues of mice were collected to determine the total cholesterol content in the small intestine. The results are shown in Figures 9A-9B.
  • Npc1l1 knockout mice were constructed. Immunoblotting confirmed that the absence of Npc1l1 did not affect the expression of Cholesin (Figure 9C). Wild-type (Npc1l1 +/+ ) and Npc1l1 knockout (Npc1l1 -/- ) mice were then fasted overnight, and then orally gavaged with 150 ⁇ l corn oil solvent control (chol-) or cholesterol (chol+, 200 mg kg -1 body weight). One hour later, whole blood was collected from the mice to separate plasma to detect plasma Cholesin and TC levels, and small intestinal tissue was collected to determine TC content. The results are shown in Figures 9D-9E.
  • Example 2 The above in vitro and in vivo results of Example 2 indicate that NPC1L1-mediated cholesterol absorption promotes the secretion of Cholesin.
  • the samples were incubated at 4°C overnight to allow the protein to adsorb to the bottom of the wells.
  • the microplate was removed, the liquid in the wells was dried, and 100 ⁇ l of 1 ⁇ gelatin was added to each well to block the microplate at room temperature for 2 hours. Discard the blocking solution, add 200 ⁇ l TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time.
  • Prepare the primary antibody for detection in TBST add 100 ⁇ l primary antibody to each well, and incubate overnight at 4°C on a shaker.
  • Discard the primary antibody for detection add 200 ⁇ l TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time.
  • Prepare the secondary antibody in TBST add 100 ⁇ l to each well, and incubate at room temperature on a shaker for 1 hour.
  • Discard the secondary antibody add 200 ⁇ l TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time.
  • VLDL very low density lipoprotein
  • IDL/LDL intermediate/low density lipoprotein cholesterol
  • HDL high density lipoprotein cholesterol
  • Figure 11A shows that when wild-type mice were fed with a normal diet, the plasma TC of male mice was about 100 mg/dL, and that of female mice was slightly lower than that of male mice; while under the WD feeding condition, the plasma TC of male mice reached about 170 mg/dL, and that of female mice was 140 mg/dL.
  • the plasma TC levels were about 20% higher than those of wild-type mice.
  • the TG content in mouse plasma was slightly increased in IKO mice compared with the WT group in male and female mice fed with RD or WD.
  • FIG 11B shows that in male and female mice fed RD or WD, the liver TC content of IKO mice was higher than that of WT group. There was no significant difference in liver TC content between male and female mice, and the difference in liver TC between WT and IKO was amplified under WD feeding conditions. There was no significant difference in liver TG between WT and IKO groups under RD conditions, but under the induction of WD, the liver TG of IKO group was slightly higher than that of WT group.
  • FIG 11C shows that the levels of high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and very low-density lipoprotein cholesterol (VLDL-C) were increased in Cholesin IKO mice.
  • HDL-C high-density lipoprotein cholesterol
  • LDL-C low-density lipoprotein cholesterol
  • VLDL-C very low-density lipoprotein cholesterol
  • the increase in plasma cholesterol levels may be caused by enhanced intestinal cholesterol absorption, increased hepatic cholesterol synthesis and VLDL secretion, or decreased cholesterol excretion.
  • the body weight, body fat percentage, food and water intake, small intestinal total cholesterol and triglyceride levels, and chylomicron decomposition rate of the mice in Example 4.1 were measured.
  • mice in the random feeding state were weighed and their body fat was measured by EchoMRI awake animal body composition analyzer, and the measurement was repeated 3 times for each mouse.
  • Example 4.1 In order to determine the total cholesterol and triglyceride content in the small intestine of mice, the small intestine tissues of the mice that were starved overnight and then fed for 6 hours in Example 4.1 were collected. The determination results are shown in FIG. 12E .
  • the lipid tolerance test was performed by orally administering a certain dose of olive oil to mice to evaluate the rate of chylomicron decomposition in the small intestine of mice.
  • the mice of Example 4.1 were fasted for 6 hours to synchronize the intestinal state of the mice, and then 200 ⁇ l of olive oil was orally administered.
  • Blood was collected from the tail vein at different time points (0, 1, 2, 3, and 4 hours) before and after oral administration, and then plasma was separated.
  • the triglyceride level in the plasma was determined and the triglyceride secretion rate ( ⁇ TG: plasma TG at 1, 2, 3, and 4 hours of each mouse minus the plasma TG at 0 of the mouse) was calculated to evaluate the rate of chylomicron decomposition.
  • the results are shown in Figure 12F.
  • Figure 12A shows that there was no significant difference in body weight between WT and IKO groups of male and female mice under the two dietary treatments.
  • Figure 12B shows that there was no significant difference in body fat percentage between WT and IKO groups of male and female mice under the two dietary treatments.
  • Figures 12C and 12D show that there was no significant difference in food intake and water intake between WT and IKO groups of male and female mice under the two dietary treatments.
  • Figure 12E shows that there was no significant difference in total cholesterol and triglyceride content in the small intestine between IKO mice and WT mice in male and female mice fed a normal diet or a Western diet.
  • Figure 12F shows that the rate of chylomicron decomposition in IKO mice was not affected in male and female mice fed a normal diet or a Western diet.
  • Cholesterol in mice is generally excreted through bile acid or feces.
  • the cholesterol content in bile acid and feces was measured to explore whether the plasma TC level of IKO mice was affected by changes in cholesterol excretion.
  • Mouse bile is excreted from the gallbladder during eating and is mainly stored in the gallbladder when hungry. The mice were starved overnight, and the bile in the gallbladder was collected with a syringe.
  • the volume of the gallbladder was then weighed, and the concentrations of total bile cholesterol (cholesterol determination kit (CHOD-PAP method, Zhongsheng Beikong) and bile acid (total bile acid (TBA) kit, Nanjing Jiancheng) were measured using kits. The results are shown in Figure 13A.
  • mice were collected and the total cholesterol and triglycerides in the feces were extracted by the following method: 40 mg of feces were weighed and dried in a 55°C oven, and then the dried and crushed feces were suspended in 1200 ⁇ l of a 2:1 chloroform-methanol (v/v) mixture. The suspension was then mixed with 300 ⁇ l of water and vortexed vigorously for 60 seconds.
  • v/v 2:1 chloroform-methanol
  • Figure 13A shows that there were no significant differences in the total bile volume, bile cholesterol level and bile acid level between WT and IKO mice under the two dietary treatments for male and female mice.
  • Figure 13B shows that there were no significant differences in the total cholesterol content and triglyceride content in the feces between WT and IKO mice under the two dietary treatments for male and female mice.
  • qPCR was used to detect the mRNA levels of cholesterol synthesis-related genes in the liver.
  • the specific experimental procedures are as follows: 1) About 20 mg of liver tissue was weighed and the total liver RNA was extracted using HP total RNA Kit (Omega); 2) 1 ⁇ g RNA was taken and reverse transcribed using REVERTAID 1ST CDNA SYNTH KIT (thermo); 3) The cDNA obtained by reverse transcription was subjected to qPCR. The results are shown in Figure 14A.
  • Immunoblotting was used to detect the protein expression level of cholesterol synthesis genes in the liver.
  • the specific operation is as follows: 1) Weigh 20 mg of liver tissue, add 500 ⁇ l RIPA lysis buffer and magnetic beads, and grind at 60 Hz for 1 minute in a tissue grinder; 2) Centrifuge at maximum speed at 4°C for 10 minutes; 3) Transfer the supernatant to a new EP tube, be careful not to absorb the fat layer, add 5 ⁇ loading buffer, mix well, and boil the sample at 100°C for 10 minutes; 4) Use the BCA method for protein quantification; 5) Detect the expression levels of SREBP2 (including pSREBP2 and mSREBP2), HMGCR, HMGCS1 and LDLR in the tissue by immunoblotting. The results are shown in Figure 14B.
  • Figures 14A and 14B show that compared with wild-type mice, the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, Pmvk, sqle in the liver of IKO mice fed with a normal diet and a Western diet, as well as the transcription factor SREBP2 that regulates the expression of these cholesterol synthesis-related genes and its downstream regulated LDLR were significantly increased. It is worth noting that the expression of SREBP2 and cholesterol synthesis-related genes was lower in mice fed with a Western diet, indicating that the absorption of intestinal cholesterol inhibits the synthesis of cholesterol in the liver.
  • mice were injected with poloxamer 407, an inhibitor of lipoprotein lipase and VLDL metabolism, and plasma cholesterol and triglyceride levels were monitored to assess hepatic VLDL secretion.
  • mice were fasted for 4 hours and then intraperitoneally injected with 1 g kg-1 body weight of Poloxamer 407. Blood samples were collected from the tail vein before, 1 hour and 2 hours after the injection of Poloxamer 407. Plasma total bile duct was measured at each time point. The levels of sterol (TC) and triglyceride (TG) were measured and the secretion rates of TC and TG were calculated (the calculation method was the TC or TG level of each mouse at 1 or 2 hours minus the TC or TG level of the mouse at 0 hours). The results are shown in FIG15 .
  • TC sterol
  • TG triglyceride
  • Example 4 show that knockout of Cholesin increases cholesterol synthesis and VLDL secretion in the liver, thereby increasing plasma cholesterol and triglyceride levels. This suggests that administration of Cholesin can reduce cholesterol synthesis and VLDL secretion in the liver, thereby reducing plasma cholesterol and triglyceride levels.
  • mice with a genetic background of C57BL/6J were selected, and purified GST-Cholesin was incubated with different tissues of mice for in vitro binding experiments on frozen tissue sections.
  • mice According to the experimental design, the mouse adipose tissue (epididymal white adipose tissue (eWAT) and brown adipose tissue (BAT)), muscle, liver, pancreas, small intestine and kidney tissues were collected and immediately placed in a 4% paraformaldehyde solution prepared in PBS and fixed overnight at 4°C; 2) Tissue dehydration and embedding: Different dehydration treatments were performed according to the different OCT and paraffin embedding agents.
  • eWAT epidymal white adipose tissue
  • BAT brown adipose tissue
  • Tissue sectioning The thickness of the sections was 10 ⁇ m using a freezing microtome, and the thickness of the paraffin sections was 5 ⁇ m; 4) The frozen sections were placed in an immunofluorescence humidified box and thawed at room temperature. The paraffin sections required antigen repair.
  • the pre-treated sections were drawn with an immunohistochemistry pen to draw a closed circle around the tissue and washed twice with PBS; 5) Protein binding: Prepare binding proteins (GST-Cholesin 200nM; GST 200nM), incubate at room temperature for 30 minutes; 6) After the protein incubation, wash twice with PBS, and block with 5% BSA blocking solution prepared in PBS at room temperature for 1 hour; 7) Discard the blocking solution, incubate with primary antibody diluted in blocking solution at 4°C overnight; 8) Discard the primary antibody, wash three times with TBST, and incubate with secondary antibody diluted in blocking solution and DAPI with a final concentration of 0.5mg/ml at room temperature for 1h; 10) Discard the secondary antibody and DAPI, and wash three times with TBST; 11) Try to dry the solution on the surface of the tissue, add an appropriate amount of mounting medium on the surface of the slide according to the size of the tissue, carefully cover with a coverslip to avoid bubbles, and place at room
  • GST-Cholesin has binding signals in muscle, liver, kidney and adipose tissue, and the binding signals in muscle, liver, kidney and adipose tissue are relatively stronger ( Figure 16A). This indicates that muscle, liver, kidney and adipose tissue are the target tissues of Cholesin.
  • GPR146 was identified as a candidate receptor for Cholesin.
  • GPR146 is a conserved orphan G protein-coupled receptor involved in the regulation of cholesterol synthesis.
  • GPR146 is a receptor for Cholesin
  • 8-10 week old Gpr146 knockout mice with a genetic background of C57BL/6J were selected to perform in vitro binding experiments as described in Example 5.1.
  • the immunofluorescence results are shown in FIG16A .
  • GPR146 is a specific receptor for Cholesin
  • mutants of GPR146 were designed based on the conservation and acid-base properties of the amino acids in the extracellular region of GPR146. Since the liver is the main target tissue of Cholesin, primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 -/- ) mice were isolated for binding staining. For primary hepatocytes of Gpr146 -/- mice, plasmids were transfected to overexpress wild-type GPR146 (WT) and mutant GPR146 (Mut).
  • the specific experimental method is as follows: 1) According to the experimental needs, a suitable number of cell slides with a diameter of 12 mm were spread in a 6-well plate in advance, and primary hepatocytes of Gpr146 +/+ and Gpr146 -/- mice were isolated at the same time, and the cells were diluted to a suitable density and spread in a 6-well plate; 2) After the cells of Gpr146 -/- mice adhered for 6 hours, plasmids were transfected to overexpress wild-type GPR146 (WT) and mutant GPR146 (Mut); 3) After 36 to 48 hours of cell culture, the cell slides in the 6-well plate were transferred to a 24-well plate and washed once with PBS buffer.
  • GST and GST-Cholesin were diluted to 200nM with fresh culture medium, and Cholesin-His was diluted to 20 ⁇ M.
  • WT cells were divided into 3 groups: GST negative control group, GST-Cholesin binding group, and GST-Cholesin and Cholesin-His co-incubation group. Gpr146 -/- cells were only co-incubated with 200nM GST-Cholesin.
  • FIG. 16A The results of Figure 16A show that the binding signals of GST-Cholesin in the muscle, liver, kidney and adipose tissue of Gpr146 -/ - mice are significantly weakened compared with those of wild-type mice.
  • GST-Cholesin can bind to the cell membrane of WT hepatocytes, and this binding can be competed out by Cholesin-His; the binding of GST-Cholesin to Gpr146 -/- cells is significantly weakened, and cells complemented with wild-type GPR146 have restored the binding to GST-Cholesin, while cells complemented with mutant GPR146 still cannot bind to GST-Cholesin.
  • the saturation binding degree of Gpr146 -/- cells is significantly reduced, and cells complemented with wild-type GPR146 have restored the binding to Cholesin, while cells complemented with mutant GPR146 still cannot bind to Cholesin.
  • Microthermophoresis is a technique that quantifies the interactions between biomacromolecules based on the thermophoresis properties of molecules. This technique was used in this example to measure the binding affinity of Cholesin to wild-type (WT) and mutant (Mut) GPR146.
  • the specific experimental steps are as follows: 1) Receptor fluorescence labeling: We purified wild-type and mutant GPR146 proteins from Hi5 cells and concentrated them to about 100ul with a concentration of about 0.4ug/ul. We took 90ul of protein sample and used the RED-NHS protein amino labeling kit to fluorescently label it. After labeling, its concentration was 1/5 of that before labeling. The volume was expanded to 450ul, and the labeled sample was subjected to fluorescence signal detection.
  • the final receptor concentration used in the MST experiment was determined to be 50nM; 2)
  • the Cholesin protein was purified by Superdex 200Incresae 5/15GL molecular sieve chromatography column. Replace with HEPES-NaCl buffer containing DDM and CHS, dilute Cholesin to 6uM with buffer, and then dilute it with buffer in a 1:1 gradient to form 16 concentration gradients, take 10ul 50nM labeled wild-type and mutant GPR146 and mix them thoroughly with 10ul Cholesin of different concentrations, and let it stand at room temperature for 20min; 3) Use Monolith NT.115 capillary to absorb samples, place them on the sample stage in order from high to low Cholesin concentration, put them into micro-thermophoresis instrument for measurement, set the measurement parameters to 10% LED power and 40% MST power, use NanoTemper analysis software to analyze and calculate the experimental results to obtain the equilibrium dissociation constant kd value between Cholesin and wild-type and mutant GPR146 proteins.
  • Example 6 Cholesin inhibits cAMP production via the GPR146-coupled G ⁇ i signaling pathway
  • GPR146 is a GPCR
  • the G protein associated with the GPCR is a heterotrimer containing three different subunits: ⁇ , ⁇ , and ⁇ .
  • the binding of ligands to the GPCR causes a conformational change in the receptor, which in turn changes and binds to and activates the G protein.
  • the active form of the G protein is then released from the receptor surface, dissociating into its ⁇ and ⁇ / ⁇ subunits. Both subunits will then activate their specific effectors, which release the second messenger.
  • G ⁇ s and G ⁇ i isoforms activate or inactivate adenylate cyclase, respectively, which converts adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP), releasing inorganic pyrophosphate in the process.
  • Guanine nucleotide binding protein G(i) subunit ⁇ 1 (GNAI1) is a G protein inhibitory subunit, and the dissociation of G ⁇ i inhibits the downstream cAMP signal. It is speculated that Cholesin promotes the dissociation of G ⁇ i after binding to GPR146, thereby inhibiting downstream cholesterol synthesis by inhibiting cAMP. In order to verify the above hypothesis, an immunoprecipitation experiment was performed.
  • hepatocytes from wild-type mice were isolated and adenovirus overexpressing GNAI1 with HA tag was added after the cells were attached to the wall. The cells were then divided into three groups, with empty adenovirus, wild-type GPR146 (WT) adenovirus with Flag tag, and mutant GPR146 (Mut) adenovirus with Flag tag added. 24 hours after adenovirus infection, each group was treated with 0nM and 200nM Cholesin for 10 minutes, and then immediately added RIPA lysis buffer to lyse the cells.
  • WT wild-type GPR146
  • Mot mutant GPR146
  • 40 ⁇ l of the lysed sample was added to 5 ⁇ SDS loading buffer and boiled at 100°C for 10min as the input sample, and the Flag-adsorbed beads were added to the cell lysate and incubated overnight on a 4°C rotating mixer. After binding, centrifuge at 5000 rpm and 4°C for 3 min, discard the supernatant, add 600 ⁇ l of pre-cooled RIPA lysis buffer and vortex to remove the remaining liquid sample and proteins non-specifically bound to the beads, centrifuge and discard the supernatant, repeat washing 6 times to fully remove the residual liquid. 40 ⁇ l 1 ⁇ SDS loading buffer was added and the sample was boiled at 100°C for 10 min, and then subjected to SDS-PAGE and immunoblotting. The immunoprecipitation results are shown in FIG18 .
  • Gpr146 +/+ mice Primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 -/- ) mice were isolated and placed in 6-well plates. Four groups were set up: Gpr146 +/+ cells, Gpr146 -/- complemented empty vector (Vec) cells, Gpr146 -/- complemented wild-type GPR146 (WT) cells, and Gpr146 -/- complemented mutant GPR146 (Mut) cells. After primary hepatocytes adhered to the wall, adenoviruses complemented with wild-type and mutant GPR146 were added.
  • Example 6 The above results of Example 6 indicate that Cholesin inhibits the production of cAMP via the GPR146-coupled G ⁇ i signaling pathway.
  • Example 7 Intraperitoneal injection of Cholesin reduces Hmgcr mRNA levels and plasma and liver cholesterol levels
  • mice 10-12 week old wild-type male mice with a genetic background of C57BL/6J were randomly divided into 4 groups, with 10 mice in each group. The mice were fasted overnight, and injected with 0 mg/kg, 1 mg/kg, 5 mg/kg and 10 mg/kg of eukaryotic purified Cholesin-his protein on the second day. The mouse liver tissue was collected 6 hours after the mice were fed. Total RNA of mouse liver tissue was extracted, and qPCR was performed to detect the expression level of Hmgcr, and statistical analysis was performed with Rpl32 as an internal reference.
  • mice with a genetic background of C57BL/6J were selected and divided into two groups. They were injected with 0 mg/kg and 5 mg/kg of eukaryotic purified Cholesin-his protein, respectively. The injections were given twice a week for two weeks. When the mice were 10 weeks old, the liver tissue and whole blood were collected after the mice were starved and fed for 6 hours. The operation process is shown in Figure 21A. Liver, total RNA of mouse liver tissue was extracted for qPCR to detect the expression level of Hmgcr; total cholesterol (TC) of the liver was also extracted to detect the level of TC in the extract. For whole blood of mice, plasma was separated by low-speed centrifugation, and the level of TC in plasma was detected. The Hmgcr mRNA level in mouse liver, the TC level in mouse plasma and liver are shown in Figures 21B, 21C and 21D, respectively.
  • Example 8 Cholesin inhibits cholesterol synthesis via GPR146
  • Gpr146 LKO mice were divided into 3 groups at 10-12 weeks, with 12 mice in each group.
  • One group was injected with adenovirus control (Vec) through the tail vein, one group was injected with adenovirus expressing wild-type GPR146 (WT), and the other group was injected with adenovirus expressing mutant GPR146 (Mut).
  • the total amount of adenovirus injected into each mouse through the tail vein was 5 ⁇ 10 ⁇ 8.
  • Each group was then divided into two small groups, with 6 mice in each group, and 0 mg/kg or 5 mg/kg Cholesin was injected twice a week for 4 consecutive times.
  • the liver tissue and whole blood were collected after the mice were starved and fed for 6 hours at 14.5 weeks.
  • the operation process is shown in Figure 22A.
  • mice liver total liver RNA was extracted and then qPCR was performed to detect the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk and Pmvk, and the results are shown in Figure 22B.
  • total liver protein was extracted and immunoblotted with antibodies to SREBP2, HMGCR, HMGCS1, LDLR, pKA Sub and HSP90, with HSP90 as the internal reference, and the results are shown in Figure 22C.
  • Total cholesterol (TC) in the liver was also extracted to detect the level of TC in the extract, and the results are shown in Figure 22D.
  • low-speed centrifugation was performed to separate plasma, and the level of TC in plasma was detected, and the results are shown in Figure 22E.
  • Ldlr -/- mice are currently a commonly used disease model for studying atherosclerosis .
  • the plasma cholesterol of Ldlr-/- mice was significantly higher than that of wild-type mice, and atherosclerotic plaques were formed under the induction of Western diet (WD).
  • WD Western diet
  • Five-week-old Ldlr -/- mice were first fed a Western diet for 4 weeks, and then the mice were divided into four groups, with 6-7 mice in each group.
  • the first group was administered with Cholesin alone by intraperitoneal injection (5 mg kg -1 , twice a week); the second group was administered with rosuvastatin (Ros) alone by drinking water (10 mg kg - 1day -1 ); the third group was administered with Cholesin (5 mg kg -1 , twice a week) and rosuvastatin (10 mg kg - 1day -1 ) in combination (Cholesin+Ros); the fourth group was the vehicle control group (Veh), and the specific operation is shown in Figure 23. During the period, the tail vein blood of mice was collected every two weeks to detect the plasma total cholesterol (TC) and triglyceride (TG) levels, and the weight and food intake of mice were detected.
  • TC total cholesterol
  • TG triglyceride
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • mice were able to significantly reduce the TC levels in the plasma of Ldlr -/- mice when they were given rosuvastatin or Cholesin alone.
  • rosuvastatin was observed to slightly increase the plasma TC level when the drug dosage remained unchanged after 4 weeks of administration, indicating that there was a drug resistance problem in the later stage of rosuvastatin administration.
  • the combined administration of rosuvastatin and Cholesin had the best effect on reducing plasma TC, which was better than the single administration of rosuvastatin or Cholesin. The combined administration could achieve a good inhibitory effect in the early stage of administration and maintain the inhibitory effect for a long time.
  • the area under the curve (AUC) of the changes in plasma TC within 8 weeks was statistically analyzed in order to more intuitively compare the effects of the three treatment regimens. Among them, the single treatment of Cholesin and rosuvastatin could significantly reduce the TC level in plasma, and the inhibitory effect of the combined treatment of the two was more prominent, achieving a synergistic effect.
  • the TC level in the liver of mice after 8 weeks of administration is shown in Figure 24B.
  • the TC level in the liver of mice was significantly reduced after administration of rosuvastatin or Cholesin.
  • the combined administration of rosuvastatin and Cholesin had the best effect on reducing liver TC, which was better than the single administration of rosuvastatin or Cholesin, achieving a synergistic effect.
  • TG levels in mouse plasma and liver are shown in Figures 24C and 24D. Cholesin and rosuvastatin alone can reduce the TG levels in plasma or liver, and the combined use of the two has the most significant inhibitory effect on TG in plasma or liver, indicating that the combination of the two achieves a synergistic effect.
  • the reduction in total cholesterol may be due to the inhibition of cholesterol synthesis, so the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, Pmvk and Sqle in mouse liver and the transcription factor SREBP2 that regulates the expression of these genes were detected by qPCR and immunoblotting.
  • the results are shown in Figures 25A and 25B.
  • Oil Red O is a strong lipid staining agent that can specifically stain neutral triglycerides, lipids and lipoproteins red. Since atherosclerotic plaques are rich in lipids, the plaque part can be stained red. The proportion of the stained plaque area to the entire aorta is calculated to evaluate the pathological condition.
  • the specific experimental steps are as follows: 1) Anesthetize the mouse, fix it in supine position on a foam board, open the chest cavity to expose the heart, draw 20 ml of PBS into the syringe, insert the syringe needle into the left ventricle from the apex of the heart, cut a small hole in the right atrium, and manually push PBS for systemic perfusion. It can be seen that the color of the liver changes from red to yellow after perfusion. Leave the needle in place and draw 10 ml of 4% paraformaldehyde/PBS into the syringe to continue perfusion.
  • the mouse tail After the fixative is injected, the mouse tail can be seen swinging slightly, indicating that the perfusion fixation effect is good; 2) Remove the mouse viscera, expose the spine, carefully peel off the heart and the entire aorta under a stereo microscope, cut the aorta below the bifurcation of the iliac artery, and cut the ascending aorta near the heart. After the entire aorta is peeled off, the fat tissue on the adventitia of the blood vessels must be carefully removed. Blunt peeling is mainly used to avoid damaging the blood vessels.
  • the processed whole aorta is stored in 4% paraformaldehyde/PBS and wait for staining; 3) Take out the fully fixed aorta, use Vannas scissors to extend into the blood vessels from the fracture of the ascending aorta under a stereo microscope, and longitudinally dissect the aortic arch along the greater curvature of the aortic arch to the distal end, leaving the three branches untreated.
  • the good aortic intima was unfolded upwards and fixed on a black-bottomed plastic plate with the smallest insect pin; 4) Prepare Oil Red O dye solution: weigh 0.5g of Oil Red O powder, add 100ml of isopropanol, and place in a water bath at 90°C for 1 hour.
  • a saturated Oil Red dye solution was obtained.
  • the saturated solution and double distilled water were mixed in a ratio of 3:2, and then filtered again after a water bath at 42°C for 10 minutes. This is the Oil Red working solution. Both the saturated solution and the working solution need to be stored at low temperature and away from light; 5)
  • staining the sample was first pretreated in 60% isopropanol for 10 minutes, and then stained in the Oil Red working solution for 15 minutes. After staining, the sample was decolorized in 60% isopropanol for 5 minutes to reduce the background.
  • the stained sample can be stored in the fixative for a long time; 6) Then, a Zeiss Stemi 508 stereo microscope was used to take images of the stained aorta. Image J software was used to quantify the lesion area of the thoracic aorta. ImageJ was used to measure the plaque area and the entire aortic intima area, and then the plaque area was divided by the entire intima area ⁇ 100% to obtain the relative percentage of plaques. Dividing by the intima area was mainly to eliminate the effects of differences in mouse body size.
  • the accumulation of lipids in the liver was observed by HE staining of liver tissue.
  • the specific operation was as follows: 1) A piece of mouse liver tissue was taken and fixed in 4% paraformaldehyde overnight; 2) Alcohol was used as a dehydrating agent from low concentration to high concentration to gradually remove the water in the tissue block, and then the tissue block was placed in xylene to make it transparent, and the alcohol in the tissue block was replaced with xylene, and then immersed in wax and embedded; 3) The transparent tissue block was placed in the melted paraffin, placed in a wax melting box for insulation, and embedded after the paraffin was completely immersed in the tissue block; 4) The embedded wax block was fixed on a microtome and cut into thin slices, generally 5 ⁇ m, and placed in a 45°C constant temperature box for drying after pasting; 5) The slices were placed in xylene I for 10min-xylene II for 10min-anhydrous ethanol I for 5min-anhydrous ethanol I for 5min Ethanol II 5min-9
  • the HE staining results are shown in Figure 28.
  • the white area in the cells is lipid accumulation. It can be observed that the control group has the most white lipid droplets in the cells, while the experimental groups have significantly weakened them, indicating that the lipid accumulation in the liver of mice after Cholesin administration is significantly reduced.
  • Example 9 The above results of Example 9 indicate that Cholesin, whether used alone or in combination with rosuvastatin, has a significant protective effect on hypercholesterolemia and atherosclerosis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present disclosure provides use of cholesin in modulating cholesterol homeostasis and a composition comprising cholesin, a nucleic acid encoding cholesin, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of endogenous cholesin protein in a subject. Specifically, the present disclosure relates to a method for treating or preventing a disease associated with an elevated plasma cholesterol level in a subject. The method comprises: administering to the subject an effective amount of cholesin, a nucleic acid encoding cholesin, or an expression vector comprising the nucleic acid, or enhancing the expression or activity of endogenous cholesin protein in the subject.

Description

Cholesin在调节胆固醇稳态中的用途Use of Cholesin in regulating cholesterol homeostasis

本国际专利申请要求2023年10月24日提交的中国专利申请号202311385254.5的优先权,其全部内容通过引用并入本文用于所有目的。This international patent application claims priority to Chinese patent application No. 202311385254.5 filed on October 24, 2023, the entire contents of which are incorporated herein by reference for all purposes.

技术领域Technical Field

本发明属于生物医药领域,特别涉及肠分泌激素Cholesin在调节胆固醇稳态中的用途,及其与其他降胆固醇药物联用后调节胆固醇稳态的用途。The present invention belongs to the field of biomedicine, and particularly relates to the use of the intestinal secretory hormone Cholesin in regulating cholesterol homeostasis, and the use of the intestinal secretory hormone Cholesin in regulating cholesterol homeostasis after being used in combination with other cholesterol-lowering drugs.

背景技术Background Art

胆固醇的平衡是由各种组织协调的,以维持饮食中胆固醇的吸收、新的合成和胆汁的清除和排泄之间的平衡。血液中胆固醇水平的升高,特别是低密度脂蛋白胆固醇(LDL-C),有助于动脉粥样硬化的发展,被认为是心血管疾病(CVD)的重要风险因素。CVD是一个紧迫的公共卫生问题,占全球死亡人数的30%以上。饮食中的胆固醇和胆汁中的胆固醇都被肠道中肠细胞顶端表面的Niemann-Pick型C1 like 1(NPC1L1)蛋白吸收。吸收后,饮食中的胆固醇以乳糜微粒的形式释放出来,然后被肝脏(胆固醇合成的主要场所)吸收。来自新合成和肠道吸收的胆固醇分子通过循环被运送到细胞中使用。为了维持胆固醇平衡,多余的胆固醇通过胆汁分泌进入粪便排出体外。Cholesterol homeostasis is coordinated by various tissues to maintain a balance between dietary cholesterol absorption, new synthesis, and bile clearance and excretion. Elevated blood cholesterol levels, especially low-density lipoprotein cholesterol (LDL-C), contribute to the development of atherosclerosis and are considered an important risk factor for cardiovascular disease (CVD). CVD is a pressing public health problem, accounting for more than 30% of deaths worldwide. Both dietary cholesterol and bile cholesterol are taken up by Niemann-Pick type C1 like 1 (NPC1L1) proteins on the apical surface of enterocytes in the intestine. After absorption, dietary cholesterol is released in the form of chylomicrons, which are then taken up by the liver, the major site of cholesterol synthesis. Cholesterol molecules from new synthesis and intestinal absorption are transported to cells for use via the circulation. To maintain cholesterol homeostasis, excess cholesterol is excreted from the body via bile secretion into the feces.

新的胆固醇合成是一个消耗能量的过程,在一系列酶的控制下,胆固醇从乙酰CoA生成。其中,HMGCR(3-羟基-3-甲基戊二酰辅酶A(HMG-CoA)还原酶)是限速酶,其活性在转录和转录后水平上受到控制。SREBP2(甾醇调节元件结合蛋白2)作为主转录因子,支配HMGCR和其他参与胆固醇合成和吸收的基因的表达。胆固醇的生物合成和摄取是通过负反馈机制进行严格调控的,该机制可以感知细胞的胆固醇水平。当细胞缺乏胆固醇时,SREBP2与其护送蛋白SCAP(SREBP裂解激活蛋白)一起,在COPII小泡中从内质网(ER)被运输到高尔基体。在高尔基体中,SREBP2依次被位点1和位点2蛋白酶裂解。通过这种裂解释放的SREBP2的N端结构前往细胞核,在那里它作为一个转录因子,增强参与胆固醇合成和吸收的基因的表达。相反,当细胞胆固醇水平上升时,胆固醇分子与SCAP结合,引发其与INSIG(胰岛素诱导基因)的相互作用。这种相互作用将SREBP保留在ER中,并阻止随后SREBP的激活和参与胆固醇代谢的基因的表达。这种机制有助于维持胆固醇的平衡。内源性合成的和外源性获得的胆固醇都以极低密度脂蛋白(VLDL)的形式分泌到血液中。在血液中处理后,VLDL产生循环的LDL, 它们可以通过LDL受体(LDLR)介导的内吞作用被细胞吸收。虽然对胆固醇水平反应的胆固醇平衡调节已被充分理解,但对胆固醇以外的其他信号如何影响胆固醇合成的理解仍然有限。New cholesterol synthesis is an energy-consuming process in which cholesterol is generated from acetyl CoA under the control of a series of enzymes. Among them, HMGCR (3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase) is the rate-limiting enzyme, and its activity is controlled at the transcriptional and post-transcriptional levels. SREBP2 (sterol regulatory element binding protein 2) acts as a master transcription factor, governing the expression of HMGCR and other genes involved in cholesterol synthesis and uptake. Cholesterol biosynthesis and uptake are tightly regulated by a negative feedback mechanism that senses the cholesterol level of the cell. When cells are cholesterol-deficient, SREBP2, together with its escort protein SCAP (SREBP cleavage-activating protein), is transported from the endoplasmic reticulum (ER) to the Golgi apparatus in COPII vesicles. In the Golgi apparatus, SREBP2 is cleaved by site 1 and site 2 proteases in sequence. The N-terminal structure of SREBP2 released by this cleavage travels to the nucleus, where it acts as a transcription factor, enhancing the expression of genes involved in cholesterol synthesis and uptake. Conversely, when cellular cholesterol levels rise, cholesterol molecules bind to SCAP, triggering its interaction with INSIG (insulin-induced gene). This interaction retains SREBP in the ER and prevents subsequent activation of SREBP and expression of genes involved in cholesterol metabolism. This mechanism helps maintain cholesterol homeostasis. Both endogenously synthesized and exogenously acquired cholesterol are secreted into the blood in the form of very low-density lipoproteins (VLDL). After processing in the blood, VLDL produces circulating LDL, They can be taken up by cells via LDL receptor (LDLR)-mediated endocytosis. While the regulation of cholesterol homeostasis in response to cholesterol levels is well understood, the understanding of how signals other than cholesterol influence cholesterol synthesis remains limited.

已知胆固醇合成和吸收之间的相互关系可以调节循环胆固醇对饮食或胆固醇干预的反应。然而,负责协调胆固醇吸收和合成之间平衡的具体因素,以及这些因素如何精确地调节这种平衡,仍然知之甚少。The reciprocal relationship between cholesterol synthesis and absorption is known to regulate the response of circulating cholesterol to dietary or cholesterol interventions. However, the specific factors responsible for coordinating the balance between cholesterol absorption and synthesis, and how these factors precisely regulate this balance, remain poorly understood.

目前有三大类降胆固醇药物常用于防治高胆固醇血症。第一类药物,依折麦布抑制NPC1L1的内吞作用,从而阻断胆固醇的摄入。第二类药物,他汀类药物通过抑制HMGCR减少胆固醇的合成,并通过上调LDLR增加LDL的摄取。第三类药物,PCSK9抑制剂,能够稳定LDLR,促进肝脏对LDL的吸收。虽然依折麦布能减少肠道胆固醇的吸收,但其增强胆固醇合成的补偿作用限制了其降低血浆胆固醇水平的效果。他汀类药物和PCSK9抑制剂的降胆固醇作用都依赖于LDLR,这给LDLR有缺陷或不足的患者带来了挑战。此外,HMGCR水平的代偿性增加使某些长期治疗后停止使用他汀类药物的患者的结果恶化。并且他汀类药物能够极大地增加Hmgcr和其他致胆固醇基因的表达,其疗效受到质疑。There are three major classes of cholesterol-lowering drugs commonly used to prevent and treat hypercholesterolemia. The first class of drugs, ezetimibe, inhibits NPC1L1 endocytosis, thereby blocking cholesterol uptake. The second class of drugs, statins, reduce cholesterol synthesis by inhibiting HMGCR and increase LDL uptake by upregulating LDLR. The third class of drugs, PCSK9 inhibitors, stabilize LDLR and promote liver absorption of LDL. Although ezetimibe can reduce intestinal cholesterol absorption, its compensatory effect of enhancing cholesterol synthesis limits its effectiveness in lowering plasma cholesterol levels. The cholesterol-lowering effects of statins and PCSK9 inhibitors are both LDLR-dependent, which poses a challenge for patients with defective or insufficient LDLR. In addition, compensatory increases in HMGCR levels worsen the outcomes of some patients who stop taking statins after long-term treatment. And statins can greatly increase the expression of Hmgcr and other cholesterol-causing genes, and their efficacy has been questioned.

因此,寻找新型降胆固醇药物对于治疗包括高胆固醇血症和动脉粥样硬化在内的与血浆胆固醇水平升高相关的疾病具有重要的生物学意义。Therefore, the search for new cholesterol-lowering drugs is of great biological significance for the treatment of diseases associated with elevated plasma cholesterol levels, including hypercholesterolemia and atherosclerosis.

发明内容Summary of the invention

本发明旨在提供治疗包括高胆固醇血症和动脉粥样硬化在内的与血浆胆固醇水平升高相关的疾病的方法。发明人出人意料地发现,Cholesin是一种由肠道分泌的响应胆固醇吸收的激素,在此之前并未被报道过,其能够抑制胆固醇的合成和肝脏中VLDL的分泌,从而降低血浆胆固醇的水平,以防止高胆固醇血症和动脉硬化等与血浆胆固醇水平升高相关的疾病。机制研究表明,Cholesin通过与其受体GPR146结合,抑制PKA信号和SREBP2控制的胆固醇合成(图30)。因此,Cholesin-GPR146轴是肠道吸收胆固醇和抑制肝脏中胆固醇合成的分子联系。因此,外源性Cholesin或促进受试者内源性Cholesin蛋白表达或活性的方法或试剂均能降低血浆胆固醇和/或甘油三酯的水平,从而用于预防或治疗高胆固醇血症和动脉硬化等与血浆胆固醇水平升高相关的疾病。与现有的三大类降胆固醇药物相比,Cholesin对参与胆固醇合成的基因的表达产生不依赖于LDLR的下调作用,并降低血浆胆固醇水平,这意味着Cholesin治疗是对抗高胆固醇血症和动脉粥样硬化等与血浆胆固醇水平升高相关的疾病的有效策略。此外,由于Cholesin能够降低 体重增加和TG水平,将Cholesin与他汀类等现有降胆固醇药物组合也是治疗包括高脂血症和动脉粥样硬化在内的与血浆胆固醇水平升高相关的疾病的有前途的策略。The present invention aims to provide a method for treating diseases associated with elevated plasma cholesterol levels, including hypercholesterolemia and atherosclerosis. The inventors unexpectedly discovered that Cholesin is a hormone secreted by the intestine in response to cholesterol absorption, which has not been reported before, and can inhibit the synthesis of cholesterol and the secretion of VLDL in the liver, thereby reducing the level of plasma cholesterol to prevent diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. Mechanistic studies have shown that Cholesin inhibits cholesterol synthesis controlled by PKA signaling and SREBP2 by binding to its receptor GPR146 (Figure 30). Therefore, the Cholesin-GPR146 axis is a molecular link between intestinal absorption of cholesterol and inhibition of cholesterol synthesis in the liver. Therefore, exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin protein in a subject can reduce the level of plasma cholesterol and/or triglycerides, thereby preventing or treating diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. Compared with the three existing cholesterol-lowering drugs, Cholesin has an LDLR-independent downregulation effect on the expression of genes involved in cholesterol synthesis and reduces plasma cholesterol levels, which means that Cholesin treatment is an effective strategy to combat diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. In addition, because Cholesin can reduce Combining Cholesin with existing cholesterol-lowering drugs such as statins is also a promising strategy for treating diseases associated with elevated plasma cholesterol levels, including hyperlipidemia and atherosclerosis.

相应地,在一方面,本发明提供了一种降低受试者中的血浆胆固醇水平和/或甘油三酯水平的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。Accordingly, in one aspect, the present invention provides a method for lowering plasma cholesterol level and/or triglyceride level in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在一些实施方案中,所述血浆胆固醇选自血浆总胆固醇和血浆LDL-胆固醇。In some embodiments, the plasma cholesterol is selected from plasma total cholesterol and plasma LDL-cholesterol.

在另一方面,本发明提供了一种抑制受试者中的肝脏胆固醇合成的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In another aspect, the present invention provides a method for inhibiting hepatic cholesterol synthesis in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在一些实施方案中,所述方法抑制与胆固醇合成相关基因的表达,并且所述基因选自Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk和Sqle。在一些实施方案中,所述方法抑制与胆固醇摄取相关基因的表达,并且所述基因为Ldlr。In some embodiments, the method inhibits the expression of a gene associated with cholesterol synthesis, and the gene is selected from Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle. In some embodiments, the method inhibits the expression of a gene associated with cholesterol uptake, and the gene is Ldlr.

在一些实施方案中,所述方法抑制促进胆固醇合成或摄取相关基因表达的转录因子的表达,并且所述转录因子为SREBP2。In some embodiments, the method inhibits the expression of a transcription factor that promotes the expression of genes involved in cholesterol synthesis or uptake, and the transcription factor is SREBP2.

在又一方面,本发明提供了一种抑制受试者中的肝脏VLDL-胆固醇分泌的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In yet another aspect, the present invention provides a method for inhibiting hepatic VLDL-cholesterol secretion in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在另一方面,本发明提供了一种治疗或预防受试者中与血浆胆固醇水平升高相关的疾病的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In another aspect, the present invention provides a method for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在本发明的方法的实施方案中,所述Cholesin蛋白是人Cholesin蛋白。In an embodiment of the method of the present invention, the Cholesin protein is a human Cholesin protein.

在一些实施方案中,所述核酸是DNA或RNA。In some embodiments, the nucleic acid is DNA or RNA.

在一些实施方案中,所述表达载体选自慢病毒载体、腺病毒载体、腺相关病毒(AAV)载体、逆转录病毒载体、质粒、DNA载体、mRNA载体、基于转座子的载体和人工染色体。In some embodiments, the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。 In some embodiments, the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为高胆固醇血症。In some embodiments, the disease associated with elevated plasma cholesterol levels is hypercholesterolemia.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。In some embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.

在一些实施方案中,所述方法还包括施用第二治疗剂的步骤,优选地,所述第二治疗剂选自蛋白质或肽、核酸和小分子药物。In some embodiments, the method further comprises the step of administering a second therapeutic agent, preferably, the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.

在一些实施方案中,所述第二治疗剂为选自以下的降胆固醇药物:HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。In some embodiments, the second therapeutic agent is a cholesterol-lowering drug selected from the group consisting of an HMGCR inhibitor, a PCSK9 inhibitor, and a cholesterol absorption inhibitor.

在一些实施方案中,所述HMGCR抑制剂为他汀类药物。In some embodiments, the HMGCR inhibitor is a statin.

在一些实施方案中,所述他汀类药物选自瑞舒伐他汀(Rosuvastatin)、洛伐他汀(Lovastatin)、辛伐他汀(Simvastatin)、阿托伐他汀(Atorvastatin)、普伐他汀(Pravastatin)、氟伐他汀(Fluvastatin)、西立伐他汀(Cerivastatin)和匹伐他汀(Pitavastatin),优选瑞舒伐他汀。In some embodiments, the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin.

在一些实施方案中,所述PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。In some embodiments, the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.

在一些实施方案中,所述胆固醇吸收抑制剂为NPC1L1抑制剂,优选依折麦布或海博麦布。In some embodiments, the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.

在一些实施方案中,所述第二治疗剂在所述Cholesin蛋白、所述核酸或所述表达载体之前、之后或同时施用。In some embodiments, the second therapeutic agent is administered before, after, or simultaneously with the Cholesin protein, the nucleic acid, or the expression vector.

在一些实施方案中,所述受试者为人。In some embodiments, the subject is a human.

在一些实施方案中,所述受试者对胆固醇吸收抑制剂、HMGCR抑制剂和PCSK9抑制剂中的一种或多种无应答或不耐受,例如所述受试者具有LDL受体(LDLR)缺陷。In some embodiments, the subject is unresponsive or intolerant to one or more of a cholesterol absorption inhibitor, an HMGCR inhibitor, and a PCSK9 inhibitor, for example, the subject has an LDL receptor (LDLR) defect.

在另一方面,本发明提供了组合物,其包含Cholesin蛋白、编码所述Cholesin蛋白的核酸、包含所述核酸的表达载体、或增强人内源性Cholesin蛋白的表达或活性的试剂,和药学上可接受的载剂或赋形剂。In another aspect, the present invention provides a composition comprising a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of human endogenous Cholesin protein, and a pharmaceutically acceptable carrier or excipient.

在一些实施方案中,所述组合物用于以下中的一种或多种:In some embodiments, the composition is used for one or more of the following:

1)降低受试者中的血浆胆固醇水平;1) reducing plasma cholesterol levels in a subject;

2)降低受试者中的血浆总胆固醇或血浆LDL-胆固醇水平;2) reducing plasma total cholesterol or plasma LDL-cholesterol levels in a subject;

3)降低受试者中的血浆甘油三酯水平;3) reducing plasma triglyceride levels in a subject;

4)抑制受试者中的肝脏胆固醇合成;4) inhibiting hepatic cholesterol synthesis in a subject;

5)抑制受试者中的肝脏VLDL-胆固醇分泌;和5) inhibiting hepatic VLDL-cholesterol secretion in a subject; and

6)治疗或预防受试者中与血浆胆固醇水平升高相关的疾病。6) Treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.

在一些实施方案中,所述Cholesin蛋白是人Cholesin蛋白。In some embodiments, the Cholesin protein is a human Cholesin protein.

在一些实施方案中,所述组合物还包含第二治疗剂,优选地,所述第二治疗剂选自蛋 白质或肽、核酸和小分子药物。In some embodiments, the composition further comprises a second therapeutic agent, preferably, the second therapeutic agent is selected from protein Proteins or peptides, nucleic acids and small molecule drugs.

在一些实施方案中,所述第二治疗剂选自降胆固醇药物。In some embodiments, the second therapeutic agent is selected from a cholesterol-lowering drug.

在一些实施方案中,所述降胆固醇药物选自HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。In some embodiments, the cholesterol-lowering drug is selected from the group consisting of an HMGCR inhibitor, a PCSK9 inhibitor, and a cholesterol absorption inhibitor.

在一些实施方案中,所述HMGCR抑制剂为他汀类药物。In some embodiments, the HMGCR inhibitor is a statin.

在一些实施方案中,所述他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀,优选瑞舒伐他汀。In some embodiments, the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin.

在一些实施方案中,所述PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。In some embodiments, the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.

在一些实施方案中,所述胆固醇吸收抑制剂为NPC1L1抑制剂,优选依折麦布或海博麦布。In some embodiments, the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe.

在一些实施方案中,所述组合物被配制为注射用制剂或口服制剂,例如片剂、胶囊剂、颗粒剂、混悬剂。In some embodiments, the composition is formulated as an injection preparation or an oral preparation, such as a tablet, a capsule, a granule, a suspension.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

图1:喂食普通饮食(RD)和西方饮食(WD)的小鼠血清蛋白的银染和免疫印迹结果。Figure 1: Silver staining and immunoblotting results of serum proteins from mice fed a normal diet (RD) and a Western diet (WD).

图2A:通过免疫荧光实验检测的人源Cholesin(hCholesin)和鼠源Cholesin(mCholesin)在细胞中的定位。Figure 2A: Localization of human Cholesin (hCholesin) and mouse Cholesin (mCholesin) in cells detected by immunofluorescence experiments.

图2B:通过免疫印迹实验检测的人源Cholesin和鼠源Cholesin在培养基中的分泌。Figure 2B: Secretion of human and mouse Cholesin in the culture medium detected by immunoblotting.

图3A:通过ELISA测定的禁食组、RD组和WD组小鼠的血浆Cholesin水平。FIG. 3A : Plasma Cholesin levels of fasting, RD, and WD mice measured by ELISA.

图3B:人类临床样品中禁食和进食1h后的浆Cholesin水平变化。FIG. 3B : Changes in plasma Cholesin levels in human clinical samples after fasting and 1 h of feeding.

图4:小鼠不同组织的Cholesin蛋白水平。Cholesin全身敲除(KO)小鼠作为对照。Figure 4: Cholesin protein levels in different mouse tissues. Cholesin whole body knockout (KO) mice were used as controls.

图5A:禁食和进食WD的WT小鼠和IKO小鼠的血浆Cholesin水平和小肠总胆固醇水平。Fig. 5A: Plasma Cholesin levels and small intestinal total cholesterol levels in fasted and WD-fed WT and IKO mice.

图5B:禁食和进食WD的WT小鼠和IKO小鼠的血浆总胆固醇水平和血浆甘油三酯水平。FIG. 5B : Plasma total cholesterol levels and plasma triglyceride levels in fasted and WD-fed WT and IKO mice.

图5C:灌胃玉米油(chol-)和灌胃胆固醇(chol+)的WT小鼠和IKO小鼠的血浆Cholesin水平和小肠总胆固醇水平。FIG. 5C : Plasma Cholesin levels and small intestinal total cholesterol levels in WT mice and IKO mice gavaged with corn oil (chol-) and cholesterol (chol+).

图5D:灌胃玉米油(chol-)和灌胃胆固醇(chol+)的WT小鼠和IKO小鼠的血浆总胆固醇水平和血浆甘油三酯水平。 FIG. 5D : Plasma total cholesterol levels and plasma triglyceride levels in WT mice and IKO mice gavaged with corn oil (chol-) and cholesterol (chol+).

图6:通过免疫荧光实验观察到的Cholesin的细胞定位。Fig. 6: Cellular localization of Cholesin observed by immunofluorescence experiments.

图7:过表达Cholesin的HCT116细胞经胆固醇刺激后在培养基中分泌的Cholesin水平和细胞中的总胆固醇水平。Figure 7: Cholesin levels secreted in the culture medium and total cholesterol levels in cells after cholesterol stimulation of HCT116 cells overexpressing Cholesin.

图8A:通过免疫印迹检测的NPC1L1敲低(NPC1L1 KD)的过表达Cholesin的HCT116细胞分泌的Cholesin。NPC1L1未敲低(KD)的过表达Cholesin的HCT116细胞作为对照。Fig. 8A: Cholesin secretion from Cholesin-overexpressing HCT116 cells with NPC1L1 knockdown (NPC1L1 KD) detected by immunoblotting. Cholesin-overexpressing HCT116 cells without NPC1L1 knockdown (KD) served as a control.

图8B:NPC1L1敲低(NPC1L1 KD)的过表达Cholesin的HCT116细胞与胆固醇共培养后在培养基中分泌的Cholesin水平和细胞中的总胆固醇水平。NPC1L1未敲低(NT)的过表达Cholesin的HCT116细胞作为对照。Fig. 8B: The secreted Cholesin level in the culture medium and the total cholesterol level in the cells after co-culture with cholesterol in NPC1L1 knockdown (NPC1L1 KD) Cholesin-overexpressing HCT116 cells. NPC1L1 non-knockdown (NT) Cholesin-overexpressing HCT116 cells served as a control.

图9A:经依折麦布处理的小鼠(Ezetimlbe+)在灌胃胆固醇(chol+)后血浆中的Cholesin水平和小肠中的总胆固醇水平。未依折麦布处理的小鼠(Ezetimlbe-)作为对照。Figure 9A: Cholesin levels in plasma and total cholesterol levels in the small intestine of mice treated with ezetimibe (Ezetimlbe+) after oral administration of cholesterol (chol+). Mice not treated with ezetimibe (Ezetimlbe-) served as controls.

图9B:经依折麦布处理的小鼠在灌胃胆固醇后血浆中的总胆固醇水平。未依折麦布处理的小鼠作为对照。Figure 9B: Total cholesterol levels in plasma of mice treated with ezetimibe after oral administration of cholesterol. Mice not treated with ezetimibe served as controls.

图9C:通过免疫印迹检测的Npc1l1基因敲除(Npc1l1-/-)小鼠分泌的Cholesin。Npc1l1基因未敲除的野生型(Npc1l1+/+)小鼠作为对照。Fig. 9C: Cholesin secreted by Npc1l1 knockout (Npc1l1 -/- ) mice detected by immunoblotting. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout served as controls.

图9D:Npc1l1基因敲除(Npc1l1-/-)小鼠在灌胃胆固醇后血浆中的Cholesin水平和小肠中的总胆固醇水平。Npc1l1基因未敲除的野生型(Npc1l1+/+)小鼠作为对照。Fig. 9D shows the level of Cholesin in plasma and the level of total cholesterol in small intestine of Npc1l1 knockout (Npc1l1 -/- ) mice after oral administration of cholesterol. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout were used as controls.

图9E:Npc1l1基因敲除(Npc1l1-/-)小鼠在灌胃胆固醇后血浆中的总胆固醇水平。Npc1l1基因未敲除的野生型(Npc1l1+/+)小鼠作为对照。Fig. 9E shows the total cholesterol level in plasma of Npc1l1 knockout (Npc1l1 -/- ) mice after oral administration of cholesterol. Wild-type (Npc1l1 +/+ ) mice without Npc1l1 knockout were used as controls.

图10A:人体中血浆Cholesin水平与血浆总胆固醇(TC)水平的相关性。FIG. 10A : Correlation between plasma Cholesin level and plasma total cholesterol (TC) level in humans.

图10B:血浆Cholesin水平与血浆低密度脂蛋白胆固醇(LDL-C)水平的相关性。FIG. 10B : Correlation between plasma Cholesin level and plasma low-density lipoprotein cholesterol (LDL-C) level.

图11A:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的血浆总胆固醇和甘油三酯的水平。FIG. 11A : Plasma total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.

图11B:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的肝脏总胆固醇和甘油三酯的水平。FIG. 11B : Levels of total cholesterol and triglycerides in the liver of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.

图11C:通过快速蛋白液相色谱法测定的野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的血浆总胆固醇水平以及HDL、IDL/LDL、VLDL的水平。FIG. 11C : Plasma total cholesterol levels and levels of HDL, IDL/LDL, and VLDL in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD as measured by fast protein liquid chromatography.

图12A:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的体重。FIG. 12A : Body weight of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.

图12B:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD 后的体脂率。Figure 12B: Wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD After body fat percentage.

图12C:喂食RD和WD的野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠的采食量。FIG. 12C : Food intake of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.

图12D:喂食RD和WD的野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠的饮水量。FIG. 12D : Water intake of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice fed RD and WD.

图12E:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的小肠总胆固醇和甘油三酯水平。FIG. 12E : Small intestinal total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.

图12F:通过脂质耐受实验评估的野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后小肠的乳糜微粒分解速率。FIG. 12F : Chylomicron breakdown rates in the small intestine of wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD, as assessed by lipid tolerance assay.

图13A:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后的胆囊体积、胆汁总胆固醇浓度和胆汁酸浓度。FIG. 13A : Gallbladder volume, bile total cholesterol concentration, and bile acid concentration in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.

图13B:野生型(WT)和肠道Cholesin特异性敲除(IKO)小鼠在喂食RD和WD后粪便中的总胆固醇和甘油三酯水平。FIG. 13B : Fecal total cholesterol and triglyceride levels in wild-type (WT) and intestinal Cholesin-specific knockout (IKO) mice after feeding RD and WD.

图14A:通过qPCR测定的喂食RD和WD的WT和IKO小鼠肝脏中胆固醇合成相关基因的mRNA水平。FIG. 14A : mRNA levels of cholesterol synthesis-related genes in the liver of WT and IKO mice fed RD and WD as determined by qPCR.

图14B:通过免疫印迹测定的喂食RD和WD的WT和IKO小鼠肝脏中SREBP2、HMGCR、HMGCS1和LDLR的蛋白表达水平。FIG. 14B : Protein expression levels of SREBP2, HMGCR, HMGCS1, and LDLR in the livers of WT and IKO mice fed RD and WD as determined by immunoblotting.

图15:注射泊洛沙姆407的喂食RD和WD的WT和IKO小鼠血浆VLDL的分泌率。FIG. 15 : Poloxamer 407-injected plasma VLDL secretion rate in WT and IKO mice fed RD and WD.

图16A:Cholesin与野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠各组织的结合的免疫荧光染色图。FIG. 16A : Immunofluorescence staining of Cholesin binding to various tissues of wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 −/− ) mice.

图16B:野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠原代肝细胞与Cholesin的结合的免疫荧光染色图。FIG. 16B : Immunofluorescence staining of Cholesin binding in primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 −/− ) mice.

图16C:野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠原代肝细胞与Cholesin的结合饱和曲线。FIG. 16C : Saturation curves of Cholesin binding to wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 −/− ) mouse primary hepatocytes.

图17:Cholesin与野生型(WT)和突变型(Mut)GPR146的结合亲和力曲线。FIG. 17 : Binding affinity curves of Cholesin to wild-type (WT) and mutant (Mut) GPR146.

图18:在存在和不存在Cholesin的情况下通过免疫共沉淀检测的GPR146与GNAI1的相互作用。FIG. 18 : Interaction of GPR146 with GNAI1 detected by co-immunoprecipitation in the presence and absence of Cholesin.

图19:经Cholesin处理后的野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠原代肝细胞的cAMP水平。FIG. 19 : cAMP levels in primary hepatocytes of wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 −/− ) mice after Cholesin treatment.

图20:腹腔注射Cholesin后小鼠肝脏中的Hmgcr mRNA水平。 FIG. 20 : Hmgcr mRNA levels in mouse liver after intraperitoneal injection of Cholesin.

图21A:向小鼠腹腔注射Cholesin的实验方案。FIG. 21A : Experimental scheme for intraperitoneal injection of Cholesin into mice.

图21B:腹腔注射Cholesin的小鼠肝脏中的Hmgcr mRNA水平。Figure 21B: Hmgcr mRNA levels in the liver of mice injected intraperitoneally with Cholesin.

图21C:腹腔注射Cholesin的小鼠血浆中的总胆固醇水平。FIG. 21C : Total cholesterol levels in plasma of mice injected intraperitoneally with Cholesin.

图21D:腹腔注射Cholesin的小鼠肝脏中的总胆固醇水平。FIG. 21D : Total cholesterol levels in the liver of mice injected intraperitoneally with Cholesin.

图22A:向小鼠注射腺病毒和Cholesin的实验方案。FIG. 22A : Experimental scheme for injection of adenovirus and Cholesin into mice.

图22B:在注射或不注射Cholesin的情况下,野生型(Gpr146fl/fl)小鼠和Gpr146基因敲除(Gpr146 LKO)小鼠注射腺病毒对照(Vec),表达野生型GPR146(WT)的腺病毒,或表达突变型GPR146(Mut)的腺病毒后,肝脏中的胆固醇合成相关基因Hmgcs1、Hmgcr、Mvd、Mvk和Pmvk的mRNA水平。Figure 22B: mRNA levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, and Pmvk in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut), with or without Cholesin injection.

图22C:在注射或不注射Cholesin的情况下,野生型(Gpr146fl/fl)小鼠和Gpr146基因敲除(Gpr146 LKO)小鼠注射腺病毒对照(Vec),表达野生型GPR146(WT)的腺病毒,或表达突变型GPR146(Mut)的腺病毒后,肝脏中的SREBP2、HMGCR、HMGCS1、LDLR和pKA Sub的蛋白水平。HSP90的表达水平作为内参。FIG22C : Protein levels of SREBP2, HMGCR, HMGCS1, LDLR, and pKA Sub in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut) with or without Cholesin injection. The expression level of HSP90 was used as an internal control.

图22D::在注射或不注射Cholesin的情况下,野生型(Gpr146fl/fl)小鼠和Gpr146基因敲除(Gpr146 LKO)小鼠注射腺病毒对照(Vec),表达野生型GPR146(WT)的腺病毒,或表达突变型GPR146(Mut)的腺病毒后,肝脏中的总胆固醇水平。FIG. 22D : Total cholesterol levels in the liver of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut), with or without Cholesin injection.

图22E:在注射或不注射Cholesin的情况下,野生型(Gpr146fl/fl)小鼠和Gpr146基因敲除(Gpr146 LKO)小鼠注射腺病毒对照(Vec),表达野生型GPR146(WT)的腺病毒,或表达突变型GPR146(Mut)的腺病毒后,血浆中的总胆固醇水平。22E : Total cholesterol levels in plasma of wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice injected with adenovirus control (Vec), adenovirus expressing wild-type GPR146 (WT), or adenovirus expressing mutant GPR146 (Mut), with or without Cholesin injection.

图23:Ldlr基因敲除(Ldlr-/-)小鼠给药方案示意图。FIG. 23 : Schematic diagram of the dosing regimen for Ldlr knockout (Ldlr −/− ) mice.

图24A:不同给药方案下Ldlr-/-小鼠血浆总胆固醇水平的变化及其曲线下面积(AUC)统计。FIG. 24A : Changes in plasma total cholesterol levels in Ldlr −/− mice under different dosing regimens and statistics of the area under the curve (AUC).

图24B:不同给药方案下Ldlr-/-小鼠给药8周后的肝脏总胆固醇水平。FIG. 24B : Total cholesterol levels in the liver of Ldlr −/− mice after 8 weeks of drug administration under different dosing regimens.

图24C:不同给药方案下Ldlr-/-小鼠血浆甘油三酯水平的变化及其曲线下面积(AUC)统计。FIG. 24C : Changes in plasma triglyceride levels in Ldlr −/− mice under different dosing regimens and statistics of the area under the curve (AUC).

图24D:不同给药方案下Ldlr-/-小鼠给药8周后的肝脏甘油三酯水平。FIG. 24D : Hepatic triglyceride levels in Ldlr −/− mice after 8 weeks of drug administration under different dosing regimens.

图25A:不同给药方案下Ldlr-/-小鼠给药8周后的肝脏胆固醇合成相关基因的mRNA水平。FIG. 25A : mRNA levels of genes related to cholesterol synthesis in the liver of Ldlr −/− mice after 8 weeks of drug administration under different drug administration regimens.

图25B:不同给药方案下Ldlr-/-小鼠给药8周后的肝脏胆固醇合成相关基因Hmgcs1、Hmgcr以及转录因子SREBP2的表达水平。FIG. 25B : Expression levels of liver cholesterol synthesis-related genes Hmgcs1, Hmgcr and transcription factor SREBP2 in Ldlr -/- mice after 8 weeks of drug administration under different dosing regimens.

图26:不同给药方案下Ldlr-/-小鼠给药8周后的全主动脉油红染色图及其统计结果。 Figure 26: Oil red staining of the whole aorta of Ldlr -/- mice after 8 weeks of drug administration under different drug administration regimens and their statistical results.

图27A:不同给药方案下Ldlr-/-小鼠的体重变化。FIG. 27A : Body weight changes of Ldlr −/− mice under different dosing regimens.

图27B:不同给药方案下Ldlr-/-小鼠的采食量变化。FIG. 27B : Changes in food intake of Ldlr −/− mice under different dosing regimens.

图28:不同给药方案下Ldlr-/-小鼠给药8周后的肝脏组织HE染色图。Figure 28: HE staining of liver tissue of Ldlr -/- mice after 8 weeks of drug administration under different dosing regimens.

图29A:不同给药方案下Ldlr-/-小鼠给药8周后的血浆丙氨酸氨基转移酶(ALT)水平。FIG. 29A : Plasma alanine aminotransferase (ALT) levels in Ldlr −/− mice after 8 weeks of drug administration under different dosing regimens.

图29B:不同给药方案下Ldlr-/-小鼠给药8周后的血浆天冬氨酸氨基转移酶(AST)水平。FIG. 29B : Plasma aspartate aminotransferase (AST) levels in Ldlr −/− mice after 8 weeks of administration under different dosing regimens.

图30:Cholesin通过其受体GPR146调控胆固醇代谢的作用机制示意图。Figure 30: Schematic diagram of the mechanism by which Cholesin regulates cholesterol metabolism through its receptor GPR146.

具体实施方式DETAILED DESCRIPTION

通过结合附图和以下实施方案的详细描述,本发明的上述特征和优点及其附加特征和优点将在下文中得到更清楚的理解。此处参照附图描述的实施方案是解释性的、说明性的,并用于普遍理解本发明。实施方案不应解释为限制本发明的范围。The above-mentioned features and advantages of the present invention and their additional features and advantages will be more clearly understood below by combining the accompanying drawings and the detailed description of the following embodiments. The embodiments described herein with reference to the accompanying drawings are explanatory, illustrative, and are used for a general understanding of the present invention. The embodiments should not be interpreted as limiting the scope of the present invention.

除非另有限定,否则本文所使用的全部技术和科学术语的含义与本领域普通技术人员通常所理解的含义相同。例如,本文所使用的术语如“A multilingual glossary of biotechnological terms:(IUPAC Recommendations)”,Leuenberger,H.G.W,Nagel,B.和H.编辑(1995),Helvetica Chimica Acta,CH-4010Basel,Switzerland中所述的定义。Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art. For example, terms used herein such as "A multilingual glossary of biotechnological terms: (IUPAC Recommendations)", Leuenberger, HGW, Nagel, B. and Definitions as described in H. E., ed. (1995), Helvetica Chimica Acta, CH-4010 Basel, Switzerland.

应当注意,如本文中及所附权利要求书中使用的,单数形式“一个”、“一种”和“该/所述”包括复数提及物,除非上下文另有明确规定。因此,术语“一个”、“一种”、“一个/种或多个/种”和“至少一个/种”可以互换使用。类似地,术语“包含”、“包括”和“具有”可以互换使用。It should be noted that as used herein and in the appended claims, the singular forms "a," "an," and "the" include plural references unless the context clearly dictates otherwise. Thus, the terms "a," "an," "one or more," and "at least one" may be used interchangeably. Similarly, the terms "comprising," "including," and "having" may be used interchangeably.

在本文中及所附权利要求书中使用术语“包含”时,其不排除其它元素。为了本发明的目的,术语“由...组成”被认为是术语“包含”的优选实施方案。如果在下文中将组定义为包括或包含至少一定数量的实施方案,则还应被理解为公开了优选仅由这些实施方案组成的组。When the term "comprising" is used herein and in the appended claims, it does not exclude other elements. For the purpose of the present invention, the term "consisting of" is considered to be a preferred embodiment of the term "comprising". If a group is defined hereinafter as comprising or containing at least a certain number of embodiments, it should also be understood to disclose a group that preferably consists of only these embodiments.

如本文使用的术语“和/或”视为具有或不具有另一个的两个指定特征或组件中的每一个的具体公开。因此,如在短语例如“A和/或B”中所使用的术语“和/或”旨在包括A和B;A或B;A(单独);和B(单独)。同样地,如在短语例如“A、B和/或C”中所使用的术语“和/或”旨在涵盖以下方面的每一个:A、B和C;A、B或C;A或C;A或B;B或C;A和C;A和B;B和C;A(单独);B(单独);和C(单独)。As used herein, the term "and/or" is considered a specific disclosure of each of the two specified features or components with or without the other. Thus, the term "and/or" as used in phrases such as "A and/or B" is intended to include A and B; A or B; A (alone); and B (alone). Similarly, the term "and/or" as used in phrases such as "A, B, and/or C" is intended to cover each of the following: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).

在本文中列举的范围应理解为所述范围内的所有值(包括所列举的端点值)。例如, 范围1至100应理解为包括1至100之间的任何数值、数值组合或子范围。The ranges listed in this article should be understood to include all values within the range (including the listed endpoint values). The range of 1 to 100 is understood to include any value, combination of values, or sub-range therebetween.

如本文所用的术语“Cholesin”或“Cholesin蛋白”为未知人类蛋白C7orf50的同源物,人Cholesin基因位于人类染色体7的p22区域,编码人源Cholesin的基因在NCBI中的基因ID为84310,但其具体功能尚未报道。发明人通过对该蛋白的表征发现其在胆固醇合成中的抑制作用,因此将其命名为“Cholesin”。Cholesin蛋白的氨基酸序列在各物种中具有高度保守性,不同物种中Cholesin蛋白的氨基酸序列如下所示:As used herein, the term "Cholesin" or "Cholesin protein" is a homolog of the unknown human protein C7orf50. The human Cholesin gene is located in the p22 region of human chromosome 7. The gene encoding human Cholesin has a gene ID of 84310 in NCBI, but its specific function has not yet been reported. The inventors discovered its inhibitory effect on cholesterol synthesis through characterization of the protein, so it was named "Cholesin". The amino acid sequence of Cholesin protein is highly conserved in various species. The amino acid sequences of Cholesin proteins in different species are as follows:

人(Homo sapiens)(基因ID为84310)
Homo sapiens (Gene ID 84310)

小鼠(Mus musculus)(基因ID为73212)
Mouse (Mus musculus) (Gene ID 73212)

猕猴(Macaca mulatta)(基因ID为696867)
Rhesus monkey (Macaca mulatta) (Gene ID 696867)

家牛(Bos taurus)(基因ID为522840)
Domestic cattle (Bos taurus) (Gene ID 522840)

褐家鼠(Rattus norvegicus)(基因ID为498154)
Brown rat (Rattus norvegicus) (Gene ID 498154)

原鸡(Gallus gallus)(基因ID为416455)
Junglefowl (Gallus gallus) (Gene ID 416455)

在本文中,Cholesin蛋白应以其最广泛的意义来理解,包括野生型的、分离或纯化的、或重组的Cholesin蛋白、其变体或衍生物,只要保留期望的生物活性(例如降低受试者中的血浆胆固醇水平和/或甘油三酯水平的活性)即可。Cholesin蛋白的变体或衍生物涵盖例如保留了以与野生型Cholesin蛋白类似的程度、以与野生型Cholesin蛋白相同的程度或以比野生型Cholesin蛋白更高的程度降低受试者中的血浆胆固醇水平和/或甘油三酯水平的能力的本文所述Cholesin蛋白的变体或衍生物。Cholesin蛋白的变体或衍生物保留了野生型Cholesin蛋白的至少50%、至少60%、至少70%、至少75%、至少80%、至少85%、至少90%、至少91%、至少92%、至少93%、至少94%、至少95%、至少96%、至少97%、至少98%、至少99%、至少99.1%、至少99.2%、至少99.3%、至少99.4%、至少99.5%、至少99.6%、至少99.7%、至少99.8%、至少99.9%、或100%的生物活性,或者具有比野生型Cholesin蛋白更高的生物活性,特别是降低受试者中的血浆胆固醇水平和/或甘油三酯水平的活性。In this article, Cholesin protein should be understood in its broadest sense, including wild-type, isolated or purified, or recombinant Cholesin protein, its variants or derivatives, as long as the desired biological activity (e.g., activity of reducing plasma cholesterol level and/or triglyceride level in a subject) is retained. Variants or derivatives of Cholesin protein encompass, for example, variants or derivatives of Cholesin protein described herein that retain the ability to reduce plasma cholesterol level and/or triglyceride level in a subject to a degree similar to that of wild-type Cholesin protein, to the same degree as that of wild-type Cholesin protein, or to a degree higher than that of wild-type Cholesin protein. The variant or derivative of the Cholesin protein retains at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.1%, at least 99.2%, at least 99.3%, at least 99.4%, at least 99.5%, at least 99.6%, at least 99.7%, at least 99.8%, at least 99.9%, or 100% of the biological activity of the wild-type Cholesin protein, or has a higher biological activity than the wild-type Cholesin protein, particularly the activity of reducing the plasma cholesterol level and/or triglyceride level in a subject.

Cholesin蛋白的变体包括与野生型Cholesin蛋白具有显著的序列同一性并且保留了Cholesin蛋白的生物活性(例如降低受试者中的血浆胆固醇水平和/或甘油三酯水平的活性)的多肽。变体与野生型Cholesin蛋白相比,可以具有一个或多个氨基酸突变。突变可以包括例如氨基酸插入、缺失或取代。变体的氨基酸序列与野生型Cholesin蛋白的氨基酸序列可以例如,具有至少约65%、70%、75%、80%、90%、95%、96%、97%、98%、98.2%、98.4%、98.6%、98.8%、99%、99.1%、99.2%、99.3%、99.4%、99.5%、99.6%、99.7%、99.8%、99.9%或更高的同一性。Variants of Cholesin protein include polypeptides that have significant sequence identity to wild-type Cholesin protein and retain the biological activity of Cholesin protein (e.g., activity that reduces plasma cholesterol levels and/or triglyceride levels in a subject). Variants may have one or more amino acid mutations compared to wild-type Cholesin protein. Mutations may include, for example, amino acid insertions, deletions, or substitutions. The amino acid sequence of the variant may, for example, have at least about 65%, 70%, 75%, 80%, 90%, 95%, 96%, 97%, 98%, 98.2%, 98.4%, 98.6%, 98.8%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9% or higher identity to the amino acid sequence of wild-type Cholesin protein.

Cholesin蛋白的衍生物通常是指可以通过化学修饰,特别是通过一个或多个取代基的 共价连接,由野生型Cholesin蛋白或其变体制备的化合物。因此,Cholesin蛋白的衍生物是指经化学修饰的Cholesin蛋白或其变体。典型的衍生物例如可以是酰胺化、糖基化、烷基化、酰基化、酯化、聚乙二醇化的Cholesin蛋白或其变体。Derivatives of cholesin proteins generally refer to those that have been chemically modified, especially by one or more substituents. Covalently linked compounds prepared from wild-type Cholesin protein or its variants. Therefore, the derivatives of Cholesin protein refer to chemically modified Cholesin protein or its variants. Typical derivatives can be, for example, amidated, glycosylated, alkylated, acylated, esterified, pegylated Cholesin protein or its variants.

在本文中,“增强受试者的内源性Cholesin蛋白的表达或活性的步骤”、“增强受试者的内源性Cholesin蛋白的表达或活性的方法”和“增强受试者的内源性Cholesin蛋白的表达或活性的试剂”是指任何可增强编码Cholesin蛋白的核酸的稳定性或表达、增强Cholesin的转录和翻译、增加Cholesin蛋白水平、增强Cholesin蛋白的活性、增强Cholesin蛋白的稳定性、增加Cholesin蛋白的有效作用时间的方法或物质。例如,增强受试者的内源性Cholesin蛋白的表达或活性的试剂可以是小分子、蛋白、多肽、核酸等。Herein, "a step of enhancing the expression or activity of an endogenous Cholesin protein in a subject", "a method of enhancing the expression or activity of an endogenous Cholesin protein in a subject" and "an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject" refer to any method or substance that can enhance the stability or expression of a nucleic acid encoding a Cholesin protein, enhance the transcription and translation of Cholesin, increase the level of Cholesin protein, enhance the activity of Cholesin protein, enhance the stability of Cholesin protein, and increase the effective action time of Cholesin protein. For example, an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject can be a small molecule, a protein, a polypeptide, a nucleic acid, etc.

如本文所用的术语“GPR146”是一种G蛋白偶联受体(GPCR),参与胆固醇代谢的调节,并抑制PKA信号和SREBP2控制的胆固醇合成。先前的研究发现,GPR146通过激活细胞外信号调节激酶(ERK)信号通路,促进肝脏固醇调节元件结合蛋白2(SREBP2)的活性,从而调节肝脏极低密度脂蛋白(VLDL)的分泌,进而调节循环中的低密度脂蛋白胆固醇(LDL-C)和甘油三酯(TG)水平。GPR146/ERK轴在全身胆固醇代谢中具有调节作用,抑制GPR146可能是降低血浆胆固醇水平和动脉粥样硬化的有效策略。The term "GPR146" as used herein is a G protein-coupled receptor (GPCR) that is involved in the regulation of cholesterol metabolism and inhibits cholesterol synthesis controlled by PKA signaling and SREBP2. Previous studies have found that GPR146 regulates the secretion of very low-density lipoprotein (VLDL) in the liver by activating the extracellular signal-regulated kinase (ERK) signaling pathway and promoting the activity of liver sterol regulatory element binding protein 2 (SREBP2), thereby regulating the levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) in the circulation. The GPR146/ERK axis has a regulatory role in systemic cholesterol metabolism, and inhibiting GPR146 may be an effective strategy to reduce plasma cholesterol levels and atherosclerosis.

人源GPR146基因ID为115330,氨基酸序列为:MWSCSWFNGTGLVEELPACQDLQLGLSLLSLLGLVVGVPVGLCYNALLVLANLHSKASMTMPDVYFVNMAVAGLVLSALAPVHLLGPPSSRWALWSVGGEVHVALQIPFNVSSLVAMYSTALLSLDHYIERALPRTYMASVYNTRHVCGFVWGGALLTSFSSLLFYICSHVSTRALECAKMQNAEAADATLVFIGYVVPALATLYALVLLSRVRREDTPLDRDTGRLEPSAHRLLVATVCTQFGLWTPHYLILLGHTVIISRGKPVDAHYLGLLHFVKDFSKLLAFSSSFVTPLLYRYMNQSFPSKLQRLMKKLPCGDRHCSPDHMGVQQVLA(SEQ ID NO:7)The human GPR146 gene ID is 115330, and the amino acid sequence is: MWSCSWFNGTGLVEELPACQDLQLGLSLLSLLGLVVGVPVGLCYNALLVLANLHSKASMTMPDVYFVNMAVAGLVLSALAPVHLLGPPSSRWALWSVGGEVHVALQIPFNVSSLVAMYSTALLSLDHYIERALPRTYMASVYNTRHVCGFVWGGALLTS FSSLLFYICSHVSTRALECAKMQNAEAADATLVFIGYVVPALATLYALVLLSRVRREDTPLDRDTGRLEPSAHRLLVATVCTQFGLWTPHYLILLGHTVIISRGKPVDAHYLGLLHFVKDFSKLLAFSSSFVTPLLYRYMNQSFPSKLQRLMKKLPCGDRHCSPDHMGVQQVLA(SEQ ID NO:7)

鼠源GPR146基因ID为80290,氨基酸序列为:MWSCGPLNSTAWAEEPLCRNLRLGLWVLSLLYLGAGVPVSLGYNALLVLANLASKNTMTMPDVYFVNMAVAGLVLTALAPAYLLGPAHSRWALWSLSSEAHVTLLILFNVASLVTMYSTALLSLDYYIERALPRTYMASVYNTRHVCGFVWGGAVLTSFSSLLFYICSHVSSRIAECARMQNTEAADAILVLIGYVVPGLAVLYALALISRIGKEDTPLDQDTSRLDPSVHRLLVATVCTQFGLWTPYYLSLGHTVLTSRGRTVEGHYLGILQVAKDLAKFLAFSSSSVTPLLYRYINKAFPGKLRRLMKKMHCGRRHCSPDPSGIQQVMAQA(SEQ ID NO:8)。The mouse GPR146 gene ID is 80290, and the amino acid sequence is: MWSCGPLNSTAWAEEPLCRNLRLGLWVLSLLYLGAGVPVSLGYNALLVLANLASKNTMTMPDVYFVNMAVAGLVLTALAPAYLLGPAHSRWALWSLSSEAHVTLLILFNVASLVTMYSTALLSLDYYIERALPRTYMASVYNTRHVCGFVWGGAVLTSFS SLLFYICSHVSSRIAECARMQNTEAADAILVLIGYVVPGLAVLYALALISRIGKEDTPLDQDTSRLDPSVHRLLVATVCTQFGLWTPYYLSLGHTVLTSRGRTVEGHYLGILQVAKDLAKFLAFSSSSVTPLLYRYINKAFPGKLRRLMKKMHCGRRHCSPDPSGIQQVMAQA(SEQ ID NO: 8).

胆固醇是细胞膜的主要成分,同时也是合成肾上腺皮质激素、性激素、胆汁酸及维生 素D等生理活性物质的重要原料。人体血液中胆固醇(包括总胆固醇和/或低密度脂蛋白胆固醇)浓度可作为脂代谢的指标,用于评估动心脑血管疾病、高脂血症、脉粥样硬化等疾病的发病风险。Cholesterol is the main component of cell membranes and is also the main component of the synthesis of adrenal cortex hormones, sex hormones, bile acids and vitamins. The concentration of cholesterol (including total cholesterol and/or low-density lipoprotein cholesterol) in human blood can be used as an indicator of lipid metabolism to assess the risk of cardiovascular and cerebrovascular diseases, hyperlipidemia, atherosclerosis and other diseases.

胆固醇在血液中常以脂蛋白的形式存在。脂蛋白根据颗粒大小、密度高低分为五种,即乳糜微粒、极低密度脂蛋白、中间密度脂蛋白、低密度脂蛋白和高密度脂蛋白。肝脏合成的极低密度脂蛋白运输着甘油三酯进入血液,在血液中被脂肪酶所分解,产生了极低密度脂蛋白残粒,也叫中间密度脂蛋白。中间密度脂蛋白一部分被肝脏所代谢;另一部分在血液当中被脂肪酶和肝脂酶进一步转化为低密度脂蛋白。低密度脂蛋白作用于全身,与细胞、组织的低密度脂蛋白受体结合被利用。Cholesterol often exists in the form of lipoproteins in the blood. Lipoproteins are divided into five types according to particle size and density, namely chylomicrons, very low-density lipoproteins, intermediate-density lipoproteins, low-density lipoproteins and high-density lipoproteins. The very low-density lipoproteins synthesized by the liver transport triglycerides into the blood, where they are broken down by lipase to produce very low-density lipoprotein remnants, also called intermediate-density lipoproteins. Part of the intermediate-density lipoproteins is metabolized by the liver; the other part is further converted into low-density lipoproteins by lipase and hepatic lipase in the blood. Low-density lipoproteins act on the whole body and are used by binding to low-density lipoprotein receptors of cells and tissues.

血浆中低密度脂蛋白(LDL)是运输内源性胆固醇的主要载体,其通过结合其细胞膜上的低密度脂蛋白受体(LDLR)被降解和转化。LDLR功能缺陷会造成血浆LDL的清除能力降低,最终导致动脉内膜粥样斑块形成。低密度脂蛋白胆固醇(LDL-C)是空腹血浆中的主要脂蛋白,约占血浆脂蛋白的2/3,是运输胆固醇到肝外组织的主要运载工具。因此,LDL-C的含量与心血管疾病、高脂血症、动脉粥样硬化等疾病的发病率及病变程度相关,被认为是动脉粥样硬化的主要致病因子,其浓度与冠心病的发病率有明显正相关,也是评价个体冠心病发生的危险因素的一个重要指标。在本文中,“LDL”和“LDL-C”可以互换使用。Low-density lipoprotein (LDL) in plasma is the main carrier for transporting endogenous cholesterol, which is degraded and converted by binding to the low-density lipoprotein receptor (LDLR) on its cell membrane. LDLR functional defects will reduce the clearance ability of plasma LDL, ultimately leading to the formation of atherosclerotic plaques in the arterial intima. Low-density lipoprotein cholesterol (LDL-C) is the main lipoprotein in fasting plasma, accounting for about 2/3 of plasma lipoproteins, and is the main carrier for transporting cholesterol to extrahepatic tissues. Therefore, the content of LDL-C is related to the incidence and degree of cardiovascular disease, hyperlipidemia, atherosclerosis and other diseases. It is considered to be the main pathogenic factor of atherosclerosis. Its concentration has a significant positive correlation with the incidence of coronary heart disease and is also an important indicator for evaluating the risk factors for the occurrence of coronary heart disease in individuals. In this article, "LDL" and "LDL-C" can be used interchangeably.

极低密度脂蛋白(VLDL)主要包含甘油三酯、胆固醇和脂蛋白。VLDL在肝脏中合成,并通过血液运输甘油三酯和胆固醇到组织中。这类脂蛋白由于携带胆固醇数量相对较少,且它们的颗粒相对较大,不易透过血管内膜,因此,正常的极低密度脂蛋白一般没有致动脉粥样硬化的作用。但由于极低密度脂蛋白中甘油三酯占50%-70%,胆固醇占8%-12%,所以一旦极低密度脂蛋白水平明显增高时,血浆中除甘油三酯升高外,胆固醇水平也随之增高,具有致动脉粥样硬化的作用。在本文中,“VLDL”和“VLDL-C”可以互换使用。Very low-density lipoprotein (VLDL) mainly contains triglycerides, cholesterol and lipoproteins. VLDL is synthesized in the liver and transports triglycerides and cholesterol to tissues through the blood. Since this type of lipoprotein carries a relatively small amount of cholesterol and their particles are relatively large, it is not easy to penetrate the vascular endothelium. Therefore, normal very low-density lipoprotein generally does not cause atherosclerosis. However, since triglycerides account for 50%-70% and cholesterol accounts for 8%-12% in very low-density lipoprotein, once the level of very low-density lipoprotein increases significantly, in addition to the increase in triglycerides in the plasma, the cholesterol level also increases, which has the effect of causing atherosclerosis. In this article, "VLDL" and "VLDL-C" can be used interchangeably.

中间密度脂蛋白(IDL)主要是极低密度脂蛋白(VLDL)异化的中间代谢产物,所以也称为残余的VLDL。IDL也可直接由肝脏分泌,但其量微小。其在含有载脂蛋白B的脂蛋白中是被脂蛋白脂酶(LPL)最后分解的脂蛋白。Intermediate density lipoprotein (IDL) is mainly an intermediate metabolite of very low density lipoprotein (VLDL), so it is also called residual VLDL. IDL can also be directly secreted by the liver, but its amount is very small. It is the last lipoprotein to be decomposed by lipoprotein lipase (LPL) among lipoproteins containing apolipoprotein B.

高密度脂蛋白(HDL)是直径最小但密度最大的一组血浆脂蛋白。在正常血浆中,大多数高密度脂蛋白颗粒呈球形,由疏水核心组成,周围环绕着磷脂、未酰基化的胆固醇和载脂蛋白。高密度脂蛋白胆固醇(HDL-C)可将胆固醇从肝外组织转运到肝脏进行代谢,由胆汁排除体外。在本文中,“HDL”和“HDL-C”可以互换使用。 High-density lipoprotein (HDL) is a group of plasma lipoproteins with the smallest diameter but the highest density. In normal plasma, most HDL particles are spherical and consist of a hydrophobic core surrounded by phospholipids, unacylated cholesterol, and apolipoproteins. High-density lipoprotein cholesterol (HDL-C) can transport cholesterol from extrahepatic tissues to the liver for metabolism and excretion from the body by bile. In this article, "HDL" and "HDL-C" can be used interchangeably.

乳糜微粒(CM)是血浆中最大的脂蛋白颗粒,其是运输外源性甘油三酯的主要脂蛋白。食物中包含的胆固醇和甘油三酯在小肠中被吸收,经过淋巴管,通过乳糜微粒被吸收。进入血液后,乳糜微粒要变成成熟的乳糜微粒,在脂肪酶的作用下,转变为游离脂肪酸,游离脂肪酸一方面被人体转变为能量,同时也被进一步合成,储存在脂肪中。Chylomicrons (CM) are the largest lipoprotein particles in plasma and are the main lipoproteins for transporting exogenous triglycerides. Cholesterol and triglycerides contained in food are absorbed in the small intestine, through the lymphatic vessels, and absorbed through chylomicrons. After entering the blood, chylomicrons will become mature chylomicrons and, under the action of lipase, will be converted into free fatty acids. Free fatty acids are converted into energy by the human body on the one hand, and are also further synthesized and stored in fat.

胆固醇的合成主要在肝脏中进行。在一系列酶的控制下,胆固醇从乙酰CoA生成。其中,HMGCR(3-羟基-3-甲基戊二酰辅酶A还原酶)是限速酶,其活性在转录和转录后水平上受到控制。SREBP2(甾醇调节元件结合蛋白2)作为主转录因子,支配HMGCR和其他参与胆固醇合成和吸收的基因的表达。胆固醇的生物合成和摄取是通过负反馈机制进行严格调控的,该机制可以感知细胞的胆固醇水平。当细胞缺乏胆固醇时,SREBP2与其护送蛋白SCAP(SREBP裂解激活蛋白)一起,在COPII小泡中从内质网(ER)被运输到高尔基体。在高尔基体中,SREBP2依次被位点1和位点2蛋白酶裂解。通过这种裂解释放的SREBP2的N端结构前往细胞核,在那里它作为一个转录因子,增强参与胆固醇合成相关基因(例如Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk和Sqle)的表达。相反,当细胞胆固醇水平上升时,胆固醇分子与SCAP结合,引发其与INSIG(胰岛素诱导基因)的相互作用。这种相互作用将SREBP保留在ER中,并阻止随后SREBP的激活和参与胆固醇代谢的基因的表达。这种机制有助于维持胆固醇的平衡。Cholesterol synthesis occurs mainly in the liver. Cholesterol is generated from acetyl CoA under the control of a series of enzymes. Among them, HMGCR (3-hydroxy-3-methylglutaryl CoA reductase) is the rate-limiting enzyme, and its activity is controlled at the transcriptional and post-transcriptional levels. SREBP2 (sterol regulatory element binding protein 2) acts as a master transcription factor, dominating the expression of HMGCR and other genes involved in cholesterol synthesis and uptake. Cholesterol biosynthesis and uptake are strictly regulated by a negative feedback mechanism that senses the cholesterol level of the cell. When cells are cholesterol-deficient, SREBP2, together with its escort protein SCAP (SREBP cleavage-activating protein), is transported from the endoplasmic reticulum (ER) to the Golgi apparatus in COPII vesicles. In the Golgi apparatus, SREBP2 is cleaved by site 1 and site 2 proteases in sequence. The N-terminal structure of SREBP2 released by this cleavage travels to the nucleus, where it acts as a transcription factor, enhancing the expression of genes involved in cholesterol synthesis (e.g., Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk, and Sqle). Conversely, when cellular cholesterol levels rise, cholesterol molecules bind to SCAP, triggering its interaction with INSIG (insulin-induced gene). This interaction retains SREBP in the ER and prevents subsequent activation of SREBP and expression of genes involved in cholesterol metabolism. This mechanism helps maintain cholesterol homeostasis.

内源性合成的和外源性获得的胆固醇都以极低密度脂蛋白(VLDL)的形式分泌到血液中。在血液中处理后,VLDL产生循环的LDL,它们可以通过LDL受体(LDLR)介导的内吞作用被细胞吸收。Both endogenously synthesized and exogenously acquired cholesterol are secreted into the blood in the form of very low-density lipoprotein (VLDL). After processing in the blood, VLDL generates circulating LDL, which can be taken up by cells via LDL receptor (LDLR)-mediated endocytosis.

如本文所用的术语“表达载体”是用作将(外源)遗传材料转移到宿主细胞中的媒介的核酸分子,在该宿主细胞中作为载体的所述核酸分子可以例如复制和/或表达。术语“表达载体”涵盖但不限于质粒、病毒载体(包括例如逆转录病毒载体、慢病毒载体、腺病毒载体、牛痘病毒载体、多瘤病毒载体和腺病毒相关载体(AAV))、噬菌体、噬菌粒、粘粒和人工染色体(包括例如BAC和YAC)。As used herein, the term "expression vector" is a nucleic acid molecule used as a medium for transferring (exogenous) genetic material into a host cell, where the nucleic acid molecule as a vector can, for example, be replicated and/or expressed. The term "expression vector" encompasses, but is not limited to, plasmids, viral vectors (including, for example, retroviral vectors, lentiviral vectors, adenoviral vectors, vaccinia virus vectors, polyoma virus vectors, and adenovirus-associated vectors (AAV)), phages, phagemids, cosmids, and artificial chromosomes (including, for example, BACs and YACs).

如本文所用的术语“治疗”包括在有需要的受试者中的治疗性或预防性治疗。“治疗性或预防性治疗”包括旨在完全预防临床和/或病理表现的预防性治疗或旨在改善或缓解临床和/或病理表现的治疗性治疗。因此,术语“治疗”还包括改善或预防疾病。As used herein, the term "treatment" includes therapeutic or prophylactic treatment in a subject in need thereof. "Therapeutic or prophylactic treatment" includes prophylactic treatment aimed at completely preventing clinical and/or pathological manifestations or therapeutic treatment aimed at improving or alleviating clinical and/or pathological manifestations. Therefore, the term "treatment" also includes ameliorating or preventing a disease.

如本文所用的术语“有效量”意指当施用到受试者用于治疗或预防疾病时足以实现这样的治疗或预防的治疗剂的量。“有效量”可根据化合物、疾病及其严重度、以及待治疗的受试者的年龄、体重等改变。“治疗有效量”是指用于治疗性治疗的有效量。“预防有效量”是指用于预防性治疗的有效量。 As used herein, the term "effective amount" means an amount of a therapeutic agent that, when administered to a subject for the treatment or prevention of a disease, is sufficient to achieve such treatment or prevention. The "effective amount" may vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated. A "therapeutically effective amount" refers to an effective amount for therapeutic treatment. A "prophylactically effective amount" refers to an effective amount for prophylactic treatment.

如本文所用的术语“受试者”或“个体”或“动物”或“患者”在本文可互换使用,指需要治疗的任何受试者,特别是哺乳动物受试者。一般而言,哺乳动物受试者包括人、非人灵长类动物、狗、猫、豚鼠、兔、大鼠、小鼠、马、牛、乳牛等。As used herein, the terms "subject" or "individual" or "animal" or "patient" are used interchangeably herein and refer to any subject in need of treatment, particularly a mammalian subject. Generally speaking, mammalian subjects include humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, cows, dairy cows, etc.

如本文所用的术语“与血浆胆固醇水平升高相关的疾病”是指由血浆胆固醇水平升高引起的疾病、病况或障碍,其包括但不限于由血脂异常导致的疾病和脂肪代谢障碍相关疾病。As used herein, the term "disease associated with elevated plasma cholesterol levels" refers to a disease, condition or disorder caused by elevated plasma cholesterol levels, which includes but is not limited to diseases caused by dyslipidemia and lipodystrophy-related diseases.

血脂异常是人体内脂蛋白的代谢异常,主要包括总胆固醇和低密度脂蛋白胆固醇、甘油三酯升高和/或高密度脂蛋白胆固醇降低等。血脂异常可导致高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病、心绞痛、恶性心律失常)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肝功能衰竭、肾病综合征、胆结石、胰腺炎等疾病或症状。Dyslipidemia is an abnormal metabolism of lipoproteins in the human body, mainly including elevated total cholesterol, low-density lipoprotein cholesterol, triglycerides and/or reduced high-density lipoprotein cholesterol, etc. Dyslipidemia can lead to hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease, angina pectoris, malignant arrhythmia), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, liver failure, nephrotic syndrome, gallstones, pancreatitis and other diseases or symptoms.

脂肪代谢障碍相关疾病为脂肪代谢紊乱导致的相关疾病的总称,包括例如:高脂血症、高血压、动脉粥样硬化、肥胖症、脂肪肝、肝硬化、冠心病、心绞痛、心肌梗死、炎性肠病、消化不良和胃肠道溃疡等。Diseases related to fat metabolism disorder are a general term for related diseases caused by fat metabolism disorders, including, for example: hyperlipidemia, hypertension, atherosclerosis, obesity, fatty liver, cirrhosis, coronary heart disease, angina pectoris, myocardial infarction, inflammatory bowel disease, indigestion and gastrointestinal ulcers.

高脂血症通常是指血浆胆固醇和/或甘油三酯水平升高。高脂血症是人体脂肪代谢异常的表现,主要分为三类:高胆固醇血症、高甘油三酯血症、和混合性高脂血症。其中,高胆固醇血症是指血浆总胆固醇或者低密度脂蛋白胆固醇水平增高。高胆固醇血症可导致各种危及生命的心血管疾病、动脉粥样硬化等并发症出现。Hyperlipidemia usually refers to elevated levels of plasma cholesterol and/or triglycerides. Hyperlipidemia is a manifestation of abnormal fat metabolism in the human body and is mainly divided into three categories: hypercholesterolemia, hypertriglyceridemia, and mixed hyperlipidemia. Among them, hypercholesterolemia refers to elevated levels of total plasma cholesterol or low-density lipoprotein cholesterol. Hypercholesterolemia can lead to various life-threatening cardiovascular diseases, atherosclerosis and other complications.

动脉粥样硬化是动脉硬化的血管病中常见的最重要的一种,脂质代谢障碍是动脉粥样硬化的病变基础,其特点是受累动脉病变从内膜开始。一般先有脂质和复合糖类积聚、出血及血栓形成,纤维组织增生及钙质沉着,并有动脉中层的逐渐蜕变和钙化,病变常累及弹性及大中等肌性动脉,一旦发展到足以阻塞动脉腔,则该动脉所供应的组织或器官将缺血或坏死。由于在动脉内膜积聚的脂质外观呈黄色粥样,因此称为动脉粥样硬化。动脉粥样硬化常伴随高血压、糖尿病、肥胖等疾病。Atherosclerosis is the most common and important type of vascular disease of arteriosclerosis. Lipid metabolism disorder is the pathological basis of atherosclerosis, and its characteristic is that the lesions of the affected arteries start from the intima. Generally, there is accumulation of lipids and complex carbohydrates, bleeding and thrombosis, fibrosis and calcification, and gradual degeneration and calcification of the middle layer of the artery. The lesions often involve elastic and large and medium muscular arteries. Once they develop to the point of blocking the arterial lumen, the tissues or organs supplied by the artery will be ischemic or necrotic. Because the lipids accumulated in the intima of the artery appear yellow and porridge-like, it is called atherosclerosis. Atherosclerosis is often accompanied by diseases such as hypertension, diabetes, and obesity.

如本文所用的术语“HMGCR抑制剂”也称为HMG-CoA还原酶抑制剂,是指能够抑制HMG-CoA还原酶的化合物。其通过抑制HMG-CoA还原酶来抑制类异戊二烯的生物合成,阻断蛋白的异戊二烯化,从而降低血浆胆固醇水平。As used herein, the term "HMGCR inhibitor" is also referred to as an HMG-CoA reductase inhibitor, and refers to a compound capable of inhibiting HMG-CoA reductase. It inhibits the biosynthesis of isoprenoids by inhibiting HMG-CoA reductase, blocks the isoprenylation of proteins, and thereby reduces plasma cholesterol levels.

如本文所用的术语“PCSK9抑制剂”是一类抑制PCSK9(Kexin样前转化酶枯草杆菌蛋白酶家族的第9个成员)的化合物。PCSK9是一种肝源性分泌蛋白,能通过降低肝细胞上LDLR的数量,影响LDL内化,使血液中LDL不能清除,从而导致高胆固醇血 症。因此,抑制PCSK9能够稳定LDLR,促进肝脏对LDL的吸收,从而降低血液中LDL的水平。As used herein, the term "PCSK9 inhibitor" refers to a class of compounds that inhibit PCSK9 (the ninth member of the Kexin-like proconvertase subtilisin family). PCSK9 is a liver-derived secretory protein that can reduce the number of LDLRs on hepatocytes, affect LDL internalization, and prevent LDL from being cleared from the blood, thereby leading to hypercholesterolemia. Therefore, inhibiting PCSK9 can stabilize LDLR and promote the liver's absorption of LDL, thereby reducing the level of LDL in the blood.

如本文所用的术语“胆固醇吸收抑制剂”是指能够抑制胆固醇的吸收,例如通过抑制NPC1L1介导的内吞作用从而阻断胆固醇的摄入的化合物。As used herein, the term "cholesterol absorption inhibitor" refers to a compound that is capable of inhibiting the absorption of cholesterol, for example, by inhibiting NPC1L1-mediated endocytosis, thereby blocking the uptake of cholesterol.

术语“组合物”特别是指适合施用于人的组合物。然而,该术语通常也涵盖适合施用于非人动物的组合物。所述组合物及其组分(即活性剂和任选的载剂或赋形剂)优选为药物上可接受的,即在接受者中能够引发所需的治疗效果而不会引起任何不希望的局部或全身作用。The term "composition" refers in particular to a composition suitable for administration to humans. However, the term also generally encompasses compositions suitable for administration to non-human animals. The composition and its components (i.e., active agent and optional carrier or excipient) are preferably pharmaceutically acceptable, i.e., capable of inducing the desired therapeutic effect in the recipient without causing any undesirable local or systemic effects.

术语“药学上可接受”是指载剂或赋形剂与组合物的其他成分相容并且对其接受者没有大量毒害,和/或此类载剂或赋形剂被批准或可用于包含在组合物中。The term "pharmaceutically acceptable" means that the carrier or excipient is compatible with the other ingredients of the composition and not substantially toxic to the recipient thereof, and/or such carrier or excipient is approved or available for inclusion in the composition.

发明人出人意料地发现,Cholesin是一种由肠道分泌的响应胆固醇吸收的激素,其能够抑制肝脏中的胆固醇合成和VLDL的分泌,从而降低血浆胆固醇和/或甘油三酯的水平,以防止高胆固醇血症和动脉硬化等与血浆胆固醇水平升高相关的疾病。机制研究表明,Cholesin通过与其受体GPR146结合,抑制PKA信号和SREBP2控制的胆固醇合成。Cholesin对参与胆固醇合成的基因的表达产生不依赖于LDLR的下调作用,并降低血浆胆固醇水平,这意味着Cholesin治疗是对抗高胆固醇血症和动脉粥样硬化等与血浆胆固醇水平升高相关的疾病的有效策略。因此,外源性Cholesin或促进受试者内源性Cholesin蛋白表达或活性的方法或试剂均能降低血浆胆固醇和/或甘油三酯的水平,从而用于预防或治疗高胆固醇血症和动脉硬化等与血浆胆固醇水平升高相关的疾病。此外,将外源性Cholesin或促进受试者内源性Cholesin蛋白表达或活性的方法或试剂与他汀类等现有降胆固醇药物组合也是治疗包括高脂血症和动脉粥样硬化在内的与血浆胆固醇水平升高相关的疾病的有前途的策略。The inventors unexpectedly discovered that Cholesin is a hormone secreted by the intestine in response to cholesterol absorption, which can inhibit cholesterol synthesis and VLDL secretion in the liver, thereby reducing the levels of plasma cholesterol and/or triglycerides to prevent diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. Mechanistic studies have shown that Cholesin inhibits cholesterol synthesis controlled by PKA signaling and SREBP2 by binding to its receptor GPR146. Cholesin has an LDLR-independent downregulation effect on the expression of genes involved in cholesterol synthesis and reduces plasma cholesterol levels, which means that Cholesin treatment is an effective strategy to combat diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. Therefore, exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin proteins in subjects can reduce the levels of plasma cholesterol and/or triglycerides, thereby preventing or treating diseases associated with elevated plasma cholesterol levels, such as hypercholesterolemia and atherosclerosis. In addition, combining exogenous Cholesin or methods or agents that promote the expression or activity of endogenous Cholesin protein in a subject with existing cholesterol-lowering drugs such as statins is also a promising strategy for treating diseases associated with elevated plasma cholesterol levels, including hyperlipidemia and atherosclerosis.

相应地,在一方面,本发明提供了一种降低受试者中的血浆胆固醇水平和/或甘油三酯水平的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。Accordingly, in one aspect, the present invention provides a method for lowering plasma cholesterol level and/or triglyceride level in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在一些实施方案中,血浆胆固醇选自血浆总胆固醇和血浆LDL-胆固醇。在一些实施方案中,血浆胆固醇是血浆总胆固醇。在一些实施方案中,血浆胆固醇是血浆LDL-胆固醇。在一些实施方案中,血浆胆固醇是血浆总胆固醇和血浆LDL-胆固醇。In some embodiments, plasma cholesterol is selected from plasma total cholesterol and plasma LDL-cholesterol. In some embodiments, plasma cholesterol is plasma total cholesterol. In some embodiments, plasma cholesterol is plasma LDL-cholesterol. In some embodiments, plasma cholesterol is plasma total cholesterol and plasma LDL-cholesterol.

在另一方面,本发明提供了一种抑制受试者中的肝脏胆固醇合成的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸 的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In another aspect, the present invention provides a method for inhibiting liver cholesterol synthesis in a subject, comprising administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or a nucleic acid comprising the Cholesin protein. The step of providing an expression vector for the subject, or the step of enhancing the expression or activity of the endogenous Cholesin protein of the subject.

在一些实施方案中,所述方法抑制与胆固醇合成相关基因的表达。在一些实施方案中,与胆固醇合成相关基因选自Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk和Sqle中的一种或多种。在一些实施方案中,与胆固醇合成相关基因为Hmgcr。在一些实施方案中,与胆固醇合成相关基因为Hmgcs1。在一些实施方案中,与胆固醇合成相关基因为Mvd。在一些实施方案中,与胆固醇合成相关基因为Mvk。在一些实施方案中,与胆固醇合成相关基因为Pmvk。在一些实施方案中,与胆固醇合成相关基因为Sqle。在一些实施方案中,与胆固醇合成相关基因为Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk和Sqle。In some embodiments, the method inhibits the expression of genes related to cholesterol synthesis. In some embodiments, the genes related to cholesterol synthesis are selected from one or more of Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle. In some embodiments, the gene related to cholesterol synthesis is Hmgcr. In some embodiments, the gene related to cholesterol synthesis is Hmgcs1. In some embodiments, the gene related to cholesterol synthesis is Mvd. In some embodiments, the gene related to cholesterol synthesis is Mvk. In some embodiments, the gene related to cholesterol synthesis is Pmvk. In some embodiments, the gene related to cholesterol synthesis is Sqle. In some embodiments, the gene related to cholesterol synthesis is Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle.

在一些实施方案中,所述方法抑制与胆固醇摄取相关基因的表达。在一些实施方案中,与胆固醇合成摄取相关基因为Ldlr。In some embodiments, the method inhibits the expression of a gene associated with cholesterol uptake. In some embodiments, the gene associated with cholesterol synthesis and uptake is Ldlr.

在一些实施方案中,所述方法抑制与胆固醇合成和摄取相关基因的表达。在一些实施方案中,与胆固醇合成相关基因为Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk和Sqle;并且与胆固醇合成摄取相关基因为Ldlr。In some embodiments, the method inhibits the expression of genes associated with cholesterol synthesis and uptake. In some embodiments, the genes associated with cholesterol synthesis are Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk and Sqle; and the gene associated with cholesterol synthesis and uptake is Ldlr.

在一些实施方案中,所述方法抑制促进胆固醇合成相关基因表达的转录因子的表达。在一些实施方案中,所述方法抑制促进胆固醇摄取相关基因表达的转录因子的表达。在一些实施方案中,促进胆固醇合成和/或摄取相关基因表达的转录因子为SREBP2。In some embodiments, the method inhibits the expression of transcription factors that promote the expression of cholesterol synthesis-related genes. In some embodiments, the method inhibits the expression of transcription factors that promote the expression of cholesterol uptake-related genes. In some embodiments, the transcription factor that promotes the expression of cholesterol synthesis and/or uptake-related genes is SREBP2.

在又一方面,本发明提供了一种抑制受试者中的肝脏VLDL-胆固醇分泌的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In yet another aspect, the present invention provides a method for inhibiting hepatic VLDL-cholesterol secretion in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在另一方面,本发明提供了一种治疗或预防受试者中与血浆胆固醇水平升高相关的疾病的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。In another aspect, the present invention provides a method for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein.

在本发明的方法的实施方案中,Cholesin蛋白是分离或纯化的。在一些实施方案中,Cholesin蛋白是哺乳动物Cholesin蛋白。哺乳动物Cholesin蛋白的实例包括但限于人、非人灵长类动物、狗、猫、豚鼠、兔、大鼠、小鼠、马、牛的Cholesin蛋白。在一些实施方案中,Cholesin蛋白是非人灵长类动物Cholesin蛋白。在一些实施方案中,Cholesin蛋白是人Cholesin蛋白。在一些实施方案中,人Cholesin蛋白包含如SEQ ID NO:1所示的氨基酸序列。In an embodiment of the method of the present invention, the Cholesin protein is isolated or purified. In some embodiments, the Cholesin protein is a mammalian Cholesin protein. Examples of mammalian Cholesin proteins include but are not limited to Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cows. In some embodiments, the Cholesin protein is a non-human primate Cholesin protein. In some embodiments, the Cholesin protein is a human Cholesin protein. In some embodiments, the human Cholesin protein comprises an amino acid sequence as shown in SEQ ID NO:1.

在其他实施方案中,Cholesin蛋白是小鼠Cholesin蛋白。在一些实施方案中,小鼠Cholesin蛋白包含如SEQ ID NO:2所示的氨基酸序列。 In other embodiments, the Cholesin protein is a mouse Cholesin protein. In some embodiments, the mouse Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2.

在其他实施方案中,Cholesin蛋白是猕猴Cholesin蛋白。在一些实施方案中,猕猴Cholesin蛋白包含如SEQ ID NO:3所示的氨基酸序列。In other embodiments, the Cholesin protein is a macaque Cholesin protein. In some embodiments, the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3.

在其他实施方案中,Cholesin蛋白是家牛Cholesin蛋白。在一些实施方案中,家牛Cholesin蛋白包含如SEQ ID NO:4所示的氨基酸序列。In other embodiments, the Cholesin protein is a bovine Cholesin protein. In some embodiments, the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4.

在其他实施方案中,Cholesin蛋白是褐家鼠Cholesin蛋白。在一些实施方案中,褐家鼠Cholesin蛋白包含如SEQ ID NO:5所示的氨基酸序列。In other embodiments, the Cholesin protein is a Rattus norvegicus Cholesin protein. In some embodiments, the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5.

在其他实施方案中,Cholesin蛋白是原鸡Cholesin蛋白。在一些实施方案中,原鸡Cholesin蛋白包含如SEQ ID NO:6所示的氨基酸序列。In other embodiments, the Cholesin protein is a Gallus gallus Cholesin protein. In some embodiments, the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.

在一些实施方案中,所述核酸是DNA或RNA。在一些实施方案中,所述核酸是DNA。在一些实施方案中,所述核酸编码人Cholesin蛋白。In some embodiments, the nucleic acid is DNA or RNA. In some embodiments, the nucleic acid is DNA. In some embodiments, the nucleic acid encodes human Cholesin protein.

在一些实施方案中,所述表达载体选自慢病毒载体、腺病毒载体、腺相关病毒(AAV)载体、逆转录病毒载体、质粒、DNA载体、mRNA载体、基于转座子的载体和人工染色体。In some embodiments, the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.

表达载体本身通常是核苷酸序列,通常是包含插入物(转基因)的DNA序列和作为载体“骨架”的较大序列。工程化载体通常包含在宿主细胞中自主复制的起点(如果需要多核苷酸的稳定表达)、选择标记和限制酶切割位点(如多克隆位点,MCS)。载体可另外包含启动子、遗传标记、报告基因、靶向序列和/或蛋白质纯化标签。如本领域技术人员已知的,大量合适的表达载体是本领域技术人员已知的,并且许多可商购获得。The expression vector itself is usually a nucleotide sequence, usually a DNA sequence containing an insert (transgene) and a larger sequence as a vector "backbone". Engineered vectors usually include an origin of autonomous replication in a host cell (if stable expression of the polynucleotide is desired), a selection marker, and a restriction enzyme cleavage site (such as a multiple cloning site, MCS). The vector may additionally include a promoter, a genetic marker, a reporter gene, a targeting sequence, and/or a protein purification tag. As known to those skilled in the art, a large number of suitable expression vectors are known to those skilled in the art, and many are commercially available.

本文公开的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂可用于治疗和或/预防与血浆胆固醇水平升高相关的疾病,即由血浆胆固醇水平升高引导的疾病、病况或障碍,例如适用于治疗和/或预防:The Cholesin protein disclosed herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent for enhancing the expression or activity of the endogenous Cholesin protein of a subject can be used for treating and/or preventing diseases associated with elevated plasma cholesterol levels, i.e., diseases, conditions or disorders induced by elevated plasma cholesterol levels, for example, for treating and/or preventing:

1.血脂异常及其后遗症,例如动脉粥样硬化、心脑血管疾病等,尤其涉及下述一个或多个因素所表征的那些疾病(但不限于此):1. Dyslipidemia and its sequelae, such as atherosclerosis, cardiovascular and cerebrovascular diseases, etc., especially those diseases characterized by one or more of the following factors (but not limited to):

-高血浆总胆固醇浓度,- high plasma total cholesterol concentration,

-高血浆甘油三酯浓度、高餐后血浆甘油三酯浓度,- High plasma triglyceride concentrations, high postprandial plasma triglyceride concentrations,

-低HDL胆固醇浓度,- low HDL cholesterol concentration,

-低ApoA脂蛋白浓度,- low ApoA lipoprotein concentration,

-高LDL胆固醇浓度,- high LDL cholesterol concentration,

-低密度LDL胆固醇颗粒,- low density LDL cholesterol particles,

-高ApoB脂蛋白浓度; - High ApoB lipoprotein concentration;

2.可能与代谢综合征有关的多种其它病症,如:2. A variety of other conditions that may be associated with metabolic syndrome, such as:

-肥胖症(超重),包括向心性肥胖,- Obesity (overweight), including central obesity,

-血栓形成、凝固性过高和血栓形成前状态(动脉和静脉),-Thrombosis, hypercoagulability and prothrombotic states (arterial and venous),

-高血压,-hypertension,

-心力衰竭,例如(但不限于)心肌梗死、高血压性心脏病或心肌病后的心力衰竭;- Heart failure, such as (but not limited to) heart failure following myocardial infarction, hypertensive heart disease or cardiomyopathy;

3.脂肪代谢障碍3. Lipodystrophy

-高脂血症,- Hyperlipidemia,

-动脉粥样硬化,- Atherosclerosis,

-高血压,-hypertension,

-肥胖症,- Obesity,

-脂肪肝、肝硬化、肝功能衰竭,- Fatty liver, cirrhosis, liver failure,

4.糖尿病,尤其是II型糖尿病,包括与之有关的后遗症4. Diabetes, especially type 2 diabetes, including its related sequelae

其中具体涉及:Specifically, it involves:

-糖尿病以及糖尿病并发症,例如糖尿病血管病变,例如大血管和微血管受损导致的心、脑、肾、周围神经、眼睛、足等组织、器官的病变,包括糖尿病性眼病,糖尿病性心脏病、糖尿病性肾病、糖尿病性神经病变和下肢远端肢体坏死等,- Diabetes and diabetic complications, such as diabetic vascular disease, such as damage to large and small blood vessels leading to lesions of the heart, brain, kidneys, peripheral nerves, eyes, feet and other tissues and organs, including diabetic eye disease, diabetic heart disease, diabetic nephropathy, diabetic neuropathy and distal limb necrosis of the lower limbs, etc.

-在糖尿病情况下高发、伴随糖尿病发生或由于糖尿病加重的疾病,例如动脉粥样硬化、高血压、冠心病、心肌梗塞、脑血栓、脑出血、脑栓塞、骨质疏松等,- Diseases that are common in diabetes, occur with diabetes, or are aggravated by diabetes, such as atherosclerosis, hypertension, coronary heart disease, myocardial infarction, cerebral thrombosis, cerebral hemorrhage, cerebral embolism, osteoporosis, etc.,

-在糖尿病情况下高发、伴随糖尿病发生或由于糖尿病加重的脂肪代谢紊乱及其相关疾病,包括高脂血症,高血压、动脉粥样硬化、肥胖症、脂肪肝、肝硬化;- Fat metabolism disorders and related diseases that are common in diabetes, occur with diabetes, or are aggravated by diabetes, including hyperlipidemia, hypertension, atherosclerosis, obesity, fatty liver, and cirrhosis;

5.涉及炎性反应的疾病或病症:5. Diseases or conditions involving inflammatory responses:

-动脉粥样硬化,例如(但不限于)冠状动脉硬化,包括心绞痛或心肌梗塞、中风,- atherosclerosis, such as (but not limited to) coronary artery atherosclerosis, including angina pectoris or myocardial infarction, stroke,

-血管再狭窄或再闭塞,- Restenosis or reocclusion of the blood vessel,

-慢性炎性肠病,例如克罗恩病和溃疡性结肠炎,- Chronic inflammatory bowel disease, such as Crohn's disease and ulcerative colitis,

-哮喘,-asthma,

-其它炎性状态;- Other inflammatory conditions;

6.其它障碍:6. Other obstacles:

-肾病综合征,- Nephrotic syndrome,

-胰腺炎,-pancreatitis,

-胆结石,- Gallstones,

-脉管炎, - vasculitis,

-缺血/再灌注综合征。-Ischemia/reperfusion syndrome.

在一些实施方案中,与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。In some embodiments, the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.

在优选实施方案中,与血浆胆固醇水平升高相关的疾病为高胆固醇血症。在其他优选的实施方案中,与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。In a preferred embodiment, the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In other preferred embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.

在一些实施方案中,所述方法还包括施用第二治疗剂的步骤。在优选的实施方案中,第二治疗剂选自蛋白质或肽、核酸和小分子药物。In some embodiments, the method further comprises the step of administering a second therapeutic agent. In a preferred embodiment, the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.

在一些实施方案中,第二治疗剂为降胆固醇药物。在一些实施方案中,降胆固醇药物选自:HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。In some embodiments, the second therapeutic agent is a cholesterol-lowering drug. In some embodiments, the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.

在一些实施方案中,降胆固醇药物为HMGCR抑制剂。在一些实施方案中,HMGCR抑制剂为他汀类药物。在一些实施方案中,他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀。在优选的实施方案中,第二治疗剂为瑞舒伐他汀。In some embodiments, the cholesterol-lowering drug is an HMGCR inhibitor. In some embodiments, the HMGCR inhibitor is a statin. In some embodiments, the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin. In a preferred embodiment, the second therapeutic agent is rosuvastatin.

在一些实施方案中,降胆固醇药物为PCSK9抑制剂。在一些实施方案中,PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。在一些实施方案中,第二治疗剂为Evolocumab。在一些实施方案中,第二治疗剂为Alirocumab。在一些实施方案中,第二治疗剂为Inclisiran。在一些实施方案中,第二治疗剂为Tafolecimab。In some embodiments, the cholesterol-lowering drug is a PCSK9 inhibitor. In some embodiments, the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab. In some embodiments, the second therapeutic agent is Evolocumab. In some embodiments, the second therapeutic agent is Alirocumab. In some embodiments, the second therapeutic agent is Inclisiran. In some embodiments, the second therapeutic agent is Tafolecimab.

Evolocumab(依洛尤单抗,商品名为)是一种全人源IgG2型单克隆抗体,Evolocumab能结合PCSK9并抑制循环型PCSK9与低密度脂蛋白受体(LDLR)的结合,从而阻止PCSK9介导的低密度脂蛋白受体降解,其被批准的适应症为高胆固醇血症和混合血脂异常。Evolocumab (Evolocumab, trade name ) is a fully human IgG2 monoclonal antibody. Evolocumab can bind to PCSK9 and inhibit the binding of circulating PCSK9 to the low-density lipoprotein receptor (LDLR), thereby preventing PCSK9-mediated low-density lipoprotein receptor degradation. Its approved indications are hypercholesterolemia and mixed dyslipidemia.

Alirocumab(阿利西尤单抗,商品名为)是一种全人源IgG1型单克隆抗体,其能结合PCSK9并抑制循环型PCSK9与低密度脂蛋白受体(LDLR)的结合,从而阻止PCSK9介导的低密度脂蛋白受体降解。该药用于治疗成人杂合子型家族性高胆甾醇血症和临床动脉粥样硬化心血管疾病(例如,需降低低密度胆固醇的心脏病或中风)。Alirocumab (Alirocumab, trade name ) is a fully human IgG1 monoclonal antibody that binds to PCSK9 and inhibits the binding of circulating PCSK9 to the low-density lipoprotein receptor (LDLR), thereby preventing PCSK9-mediated degradation of the low-density lipoprotein receptor. The drug is used to treat adults with heterozygous familial hypercholesterolemia and clinical atherosclerotic cardiovascular disease (e.g., heart disease or stroke requiring low-density cholesterol reduction).

Inclisiran(英克司兰,商品名为)是一种靶向PCSK9、用于降低低密度脂蛋白胆固醇的小干扰核酸(siRNA)药物,其已被批注用于成人原发性高胆固醇血症(杂合子型家族性和非家族性)或混合型血脂异常患者的治疗。Inclisiran (Inclisiran, trade name ) is a small interfering nucleic acid (siRNA) drug targeting PCSK9 for lowering low-density lipoprotein cholesterol. It has been approved for the treatment of adult patients with primary hypercholesterolemia (heterozygous familial and non-familial) or mixed dyslipidemia.

Tafolecimab(托莱西单抗,商品名为)是一种全人源IgG2单克隆抗体, 能特异性结合PCSK9分子,通过减少PCSK9介导的低密度脂蛋白受体(LDLR)内吞来增加LDLR水平,继而增加低密度脂蛋白胆固醇(LDL-C)清除,降低LDL-C水平,其用于在接受中等剂量或中等剂量以上他汀类药物治疗仍无法达到LDL-C目标的原发性高胆固醇血症(包括杂合子型家族性和非家族性高胆固醇血症)和混合型血脂异常的成人患者,以降低LDL-C、总胆固醇(TC)和载脂蛋白B(ApoB)水平。Tafolecimab (Tafolecimab, trade name ) is a fully human IgG2 monoclonal antibody. It can specifically bind to the PCSK9 molecule, increase the low-density lipoprotein receptor (LDLR) level by reducing PCSK9-mediated endocytosis of the LDLR, and then increase the clearance of low-density lipoprotein cholesterol (LDL-C) and reduce the LDL-C level. It is used for adult patients with primary hypercholesterolemia (including heterozygous familial and non-familial hypercholesterolemia) and mixed dyslipidemia who are unable to achieve the LDL-C target after receiving moderate or higher doses of statins, to reduce LDL-C, total cholesterol (TC) and apolipoprotein B (ApoB) levels.

在一些实施方案中,降胆固醇药物为胆固醇吸收抑制剂。在一些实施方案中,胆固醇吸收抑制剂为NPC1L1抑制剂。NPC1L1抑制剂以胆固醇转运蛋白NPC1L1为作用靶点,通过作用与小肠绒毛刷状缘上胆固醇摄取与吸收的关键转运蛋白NPC1L1,抑制胆固醇的胞内转运,减少小肠中食物和胆汁中胆固醇的吸收,阻碍胆固醇经小肠向肝脏转运途径,降低肝脏胆固醇贮量,加速胆固醇的清除。In some embodiments, the cholesterol-lowering drug is a cholesterol absorption inhibitor. In some embodiments, the cholesterol absorption inhibitor is an NPC1L1 inhibitor. NPC1L1 inhibitors target the cholesterol transporter NPC1L1, and inhibit the intracellular transport of cholesterol by acting on the key transporter NPC1L1 for cholesterol uptake and absorption on the brush border of the small intestinal villi, thereby reducing the absorption of cholesterol in food and bile in the small intestine, hindering the transport pathway of cholesterol from the small intestine to the liver, reducing the cholesterol storage in the liver, and accelerating the clearance of cholesterol.

在一些实施方案中,NPC1L1抑制剂选自依折麦布(Ezetimibe)和海博麦布(Hybutimibe)。在一些实施方案中,第二治疗剂为依折麦布。在一些实施方案中,第二治疗剂为海博麦布。In some embodiments, the NPC1L1 inhibitor is selected from Ezetimibe and Hybutimibe. In some embodiments, the second therapeutic agent is Ezetimibe. In some embodiments, the second therapeutic agent is Hybutimibe.

在一些实施方案中,所述第二治疗剂在所述Cholesin蛋白、所述核酸或所述表达载体、或所述增强受试者的内源性Cholesin蛋白的表达或活性的试剂之前、之后或同时施用。在一些实施方案中,所述第二治疗剂在所述Cholesin蛋白、所述核酸或所述表达载体、或所述增强受试者的内源性Cholesin蛋白的表达或活性的试剂之前施用。在一些实施方案中,所述第二治疗剂在所述Cholesin蛋白、所述核酸或所述表达载体、或所述增强受试者的内源性Cholesin蛋白的表达或活性的试剂之后施用。在一些实施方案中,所述第二治疗剂与所述Cholesin蛋白、所述核酸或所述表达载体、或所述增强受试者的内源性Cholesin蛋白的表达或活性的试剂同时施用。In some embodiments, the second therapeutic agent is administered before, after, or simultaneously with the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered before the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered after the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein. In some embodiments, the second therapeutic agent is administered simultaneously with the Cholesin protein, the nucleic acid, or the expression vector, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein.

应认识到,治疗可能需要单次施用治疗有效剂量或多次施用治疗有效剂量的本发明的活性剂。例如,根据活性剂的类型、半衰期和清除率,活性剂可以每天施用一至三次、每两天施用一次、每3至4天施用一次、每周施用一次,或每两周施用一次,或在一个月内施用一次。It should be recognized that treatment may require a single administration of a therapeutically effective dose or multiple administrations of a therapeutically effective dose of an active agent of the invention. For example, depending on the type, half-life, and clearance rate of the active agent, the active agent may be administered one to three times a day, once every two days, once every 3 to 4 days, once a week, or once every two weeks, or once in a month.

本文公开的活性剂(Cholesin蛋白、编码Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂)可适用于多种途径施用。通常,通过胃肠外完成施用。胃肠外递送方法包括局部、静脉内、动脉内、肌内、皮下、髓内、鞘内、心室内、腹膜内、子宫内、阴道内、舌下或鼻内施用。The active agents disclosed herein (Cholesin protein, nucleic acid encoding Cholesin protein, or expression vector comprising the nucleic acid, or agent that enhances the expression or activity of endogenous Cholesin protein of a subject) can be applied to administration by various routes. Typically, administration is completed parenterally. Parenteral delivery methods include topical, intravenous, intraarterial, intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intraperitoneal, intrauterine, intravaginal, sublingual or intranasal administration.

在一些实施方案中,所述受试者为哺乳动物。在优选的实施方案中,所述受试者为人。在一些实施方案中,所述受试者对胆固醇吸收抑制剂、HMGCR抑制剂和PCSK9抑制 剂中的一种或多种无应答或不耐受,例如所述受试者具有LDL受体(LDLR)缺陷。In some embodiments, the subject is a mammal. In a preferred embodiment, the subject is a human. In some embodiments, the subject is resistant to cholesterol absorption inhibitors, HMGCR inhibitors, and PCSK9 inhibitors. The subject may be unresponsive or intolerant to one or more of the agents, for example, the subject has an LDL receptor (LDLR) defect.

本文公开的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂的有效量可以由本领域技术人员采用已知技术来确定。合适的剂量提供足够量的本发明活性剂,并且优选是治疗有效的,即足以在合理的时间范围内引起受试者或动物的例如治疗性或预防性响应。例如,本发明的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂的剂量应在从施用时起约20分钟、30分钟、1小时、2小时或更久,例如12小时至24小时或更久时间(例如,1个月、2个月、3个月、6个月、12个月、24个月等)的时段内足以产生治疗性或预防性响应。在某些实施方案中,时间段甚至可以更久。如本领域已知,针对治疗目的,给药途径、时间和频率,给药制剂的时间和频率,年龄,体重,一般健康状况,性别,饮食,疾病状态的严重程度,药物组合,反应敏感性和对治疗的耐受性/响应的调整可能是必要的。The effective amount of the Cholesin protein disclosed herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the endogenous Cholesin protein of the subject can be determined by those skilled in the art using known techniques. The appropriate dosage provides a sufficient amount of the active agent of the present invention, and is preferably therapeutically effective, that is, sufficient to cause, for example, a therapeutic or preventive response in the subject or animal within a reasonable time frame. For example, the dosage of the Cholesin protein of the present invention, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the endogenous Cholesin protein of the subject should be sufficient to produce a therapeutic or preventive response within a period of about 20 minutes, 30 minutes, 1 hour, 2 hours or longer, such as 12 hours to 24 hours or longer (e.g., 1 month, 2 months, 3 months, 6 months, 12 months, 24 months, etc.) from the time of administration. In certain embodiments, the time period can be even longer. As is known in the art, adjustments for the route, timing and frequency of administration, timing and frequency of administration of formulations, age, weight, general health, sex, diet, severity of the disease state, drug combinations, reaction sensitivities and tolerance/response to treatment may be necessary for therapeutic purposes.

用于确定施用剂量的许多测定为本领域已知的。通常,主治医师决定施用于每个个体患者的本公开活性剂的剂量,这考虑到多种因素,如年龄、体重、一般健康、饮食、性别、待施用的活性剂、施用途径以及治疗病况的严重程度。Many assays for determining the dosage to be administered are known in the art. Typically, the attending physician determines the dosage of the disclosed active agent to be administered to each individual patient, taking into account a variety of factors such as age, weight, general health, diet, sex, the active agent to be administered, the route of administration, and the severity of the condition being treated.

在另一方面,本发明提供了组合物,其包含Cholesin蛋白、编码所述Cholesin蛋白的核酸、包含所述核酸的表达载体、或增强人内源性Cholesin蛋白的表达或活性的试剂,和药学上可接受的载剂或赋形剂。In another aspect, the present invention provides a composition comprising a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of human endogenous Cholesin protein, and a pharmaceutically acceptable carrier or excipient.

在一些实施方案中,所述组合物用于以下中的一种或多种:In some embodiments, the composition is used for one or more of the following:

1)降低受试者中的血浆胆固醇水平;1) reducing plasma cholesterol levels in a subject;

2)降低受试者中的血浆总胆固醇或血浆LDL-胆固醇水平;2) reducing plasma total cholesterol or plasma LDL-cholesterol levels in a subject;

3)降低受试者中的血浆甘油三酯水平;3) reducing plasma triglyceride levels in a subject;

4)抑制受试者中的肝脏胆固醇合成;4) inhibiting hepatic cholesterol synthesis in a subject;

5)抑制受试者中的肝脏VLDL-胆固醇分泌;和5) inhibiting hepatic VLDL-cholesterol secretion in a subject; and

6)治疗或预防受试者中与血浆胆固醇水平升高相关的疾病。6) Treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.

在本发明的组合物的实施方案中,Cholesin蛋白是分离或纯化的。在一些实施方案中,Cholesin蛋白是哺乳动物Cholesin蛋白。哺乳动物Cholesin蛋白的实例包括但限于人、非人灵长类动物、狗、猫、豚鼠、兔、大鼠、小鼠、马、牛的Cholesin蛋白。在一些实施方案中,Cholesin蛋白是非人灵长类动物Cholesin蛋白。在一些实施方案中,Cholesin蛋白是人Cholesin蛋白。在一些实施方案中,人Cholesin蛋白包含如SEQ ID NO:1所示的氨基酸序列。 In embodiments of the compositions of the present invention, the Cholesin protein is isolated or purified. In some embodiments, the Cholesin protein is a mammalian Cholesin protein. Examples of mammalian Cholesin proteins include, but are not limited to, Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cattle. In some embodiments, the Cholesin protein is a non-human primate Cholesin protein. In some embodiments, the Cholesin protein is a human Cholesin protein. In some embodiments, the human Cholesin protein comprises the amino acid sequence shown in SEQ ID NO: 1.

在其他实施方案中,Cholesin蛋白是小鼠Cholesin蛋白。在一些实施方案中,小鼠Cholesin蛋白包含如SEQ ID NO:2所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是猕猴Cholesin蛋白。在一些实施方案中,猕猴Cholesin蛋白包含如SEQ ID NO:3所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是家牛Cholesin蛋白。在一些实施方案中,家牛Cholesin蛋白包含如SEQ ID NO:4所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是褐家鼠Cholesin蛋白。在一些实施方案中,褐家鼠Cholesin蛋白包含如SEQ ID NO:5所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是原鸡Cholesin蛋白。在一些实施方案中,原鸡Cholesin蛋白包含如SEQ ID NO:6所示的氨基酸序列。In other embodiments, the Cholesin protein is a mouse Cholesin protein. In some embodiments, the mouse Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2. In other embodiments, the Cholesin protein is a macaque Cholesin protein. In some embodiments, the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3. In other embodiments, the Cholesin protein is a bovine Cholesin protein. In some embodiments, the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4. In other embodiments, the Cholesin protein is a Rattus norvegicus Cholesin protein. In some embodiments, the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5. In other embodiments, the Cholesin protein is a Gallus gallus Cholesin protein. In some embodiments, the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.

在一些实施方案中,所述组合物还包含第二治疗剂。在优选的实施方案中,所述第二治疗剂选自蛋白质或肽、核酸和小分子药物。In some embodiments, the composition further comprises a second therapeutic agent. In a preferred embodiment, the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.

在一些实施方案中,第二治疗剂为降胆固醇药物。在一些实施方案中,降胆固醇药物选自:HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。In some embodiments, the second therapeutic agent is a cholesterol-lowering drug. In some embodiments, the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.

在一些实施方案中,HMGCR抑制剂为他汀类药物。在一些实施方案中,所述他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀。在优选的实施方案中,第二治疗剂为瑞舒伐他汀。In some embodiments, the HMGCR inhibitor is a statin. In some embodiments, the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin. In a preferred embodiment, the second therapeutic agent is rosuvastatin.

在一些实施方案中,PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。In some embodiments, the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.

在一些实施方案中,胆固醇吸收抑制剂为NPC1L1抑制剂,优选依折麦布或海博麦布。在优选的实施方案中,第二治疗剂为依折麦布。In some embodiments, the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe. In a preferred embodiment, the second therapeutic agent is ezetimibe.

根据所采用的活性剂(如Cholesin蛋白),可将本公开的组合物制备成各种形式,如固态、液态、气态或冻干形式,例如可以是软膏剂、乳膏剂、透皮贴剂、凝胶剂、粉剂、片剂、溶液剂、气雾剂、颗粒剂、丸剂、混悬剂、乳剂、胶囊剂、糖浆剂、液体剂、酏剂、浸膏剂、酊剂或流浸膏提取物的形式,或者是特别适用于所需施用方法的形式。本发明已知的用于生产药物的过程在第22版的Remington’s Pharmaceutical Sciences(Ed.MaackPublishing Co,Easton,Pa.,2012)中显示,并可包括例如常规混合、溶解、制粒、制糖衣、研磨、乳化、包封、包埋或冻干过程。包含例如本文所述的Cholesin蛋白、编码所述Cholesin蛋白、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂的组合物通常以液体形式提供,并且优选包含药学上可接受的缓冲剂。液体制剂可以是溶液或悬浮液。Depending on the active agent used (such as Cholesin protein), the compositions of the present disclosure can be prepared in various forms, such as solid, liquid, gaseous or lyophilized forms, for example, in the form of ointments, creams, transdermal patches, gels, powders, tablets, solutions, aerosols, granules, pills, suspensions, emulsions, capsules, syrups, liquids, elixirs, extracts, tinctures or fluid extracts, or in a form particularly suitable for the desired method of administration. The processes known in the present invention for producing drugs are shown in the 22nd edition of Remington’s Pharmaceutical Sciences (Ed. Maack Publishing Co, Easton, Pa., 2012) and may include, for example, conventional mixing, dissolving, granulating, sugar coating, grinding, emulsifying, encapsulating, embedding or lyophilizing processes. Compositions comprising, for example, the Cholesin protein described herein, an expression vector encoding the Cholesin protein, or comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein of a subject are generally provided in liquid form, and preferably contain a pharmaceutically acceptable buffer. The liquid preparation may be a solution or a suspension.

在一些实施方案中,本发明的组合物被配制为注射用制剂或口服制剂。口服制剂的实例包括但不限于片剂、胶囊剂、颗粒剂、混悬剂。在一些实施方案中,本发明的组合物 可用于多种途径施用,例如肠胃外施用。在一些实施方案中,本发明的组合物可以通过静脉内、肌内、皮下、腹膜内等途径施用。在一些实施方案中,本发明的组合物可以通过口服施用。In some embodiments, the composition of the present invention is formulated as an injection preparation or an oral preparation. Examples of oral preparations include, but are not limited to, tablets, capsules, granules, suspensions. In some embodiments, the composition of the present invention It can be used for various routes of administration, such as parenteral administration. In some embodiments, the composition of the present invention can be administered intravenously, intramuscularly, subcutaneously, intraperitoneally, etc. In some embodiments, the composition of the present invention can be administered orally.

在本发明的组合物中,载剂和赋形剂的实例包括但不限于填充剂、粘合剂、崩解剂、包衣剂、吸附剂、抗粘附剂、助流剂、防腐剂、抗氧化剂、调味剂、着色剂、甜味剂、溶剂、共溶剂、缓冲剂、螯合剂、粘度赋予剂、表面活性剂、稀释剂、润湿剂、稀释剂、防腐剂、乳化剂、稳定剂和张力调节剂。本领域技术人员已知选择合适的赋形剂以制备本发明的组合物。用于本发明的组合物中的示例性载剂包括盐水、缓冲盐水、葡萄糖和水。通常,合适的赋形剂的选择尤其取决于所使用的活性剂、待治疗的疾病和组合物的期望剂型。In the compositions of the present invention, examples of carriers and excipients include, but are not limited to, fillers, adhesives, disintegrants, coating agents, adsorbents, antiadhesives, glidants, preservatives, antioxidants, flavoring agents, coloring agents, sweeteners, solvents, cosolvents, buffers, chelating agents, viscosity imparting agents, surfactants, diluents, wetting agents, diluents, preservatives, emulsifiers, stabilizers and tension regulators. It is known to those skilled in the art that suitable excipients are selected to prepare the compositions of the present invention. Exemplary carriers used in the compositions of the present invention include saline, buffered saline, glucose and water. Typically, the selection of suitable excipients depends especially on the desired dosage form of the active agent used, the disease to be treated and the compositions.

在另一方面,本发明提供了本文所述的组合物在制备用于以下中的一种或多种的药物中的用途:In another aspect, the present invention provides the use of a composition as described herein in the preparation of a medicament for one or more of:

1)降低受试者中的血浆胆固醇水平;1) reducing plasma cholesterol levels in a subject;

2)降低受试者中的血浆总胆固醇或血浆LDL-胆固醇水平;2) reducing plasma total cholesterol or plasma LDL-cholesterol levels in a subject;

3)降低受试者中的血浆甘油三酯水平;3) reducing plasma triglyceride levels in a subject;

4)抑制受试者中的肝脏胆固醇合成;4) inhibiting hepatic cholesterol synthesis in a subject;

5)抑制受试者中的肝脏VLDL-胆固醇分泌;和5) inhibiting hepatic VLDL-cholesterol secretion in a subject; and

6)治疗或预防受试者中与血浆胆固醇水平升高相关的疾病。6) Treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.

在又一方面,本发明提供了用于治疗受试者中与血浆胆固醇水平升高相关的疾病的方法,其包括向所述受试者施用有效量的本发明的组合物的步骤。In yet another aspect, the present invention provides a method for treating a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of the composition of the present invention.

在另一方面,本发明提供了一种诊断受试者的与血浆胆固醇水平升高相关的疾病的方法,其包括以下步骤:In another aspect, the present invention provides a method for diagnosing a disease associated with elevated plasma cholesterol levels in a subject, comprising the steps of:

a.从所述受试者获得血液样品,a. obtaining a blood sample from the subject,

b.确定所述样品中Cholesin蛋白的水平,b. determining the level of Cholesin protein in the sample,

其中与健康对照相比,所述样品中Cholesin蛋白水平的升高指示所述受试者具有患与血浆胆固醇水平升高相关的疾病的风险。Wherein an increased level of Cholesin protein in the sample compared to a healthy control indicates that the subject is at risk for a disease associated with increased plasma cholesterol levels.

在本发明的诊断方法的实施方案中,所述与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。In an embodiment of the diagnostic method of the present invention, the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为高胆固醇血症。在一 些实施方案中,所述与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。In some embodiments, the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In some embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.

在一些实施方案中,所述血液样品选自血液、血清和血浆。在某些实施方案中,所述方法在体外进行。在一些实施方案中,所述方法还包括从样品中分离Cholesin蛋白的步骤。分离Cholesin蛋白的方法是本领域已知的。In some embodiments, the blood sample is selected from blood, serum and plasma. In certain embodiments, the method is performed in vitro. In some embodiments, the method further comprises the step of isolating Cholesin protein from the sample. Methods for isolating Cholesin protein are known in the art.

确定样品中Cholesin蛋白的水平的方法是本领域已知的,例如可以在步骤b中,通过选自以下的方法来确定所述样品中Cholesin蛋白的水平:质谱方法、荧光检测方法、化学发光方法、ELISA测定、Western印迹、放射免疫分析、免疫组化染色、多重免疫试验和斑点印迹试验。Methods for determining the level of Cholesin protein in a sample are known in the art. For example, in step b, the level of Cholesin protein in the sample can be determined by a method selected from the following methods: mass spectrometry, fluorescence detection, chemiluminescence, ELISA, Western blotting, radioimmunoassay, immunohistochemical staining, multiple immunoassay and dot blot assay.

在一些实施方案中,可以通过抗Cholesin蛋白抗体来确定所述样品中Cholesin蛋白的水平。所述抗体优选是单克隆抗体,但也可以是Fab、Fab’、(Fab’)2、Fv、scFv、双scFv、双特异性抗体、三特异性抗体、四特异性抗体等。In some embodiments, the level of Cholesin protein in the sample can be determined by anti-Cholesin protein antibodies. The antibody is preferably a monoclonal antibody, but can also be Fab, Fab', (Fab') 2 , Fv, scFv, bi-scFv, bispecific antibody, trispecific antibody, tetraspecific antibody, etc.

在又一方面,本发明提供了用于确定来自受试者的血液样品中Cholesin蛋白的水平的试剂在制备用于在所述受试者中诊断与血浆胆固醇水平升高相关的疾病的试剂盒中的用途。In yet another aspect, the present invention provides use of a reagent for determining the level of Cholesin protein in a blood sample from a subject in the preparation of a kit for diagnosing a disease associated with elevated plasma cholesterol levels in the subject.

在另一方面,本发明提供了用于在受试者中诊断与血浆胆固醇水平升高相关的疾病的试剂盒,其包含用于确定来自所述受试者的血液样品中Cholesin蛋白的水平的试剂。In another aspect, the present invention provides a kit for diagnosing a disease associated with elevated plasma cholesterol levels in a subject, comprising reagents for determining the level of Cholesin protein in a blood sample from the subject.

在本发明的诊断用途和试剂盒的实施方案中,所述试剂用于在选自以下的方法中确定所述样品中Cholesin蛋白的水平:质谱方法、荧光检测方法、化学发光方法、ELISA测定、Western印迹、放射免疫分析、免疫组化染色、多重免疫试验和斑点印迹试验。在一些实施方案中,所述试剂为抗Cholesin蛋白抗体。所述抗体优选是单克隆抗体,但也可以是Fab、Fab’、(Fab’)2、Fv、scFv、双scFv、双特异性抗体、三特异性抗体、四特异性抗体等。In the embodiments of the diagnostic use and kit of the present invention, the reagent is used to determine the level of Cholesin protein in the sample in a method selected from the following: mass spectrometry, fluorescence detection, chemiluminescence, ELISA, Western blot, radioimmunoassay, immunohistochemical staining, multiple immunoassay and dot blot assay. In some embodiments, the reagent is an anti-Cholesin protein antibody. The antibody is preferably a monoclonal antibody, but can also be Fab, Fab', (Fab') 2 , Fv, scFv, bi-scFv, bispecific antibody, trispecific antibody, tetraspecific antibody, etc.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为高胆固醇血症。在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。In some embodiments, the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones. In some embodiments, the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In some embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.

在一些实施方案中,所述血液样品选自血液、血清和血浆。In some embodiments, the blood sample is selected from the group consisting of blood, serum, and plasma.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂在制备用于 降低受试者中的血浆胆固醇和/或甘油三酯水平的组合物中的用途。在一些实施方案中,血浆胆固醇为血浆总胆固醇或血浆LDL-胆固醇。In another aspect, the present disclosure provides a method for preparing a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject. Use in a composition for reducing plasma cholesterol and/or triglyceride levels in a subject. In some embodiments, the plasma cholesterol is plasma total cholesterol or plasma LDL-cholesterol.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂在制备用于抑制受试者中的肝脏胆固醇合成的组合物中的用途。In yet another aspect, the present disclosure provides use of a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject in the preparation of a composition for inhibiting liver cholesterol synthesis in a subject.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂在制备用于抑制受试者中的肝脏VLDL-胆固醇分泌的组合物中的用途。In yet another aspect, the present disclosure provides use of a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject in the preparation of a composition for inhibiting hepatic VLDL-cholesterol secretion in a subject.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂在制备用于治疗或预防受试者中与血浆胆固醇水平升高相关的疾病的组合物中的用途。In yet another aspect, the present disclosure provides use of a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject in the preparation of a composition for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂,其用于降低受试者中的血浆胆固醇和/或甘油三酯水平。在一些实施方案中,血浆胆固醇为血浆总胆固醇或血浆LDL-胆固醇。In another aspect, the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject, for use in reducing plasma cholesterol and/or triglyceride levels in a subject. In some embodiments, plasma cholesterol is plasma total cholesterol or plasma LDL-cholesterol.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂,其用于抑制受试者中的肝脏胆固醇合成。In yet another aspect, the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent that enhances the expression or activity of an endogenous Cholesin protein in a subject, for use in inhibiting liver cholesterol synthesis in a subject.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂,其用于抑制受试者中的肝脏VLDL-胆固醇分泌。In yet another aspect, the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject, for use in inhibiting hepatic VLDL-cholesterol secretion in a subject.

在又一方面,本公开提供了Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂,其用于治疗或预防受试者中与血浆胆固醇水平升高相关的疾病。In another aspect, the present disclosure provides a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of an endogenous Cholesin protein in a subject, for use in treating or preventing a disease associated with elevated plasma cholesterol levels in a subject.

在本发明的用途的实施方案中,Cholesin蛋白是分离或纯化的。在一些实施方案中,Cholesin蛋白是哺乳动物Cholesin蛋白。哺乳动物Cholesin蛋白的实例包括但限于人、非人灵长类动物、狗、猫、豚鼠、兔、大鼠、小鼠、马、牛的Cholesin蛋白。在一些实施方案中,Cholesin蛋白是非人灵长类动物Cholesin蛋白。在一些实施方案中,Cholesin蛋白是人Cholesin蛋白。在一些实施方案中,人Cholesin蛋白包含如SEQ ID NO:1所示的氨基酸序列。In embodiments of the use of the present invention, the Cholesin protein is isolated or purified. In some embodiments, the Cholesin protein is a mammalian Cholesin protein. Examples of mammalian Cholesin proteins include but are not limited to Cholesin proteins of humans, non-human primates, dogs, cats, guinea pigs, rabbits, rats, mice, horses, and cows. In some embodiments, the Cholesin protein is a non-human primate Cholesin protein. In some embodiments, the Cholesin protein is a human Cholesin protein. In some embodiments, the human Cholesin protein comprises an amino acid sequence as shown in SEQ ID NO:1.

在其他实施方案中,Cholesin蛋白是小鼠Cholesin蛋白。在一些实施方案中,小鼠 Cholesin蛋白包含如SEQ ID NO:2所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是猕猴Cholesin蛋白。在一些实施方案中,猕猴Cholesin蛋白包含如SEQ ID NO:3所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是家牛Cholesin蛋白。在一些实施方案中,家牛Cholesin蛋白包含如SEQ ID NO:4所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是褐家鼠Cholesin蛋白。在一些实施方案中,褐家鼠Cholesin蛋白包含如SEQ ID NO:5所示的氨基酸序列。在其他实施方案中,Cholesin蛋白是原鸡Cholesin蛋白。在一些实施方案中,原鸡Cholesin蛋白包含如SEQ ID NO:6所示的氨基酸序列。In other embodiments, the Cholesin protein is a mouse Cholesin protein. The Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:2. In other embodiments, the Cholesin protein is a macaque Cholesin protein. In some embodiments, the macaque Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:3. In other embodiments, the Cholesin protein is a bovine Cholesin protein. In some embodiments, the bovine Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:4. In other embodiments, the Cholesin protein is a Rattus norvegicus Cholesin protein. In some embodiments, the Rattus norvegicus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:5. In other embodiments, the Cholesin protein is a Gallus gallus Cholesin protein. In some embodiments, the Gallus gallus Cholesin protein comprises the amino acid sequence shown in SEQ ID NO:6.

在一些实施方案中,所述核酸是DNA或RNA。在一些实施方案中,所述核酸是DNA。In some embodiments, the nucleic acid is DNA or RNA. In some embodiments, the nucleic acid is DNA.

在一些实施方案中,所述表达载体选自慢病毒载体、腺病毒载体、腺相关病毒(AAV)载体、逆转录病毒载体、质粒、DNA载体、mRNA载体、基于转座子的载体和人工染色体。In some embodiments, the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector, and an artificial chromosome.

在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为高胆固醇血症。在一些实施方案中,所述与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。In some embodiments, the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones. In some embodiments, the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. In some embodiments, the disease associated with elevated plasma cholesterol levels is atherosclerosis.

在一些实施方案中,本文所述的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体、或增强受试者的内源性Cholesin蛋白的表达或活性的试剂与第二治疗剂组合。在优选的实施方案中,第二治疗剂选自蛋白质或肽、核酸和小分子药物。In some embodiments, the Cholesin protein described herein, the nucleic acid encoding the Cholesin protein, or the expression vector comprising the nucleic acid, or the agent that enhances the expression or activity of the subject's endogenous Cholesin protein is combined with a second therapeutic agent. In a preferred embodiment, the second therapeutic agent is selected from a protein or peptide, a nucleic acid, and a small molecule drug.

在一些实施方案中,第二治疗剂为降胆固醇药物。在一些实施方案中,降胆固醇药物选自:HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。In some embodiments, the second therapeutic agent is a cholesterol-lowering drug. In some embodiments, the cholesterol-lowering drug is selected from: HMGCR inhibitors, PCSK9 inhibitors, and cholesterol absorption inhibitors.

在一些实施方案中,HMGCR抑制剂为他汀类药物。在一些实施方案中,所述他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀。在优选的实施方案中,第二治疗剂为瑞舒伐他汀。In some embodiments, the HMGCR inhibitor is a statin. In some embodiments, the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin. In a preferred embodiment, the second therapeutic agent is rosuvastatin.

在一些实施方案中,PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。In some embodiments, the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran, and Tafolecimab.

在一些实施方案中,胆固醇吸收抑制剂为NPC1L1抑制剂,优选依折麦布或海博麦布。在优选的实施方案中,第二治疗剂为依折麦布。In some embodiments, the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe. In a preferred embodiment, the second therapeutic agent is ezetimibe.

在一些实施方案中,所述受试者为哺乳动物。在优选的实施方案中,所述受试者为 人。在一些实施方案中,所述受试者对胆固醇吸收抑制剂、HMGCR抑制剂和PCSK9抑制剂中的一种或多种无应答或不耐受,例如所述受试者具有LDL受体(LDLR)缺陷。In some embodiments, the subject is a mammal. In a preferred embodiment, the subject is In some embodiments, the subject is unresponsive or intolerant to one or more of a cholesterol absorption inhibitor, an HMGCR inhibitor, and a PCSK9 inhibitor, for example, the subject has an LDL receptor (LDLR) defect.

实施例Example

通过下面的具体实施例进一步阐述本发明。应当理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。以下实施例中未注明具体条件的实验方法,通过按照本领域的常规条件,或按照制造商所建议的条件。除非另有说明,否则以下实施例中所用的实验材料和试剂均可商购获得。The present invention is further described by the following specific examples. It should be understood that these examples are only used to illustrate the present invention and are not intended to limit the scope of the present invention. The experimental methods in the following examples without specifying specific conditions are carried out according to conventional conditions in the art or according to the conditions recommended by the manufacturer. Unless otherwise stated, the experimental materials and reagents used in the following examples are all commercially available.

实施例1:Cholesin是一种胆固醇诱导的肠分泌激素Example 1: Cholesin is a cholesterol-induced intestinal secretory hormone

1.1 Cholesin的发现和鉴定1.1 Discovery and identification of Cholesin

本实施例选用遗传背景为C57BL/6J的10-12周龄野生型雄性小鼠,为了寻找具有调控胆固醇稳态作用的激素,分析了禁食过夜后喂食1小时普通饮食(RD,含0.02%胆固醇)和西方饮食(WD,含1.25%胆固醇)的小鼠血清蛋白。将小鼠随机分成2组,每组6只。其中,第一组小鼠饥饿过夜后喂食普通饮食(普通饮食组(RD组));第二组小鼠饥饿过夜后喂食西方饮食(西方饮食组(WD组))。用肝素锂处理后的抗凝管分别收集两组小鼠的血液,在4℃、4000rpm下离心15分钟,上清即为血浆,将其转移到新的离心管中。血浆用柱式白蛋白/免疫球蛋白清除试剂盒(生工生物)处理,除去其中的IgG和白蛋白。随后再用ProteoMiner Protein Enrichment Kit(BIO-RAD)稀释高丰度蛋白并浓缩低丰度蛋白。处理后的血浆与5×SDS上样缓冲液(250mM Tris-HCl,PH6.8,10%SDS,0.5%溴酚蓝,50%甘油,5%β-ME)混匀,在100℃下煮样10分钟,进行SDS-PAGE蛋白电泳。电泳完成后进行银染。结果显示,与普通饮食组相比,西方饮食组在23kDa左右观察到一条增强条带(图1)。In this example, 10-12 week old wild-type male mice with a genetic background of C57BL/6J were selected. In order to find hormones that regulate cholesterol homeostasis, serum proteins of mice fed with a normal diet (RD, containing 0.02% cholesterol) and a Western diet (WD, containing 1.25% cholesterol) for 1 hour after fasting overnight were analyzed. The mice were randomly divided into 2 groups, 6 in each group. Among them, the first group of mice were fed a normal diet after starvation overnight (normal diet group (RD group)); the second group of mice were fed a Western diet after starvation overnight (Western diet group (WD group)). The blood of the two groups of mice was collected in anticoagulant tubes treated with lithium heparin, and centrifuged at 4°C and 4000rpm for 15 minutes. The supernatant was plasma, which was transferred to a new centrifuge tube. The plasma was treated with a column albumin/immunoglobulin removal kit (Bio-Rad) to remove IgG and albumin. Subsequently, the ProteoMiner Protein Enrichment Kit (BIO-RAD) was used to dilute high-abundance proteins and concentrate low-abundance proteins. The treated plasma was mixed with 5×SDS loading buffer (250mM Tris-HCl, pH 6.8, 10% SDS, 0.5% bromophenol blue, 50% glycerol, 5% β-ME), boiled at 100°C for 10 minutes, and subjected to SDS-PAGE protein electrophoresis. Silver staining was performed after electrophoresis. The results showed that compared with the normal diet group, an enhanced band was observed at around 23kDa in the Western diet group (Figure 1).

通过对差异条带切胶进行质谱鉴定,确定差异条带是来自3110082I17Rik(C7ORF50的小鼠同源基因)基因编码的蛋白质,该蛋白在以前的文献中未报道过。基于后续工作发现其对胆固醇合成的抑制作用,发明人将其命名为Cholesin。By mass spectrometry identification of differential bands, it was determined that the differential bands were proteins encoded by 3110082I17Rik (mouse homologous gene of C7ORF50), which had not been reported in previous literature. Based on subsequent work, the inventors found that it had an inhibitory effect on cholesterol synthesis and named it Cholesin.

1.2 Cholesin是分泌蛋白1.2 Cholesin is a secreted protein

为了探究Cholesin在细胞中的定位,进行了免疫荧光实验。具体方法如下:1)在孔板中铺上适量直径12mm的细胞爬片,并将HEK293T细胞按1:6的比例铺在孔板中,细胞贴壁后过表达c端带有Flag标签的人源Cholesin(hCholesin)、鼠源Cholesin(mCholesin)和对照空质粒;2)转染36小时后将吸弃培养基,用37℃预热的PBS洗涤细胞一次,随后用平衡至室温的4%多聚甲醛/PBS溶液室温固定细胞20分钟;3)固 定结束后吸弃4%多聚甲醛/PBS溶液,用PBS清洗一次,随后用含0.2%Triton X-100的PBS溶液室温处理细胞5-7分钟,对细胞膜进行穿透;4)立即吸弃破膜液,用PBS清洗细胞一次,用5%BSA/TBST溶液室温封闭细胞30分钟;5)用5%BSA/TBST溶液稀释一抗,封闭完成后室温孵育一抗1小时;6)回收一抗,用TBST洗涤爬片3次,每次10分钟;7)用5%BSA/TBST溶液稀释荧光二抗和DAPI(终浓度0.5mg/ml),室温避光染色1小时;8)弃二抗,用TBST洗涤爬片3次,每次10分钟;9)封片,用激光共聚焦显微镜拍摄Cholesin定位结果。免疫荧光结果显示Cholesin主要定位在细胞质中(图2A)。In order to explore the localization of Cholesin in cells, an immunofluorescence experiment was performed. The specific method is as follows: 1) Place an appropriate amount of cell slides with a diameter of 12 mm in the well plate, and place HEK293T cells in the well plate at a ratio of 1:6. After the cells adhere to the well plate, overexpress human Cholesin (hCholesin) with a Flag tag at the C-terminus, mouse Cholesin (mCholesin) and a control empty plasmid; 2) After 36 hours of transfection, the culture medium was aspirated and the cells were washed once with PBS preheated at 37°C, and then fixed with a 4% paraformaldehyde/PBS solution equilibrated to room temperature for 20 minutes at room temperature; 3) Fix After the fixation, the 4% paraformaldehyde/PBS solution was aspirated and discarded, and the cells were washed once with PBS. Subsequently, the cells were treated with PBS solution containing 0.2% Triton X-100 at room temperature for 5-7 minutes to permeate the cell membrane; 4) The permeation solution was immediately aspirated and discarded, the cells were washed once with PBS, and the cells were blocked with 5% BSA/TBST solution for 30 minutes at room temperature; 5) The primary antibody was diluted with 5% BSA/TBST solution, and the primary antibody was incubated at room temperature for 1 hour after the blocking was completed; 6) The primary antibody was recovered, and the slides were washed with TBST for 3 times, each for 10 minutes; 7) The fluorescent secondary antibody and DAPI (final concentration 0.5 mg/ml) were diluted with 5% BSA/TBST solution, and stained at room temperature in the dark for 1 hour; 8) The secondary antibody was discarded, and the slides were washed with TBST for 3 times, each for 10 minutes; 9) The slides were sealed, and the results of Cholesin localization were photographed with a laser confocal microscope. The immunofluorescence results showed that Cholesin was mainly located in the cytoplasm (Figure 2A).

随后利用免疫印迹实验探索了Cholesin的分泌情况。在HEK293T细胞中过表达c端带有Flag标签的人源Cholesin、鼠源Cholesin和对照空质粒,转染48小时后更换为无血清培养基继续培养过夜,收集培养基。6孔板每孔的贴壁细胞加入200μl RIPA裂解液(25mM Tris,150mM NaCl,1% NP-40,1%脱氧胆酸钠,0.1% SDS,100×蛋白酶裂解液,pH 7.4),用细胞刮刀从皿底刮取细胞收集到EP管中,随后按照超6秒停6秒,总18秒,功率40%的程序冰上超声处理细胞,4℃最大转速离心15分钟,离心所得上清液加入5×SDS上样缓冲液,100℃煮样10分钟获得总蛋白提取液。通过免疫印迹检测培养基和裂解上清液中的Cholesin。结果显示,在过表达人源或鼠源Cholesin的细胞培养基中均能检测到Cholesin的分泌(图2B),表明Cholesin在体外条件下也具有分泌能力。Subsequently, the secretion of Cholesin was explored by immunoblotting. Human Cholesin, mouse Cholesin and control empty plasmids with a Flag tag at the C-terminus were overexpressed in HEK293T cells. After 48 hours of transfection, the cells were replaced with serum-free medium and cultured overnight, and the culture medium was collected. 200 μl RIPA lysis buffer (25 mM Tris, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, 100× protease lysis buffer, pH 7.4) was added to the adherent cells in each well of the 6-well plate. The cells were scraped from the bottom of the dish with a cell scraper and collected in an EP tube. Then, the cells were sonicated on ice according to the program of 6 seconds on, 6 seconds off, 18 seconds in total, and 40% power. The cells were centrifuged at the maximum speed of 4°C for 15 minutes, and the supernatant obtained by centrifugation was added with 5×SDS loading buffer. The samples were boiled at 100°C for 10 minutes to obtain the total protein extract. Cholesin in the culture medium and lysis supernatant was detected by immunoblotting. The results showed that the secretion of Cholesin could be detected in the culture medium of cells overexpressing human or mouse Cholesin (Figure 2B), indicating that Cholesin also has the ability to be secreted under in vitro conditions.

为了对分泌的Cholesin进行表征,构建了过表达c端带有Flag标签的鼠源Cholesin杆状病毒,用于感染昆虫细胞Hi5。具体方法如下:1)将1L Hi5细胞培养至密度为2-3×106个细胞/ml,按1:50加入杆状病毒,培养48~72小时,离心收取培养基上清,培养上清用0.22μm滤膜过滤,去除细胞碎片;2)使用平衡缓冲液(20mM NaH2PO4,0.5MNaCl,pH 7.4)平衡HisTrap excel(Cytiva)层析柱至少5个柱体积;3)使用泵头对过滤后的培养基进行上柱;4)用清洗缓冲液(20mM NaH2PO4,0.5M NaCl,30mM咪唑,pH7.4)冲洗至少20个柱体积至无蛋白流出;5)用洗脱缓冲液(20mM NaH2PO4,0.5M NaCl,500mM咪唑,pH 7.4)进行梯度洗脱,收集蛋白峰;6)用含有10kDa NMWL滤膜的浓缩管对蛋白峰进行浓缩至总体积小于2ml,用Superdex 200 Increase 10/300 GL分子筛根据蛋白质的大小和形状,对样品进一步分离纯化。对纯化所得蛋白样品进行质谱鉴定,除赖氨酸和精氨酸富集区,Cholesin的大部分序列都可以被鉴定,表明Cholesin是全长分泌的。To characterize secreted Cholesin, a baculovirus overexpressing mouse Cholesin with a C-terminal Flag tag was constructed and used to infect insect Hi5 cells. The specific method is as follows: 1) culture 1L Hi5 cells to a density of 2-3×10 6 cells/ml, add baculovirus at a ratio of 1:50, culture for 48-72 hours, collect the culture supernatant by centrifugation, and filter the culture supernatant with a 0.22 μm filter to remove cell debris; 2) use an equilibration buffer (20 mM NaH 2 PO 4 , 0.5 M NaCl, pH 7.4) to equilibrate a HisTrap excel (Cytiva) chromatography column for at least 5 column volumes; 3) use a pump head to load the filtered culture medium onto the column; 4) rinse with a wash buffer (20 mM NaH 2 PO 4 , 0.5 M NaCl, 30 mM imidazole, pH 7.4) for at least 20 column volumes until no protein flows out; 5) use an elution buffer (20 mM NaH 2 PO 4 , 0.5 M NaCl, 500 mM imidazole, pH 7.4) to wash the column for at least 20 column volumes until no protein flows out; 7.4) Perform gradient elution and collect the protein peak; 6) Use a concentrator tube containing a 10kDa NMWL filter to concentrate the protein peak to a total volume of less than 2ml, and use Superdex 200 Increase 10/300 GL molecular sieve to further separate and purify the sample based on the size and shape of the protein. Mass spectrometry identification of the purified protein sample shows that most of the sequence of Cholesin can be identified, except for the lysine and arginine-rich regions, indicating that Cholesin is secreted in its entirety.

这些结果表明,Cholesin是以全长形式被分泌出来的分泌蛋白。 These results indicate that Cholesin is a secretory protein that is secreted in its full-length form.

1.3 Cholesin是响应进食反应的激素1.3 Cholesin is a hormone that responds to eating

本实施例选用遗传背景为C57BL/6J的10-12周龄野生型雄性小鼠,将小鼠随机分成9组,每组8只。其中,第1组为饥饿过夜的小鼠(禁食组);第2组至第5组小鼠饥饿过夜后,分别喂食普通饮食(RD)1、2、4、12小时(含0.02%胆固醇的普通饮食组);第6组至第9组小鼠饥饿过夜后,分别喂食西方饮食(WD)1、2、4、12小时(含1.25%胆固醇的西方饮食组)。将这9组小鼠处死,然后分别收集血浆,通过酶联免疫吸附测定(ELISA)来检测血浆中的Cholesin水平。In this example, 10-12 week old wild-type male mice with a genetic background of C57BL/6J were randomly divided into 9 groups, 8 mice in each group. Group 1 was starved overnight (fasting group); Groups 2 to 5 were starved overnight and then fed with a normal diet (RD) for 1, 2, 4, and 12 hours (normal diet group containing 0.02% cholesterol); Groups 6 to 9 were starved overnight and then fed with a Western diet (WD) for 1, 2, 4, and 12 hours (Western diet group containing 1.25% cholesterol). The 9 groups of mice were killed, and then plasma was collected and the Cholesin level in plasma was detected by enzyme-linked immunosorbent assay (ELISA).

具体操作如下:在聚苯乙烯微孔板中加入3μl小鼠血浆和100μl包被液(0.05mol/LNa2CO3/NaHCO3,pH8.0),96孔微板中设置Cholesin全身敲除小鼠血浆作为对照组进行校准,用纯化定量过的Cholesin蛋白标品设置标准曲线,标准曲线浓度为0,12.5,25,50,100,200pmol,加样结束后4℃孵育过夜,使蛋白吸附在孔底。4℃摇床包被过夜后,取出微孔板,甩干孔内液体,每孔加1×gelatin 100μl室温封闭微孔板2小时。弃去封闭液,每孔加入200μl TBST溶液洗4次,最后一次将液体尽量甩干。将检测一抗配制于TBST中,每孔加入100μl一抗,4℃摇床孵育过夜。弃去检测一抗,每孔加入200μl TBST溶液洗4次,最后一次将液体尽量甩干。将二抗配制于TBST中,每孔加入100μl,室温摇床孵育1小时。弃去二抗,每孔加入200μl TBST溶液洗4次,最后一次将液体尽量甩干。每孔加入100μl TMB底物,室温摇床孵育10~30分钟。随后每孔加入100μl终止液(0.05mol/L H2SO4),30分钟内用酶标仪读取OD450nm的吸光值,根据标准曲线的吸光值计算得出待测样品中Cholesin的含量。The specific operation is as follows: 3 μl mouse plasma and 100 μl coating solution (0.05 mol/L Na2CO3/NaHCO3, pH 8.0) were added to the polystyrene microplate. The Cholesin whole body knockout mouse plasma was set as the control group for calibration in the 96-well microplate. The standard curve was set with the purified and quantified Cholesin protein standard. The concentration of the standard curve was 0, 12.5, 25, 50, 100, and 200 pmol. After the addition of the sample, it was incubated at 4°C overnight to allow the protein to adsorb to the bottom of the well. After overnight coating on a 4°C shaker, the microplate was removed, the liquid in the well was shaken dry, and 100 μl of 1× gelatin was added to each well to block the microplate for 2 hours at room temperature. The blocking solution was discarded, and 200 μl of TBST solution was added to each well to wash 4 times. The liquid was shaken dry as much as possible for the last time. The primary antibody for detection was prepared in TBST, 100 μl of primary antibody was added to each well, and incubated at 4°C shaker overnight. Discard the primary antibody, add 200μl TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time. Prepare the secondary antibody in TBST, add 100μl to each well, and incubate on a shaker at room temperature for 1 hour. Discard the secondary antibody, add 200μl TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time. Add 100μl TMB substrate to each well and incubate on a shaker at room temperature for 10 to 30 minutes. Then add 100μl stop solution (0.05mol/L H 2 SO 4 ) to each well, read the absorbance value of OD450nm with an enzyme reader within 30 minutes, and calculate the content of Cholesin in the sample to be tested based on the absorbance value of the standard curve.

结果显示,在饥饿后进食的小鼠中,血浆中Cholesin的含量迅速升高,并在进食西方饮食1小时后迅速达到约1200pM的峰值,随后持续下降,在进食12小时后恢复到0小时水平。普通饮食喂饲后血浆中Cholesin变化也呈现出相同的趋势,但与西方饮食组相比变化较弱,进食1小时后Cholesin的峰值仅有约700pM左右(图3A)。这些结果表明进食高胆固醇饮食促进Cholesin分泌,提示胆固醇可能是Cholesin分泌的诱导剂。The results showed that in mice that ate after starvation, the level of Cholesin in plasma increased rapidly and quickly reached a peak of about 1200pM 1 hour after eating a Western diet, then continued to decline and returned to the 0-hour level 12 hours after eating. The changes in Cholesin in plasma after feeding a normal diet also showed the same trend, but the changes were weaker than those in the Western diet group, with the peak of Cholesin only about 700pM 1 hour after eating (Figure 3A). These results indicate that eating a high-cholesterol diet promotes Cholesin secretion, suggesting that cholesterol may be an inducer of Cholesin secretion.

为了观察人群中是否也存在和小鼠同样的分泌变化,对临床数据进行了分析。比较了13对健康人饥饿过夜和进食1小时的血浆Cholesin水平。结果显示,进食后血浆Cholesin的含量显著升高(图3B)。In order to observe whether the same secretion changes as in mice also exist in humans, clinical data were analyzed. The plasma Cholesin levels of 13 pairs of healthy people who were fasted overnight and fed for 1 hour were compared. The results showed that the plasma Cholesin content increased significantly after eating (Figure 3B).

这些结果表明,Cholesin是响应进食反应的激素,胆固醇是可能的诱导剂。These results suggest that cholesin is a hormone that is responsive to feeding and that cholesterol is a possible inducer.

1.4 Cholesin的组织分布1.4 Tissue distribution of Cholesin

本实施例选用遗传背景为C57BL/6J 10-12周龄野生型(WT)和Cholesin全身敲除(KO)雄性小鼠各5只,分别取小鼠的脑、胃、小肠、结肠、肝脏、肾脏、棕色脂肪组 织(BAT)、附睾白色脂肪组织(eWAT)和骨骼肌。配制RIPA裂解液(25mM Tris,150mM NaCl,1% NP-40,1%脱氧胆酸钠,0.1% SDS,pH 7.4),根据裂解液的体积在使用前加入蛋白酶抑制剂和磷酸酶抑制剂,以配制组织蛋白提取液。称取适量组织,加入500μl RIPA裂解液和磁珠,组织研磨机60Hz低温研磨1分钟。研磨完后用4℃预冷的离心机15000rpm离心15分钟,当组织中脂质含量较多时,将第一次离心上清转移到新的离心管中,同样参数再离心15分钟。取上清用BCA法蛋白定量,加入5×SDS上样缓冲液充分混匀后置于100℃金属浴加热10分钟,得到组织的蛋白样品,然后取25μg总蛋白进行SDS-PAGE电泳,以在蛋白水平上测定Cholesin的组织分布。In this example, five wild-type (WT) and Cholesin knockout (KO) male mice of 10-12 weeks old with a genetic background of C57BL/6J were selected, and the brain, stomach, small intestine, colon, liver, kidney, and brown fat tissues of the mice were collected. RIPA lysis buffer (25mM Tris, 150mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS, pH 7.4) was prepared, and protease inhibitors and phosphatase inhibitors were added before use according to the volume of the lysis buffer to prepare tissue protein extract. Weigh an appropriate amount of tissue, add 500μl RIPA lysis buffer and magnetic beads, and grind at 60Hz for 1 minute in a tissue grinder. After grinding, centrifuge at 15000rpm for 15 minutes in a 4℃ precooled centrifuge. When the lipid content in the tissue is high, transfer the supernatant of the first centrifugation to a new centrifuge tube and centrifuge for 15 minutes under the same parameters. The supernatant was taken for protein quantification using the BCA method, 5× SDS loading buffer was added and mixed thoroughly, and then heated in a 100°C metal bath for 10 minutes to obtain a tissue protein sample. Then 25 μg of total protein was taken for SDS-PAGE electrophoresis to determine the tissue distribution of Cholesin at the protein level.

结果如图4所示,内源性Cholesin蛋白大小略小于25kDa,在小肠中表达量最高,结肠和胃次之,其他组织表达量低,具有较强的组织特异性。The results are shown in FIG4 . The size of the endogenous Cholesin protein is slightly less than 25 kDa, and its expression level is highest in the small intestine, followed by the colon and stomach, and its expression level in other tissues is low, showing strong tissue specificity.

1.5 Cholesin是响应胆固醇吸收的肠分泌激素1.5 Cholesin is an intestinal secretory hormone that responds to cholesterol absorption

由于Cholesin在肠道中表达量最高,并且小肠是外源胆固醇吸收的主要器官,因此推测Cholesin可能由小肠分泌用于调控机体胆固醇稳态。为了验证这一猜想,构建了Cholesin肠道特异性敲除(Cholesin IKO)的小鼠作为研究工具。Since Cholesin is expressed most highly in the intestine and the small intestine is the main organ for exogenous cholesterol absorption, it is speculated that Cholesin may be secreted by the small intestine to regulate cholesterol homeostasis. To verify this hypothesis, Cholesin intestinal-specific knockout (Cholesin IKO) mice were constructed as a research tool.

本实施例将WT与IKO小鼠各随机分为2组,每组8只。第一组小鼠饥饿过夜,第二组小鼠饥饿过夜后,喂食1小时含1.25%胆固醇的西方饮食(WD)。然后分别收集小鼠血浆和小肠,对小肠总胆固醇(Total cholesterol,TC)含量以及血浆Cholesin、TC和甘油三酯(Triglyceride,TG)的含量进行测定。结果如图5A和5B所示。In this example, WT and IKO mice were randomly divided into 2 groups, each with 8 mice. The first group of mice were starved overnight, and the second group of mice were starved overnight and then fed with a Western diet (WD) containing 1.25% cholesterol for 1 hour. Then the plasma and small intestine of the mice were collected respectively, and the content of total cholesterol (TC) in the small intestine and the content of cholesin, TC and triglyceride (TG) in plasma were measured. The results are shown in Figures 5A and 5B.

为进一步证实胆固醇在促进Cholesin分泌中的作用,将WT与IKO小鼠各随机分为2组,每组7只,将小鼠饥饿过夜。第一组小鼠灌胃150μl含20%无水乙醇的玉米油(chol-),第二组小鼠灌胃150μl 200mg kg-1体重的胆固醇(chol+,其中胆固醇溶于含20%无水乙醇的玉米油)。小鼠灌胃1小时后收取小鼠血浆和小肠组织。对小肠TC含量以及血浆Cholesin、TC和TG的含量进行测定。结果如图5C和5D所示。To further confirm the role of cholesterol in promoting Cholesin secretion, WT and IKO mice were randomly divided into 2 groups, 7 mice in each group, and the mice were starved overnight. The first group of mice were gavaged with 150 μl corn oil containing 20% anhydrous ethanol (chol-), and the second group of mice were gavaged with 150 μl 200 mg kg -1 body weight of cholesterol (chol+, where cholesterol is dissolved in corn oil containing 20% anhydrous ethanol). Plasma and small intestinal tissues were collected from mice 1 hour after gavage. The content of TC in the small intestine and the content of Cholesin, TC and TG in plasma were measured. The results are shown in Figures 5C and 5D.

图5A的结果显示,WT小鼠血浆中的Cholesin含量在喂食WD后显著增加,而IKO小鼠血浆中的Cholesin含量几乎没有增加,并且在饥饿状态下IKO小鼠血浆中的Cholesin含量显著低于WT小鼠。这些结果表明小肠可能是在西方饮食刺激下分泌Cholesin的主要器官。图5A的结果还显示,喂食WD后小肠中的总胆固醇含量升高,但WT和IKO组在饥饿和进食两种状态下均没有显著差异。该结果暗示WT和IKO组小鼠小肠的胆固醇吸收能力没有差异,二者对西方饮食的响应不是由于小肠吸收能力不同造成的。The results of Figure 5A show that the Cholesin content in the plasma of WT mice increased significantly after feeding WD, while the Cholesin content in the plasma of IKO mice hardly increased, and the Cholesin content in the plasma of IKO mice was significantly lower than that of WT mice under starvation. These results indicate that the small intestine may be the main organ that secretes Cholesin under the stimulation of a Western diet. The results of Figure 5A also show that the total cholesterol content in the small intestine increased after feeding WD, but there was no significant difference between the WT and IKO groups in both the starving and fed states. This result suggests that there is no difference in the cholesterol absorption capacity of the small intestine of WT and IKO group mice, and their response to the Western diet is not due to different absorption capacities of the small intestine.

图5B的结果显示,进食前后小鼠血浆TC无明显变化,但IKO小鼠的血浆TC在进食前后均高于WT组。由于收集血浆的时间点是在进食1小时后,此时外源胆固醇 尚未进入血液循环,因此血浆TC不会随着进食变化;而IKO组的血浆TC高于WT组则暗示Cholesin可能负调控了血浆TC水平。图5B还显示,进食前后小鼠血浆TG含量明显降低,WT与IKO小鼠的血浆TG在饥饿状态下无明显差异,在进食后IKO小鼠的血浆TG略高于WT组,这表明进食会降低血液中循环的总甘油三酯,而WT与IKO组之间血浆TG的差异也暗示Cholesin可能对血浆TG有影响。The results in Figure 5B show that there was no significant change in plasma TC of mice before and after eating, but the plasma TC of IKO mice was higher than that of the WT group before and after eating. It has not yet entered the blood circulation, so plasma TC will not change with eating; the higher plasma TC in the IKO group than in the WT group suggests that Cholesin may negatively regulate the plasma TC level. Figure 5B also shows that the plasma TG content of mice decreased significantly before and after eating, and there was no significant difference in plasma TG between WT and IKO mice under starvation conditions. After eating, the plasma TG of IKO mice was slightly higher than that of the WT group, indicating that eating would reduce the total triglycerides circulating in the blood, and the difference in plasma TG between the WT and IKO groups also suggests that Cholesin may have an effect on plasma TG.

与WD喂食的效果类似,图5C的结果显示,WT小鼠血浆中的Cholesin含量在灌胃胆固醇后显著升高,而IKO小鼠基本不响应胆固醇的刺激,IKO小鼠对胆固醇的吸收也不受影响。图5D的结果显示,灌胃胆固醇后小鼠血浆TC含量无明显变化,但IKO小鼠的血浆TC在对照组和胆固醇处理组中均高于WT组。灌胃胆固醇后小鼠血浆总TG含量升高,WT与IKO小鼠的血浆TG在实验组与对照组均无明显差异,这是由于胆固醇溶解在玉米油中,因此灌胃后血浆中的TG会上升。Similar to the effect of WD feeding, the results of Figure 5C show that the Cholesin content in the plasma of WT mice increased significantly after oral administration of cholesterol, while IKO mice basically did not respond to the stimulation of cholesterol, and the absorption of cholesterol by IKO mice was also unaffected. The results of Figure 5D show that there was no significant change in the plasma TC content of mice after oral administration of cholesterol, but the plasma TC of IKO mice was higher than that of the WT group in both the control group and the cholesterol-treated group. The total TG content of mouse plasma increased after oral administration of cholesterol, and there was no significant difference in the plasma TG of WT and IKO mice in the experimental group and the control group. This is because cholesterol is dissolved in corn oil, so the TG in plasma will increase after oral administration.

这些结果证明Cholesin是响应胆固醇吸收的小肠分泌的激素。These results demonstrate that Cholesin is a hormone secreted by the small intestine in response to cholesterol absorption.

实施例2:NPC1L1介导的胆固醇吸收促进了Cholesin的分泌Example 2: NPC1L1-mediated cholesterol absorption promotes Cholesin secretion

2.1 Cholesin在吸收性肠细胞中表达2.1 Cholesin expression in absorptive enterocytes

胆固醇的吸收目前被认为是肠道中吸收性肠细胞顶面的NPC1L1介导的。由于胆固醇的吸收能促进Cholesin的分泌,因此我们推测Cholesin可能主要在吸收性肠细胞中表达。为了验证以上假设,构建了Cholesin-GFP和Apoa1-mCherry(APOA1是吸收性肠细胞的标记蛋白)双基因敲入小鼠用于免疫荧光实验,以判断表达Cholesin的细胞类型。具体的实验步骤如下:1)收取小鼠小肠组织,用PBS冲洗肠内粪便并立即置于PBS配制的4%的多聚甲醛溶液中4℃固定过夜;2)将固定后的组织转移到含30%蔗糖的PBS溶液中,4℃脱水过夜;3)脱水完成后组织应沉在蔗糖溶液底部,将组织从蔗糖溶液中取出,尽量吸干组织表面液体,在包埋盒中加入OCT并放入组织块,将包埋好的组织放在-80℃冰箱中冷冻6小时;4)切片前把组织块放在-20℃冰箱平衡,用冷冻切片机切厚度为5-8μm的切片,组织切片贴在阳离子防脱片上;5)染色前将切片取出在在室温平衡20分钟,再放置在55℃烘箱烤片10分钟防止组织从玻片上脱落,用组化笔在玻片上圈出组织;6)用TBST清洗组织3次,洗去OCT,随后用0.2%Triton X-100/PBS穿透组织5-7分钟;7)穿透完成后立即吸去0.2%Triton X-100/PBS,用TBST冲洗一次,随后用5%BSA/TBST室温封闭30分钟;8)用5%BSA/TBST稀释一抗,封闭结束后吸弃封闭液,加GFP一抗,每个组织块需要一抗80-100μl,给湿盒加双蒸水,切片在4℃孵育过夜;9)弃一抗,TBST室温洗片3次,每次5分钟;10)用5%BSA/TBST稀释荧光二 抗和DAPI,室温避光孵育1小时;11)弃二抗和DAPI,TBST室温洗片3次,每次5分钟;12)封片,用激光共聚焦显微镜拍摄Cholesin定位结果。Cholesterol absorption is currently believed to be mediated by NPC1L1 on the apical surface of absorptive enterocytes in the intestine. Since cholesterol absorption can promote the secretion of Cholesin, we speculated that Cholesin may be mainly expressed in absorptive enterocytes. To verify the above hypothesis, Cholesin-GFP and Apoa1-mCherry (APOA1 is a marker protein for absorptive enterocytes) double gene knock-in mice were constructed for immunofluorescence experiments to determine the cell type expressing Cholesin. The specific experimental steps are as follows: 1) Collect the mouse small intestine tissue, rinse the intestinal feces with PBS and immediately place it in a 4% paraformaldehyde solution prepared in PBS and fix it overnight at 4°C; 2) Transfer the fixed tissue to a PBS solution containing 30% sucrose and dehydrate it overnight at 4°C; 3) After dehydration, the tissue should sink to the bottom of the sucrose solution. Take the tissue out of the sucrose solution, try to dry the liquid on the surface of the tissue, add OCT to the embedding box and put the tissue block in, and freeze the embedded tissue in a -80°C refrigerator for 6 hours; 4) Before slicing, place the tissue block in a -20°C refrigerator for equilibrium, cut slices with a thickness of 5-8μm with a freezing microtome, and stick the tissue slices on a cationic anti-shedding film; 5) Before staining, take out the slices and equilibrate them at room temperature for 20 minutes, then place them in a 55°C oven for 10 minutes to prevent the tissue from falling off the slide, and circle the tissue on the slide with a tissue pen; 6) Wash the tissue 3 times with TBST to wash off the OCT, and then use 0.2% Triton X-100/PBS penetrates the tissue for 5-7 minutes; 7) After penetration, immediately remove 0.2% Triton X-100/PBS, rinse once with TBST, and then block with 5% BSA/TBST at room temperature for 30 minutes; 8) Dilute the primary antibody with 5% BSA/TBST, discard the blocking solution after blocking, add GFP primary antibody, each tissue block requires 80-100μl of primary antibody, add double distilled water to the wet box, and incubate the slices at 4℃ overnight; 9) Discard the primary antibody, wash the slices 3 times with TBST at room temperature, 5 minutes each time; 10) Dilute the fluorescent secondary antibody with 5% BSA/TBST 11) discard the secondary antibody and DAPI, wash the slides 3 times with TBST at room temperature, 5 minutes each time; 12) seal the slides, and use a laser confocal microscope to photograph the Cholesin localization results.

如图6所示,观察到Cholesin-GFP与Apoa1-mCherry共定位,这表明Cholesin在吸收性肠细胞中表达。As shown in Figure 6, colocalization of Cholesin-GFP and Apoa1-mCherry was observed, indicating that Cholesin is expressed in absorptive enterocytes.

2.2 Cholesin水平随着胆固醇浓度的增加而呈剂量依赖性增加2.2 Cholesin levels increased in a dose-dependent manner with increasing cholesterol concentration

为了研究胆固醇是否直接影响Cholesin的分泌,构建了稳定过表达Cholesin的HCT116细胞(人结直肠癌细胞)。实验前一天对过表达Cholesin的HCT116稳转细胞系在24孔板中进行铺板,实验时细胞应处于对数生长期。对细胞进行处理前用提前预热的无血清RPMI1640培养基清洗细胞一次。用无血清的RPMI1640将外源胆固醇稀释至终浓度为0μM、2.5μM、5μM、10μM、20μM和40μM。加入含有所述终浓度的胆固醇的无血清RPMI1640培养基500μl至24孔板的孔中,37℃培养箱中静置培养1小时。培养结束后吸取400μl培养基到1.5ml EP管中,最大转速4℃离心5分钟以去除细胞碎片。每孔细胞所收培养基平行点样3次到微孔板中,每孔100μl,进行ELISA以检测Cholesin在培养基中的分泌情况。In order to study whether cholesterol directly affects the secretion of Cholesin, HCT116 cells (human colorectal cancer cells) that stably overexpress Cholesin were constructed. The HCT116 stably transfected cell line overexpressing Cholesin was plated in a 24-well plate one day before the experiment, and the cells should be in the logarithmic growth phase during the experiment. Wash the cells once with preheated serum-free RPMI1640 medium before treating the cells. Dilute exogenous cholesterol with serum-free RPMI1640 to a final concentration of 0μM, 2.5μM, 5μM, 10μM, 20μM and 40μM. Add 500μl of serum-free RPMI1640 medium containing the final concentration of cholesterol to the wells of the 24-well plate and culture in a 37°C incubator for 1 hour. After the culture is completed, aspirate 400μl of culture medium into a 1.5ml EP tube and centrifuge at a maximum speed of 4°C for 5 minutes to remove cell debris. The culture medium collected from each well of cells was spotted into the microplate three times in parallel, with 100 μl in each well, and ELISA was performed to detect the secretion of Cholesin in the culture medium.

同时还检测了细胞中的总胆固醇水平,具体方法如下:1)细胞吸取培养基检测Cholesin分泌量后,剩余细胞用Trypsin消化,PBS重悬后进行细胞计数;2)高速离心弃上清,每个样品加150μl异丙醇,冰上超声破碎细胞(40%强度,超声2秒,间隔3秒,总时间3分钟);3)超声结束后10000g,4℃离心10分钟,上清即为细胞总胆固醇提取液;4)使用CHOD-PAP法胆固醇测定试剂盒(中生北控)测定提取液中总胆固醇浓度,再根据提取液体积计算总胆固醇量,最后用总胆固醇量除以细胞总量,换算出每106个细胞的胆固醇含量。The total cholesterol level in the cells was also detected, and the specific method was as follows: 1) After the cells absorbed the culture medium to detect the amount of Cholesin secretion, the remaining cells were digested with Trypsin, and the cells were resuspended in PBS and counted; 2) The supernatant was discarded after high-speed centrifugation, 150 μl of isopropanol was added to each sample, and the cells were ultrasonically disrupted on ice (40% intensity, ultrasonic for 2 seconds, interval of 3 seconds, total time of 3 minutes); 3) After the end of ultrasonication, centrifuge at 10000g and 4°C for 10 minutes, and the supernatant was the total cholesterol extract of the cells; 4) The total cholesterol concentration in the extract was determined using the CHOD-PAP cholesterol determination kit (Sino-Biotech Holdings), and the total cholesterol amount was calculated based on the volume of the extract, and finally the total cholesterol amount was divided by the total number of cells to convert the cholesterol content per 106 cells.

结果如图7所示。结果显示,随着胆固醇浓度的增加,培养基中Cholesin的水平呈剂量依赖性增加。此外,随着胆固醇处理剂量的增加,细胞中的胆固醇总量也在逐渐上升。The results are shown in Figure 7. The results show that as the cholesterol concentration increases, the level of Cholesin in the culture medium increases in a dose-dependent manner. In addition, as the cholesterol treatment dose increases, the total amount of cholesterol in the cells also gradually increases.

2.3 HCT116细胞中NPC1L1的敲低破坏了胆固醇吸收和Cholesin分泌NPC1L1 knockdown in HCT116 cells disrupts cholesterol absorption and Cholesin secretion

为了在细胞水平上探究NPC1L1对于胆固醇促进Cholesin分泌的作用,在过表达Cholesin的HCT116稳定细胞系基础上对NPC1L1进行敲低(Npc1l1 KD)。随后通过免疫印迹在蛋白水平验证了NPC1L1敲低对Cholesin的表达没有影响(图8A)。In order to explore the role of NPC1L1 in cholesterol-induced Cholesin secretion at the cellular level, NPC1L1 was knocked down (Npc1l1 KD) based on the HCT116 stable cell line overexpressing Cholesin. Subsequently, Western blotting confirmed that NPC1L1 knockdown had no effect on Cholesin expression at the protein level (Figure 8A).

按照2.2中的方法对非靶敲低对照组细胞(NT)和NPC1L1敲低的细胞(Npc1l1 KD)在24孔板中铺板,再分别用含0μM和10μM胆固醇的无血清RPMI1640培养细胞1小时,培养结束后收取培养基用ELISA检测Cholesin的分泌情况。细胞则在计数后加入异 丙醇超声提取总胆固醇,使用CHOD-PAP法胆固醇测定试剂盒(中生北控)测定提取液中总胆固醇浓度,再根据提取液体积计算总胆固醇量,最后用总胆固醇量除以细胞总量,换算出每106个细胞的胆固醇含量。结果显示,在HCT116细胞中敲低NPC1L1会破坏胆固醇的吸收和Cholesin的分泌(图8B)。According to the method in 2.2, non-target knockdown control cells (NT) and NPC1L1 knockdown cells (Npc1l1 KD) were plated in 24-well plates, and then cultured with serum-free RPMI1640 containing 0μM and 10μM cholesterol for 1 hour. After the culture, the culture medium was collected and the secretion of Cholesin was detected by ELISA. Total cholesterol was extracted by ultrasonication with propanol, and the total cholesterol concentration in the extract was determined using the CHOD-PAP cholesterol assay kit (Zhongsheng Beikong). The total cholesterol amount was calculated based on the volume of the extract, and finally the total cholesterol amount was divided by the total number of cells to convert the cholesterol content per 10 6 cells. The results showed that knocking down NPC1L1 in HCT116 cells impaired cholesterol absorption and Cholesin secretion (Figure 8B).

实施例2.2和2.3的结果共同表明,NPC1L1介导的胆固醇吸收刺激Cholesin的分泌。The results of Examples 2.2 and 2.3 together indicate that NPC1L1-mediated cholesterol absorption stimulates the secretion of Cholesin.

2.4小鼠中NPC1L1抑制剂处理和Npc1l1基因敲除都有效阻断了小肠胆固醇吸收和Cholesin的分泌NPC1L1 inhibitor treatment and Npc1l1 gene knockout in mice effectively blocked small intestinal cholesterol absorption and cholesin secretion

为了进一步验证Cholesin在小鼠中的分泌是否依赖于NPC1L1介导的胆固醇吸收,使用了两种方法:NPC1L1抑制剂Ezetimibe(依折麦布)处理小鼠和直接使用Npc1l1基因敲除的小鼠。To further verify whether Cholesin secretion in mice is dependent on NPC1L1-mediated cholesterol absorption, two methods were used: treating mice with the NPC1L1 inhibitor Ezetimibe and directly using Npc1l1 gene knockout mice.

NPC1L1抑制剂处理NPC1L1 inhibitor treatment

10-12周龄雄鼠禁食过夜,然后以10mg kg-1的剂量灌胃依折麦布(Ezetimlbe+),以灌胃玉米油溶剂作为对照(Ezetimlbe-)。灌胃1小时后,小鼠再灌胃150μl玉米油溶剂对照(chol-)或胆固醇(chol+,200mg kg-1体重)。灌胃1小时后收集血浆,检测血浆中总胆固醇(TC)和Cholesin水平,同时收取小鼠小肠组织测定小肠总胆固醇含量。结果如图9A-9B所示。Male mice aged 10-12 weeks were fasted overnight, and then gavaged with ezetimibe (Ezetimlbe+) at a dose of 10 mg kg -1 , and gavage with corn oil solvent as a control (Ezetimlbe-). One hour after gavage, the mice were gavaged with 150 μl corn oil solvent control (chol-) or cholesterol (chol+, 200 mg kg -1 body weight). One hour after gavage, plasma was collected to detect total cholesterol (TC) and Cholesin levels in plasma, and small intestinal tissues of mice were collected to determine the total cholesterol content in the small intestine. The results are shown in Figures 9A-9B.

结果显示,禁食过夜的对照小鼠(Chol-)血浆中的Cholesin水平较低,血浆中含量为约500pM。未经依折麦布处理的小鼠在灌胃胆固醇后血浆中Cholesin水平显著上升,而依折麦布处理后的小鼠不再响应胆固醇给药刺激,血浆中Cholesin含量与饥饿状态基本持平(图9A)。与此同时,小肠的总胆固醇含量表明依折麦布的处理抑制了小肠对胆固醇的吸收(图9A)。小鼠血浆TC含量的测定表明,短时的给药刺激并不能引起血浆TC水平的变化(图9B)。The results showed that the level of Cholesin in the plasma of control mice (Chol-) that had fasted overnight was low, with a plasma content of about 500 pM. The level of Cholesin in the plasma of mice that were not treated with ezetimibe increased significantly after oral administration of cholesterol, while mice treated with ezetimibe no longer responded to the stimulation of cholesterol administration, and the level of Cholesin in the plasma was basically the same as that in the starvation state (Figure 9A). At the same time, the total cholesterol content in the small intestine indicated that the treatment with ezetimibe inhibited the absorption of cholesterol in the small intestine (Figure 9A). The determination of the plasma TC content of mice showed that short-term administration stimulation did not cause changes in plasma TC levels (Figure 9B).

Npc1l1基因敲除Npc1l1 knockout

为了进一步探究NPC1L1对于胆固醇促进Cholesin分泌的作用,构建了Npc1l1基因敲除(Npc1l1-/-)小鼠。通过免疫印迹确认Npc1l1的缺失不影响Cholesin的表达(图9C)。随后对野生型(Npc1l1+/+)和Npc1l1敲除(Npc1l1-/-)的小鼠禁食过夜,再给小鼠口服灌胃150μl玉米油溶剂对照(chol-)或胆固醇(chol+,200mg kg-1体重),1小时后收取小鼠全血分离血浆以检测血浆Cholesin和TC水平,同时收取小肠组织以测定TC含量。结果如图9D-9E所示。To further explore the role of NPC1L1 in cholesterol-induced Cholesin secretion, Npc1l1 knockout (Npc1l1 -/- ) mice were constructed. Immunoblotting confirmed that the absence of Npc1l1 did not affect the expression of Cholesin (Figure 9C). Wild-type (Npc1l1 +/+ ) and Npc1l1 knockout (Npc1l1 -/- ) mice were then fasted overnight, and then orally gavaged with 150 μl corn oil solvent control (chol-) or cholesterol (chol+, 200 mg kg -1 body weight). One hour later, whole blood was collected from the mice to separate plasma to detect plasma Cholesin and TC levels, and small intestinal tissue was collected to determine TC content. The results are shown in Figures 9D-9E.

结果显示,Npc1l1缺失能显著抑制小鼠Cholesin的分泌并抑制小肠对胆固醇的吸收 (图9D)。小鼠血浆TC含量的测定表明,短时的胆固醇处理不引起血浆TC水平的变化(图9E)。The results showed that Npc1l1 deficiency could significantly inhibit the secretion of Cholesin and the absorption of cholesterol in the small intestine of mice. (Fig. 9D) The measurement of plasma TC content in mice showed that short-term cholesterol treatment did not cause changes in plasma TC levels (Fig. 9E).

实施例2的上述体外和体内结果表明,NPC1L1介导的胆固醇吸收促进Cholesin的分泌。The above in vitro and in vivo results of Example 2 indicate that NPC1L1-mediated cholesterol absorption promotes the secretion of Cholesin.

实施例3:Cholesin在人体中调控胆固醇代谢Example 3: Cholesin regulates cholesterol metabolism in humans

为了探究Cholesin在人体中是否具有类似的对胆固醇的调控作用,收集了600例人类进食1小时后的血浆样品进行ELISA测定,以检测血浆样品中的Cholesin、总胆固醇(TC)和低密度脂蛋白胆固醇(LDL-C)水平并进行相关性分析。具体操作如下:在聚苯乙烯微孔板中加入3μl人类血浆和100μl包被液(0.05mol/L Na2CO3/NaHCO3,pH8.0),96孔微板中设置Cholesin全身敲除小鼠血浆作为对照组进行校准,用纯化定量过的Cholesin蛋白标品设置标准曲线,标准曲线浓度为0,12.5,25,50,100,200pmol,加样结束后4℃孵育过夜,使蛋白吸附在孔底。4℃摇床包被过夜后,取出微孔板,甩干孔内液体,每孔加1×gelatin 100μl室温封闭微孔板2小时。弃去封闭液,每孔加入200μlTBST溶液洗4次,最后一次将液体尽量甩干。将检测一抗配制于TBST中,每孔加入100μl一抗,4℃摇床孵育过夜。弃去检测一抗,每孔加入200μl TBST溶液洗4次,最后一次将液体尽量甩干。将二抗配制于TBST中,每孔加入100μl,室温摇床孵育1小时。弃去二抗,每孔加入200μl TBST溶液洗4次,最后一次将液体尽量甩干。每孔加入100μlTMB底物,室温摇床孵育10~30分钟。随后每孔加入100μl终止液(0.05mol/L H2SO4),30分钟内用酶标仪读取OD450nm的吸光值,根据标准曲线的吸光值计算得出待测样品中Cholesin的含量。将数据用Excel进行皮尔逊相关性分析,计算得到p值与r2In order to explore whether Cholesin has a similar regulatory effect on cholesterol in humans, 600 plasma samples were collected from humans 1 hour after eating for ELISA assay to detect the levels of Cholesin, total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in plasma samples and conduct correlation analysis. The specific operation is as follows: 3μl human plasma and 100μl coating solution (0.05mol/L Na2CO3/NaHCO3, pH8.0) were added to the polystyrene microplate, and the plasma of Cholesin knockout mice was set as the control group in the 96-well microplate for calibration. The standard curve was set with purified and quantified Cholesin protein standards, and the concentrations of the standard curve were 0, 12.5, 25, 50, 100, and 200pmol. After the addition of samples, the samples were incubated at 4℃ overnight to allow the protein to adsorb to the bottom of the wells. After overnight coating on a 4℃ shaker, the microplate was removed, the liquid in the wells was dried, and 100μl of 1×gelatin was added to each well to block the microplate at room temperature for 2 hours. Discard the blocking solution, add 200μl TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time. Prepare the primary antibody for detection in TBST, add 100μl primary antibody to each well, and incubate overnight at 4℃ on a shaker. Discard the primary antibody for detection, add 200μl TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time. Prepare the secondary antibody in TBST, add 100μl to each well, and incubate at room temperature on a shaker for 1 hour. Discard the secondary antibody, add 200μl TBST solution to each well and wash 4 times, and shake off the liquid as much as possible for the last time. Add 100μl TMB substrate to each well and incubate at room temperature on a shaker for 10 to 30 minutes. Then add 100μl stop solution (0.05mol/L H 2 SO 4 ) to each well, read the absorbance value of OD450nm with an enzyme reader within 30 minutes, and calculate the content of Cholesin in the sample to be tested based on the absorbance value of the standard curve. The data were subjected to Pearson correlation analysis using Excel, and the p value and r 2 were calculated.

相关性分析结果显示,Cholesin水平与TC和LDL-C水平呈负相关(图10A和10B)。这表明Cholesin对胆固醇水平具有负调控作用。Correlation analysis results showed that Cholesin levels were negatively correlated with TC and LDL-C levels (Figures 10A and 10B), indicating that Cholesin has a negative regulatory effect on cholesterol levels.

实施例4:Cholesin敲除增加了肝脏胆固醇合成和VLDL分泌Example 4: Cholesin knockout increases liver cholesterol synthesis and VLDL secretion

4.1 Cholesin IKO小鼠的血浆总胆固醇、HDL、LDL、VLDL和甘油三酯水平以及肝脏胆固醇和甘油三酯水平的测定4.1 Determination of plasma total cholesterol, HDL, LDL, VLDL and triglyceride levels and liver cholesterol and triglyceride levels in Cholesin IKO mice

本实施例选用遗传背景为C57BL/6J的12-14周龄野生型(WT)以及肠道Cholesin特异性敲除(Cholesin IKO)的雌雄小鼠,分别喂食8周的含0.02%胆固醇的普通饮食(RD)和含0.2%胆固醇的西方饮食(WD)。小鼠饥饿过夜后再进食6小时后收取小鼠血浆以检测血浆总胆固醇(TC)和甘油三酯(TG)水平,同时还收取了小鼠肝脏组织以 检测肝脏TC和TG水平。结果如图11A和11B所示。In this example, 12-14 week old wild-type (WT) and intestinal Cholesin-specific knockout (Cholesin IKO) male and female mice with a genetic background of C57BL/6J were selected and fed with a normal diet (RD) containing 0.02% cholesterol and a Western diet (WD) containing 0.2% cholesterol for 8 weeks. After the mice were starved overnight and then fed for 6 hours, the mouse plasma was collected to detect the plasma total cholesterol (TC) and triglyceride (TG) levels. The mouse liver tissue was also collected to detect the level of cholesterol. The levels of TC and TG in the liver were detected. The results are shown in Figures 11A and 11B.

由于胆固醇在血液中主要以脂蛋白(例如HDL、LDL和VLDL)的形式存在,因此使用快速蛋白液相色谱法分析血浆中各脂蛋白的含量,实验方法如下:合并每组中所有小鼠的血浆,混匀后置于冰上等待上样。将Superose 6 Increase 10/300 GL层析柱连接到AKTA纯化系统中,用PBS平衡层析柱,上样环准确进样1ml总血浆,在完全进样后不用等待出峰即可开始收集洗脱下的各组分,每个组分收样400μl,待出现盐峰时停止收样。使用酶学方法测定每个组分的总胆固醇含量,绘制FPLC曲线。根据FPLC的曲线下面积,计算极低密度脂蛋白(VLDL)、中间/低密度脂蛋白胆固醇(IDL/LDL)和高密度脂蛋白胆固醇(HDL)的相对水平。结果如图11C所示。Since cholesterol exists mainly in the form of lipoproteins (such as HDL, LDL and VLDL) in the blood, fast protein liquid chromatography was used to analyze the content of each lipoprotein in plasma. The experimental method is as follows: the plasma of all mice in each group is combined, mixed and placed on ice for loading. The Superose 6 Increase 10/300 GL column was connected to the AKTA purification system, and the column was balanced with PBS. The sample loop accurately injected 1 ml of total plasma. After complete injection, the eluted components can be collected without waiting for the peak. Each component is sampled 400 μl, and the sampling is stopped when the salt peak appears. The total cholesterol content of each component was determined by enzymatic method, and the FPLC curve was drawn. According to the area under the FPLC curve, the relative levels of very low density lipoprotein (VLDL), intermediate/low density lipoprotein cholesterol (IDL/LDL) and high density lipoprotein cholesterol (HDL) were calculated. The results are shown in Figure 11C.

图11A显示,野生型小鼠在普通饮食喂食条件下,雄鼠的血浆TC在100mg/dL左右,雌鼠略低于雄鼠;而在WD喂食条件下,雄鼠的血浆TC达到170mg/dL左右,雌鼠血浆TC在140mg/dL的水平。IKO雄性和雌性小鼠在喂食普通饮食或西方饮食后,血浆TC水平均比野生型小鼠血浆TC含量高20%左右。小鼠血浆中的TG含量,在喂食RD或WD的雄性和雌性小鼠中,IKO小鼠血浆TG相对比于WT组略微增加。Figure 11A shows that when wild-type mice were fed with a normal diet, the plasma TC of male mice was about 100 mg/dL, and that of female mice was slightly lower than that of male mice; while under the WD feeding condition, the plasma TC of male mice reached about 170 mg/dL, and that of female mice was 140 mg/dL. After IKO male and female mice were fed with a normal diet or a Western diet, the plasma TC levels were about 20% higher than those of wild-type mice. The TG content in mouse plasma was slightly increased in IKO mice compared with the WT group in male and female mice fed with RD or WD.

图11B显示,在喂食RD或WD的雄性和雌性小鼠中,IKO小鼠的肝脏TC含量均高于WT组。小鼠肝脏TC含量在雌雄之间没明显差异,在WD喂饲条件下,WT和IKO之间肝脏TC的差异被扩大。肝脏TG则在RD条件下,WT和IKO组没有明显差异,但在WD的诱导下IKO组肝脏TG稍高于WT组。Figure 11B shows that in male and female mice fed RD or WD, the liver TC content of IKO mice was higher than that of WT group. There was no significant difference in liver TC content between male and female mice, and the difference in liver TC between WT and IKO was amplified under WD feeding conditions. There was no significant difference in liver TG between WT and IKO groups under RD conditions, but under the induction of WD, the liver TG of IKO group was slightly higher than that of WT group.

图11C显示,Cholesin IKO小鼠的高密度脂蛋白胆固醇(HDL-C)、低密度脂蛋白胆固醇(LDL-C)和极低密度脂蛋白胆固醇(VLDL-C)均有所增加。Figure 11C shows that the levels of high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and very low-density lipoprotein cholesterol (VLDL-C) were increased in Cholesin IKO mice.

这些结果表明,Cholesin的缺乏导致血浆胆固醇和甘油三酯水平升高。These results suggest that Cholesin deficiency leads to elevated plasma cholesterol and triglyceride levels.

4.2 Cholesin IKO小鼠中升高的血浆胆固醇水平不是由小肠胆固醇吸收增强导致的4.2 Elevated plasma cholesterol levels in Cholesin IKO mice are not caused by enhanced cholesterol absorption in the small intestine

根据先前的报道,血浆胆固醇水平的升高可能是由小肠胆固醇吸收增强、肝脏胆固醇合成和VLDL分泌增加,或者胆固醇排泄减少所致。为探究IKO小鼠中升高的血浆胆固醇水平是否由小肠胆固醇吸收增强导致的,对实施例4.1的小鼠的体重、体脂率、食物和水摄入量、小肠总胆固醇和甘油三酯水平及乳糜微粒分解率进行了测量。According to previous reports, the increase in plasma cholesterol levels may be caused by enhanced intestinal cholesterol absorption, increased hepatic cholesterol synthesis and VLDL secretion, or decreased cholesterol excretion. To explore whether the elevated plasma cholesterol levels in IKO mice were caused by enhanced intestinal cholesterol absorption, the body weight, body fat percentage, food and water intake, small intestinal total cholesterol and triglyceride levels, and chylomicron decomposition rate of the mice in Example 4.1 were measured.

对随机采食状态下的小鼠,测量体重并通过EchoMRI清醒动物身体成分分析仪测量体脂,每只小鼠重复测量3次。小鼠体脂率按以下公式计算:体脂率=小鼠体脂/小鼠体重×100%。结果如图12A和12B所示。The mice in the random feeding state were weighed and their body fat was measured by EchoMRI awake animal body composition analyzer, and the measurement was repeated 3 times for each mouse. The body fat percentage of mice was calculated according to the following formula: body fat percentage = mouse body fat/mouse weight × 100%. The results are shown in Figures 12A and 12B.

使用TSE Systems公司的PhenoMaster系统对单独饲养的小鼠进行食物摄入量的测量。在测量前,小鼠至少有24小时的适应时间。食物和水在适当的装置中自由供给,并 由内置的自动仪器测量,连续采集48小时的自由采食与饮水数据。结果如图12C和12D所示。Food intake was measured in individually housed mice using the PhenoMaster system from TSE Systems. Mice were allowed at least 24 hours of acclimatization before measurement. Food and water were freely available in appropriate apparatus and The data of free food and water intake were collected continuously for 48 hours by the built-in automatic instrument. The results are shown in Figures 12C and 12D.

为测定小鼠小肠总胆固醇和甘油三酯含量,收取实施例4.1的饥饿过夜后再进食6小时小鼠的小肠组织,测定结果如图12E所示。In order to determine the total cholesterol and triglyceride content in the small intestine of mice, the small intestine tissues of the mice that were starved overnight and then fed for 6 hours in Example 4.1 were collected. The determination results are shown in FIG. 12E .

通过给小鼠口服一定剂量的橄榄油进行脂质耐受实验来评估小鼠小肠的乳糜微粒分解速率。将实施例4.1的小鼠禁食6小时使小鼠肠道状态进行同步化,随后口服200μl橄榄油,在口服前和口服后不同的时间点(0、1、2、3、4小时)从尾静脉采集血液,然后分离血浆,测定血浆中甘油三酯水平并计算甘油三酯分泌率(△TG:每只小鼠的1、2、3、4小时的血浆TG减去小鼠的0点的血浆TG),以评估乳糜微粒分解速率。结果如图12F所示。The lipid tolerance test was performed by orally administering a certain dose of olive oil to mice to evaluate the rate of chylomicron decomposition in the small intestine of mice. The mice of Example 4.1 were fasted for 6 hours to synchronize the intestinal state of the mice, and then 200 μl of olive oil was orally administered. Blood was collected from the tail vein at different time points (0, 1, 2, 3, and 4 hours) before and after oral administration, and then plasma was separated. The triglyceride level in the plasma was determined and the triglyceride secretion rate (△TG: plasma TG at 1, 2, 3, and 4 hours of each mouse minus the plasma TG at 0 of the mouse) was calculated to evaluate the rate of chylomicron decomposition. The results are shown in Figure 12F.

图12A显示,雌雄小鼠在两种饮食处理下,WT与IKO组小鼠的体重均无明显差异。图12B显示,雌雄小鼠在两种饮食处理下,WT与IKO组的体脂率均无明显差异。图12C和12D显示,雌雄小鼠在两种饮食处理下,WT与IKO组之间的食物摄入量和水摄入量无明显差异。图12E显示,喂食普通饮食或西方饮食的雄性和雌性小鼠中,IKO小鼠和WT小鼠的小肠总胆固醇和甘油三酯含量没有明显差异。图12F显示,喂食普通饮食或西方饮食的雄性和雌性小鼠中,IKO小鼠的乳糜微粒分解速率不受影响。Figure 12A shows that there was no significant difference in body weight between WT and IKO groups of male and female mice under the two dietary treatments. Figure 12B shows that there was no significant difference in body fat percentage between WT and IKO groups of male and female mice under the two dietary treatments. Figures 12C and 12D show that there was no significant difference in food intake and water intake between WT and IKO groups of male and female mice under the two dietary treatments. Figure 12E shows that there was no significant difference in total cholesterol and triglyceride content in the small intestine between IKO mice and WT mice in male and female mice fed a normal diet or a Western diet. Figure 12F shows that the rate of chylomicron decomposition in IKO mice was not affected in male and female mice fed a normal diet or a Western diet.

这些结果表明,IKO小鼠和WT小鼠的肠道吸收类似,因此IKO小鼠中升高的血浆胆固醇水平不是由小肠胆固醇吸收增强导致的。These results suggest that intestinal absorption is similar in IKO and WT mice and that the elevated plasma cholesterol levels in IKO mice are not due to enhanced cholesterol absorption in the small intestine.

4.3 Cholesin IKO小鼠中升高的血浆胆固醇水平不是由胆固醇排泄减少导致的4.3 Elevated plasma cholesterol levels in Cholesin IKO mice are not caused by decreased cholesterol excretion

为探究IKO小鼠中升高的血浆胆固醇水平是否由胆固醇排泄减少导致的,对实施例4.1的小鼠的胆汁胆固醇、胆汁酸水平以及粪便中的胆固醇水平进行了测量。To investigate whether the elevated plasma cholesterol levels in IKO mice were caused by decreased cholesterol excretion, the bile cholesterol, bile acid levels, and fecal cholesterol levels of the mice of Example 4.1 were measured.

小鼠机体胆固醇一般通过胆汁酸或者粪便排出体外,通过测定胆汁酸和粪便中的胆固醇含量来探究IKO小鼠是否由于胆固醇外排变化而影响了血浆TC水平。小鼠胆汁在进食过程中会从胆囊排出,而在饥饿时则主要储存在胆囊中。将小鼠饥饿过夜,用注射器收集胆囊中的胆汁,随后称量胆囊体积,并通过试剂盒测量胆汁总胆固醇(胆固醇测定试剂盒(CHOD-PAP法,中生北控)和胆汁酸(总胆汁酸(TBA)试剂盒,南京建成)的浓度。结果如图13A所示。Cholesterol in mice is generally excreted through bile acid or feces. The cholesterol content in bile acid and feces was measured to explore whether the plasma TC level of IKO mice was affected by changes in cholesterol excretion. Mouse bile is excreted from the gallbladder during eating and is mainly stored in the gallbladder when hungry. The mice were starved overnight, and the bile in the gallbladder was collected with a syringe. The volume of the gallbladder was then weighed, and the concentrations of total bile cholesterol (cholesterol determination kit (CHOD-PAP method, Zhongsheng Beikong) and bile acid (total bile acid (TBA) kit, Nanjing Jiancheng) were measured using kits. The results are shown in Figure 13A.

为测量粪便中的胆固醇含量,收集了小鼠的粪便并通过以下方法来提取粪便中的总胆固醇和甘油三酯:称取40mg粪便并在55℃烘箱中烘干,随后将干燥粉碎的粪便悬浮在1200μl 2:1的氯仿-甲醇(v/v)混合物中。然后将悬浮液与300μl的水混合,用力涡旋60秒。然后,再4℃以4000rpm的转速离心10分钟,并将有机层小心地转移到一个新 的试管中进行干燥。然后将干燥的脂质重悬于100μl 9:1异丙醇-Triton X-100(v/v)溶液中,得到的粪便提取液用于酶学测定以检测总胆固醇和甘油三酯的水平。结果如图13B所示。To measure the cholesterol content in feces, the feces of mice were collected and the total cholesterol and triglycerides in the feces were extracted by the following method: 40 mg of feces were weighed and dried in a 55°C oven, and then the dried and crushed feces were suspended in 1200 μl of a 2:1 chloroform-methanol (v/v) mixture. The suspension was then mixed with 300 μl of water and vortexed vigorously for 60 seconds. Then, it was centrifuged at 4000 rpm for 10 minutes at 4°C, and the organic layer was carefully transferred to a fresh The dried lipids were then resuspended in 100 μl of a 9:1 isopropanol-Triton X-100 (v/v) solution, and the resulting fecal extract was used for enzymatic assays to detect total cholesterol and triglyceride levels. The results are shown in FIG13B .

图13A显示,雌雄小鼠在两种饮食处理下,WT与IKO小鼠的胆汁总量、胆汁胆固醇水平和胆汁酸水平均无明显差异。图13B显示,雌雄小鼠在两种饮食处理下,WT与IKO小鼠的粪便中的总胆固醇含量和甘油三酯含量均无明显差异。Figure 13A shows that there were no significant differences in the total bile volume, bile cholesterol level and bile acid level between WT and IKO mice under the two dietary treatments for male and female mice. Figure 13B shows that there were no significant differences in the total cholesterol content and triglyceride content in the feces between WT and IKO mice under the two dietary treatments for male and female mice.

以上结果表明IKO小鼠中升高的血浆胆固醇水平不是由胆固醇排泄减少导致的。These results indicate that the elevated plasma cholesterol levels in IKO mice are not caused by decreased cholesterol excretion.

4.4 Cholesin IKO小鼠中升高的血浆胆固醇水平是由肝脏中的胆固醇合成增加导致的。4.4 Elevated plasma cholesterol levels in Cholesin IKO mice are caused by increased cholesterol synthesis in the liver.

为探究IKO小鼠中升高的血浆胆固醇水平是否由肝脏胆固醇合成增强导致的,在mRNA和蛋白质水平上检测了肝脏中胆固醇合成基因的表达。To investigate whether the elevated plasma cholesterol levels in IKO mice were caused by enhanced cholesterol synthesis in the liver, the expression of cholesterol synthesis genes in the liver was examined at the mRNA and protein levels.

利用qPCR检测肝脏中胆固醇合成相关基因的mRNA水平。具体实验操作如下:1)需称取20mg左右肝脏组织,用HP total RNA Kit(Omega)提取肝脏总RNA;2)取1μgRNA用REVERTAID 1ST CDNA SYNTH KIT(thermo)进行反转录;3)反转录所得cDNA进行qPCR。结果如图14A所示。qPCR was used to detect the mRNA levels of cholesterol synthesis-related genes in the liver. The specific experimental procedures are as follows: 1) About 20 mg of liver tissue was weighed and the total liver RNA was extracted using HP total RNA Kit (Omega); 2) 1 μg RNA was taken and reverse transcribed using REVERTAID 1ST CDNA SYNTH KIT (thermo); 3) The cDNA obtained by reverse transcription was subjected to qPCR. The results are shown in Figure 14A.

利用免疫印迹检测肝脏中胆固醇合成基因的蛋白表达水平。具体操作如下:1)称取20mg肝脏组织,加入500μl RIPA裂解液和磁珠,组织研磨机60Hz低温研磨1分钟;2)4℃最大转速离心10分钟;3)取上清转移到新EP管中,注意不要吸到脂肪层加入5×上样缓冲液,混匀,100℃煮样10分钟;4)用BCA法进行蛋白定量;5)通过免疫印迹检测组织中的SREBP2(包括pSREBP2和mSREBP2)、HMGCR、HMGCS1和LDLR表达水平。结果如图14B所示。Immunoblotting was used to detect the protein expression level of cholesterol synthesis genes in the liver. The specific operation is as follows: 1) Weigh 20 mg of liver tissue, add 500 μl RIPA lysis buffer and magnetic beads, and grind at 60 Hz for 1 minute in a tissue grinder; 2) Centrifuge at maximum speed at 4°C for 10 minutes; 3) Transfer the supernatant to a new EP tube, be careful not to absorb the fat layer, add 5× loading buffer, mix well, and boil the sample at 100°C for 10 minutes; 4) Use the BCA method for protein quantification; 5) Detect the expression levels of SREBP2 (including pSREBP2 and mSREBP2), HMGCR, HMGCS1 and LDLR in the tissue by immunoblotting. The results are shown in Figure 14B.

图14A和14B显示,相比较于野生型小鼠,普通饮食和西方饮食喂养的IKO小鼠肝脏中胆固醇合成相关基因Hmgcs1、Hmgcr、Mvd、Mvk、Pmvk、sqle以及调控这些胆固醇合成相关基因表达的转录因子SREBP2及其下游调控的LDLR的表达水平均显著增加。值得注意的是,SREBP2和胆固醇合成相关基因的表达在西方饮食喂养的小鼠中水平更低,这表明肠道胆固醇的吸收抑制了肝脏胆固醇的合成。Figures 14A and 14B show that compared with wild-type mice, the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, Pmvk, sqle in the liver of IKO mice fed with a normal diet and a Western diet, as well as the transcription factor SREBP2 that regulates the expression of these cholesterol synthesis-related genes and its downstream regulated LDLR were significantly increased. It is worth noting that the expression of SREBP2 and cholesterol synthesis-related genes was lower in mice fed with a Western diet, indicating that the absorption of intestinal cholesterol inhibits the synthesis of cholesterol in the liver.

4.5 Cholesin IKO小鼠中升高的血浆胆固醇水平是由肝脏的VLDL分泌增强导致的。4.5 Elevated plasma cholesterol levels in Cholesin IKO mice are caused by enhanced VLDL secretion in the liver.

为探究IKO小鼠中升高的血浆胆固醇水平是否由肝脏的VLDL分泌增强导致的,向小鼠注射泊洛沙姆407(一种抑制脂蛋白脂酶和VLDL代谢的抑制剂),并监测小鼠血浆胆固醇和甘油三酯水平,以评估肝脏VLDL的分泌。To investigate whether the elevated plasma cholesterol levels in IKO mice were caused by enhanced VLDL secretion from the liver, mice were injected with poloxamer 407, an inhibitor of lipoprotein lipase and VLDL metabolism, and plasma cholesterol and triglyceride levels were monitored to assess hepatic VLDL secretion.

将小鼠禁食4小时,然后向小鼠腹腔注射1g kg-1体重的泊洛沙姆407。在泊洛沙姆407注射前、注射后1小时和2小时分别从尾静脉采集血样。在每个时间点测量血浆总胆 固醇(TC)和甘油三酯(TG)的水平并计算TC和TG分泌率(计算方法为每只小鼠1或2小时的TC或TG水平减去该小鼠0小时的TC或TG水平)。结果如图15所示。The mice were fasted for 4 hours and then intraperitoneally injected with 1 g kg-1 body weight of Poloxamer 407. Blood samples were collected from the tail vein before, 1 hour and 2 hours after the injection of Poloxamer 407. Plasma total bile duct was measured at each time point. The levels of sterol (TC) and triglyceride (TG) were measured and the secretion rates of TC and TG were calculated (the calculation method was the TC or TG level of each mouse at 1 or 2 hours minus the TC or TG level of the mouse at 0 hours). The results are shown in FIG15 .

图15的结果显示,与WT小鼠相比,饲喂普通饮食和西方饮食的雄性和雌性的IKO小鼠的TC和TG分泌率均显著升高,这表明Cholesin的缺乏会增加肝脏VLDL的分泌,从而导致血浆胆固醇水平升高。The results in Figure 15 showed that the secretion rates of TC and TG in male and female IKO mice fed with a normal diet and a Western diet were significantly increased compared with those in WT mice, indicating that Cholesin deficiency increases the secretion of hepatic VLDL, thereby leading to increased plasma cholesterol levels.

实施例4的上述结果表明,Cholesin的敲除增加了肝脏的胆固醇合成和VLDL分泌,从而导致血浆胆固醇和甘油三酯水平增加。这暗示了通过施用Cholesin可以降低肝脏的胆固醇合成和VLDL分泌,从而降低血浆胆固醇和甘油三酯水平。The above results of Example 4 show that knockout of Cholesin increases cholesterol synthesis and VLDL secretion in the liver, thereby increasing plasma cholesterol and triglyceride levels. This suggests that administration of Cholesin can reduce cholesterol synthesis and VLDL secretion in the liver, thereby reducing plasma cholesterol and triglyceride levels.

实施例5:GPR146是Cholesin的受体Example 5: GPR146 is a receptor for Cholesin

5.1 Cholesin作用的靶组织5.1 Target tissues of Cholesin

为了鉴定Cholesin作用的靶组织,选用遗传背景为C57BL/6J的8-10周龄野生型(Gpr146+/+)小鼠,将纯化的GST-Cholesin与小鼠的不同组织孵育在冷冻组织切片上进行体外结合实验。具体过程如下:1)收取小鼠组织:根据实验设计收取小鼠的脂肪组织(附睾白色脂肪组织(eWAT)和棕色脂肪组织(BAT))、肌肉、肝脏、胰腺、小肠和肾脏组织,并立即置于PBS配制的4%的多聚甲醛溶液中4℃固定过夜;2)组织脱水与包埋:根据OCT和石蜡包埋剂的不同分别进行不同的脱水处理,除白色脂肪组织用于石蜡包埋,其他组织后续均用OCT包埋;3)组织切片:用冷冻切片机切片厚度为10μm,石蜡切片厚度为5μm;4)冷冻切片置于免疫荧光湿盒中,置于室温化冻,石蜡切片则需要进行抗原修复,预处理好的切片用免疫组化笔在组织外围画一个闭合的圈,并用PBS洗两遍;5)蛋白结合:配制结合蛋白(其中GST-Cholesin 200nM;GST 200nM),室温孵育30分钟;6)蛋白孵育结束后,用PBS洗2遍,用PBS配制的5% BSA封闭液室温封闭1小时;7)弃去封闭液,用封闭液稀释的一抗4℃孵育过夜;8)弃去一抗,TBST洗3次,用封闭液稀释的二抗和终浓度为0.5mg/ml的DAPI室温孵育1h;10)弃去二抗和DAPI,TBST洗3次;11)尽量吸干组织表面的溶液,根据组织大小在玻片表面滴加适量的封片剂,小心的盖上盖玻片,避免产生气泡,室温避光放置直至封片剂凝固,室温避光放置后进行成像观察。In order to identify the target tissues of Cholesin, 8-10 week old wild-type (Gpr146 +/+ ) mice with a genetic background of C57BL/6J were selected, and purified GST-Cholesin was incubated with different tissues of mice for in vitro binding experiments on frozen tissue sections. The specific process is as follows: 1) Collect mouse tissues: According to the experimental design, the mouse adipose tissue (epididymal white adipose tissue (eWAT) and brown adipose tissue (BAT)), muscle, liver, pancreas, small intestine and kidney tissues were collected and immediately placed in a 4% paraformaldehyde solution prepared in PBS and fixed overnight at 4°C; 2) Tissue dehydration and embedding: Different dehydration treatments were performed according to the different OCT and paraffin embedding agents. Except for white adipose tissue used for paraffin embedding, other tissues were subsequently embedded in OCT; 3) Tissue sectioning: The thickness of the sections was 10 μm using a freezing microtome, and the thickness of the paraffin sections was 5 μm; 4) The frozen sections were placed in an immunofluorescence humidified box and thawed at room temperature. The paraffin sections required antigen repair. The pre-treated sections were drawn with an immunohistochemistry pen to draw a closed circle around the tissue and washed twice with PBS; 5) Protein binding: Prepare binding proteins (GST-Cholesin 200nM; GST 200nM), incubate at room temperature for 30 minutes; 6) After the protein incubation, wash twice with PBS, and block with 5% BSA blocking solution prepared in PBS at room temperature for 1 hour; 7) Discard the blocking solution, incubate with primary antibody diluted in blocking solution at 4°C overnight; 8) Discard the primary antibody, wash three times with TBST, and incubate with secondary antibody diluted in blocking solution and DAPI with a final concentration of 0.5mg/ml at room temperature for 1h; 10) Discard the secondary antibody and DAPI, and wash three times with TBST; 11) Try to dry the solution on the surface of the tissue, add an appropriate amount of mounting medium on the surface of the slide according to the size of the tissue, carefully cover with a coverslip to avoid bubbles, and place at room temperature away from light until the mounting medium solidifies, and then place at room temperature away from light for imaging observation.

由免疫荧光的结果可知,GST-Cholesin在肌肉、肝脏、肾脏和脂肪组织中均存在结合信号,并且在肌肉、肝脏、肾脏和脂肪组织中的结合信号相对更强(图16A)。这表明肌肉、肝脏、肾脏和脂肪组织是Cholesin作用的靶组织。From the results of immunofluorescence, it can be seen that GST-Cholesin has binding signals in muscle, liver, kidney and adipose tissue, and the binding signals in muscle, liver, kidney and adipose tissue are relatively stronger (Figure 16A). This indicates that muscle, liver, kidney and adipose tissue are the target tissues of Cholesin.

5.2 Cholesin受体的鉴定 5.2 Identification of Cholesin Receptors

为了鉴定Cholesin的受体,使用包含65,383个靶向19050个人类蛋白编码基因的sgRNA、21,864个靶向miRNA的sgRNA和3000个非靶向对照sgRNA的合成病毒库进行了全基因组筛选。根据筛选结果,将GPR146确定为Cholesin候选受体。GPR146是涉及胆固醇合成调控的保守孤儿G蛋白偶联受体。To identify the receptor for Cholesin, a genome-wide screening was performed using a synthetic viral library containing 65,383 sgRNAs targeting 19,050 human protein-coding genes, 21,864 sgRNAs targeting miRNAs, and 3,000 non-targeting control sgRNAs. Based on the screening results, GPR146 was identified as a candidate receptor for Cholesin. GPR146 is a conserved orphan G protein-coupled receptor involved in the regulation of cholesterol synthesis.

为了研究GPR146是否为Cholesin的受体,选用遗传背景为C57BL/6J的8-10周龄Gpr146基因敲除(Gpr146-/-)小鼠,如实施例5.1所述进行体外结合实验。免疫荧光结果如图16A所示。In order to study whether GPR146 is a receptor for Cholesin, 8-10 week old Gpr146 knockout (Gpr146 -/- ) mice with a genetic background of C57BL/6J were selected to perform in vitro binding experiments as described in Example 5.1. The immunofluorescence results are shown in FIG16A .

为了证明GPR146是Cholesin的特异性受体,根据GPR146胞外区氨基酸的保守性和酸碱性,设计了GPR146的突变体(R90A,H101A,H169A,R179A,D187A,E265A)。由于肝脏是Cholesin的主要靶组织,分离了野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠的原代肝细胞进行结合染色。对于Gpr146-/-小鼠原代肝细胞,通过转染质粒以过表达野生型GPR146(WT)和突变体GPR146(Mut)。具体实验方法如下:1)根据实验需要提前在6孔板中铺上合适数量的直径12mm的细胞爬片,同时分离Gpr146+/+和Gpr146-/-小鼠的原代肝细胞,将细胞稀释至合适密度铺在6孔板中;2)Gpr146-/-小鼠的细胞贴壁6小时后转染质粒以过表达野生型GPR146(WT)和突变体GPR146(Mut);3)细胞培养36~48小时后,将6孔板中的细胞爬片转移至24孔板中,用PBS缓冲液清洗一次。将GST和GST-Cholesin用新鲜培养基稀释至200nM,Cholesin-His稀释至20μM。WT细胞分为3组:GST阴性对照组,GST-Cholesin结合组,GST-Cholesin和Cholesin-His共孵育组。Gpr146-/-细胞则只与200nM GST-Cholesin共孵育。每孔中加入200μl含配体的培养基,放置在37℃恒温培养箱中培养30min;4)孵育完毕后吸弃培养基,用PBS缓冲液清洗3次;5)每孔中加入250μl 4%多聚甲醛/PBS溶液置于水平摇床上室温固定30min;6)吸弃多聚甲醛溶液,用PBS清洗一次后加入250μl 5% BSA/PBS溶液在水平摇床上室温封闭30min;7)5% BSA/PBS溶液按合适的比例稀释一抗,吸弃封闭液并加入200μl一抗溶液室温孵育1h;8)回收一抗置于4℃保存,用PBS溶液洗三次,每次10min;9)用5% BSA/PBS稀释荧光二抗和DAPI,每孔加入200μl二抗和DAPI溶液,在水平摇床上室温避光孵育1h;10)吸弃二抗和DAPI溶液,用PBS溶液洗三次,每次10min;11)用无尘纸擦拭载玻片并标注实验条件,在载玻片上滴加适量封片剂,夹取细胞爬片在吸水纸上沿爬片边缘轻轻吸干残存的液体,将有细胞的一侧朝封片剂缓缓扣下并避免产生气泡,室温避光放置过夜,等待封片剂凝固后进行成像观察。实验结果如图16B所示。In order to prove that GPR146 is a specific receptor for Cholesin, mutants of GPR146 (R90A, H101A, H169A, R179A, D187A, E265A) were designed based on the conservation and acid-base properties of the amino acids in the extracellular region of GPR146. Since the liver is the main target tissue of Cholesin, primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 -/- ) mice were isolated for binding staining. For primary hepatocytes of Gpr146 -/- mice, plasmids were transfected to overexpress wild-type GPR146 (WT) and mutant GPR146 (Mut). The specific experimental method is as follows: 1) According to the experimental needs, a suitable number of cell slides with a diameter of 12 mm were spread in a 6-well plate in advance, and primary hepatocytes of Gpr146 +/+ and Gpr146 -/- mice were isolated at the same time, and the cells were diluted to a suitable density and spread in a 6-well plate; 2) After the cells of Gpr146 -/- mice adhered for 6 hours, plasmids were transfected to overexpress wild-type GPR146 (WT) and mutant GPR146 (Mut); 3) After 36 to 48 hours of cell culture, the cell slides in the 6-well plate were transferred to a 24-well plate and washed once with PBS buffer. GST and GST-Cholesin were diluted to 200nM with fresh culture medium, and Cholesin-His was diluted to 20μM. WT cells were divided into 3 groups: GST negative control group, GST-Cholesin binding group, and GST-Cholesin and Cholesin-His co-incubation group. Gpr146 -/- cells were only co-incubated with 200nM GST-Cholesin. 200μl of ligand-containing culture medium was added to each well and placed in a 37°C constant temperature incubator for 30min; 4) After incubation, the culture medium was aspirated and washed three times with PBS buffer; 5) 250μl of 4% paraformaldehyde/PBS solution was added to each well and fixed at room temperature for 30min on a horizontal shaker; 6) The paraformaldehyde solution was aspirated, washed once with PBS, and then 250μl of 5% BSA/PBS solution was added to block at room temperature for 30min on a horizontal shaker; 7) The primary antibody was diluted in a 5% BSA/PBS solution according to an appropriate ratio, the blocking solution was aspirated and 200μl of the primary antibody solution was added to incubate at room temperature for 1h; 8) The primary antibody was recovered and stored at 4°C, and washed three times with PBS solution, each for 10min; 9) 5% Dilute the fluorescent secondary antibody and DAPI with BSA/PBS, add 200 μl of secondary antibody and DAPI solution to each well, and incubate at room temperature in the dark on a horizontal shaker for 1 hour; 10) Aspirate and discard the secondary antibody and DAPI solution, wash three times with PBS solution, 10 minutes each time; 11) Wipe the slide with dust-free paper and mark the experimental conditions, drop an appropriate amount of mounting medium on the slide, clamp the cell slide and gently absorb the remaining liquid along the edge of the slide on the absorbent paper, slowly press the side with cells toward the mounting medium and avoid bubbles, place at room temperature in the dark overnight, and wait for the mounting medium to solidify before imaging observation. The experimental results are shown in Figure 16B.

同时,还在遗传背景为C57BL/6J的8-10周龄野生型小鼠以及Gpr146-/-小鼠的原代 肝细胞上进行了饱和结合实验。具体过程如下:1)分离WT和Gpr146-/-小鼠的原代肝细胞铺于12孔板,设置4组:WT细胞,Gpr146-/-细胞,Gpr146-/-回补野生型GPR146细胞,和Gpr146-/-回补突变型GPR146细胞。细胞贴壁6小时后对Gpr146-/-细胞加入过表达野生型和突变型GPR146的腺病毒;2)将纯化后的Cholesin用生物素标记试剂盒进行标记,并测定生物素标记的Cholesin浓度;3)感染24小时的细胞弃培养基,加入5%BSA(用M199培养基配制),孵育30min进行封闭;4)弃封闭液,加入不同梯度的Cholesin-biotin蛋白,分别加到4组原代肝细胞中,每个实验组设置3个重复,室温孵育30min;5)孵育后,弃去培养基,用PBS洗3次,加入用1%BSA的PBS缓冲液配制的高敏性-HRP-链霉亲和素抗体(1:8000),室温孵育1h;6)弃去培养基,加入PBS缓冲液洗三次,加入200ul细胞裂解液(50mM HEPES pH7.4,150mM NaCl,1% TritonX-100)裂解细胞,取10ul用于蛋白定量,取30ul转移到96空板中加入100ul可溶性单组份TMB底物溶液置于室温避光孵育显色,显色至合适程度后加入100ul 0.1mol/L硫酸溶液终止反应,置于酶标仪中检测OD450nm吸光值,吸光值与蛋白量的比值代表特定浓度Cholesin与细胞的结合,通过Graphpad软件分析不同浓度Cholesin与原代肝细胞结合量的关系,从而得到饱和结合曲线。实验结果如图16C所示。In addition, primary culture samples of 8-10 week-old wild-type mice and Gpr146 -/- mice with a genetic background of C57BL/6J were collected. Saturation binding experiments were performed on hepatocytes. The specific process is as follows: 1) Primary hepatocytes from WT and Gpr146 -/- mice were separated and plated on 12-well plates, and 4 groups were set up: WT cells, Gpr146 -/- cells, Gpr146 -/- complemented wild-type GPR146 cells, and Gpr146 -/- complemented mutant GPR146 cells. 1) After 6 hours of cell attachment, adenovirus overexpressing wild-type and mutant GPR146 was added to Gpr146 -/- cells; 2) the purified Cholesin was labeled with a biotin labeling kit, and the concentration of biotin-labeled Cholesin was measured; 3) the culture medium of cells infected for 24 hours was discarded, 5% BSA (prepared with M199 culture medium) was added, and incubated for 30 minutes for blocking; 4) the blocking solution was discarded, different gradients of Cholesin-biotin protein were added, and added to 4 groups of primary hepatocytes, with 3 replicates for each experimental group, and incubated at room temperature for 30 minutes; 5) after incubation, the culture medium was discarded, the cells were washed 3 times with PBS, and high-sensitivity-HRP-streptavidin antibody (1:8000) prepared with 1% BSA in PBS buffer was added, and incubated at room temperature for 1 hour; 6) the culture medium was discarded, PBS buffer was added for washing three times, and 200ul of cell lysis buffer (50mM HEPES pH7.4, 150mM NaCl, 1% TritonX-100) was used to lyse the cells, 10ul was taken for protein quantification, 30ul was transferred to a 96-well plate, 100ul of soluble single-component TMB substrate solution was added, and the plate was incubated at room temperature in the dark for color development. After the color development reached an appropriate degree, 100ul of 0.1mol/L sulfuric acid solution was added to terminate the reaction, and the plate was placed in an ELISA instrument to detect the OD450nm absorbance. The ratio of the absorbance value to the protein amount represents the binding of a specific concentration of Cholesin to the cells. The relationship between the binding amount of different concentrations of Cholesin and primary hepatocytes was analyzed by Graphpad software to obtain a saturation binding curve. The experimental results are shown in Figure 16C.

图16A的结果显示,GST-Cholesin在Gpr146-/-小鼠的肌肉、肝脏、肾脏和脂肪组织中的结合信号相比野生型小鼠明显减弱。由图16B的免疫荧光结果可知,GST-Cholesin能与WT肝细胞的细胞膜结合,并且这种结合能被Cholesin-His竞争掉;Gpr146-/-细胞对GST-Cholesin的结合明显减弱,回补野生型GPR146的细胞又恢复了对GST-Cholesin的结合,而回补突变体GPR146的细胞则依然无法结合GST-Cholesin。从图16C的饱和曲线可知,Gpr146-/-细胞的饱和结合程度明显下降,回补野生型GPR146的细胞又恢复了对Cholesin的结合,而回补突变体GPR146的细胞则依然无法结合Cholesin。The results of Figure 16A show that the binding signals of GST-Cholesin in the muscle, liver, kidney and adipose tissue of Gpr146 -/ - mice are significantly weakened compared with those of wild-type mice. As shown in the immunofluorescence results of Figure 16B, GST-Cholesin can bind to the cell membrane of WT hepatocytes, and this binding can be competed out by Cholesin-His; the binding of GST-Cholesin to Gpr146 -/- cells is significantly weakened, and cells complemented with wild-type GPR146 have restored the binding to GST-Cholesin, while cells complemented with mutant GPR146 still cannot bind to GST-Cholesin. As shown in the saturation curve of Figure 16C, the saturation binding degree of Gpr146 -/- cells is significantly reduced, and cells complemented with wild-type GPR146 have restored the binding to Cholesin, while cells complemented with mutant GPR146 still cannot bind to Cholesin.

这些体外和体内结合实验结果表明,GPR146是Cholesin的受体。These in vitro and in vivo binding assay results indicate that GPR146 is a receptor for Cholesin.

5.3 Cholesin与GPR146的结合亲和力测定5.3 Determination of the binding affinity of Cholesin to GPR146

微量热泳动技术是一项基于分子热泳动特性对生物大分子间相互作用进行量化的技术,本实施例中使用该技术来测量Cholesin与野生型(WT)和突变型(Mut)GPR146的结合亲和力,具体实验步骤如下:1)受体荧光标记:我们从Hi5细胞中纯化出野生型和突变型GPR146蛋白,将其浓缩至约100ul,浓度约为0.4ug/ul,取90ul蛋白样品使用RED-NHS蛋白氨基标记试剂盒对其进行荧光标记,标记后其浓度为标记前1/5,体积扩大至450ul,对标记后的样品进行荧光信号检测,根据荧光信号强度确定MST实验使用的受体终浓度为50nM;2)将Cholesin蛋白用Superdex 200Incresae 5/15GL分子筛层析柱 置换到含DDM和CHS的HEPES-NaCl缓冲液中,用缓冲液将Cholesin稀释至6uM,而后用缓冲液对其进行1:1梯度稀释形成16个浓度梯度,取10ul 50nM标记好的野生型和突变型GPR146分别与10ul不同浓度的Cholesin充分混匀,室温静置20min;3)用Monolith NT.115毛细管吸取样品,按照Cholesin的浓度从高至低依次置于载样台上,放入微量热泳动仪器中进行测量,设定测量参数为10%LED power以及40%MST power,使用NanoTemper分析软件对实验结果进行分析计算得到Cholesin与野生型和突变型GPR146蛋白间的平衡解离常数kd值。Microthermophoresis is a technique that quantifies the interactions between biomacromolecules based on the thermophoresis properties of molecules. This technique was used in this example to measure the binding affinity of Cholesin to wild-type (WT) and mutant (Mut) GPR146. The specific experimental steps are as follows: 1) Receptor fluorescence labeling: We purified wild-type and mutant GPR146 proteins from Hi5 cells and concentrated them to about 100ul with a concentration of about 0.4ug/ul. We took 90ul of protein sample and used the RED-NHS protein amino labeling kit to fluorescently label it. After labeling, its concentration was 1/5 of that before labeling. The volume was expanded to 450ul, and the labeled sample was subjected to fluorescence signal detection. According to the intensity of the fluorescence signal, the final receptor concentration used in the MST experiment was determined to be 50nM; 2) The Cholesin protein was purified by Superdex 200Incresae 5/15GL molecular sieve chromatography column. Replace with HEPES-NaCl buffer containing DDM and CHS, dilute Cholesin to 6uM with buffer, and then dilute it with buffer in a 1:1 gradient to form 16 concentration gradients, take 10ul 50nM labeled wild-type and mutant GPR146 and mix them thoroughly with 10ul Cholesin of different concentrations, and let it stand at room temperature for 20min; 3) Use Monolith NT.115 capillary to absorb samples, place them on the sample stage in order from high to low Cholesin concentration, put them into micro-thermophoresis instrument for measurement, set the measurement parameters to 10% LED power and 40% MST power, use NanoTemper analysis software to analyze and calculate the experimental results to obtain the equilibrium dissociation constant kd value between Cholesin and wild-type and mutant GPR146 proteins.

结果如图17所示,结果显示,Cholesin与野生型GPR146具有高结合亲和力,其中Kd值为21.34±0.98nM,而Cholesin与突变型GPR146的结合减弱,Kd值为971.0±91.5nM。The results are shown in FIG17 , which show that Cholesin has a high binding affinity to wild-type GPR146, with a Kd value of 21.34±0.98 nM, while Cholesin has a weakened binding to mutant GPR146, with a Kd value of 971.0±91.5 nM.

实施例6:Cholesin通过GPR146耦联的Gαi信号通路抑制cAMP的产生Example 6: Cholesin inhibits cAMP production via the GPR146-coupled Gαi signaling pathway

6.1 Cholesin抑制GPR146与GNAI1的相互作用6.1 Cholesin inhibits the interaction between GPR146 and GNAI1

确认了Cholesin和GPR146之间的相互作用后,在小鼠原代肝细胞中进一步调查了GPR146的下游信号。GPR146是一种GPCR,与GPCR相关的G蛋白是异源三聚体,包含三个不同的亚基:α亚基,β亚基和γ亚基。配体与GPCR的结合会引起受体构象的改变,进而改变并结合并激活G蛋白。然后,G蛋白的活性形式从受体表面释放出来,解离成其α和β/γ亚基。然后,两个亚基都将激活其特定的效应子,从而释放第二个信使。这些信使被蛋白激酶识别,从而导致其活化并触发朝向细胞事件的信号级联。Gαs和Gαi亚型分别激活或灭活腺苷酸环化酶,该酶将三磷酸腺苷(ATP)转换成环状单磷酸腺苷(cAMP),在此过程中释放出无机焦磷酸盐。鸟嘌呤核苷酸结合蛋白G(i)亚单位α1(GNAI1)是G蛋白抑制性亚单位,Gαi的解离会抑制下游的cAMP信号。推测Cholesin与GPR146结合后促进Gαi的解离,从而通过抑制cAMP来抑制下游胆固醇合成。为了验证以上猜想,进行了免疫共沉淀实验。Having confirmed the interaction between Cholesin and GPR146, the downstream signaling of GPR146 was further investigated in mouse primary hepatocytes. GPR146 is a GPCR, and the G protein associated with the GPCR is a heterotrimer containing three different subunits: α, β, and γ. The binding of ligands to the GPCR causes a conformational change in the receptor, which in turn changes and binds to and activates the G protein. The active form of the G protein is then released from the receptor surface, dissociating into its α and β/γ subunits. Both subunits will then activate their specific effectors, which release the second messenger. These messengers are recognized by protein kinases, leading to their activation and triggering a signaling cascade toward cellular events. The Gαs and Gαi isoforms activate or inactivate adenylate cyclase, respectively, which converts adenosine triphosphate (ATP) into cyclic adenosine monophosphate (cAMP), releasing inorganic pyrophosphate in the process. Guanine nucleotide binding protein G(i) subunit α1 (GNAI1) is a G protein inhibitory subunit, and the dissociation of Gαi inhibits the downstream cAMP signal. It is speculated that Cholesin promotes the dissociation of Gαi after binding to GPR146, thereby inhibiting downstream cholesterol synthesis by inhibiting cAMP. In order to verify the above hypothesis, an immunoprecipitation experiment was performed.

分离野生型小鼠的原代肝细胞,将细胞贴壁后加入过表达带HA标签的GNAI1的腺病毒,随后再将细胞分为三组,分别加腺病毒空载、带Flag标签的野生型GPR146(WT)腺病毒和带Flag标签的突变型GPR146(Mut)腺病毒。腺病毒感染24小时后每组分别加入0nM和200nM的Cholesin处理10分钟,随后立即加入RIPA裂解液裂解细胞。取裂解后的40μl样品加入5×SDS上样缓冲液100℃煮样10min作为input样品,加入吸附Flag的凝珠到细胞裂解液中,置于4℃盘旋式混合器上孵育过夜。结合完成后5000rpm,4℃离心3min,吸弃上清,加入600μl预冷的RIPA裂解液涡旋混匀去除残余的液体样品以及非特异性结合在凝珠上的蛋白,离心弃上清,重复洗涤6次,充分去除残余液体, 加入40μl 1×SDS上样缓冲液100℃煮样10min,进行SDS-PAGE和免疫印迹检测。免疫共沉淀结果如图18所示。Primary hepatocytes from wild-type mice were isolated and adenovirus overexpressing GNAI1 with HA tag was added after the cells were attached to the wall. The cells were then divided into three groups, with empty adenovirus, wild-type GPR146 (WT) adenovirus with Flag tag, and mutant GPR146 (Mut) adenovirus with Flag tag added. 24 hours after adenovirus infection, each group was treated with 0nM and 200nM Cholesin for 10 minutes, and then immediately added RIPA lysis buffer to lyse the cells. 40μl of the lysed sample was added to 5×SDS loading buffer and boiled at 100℃ for 10min as the input sample, and the Flag-adsorbed beads were added to the cell lysate and incubated overnight on a 4℃ rotating mixer. After binding, centrifuge at 5000 rpm and 4°C for 3 min, discard the supernatant, add 600 μl of pre-cooled RIPA lysis buffer and vortex to remove the remaining liquid sample and proteins non-specifically bound to the beads, centrifuge and discard the supernatant, repeat washing 6 times to fully remove the residual liquid. 40 μl 1×SDS loading buffer was added and the sample was boiled at 100°C for 10 min, and then subjected to SDS-PAGE and immunoblotting. The immunoprecipitation results are shown in FIG18 .

结果显示,Cholesin处理破坏了野生型GPR146与GNAI1的相互作用,而突变型GPR146则没有这种效应,表明Cholesin破坏了GPR146与GNAI1之间的相互作用。The results showed that Cholesin treatment disrupted the interaction between wild-type GPR146 and GNAI1, while mutant GPR146 had no such effect, indicating that Cholesin disrupts the interaction between GPR146 and GNAI1.

6.2 Cholesin处理降低野生型原代肝细胞中的cAMP水平6.2 Cholesin treatment reduces cAMP levels in wild-type primary hepatocytes

分离野生型(Gpr146+/+)和Gpr146基因敲除(Gpr146-/-)小鼠的原代肝细胞并置于6孔板中。设置4组:Gpr146+/+细胞,Gpr146-/-回补空载体(Vec)细胞,Gpr146-/-回补野生型GPR146(WT)细胞和Gpr146-/-回补突变型GPR146(Mut)细胞。原代肝细胞贴壁后加入回补的野生型和突变型GPR146的腺病毒,感染24小时后每组均用0nM或200nM的Cholesin处理细胞10分钟,随后立即吸弃细胞培养基,用Cayman的Cyclic AMP ELISA Kit检测细胞中的cAMP水平,实验结果如图19所示。Primary hepatocytes from wild-type (Gpr146 +/+ ) and Gpr146 knockout (Gpr146 -/- ) mice were isolated and placed in 6-well plates. Four groups were set up: Gpr146 +/+ cells, Gpr146 -/- complemented empty vector (Vec) cells, Gpr146 -/- complemented wild-type GPR146 (WT) cells, and Gpr146 -/- complemented mutant GPR146 (Mut) cells. After primary hepatocytes adhered to the wall, adenoviruses complemented with wild-type and mutant GPR146 were added. After 24 hours of infection, each group was treated with 0nM or 200nM Cholesin for 10 minutes, and then the cell culture medium was immediately aspirated and the cAMP level in the cells was detected using Cayman's Cyclic AMP ELISA Kit. The experimental results are shown in Figure 19.

结果显示,野生型小鼠原代肝细胞在Cholesin处理后cAMP水平下降,而Gpr146-/-原代肝细胞则不再响应Cholesin的处理,回补野生型GPR146后Gpr146-/-对Cholesin的响应恢复,回补突变型GPR146则依然不响应Cholesin对cAMP的抑制作用。这表明Cholesin处理会降低原代肝细胞中的cAMP水平。The results showed that cAMP levels in wild-type mouse primary hepatocytes decreased after Cholesin treatment, while Gpr146 -/- primary hepatocytes no longer responded to Cholesin treatment. After wild-type GPR146 was supplemented, Gpr146 -/-' s response to Cholesin was restored, while supplementation of mutant GPR146 still did not respond to Cholesin's inhibitory effect on cAMP. This indicates that Cholesin treatment reduces cAMP levels in primary hepatocytes.

实施例6的上述结果说明,Cholesin通过GPR146耦联的Gαi信号通路抑制cAMP的产生。The above results of Example 6 indicate that Cholesin inhibits the production of cAMP via the GPR146-coupled Gαi signaling pathway.

实施例7:腹腔注射Cholesin降低Hmgcr mRNA水平以及血浆和肝脏胆固醇水平Example 7: Intraperitoneal injection of Cholesin reduces Hmgcr mRNA levels and plasma and liver cholesterol levels

7.1腹腔注射Cholesin对小鼠肝脏中Hmgcr mRNA水平的剂量依赖性抑制7.1 Dose-dependent inhibition of Hmgcr mRNA levels in mouse liver by intraperitoneal injection of Cholesin

本实施例选用遗传背景为C57BL/6J的10-12周龄野生型雄鼠,将小鼠随机分成4组,每组10只小鼠。小鼠禁食过夜,第二天注射0mg/kg、1mg/kg、5mg/kg和10mg/kg的真核纯化的Cholesin-his蛋白,小鼠进食6小时后收取小鼠肝脏组织。提取小鼠肝脏组织总RNA,并进行qPCR,检测Hmgcr的表达水平,以Rpl32为内参,进行统计分析。In this example, 10-12 week old wild-type male mice with a genetic background of C57BL/6J were randomly divided into 4 groups, with 10 mice in each group. The mice were fasted overnight, and injected with 0 mg/kg, 1 mg/kg, 5 mg/kg and 10 mg/kg of eukaryotic purified Cholesin-his protein on the second day. The mouse liver tissue was collected 6 hours after the mice were fed. Total RNA of mouse liver tissue was extracted, and qPCR was performed to detect the expression level of Hmgcr, and statistical analysis was performed with Rpl32 as an internal reference.

结果如图20所示,注射外源Cholesin能抑制肝脏Hmgcr的表达,并且这种抑制作用呈现剂量依赖性。The results are shown in Figure 20. Injection of exogenous Cholesin can inhibit the expression of liver Hmgcr, and this inhibitory effect is dose-dependent.

7.2腹腔注射Cholesin对小鼠肝脏Hmgcr mRNA水平以及血浆和肝脏TC含量的影响7.2 Effects of intraperitoneal injection of Cholesin on Hmgcr mRNA levels in mouse liver and TC content in plasma and liver

选用遗传背景为C57BL/6J的8周龄野生型雄鼠,分为两组,分别注射0mg/kg和5mg/kg的真核纯化的Cholesin-his蛋白。每周注射两次,共注射两周。在小鼠10周龄大时对小鼠饥饿再进食6小时后收取肝脏组织和全血,操作流程如图21A所示。对于小鼠 肝脏,提取小鼠肝脏组织总RNA进行qPCR以检测Hmgcr的表达水平;同时还提取肝脏总胆固醇(TC)以检测提取液中TC的水平。对于小鼠全血,进行低速离心分离血浆,检测血浆中TC的水平。小鼠肝脏中的Hmgcr mRNA水平、小鼠血浆和肝脏的TC水平分别如图21B、21C和21D所示。Eight-week-old wild-type male mice with a genetic background of C57BL/6J were selected and divided into two groups. They were injected with 0 mg/kg and 5 mg/kg of eukaryotic purified Cholesin-his protein, respectively. The injections were given twice a week for two weeks. When the mice were 10 weeks old, the liver tissue and whole blood were collected after the mice were starved and fed for 6 hours. The operation process is shown in Figure 21A. Liver, total RNA of mouse liver tissue was extracted for qPCR to detect the expression level of Hmgcr; total cholesterol (TC) of the liver was also extracted to detect the level of TC in the extract. For whole blood of mice, plasma was separated by low-speed centrifugation, and the level of TC in plasma was detected. The Hmgcr mRNA level in mouse liver, the TC level in mouse plasma and liver are shown in Figures 21B, 21C and 21D, respectively.

结果显示,注射Cholesin能抑制小鼠肝脏Hmgcr的表达(图21B),注射Cholesin后小鼠血浆TC水平(图21C)和肝脏TC水平(图21D)下降。The results showed that the injection of Cholesin could inhibit the expression of Hmgcr in mouse liver ( FIG. 21B ). The plasma TC level ( FIG. 21C ) and liver TC level ( FIG. 21D ) of mice decreased after the injection of Cholesin.

实施例8:Cholesin通过GPR146抑制胆固醇合成Example 8: Cholesin inhibits cholesterol synthesis via GPR146

为了探究Cholesin是否通过GPR146抑制小鼠中的胆固醇合成,选用遗传背景为C57BL/6J的10周龄野生型(Gpr146fl/fl)小鼠和Gpr146基因敲除(Gpr146 LKO)小鼠进行实验。将Gpr146 LKO小鼠在10-12周时分成3组,每组12只,一组尾静脉注射腺病毒对照(Vec),一组尾静脉注射表达野生型GPR146(WT)的腺病毒,另一组注射表达突变型GPR146(Mut)的腺病毒。每只小鼠通过尾静脉注射腺病毒的病毒量总计为5×10^8。随后将每组再分为两小组,每小组6只,每周注射两次0mg/kg或5mg/kg Cholesin,连续注射4次,在小鼠14.5周时对小鼠饥饿再进食6小时后收取肝脏组织和全血,操作流程如图22A所示。In order to explore whether Cholesin inhibits cholesterol synthesis in mice through GPR146, 10-week-old wild-type (Gpr146 fl/fl ) mice and Gpr146 knockout (Gpr146 LKO) mice with a genetic background of C57BL/6J were selected for the experiment. Gpr146 LKO mice were divided into 3 groups at 10-12 weeks, with 12 mice in each group. One group was injected with adenovirus control (Vec) through the tail vein, one group was injected with adenovirus expressing wild-type GPR146 (WT), and the other group was injected with adenovirus expressing mutant GPR146 (Mut). The total amount of adenovirus injected into each mouse through the tail vein was 5×10^8. Each group was then divided into two small groups, with 6 mice in each group, and 0 mg/kg or 5 mg/kg Cholesin was injected twice a week for 4 consecutive times. The liver tissue and whole blood were collected after the mice were starved and fed for 6 hours at 14.5 weeks. The operation process is shown in Figure 22A.

对于小鼠肝脏,提取肝脏总RNA后进行qPCR,检测胆固醇合成相关基因Hmgcs1、Hmgcr、Mvd、Mvk和Pmvk的表达水平,结果如图22B所示。同时提取肝脏总蛋白用SREBP2、HMGCR、HMGCS1、LDLR、pKA Sub和HSP90的抗体进行免疫印迹检测,以HSP90为内参,结果如图22C所示。还提取肝脏总胆固醇(TC)以检测提取液中TC的水平,结果如图22D所示。对于小鼠全血,进行低速离心分离血浆,检测血浆中TC的水平,结果如图22E所示。For mouse liver, total liver RNA was extracted and then qPCR was performed to detect the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk and Pmvk, and the results are shown in Figure 22B. At the same time, total liver protein was extracted and immunoblotted with antibodies to SREBP2, HMGCR, HMGCS1, LDLR, pKA Sub and HSP90, with HSP90 as the internal reference, and the results are shown in Figure 22C. Total cholesterol (TC) in the liver was also extracted to detect the level of TC in the extract, and the results are shown in Figure 22D. For mouse whole blood, low-speed centrifugation was performed to separate plasma, and the level of TC in plasma was detected, and the results are shown in Figure 22E.

由图22B和22C的结果可知,Cholesin在野生型小鼠中抑制PKA和SREBP2的表达,从而降低胆固醇合成相关基因的表达并降低肝脏和血浆TC水平。然而,在GPR146敲除小鼠中,Cholesin的这种作用被消除,添加野生型GPR146后Cholesin的作用恢复,添加突变型GPR146后,Cholesin的作用依旧无法恢复。As shown in the results of Figures 22B and 22C, Cholesin inhibits the expression of PKA and SREBP2 in wild-type mice, thereby reducing the expression of cholesterol synthesis-related genes and reducing liver and plasma TC levels. However, in GPR146 knockout mice, this effect of Cholesin was eliminated, and the effect of Cholesin was restored after adding wild-type GPR146, but the effect of Cholesin could not be restored after adding mutant GPR146.

这些结果表明,Cholesin通过GPR146抑制肝脏SREBP2的表达,从而调节胆固醇稳态。These results suggest that Cholesin regulates cholesterol homeostasis by inhibiting hepatic SREBP2 expression through GPR146.

实施例9:Cholesin能够预防高胆固醇血症和动脉粥样硬化Example 9: Cholesin can prevent hypercholesterolemia and atherosclerosis

Ldlr-/-(Ldlr基因敲除)小鼠是目前常用的研究动脉粥样硬化的疾病模型,Ldlr-/-小鼠 的血浆胆固醇显著高于野生型小鼠,并在西方饮食(WD)诱导下会形成动脉硬化斑块。先对5周龄的Ldlr-/-小鼠进行4周的西方饮食喂养,随后将小鼠分为四组,每组6-7只小鼠。第一组为Cholesin单独腹腔注射给药(5mg kg-1,每周两次),;第二组为瑞舒伐他汀(Ros)单独饮水给药(10mg kg-1-1);第三组为Cholesin(5mg kg-1,每周两次)与瑞舒伐他汀(10mg kg-1-1)联合(Cholesin+Ros)给药处理;第四组为载剂对照组(Veh),具体操作如图23所示。期间每两周对小鼠进行尾静脉采血以检测血浆总胆固醇(TC)和甘油三酯(TG)水平,并检测小鼠体重和进食情况,同时还检测给药8周后小鼠血浆中的丙氨酸氨基转移酶(ALT)和天冬氨酸氨基转移酶(AST)水平。持续给药8周后处死小鼠,收取肝脏组织以检测肝脏TC和TG水平、肝脏中胆固醇合成相关基因的表达水平,并对肝脏组织进行HE染色以观察小鼠肝脏脂质堆积情况。另外,还对给药8周后的小鼠进行全主动脉油红染色以观察动脉粥样硬化斑块的变化情况。Ldlr -/- (Ldlr gene knockout) mice are currently a commonly used disease model for studying atherosclerosis . The plasma cholesterol of Ldlr-/- mice was significantly higher than that of wild-type mice, and atherosclerotic plaques were formed under the induction of Western diet (WD). Five-week-old Ldlr -/- mice were first fed a Western diet for 4 weeks, and then the mice were divided into four groups, with 6-7 mice in each group. The first group was administered with Cholesin alone by intraperitoneal injection (5 mg kg -1 , twice a week); the second group was administered with rosuvastatin (Ros) alone by drinking water (10 mg kg - 1day -1 ); the third group was administered with Cholesin (5 mg kg -1 , twice a week) and rosuvastatin (10 mg kg - 1day -1 ) in combination (Cholesin+Ros); the fourth group was the vehicle control group (Veh), and the specific operation is shown in Figure 23. During the period, the tail vein blood of mice was collected every two weeks to detect the plasma total cholesterol (TC) and triglyceride (TG) levels, and the weight and food intake of mice were detected. At the same time, the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in the mouse plasma were also detected after 8 weeks of administration. After 8 weeks of continuous administration, the mice were killed, and the liver tissues were collected to detect the liver TC and TG levels, the expression levels of cholesterol synthesis-related genes in the liver, and the liver tissues were stained with HE to observe the lipid accumulation in the mouse liver. In addition, the whole aorta of the mice after 8 weeks of administration was stained with oil red to observe the changes in atherosclerotic plaques.

小鼠血浆和肝脏TC水平以及TG水平Plasma and liver TC and TG levels in mice

小鼠血浆TC水平变化如图24A所示。小鼠在单独给药瑞舒伐他汀或Cholesin均能显著降低Ldlr-/-鼠血浆中的TC水平,其中瑞舒伐他汀在给药4周后在药物剂量不变的情况下能观察到血浆TC水平略有回升,这表明瑞舒伐他汀给药后期存在耐药性问题。瑞舒伐他汀和Cholesin联合给药对血浆TC的降低效果最好,优于单独的瑞舒伐他汀或Cholesin给药,联合给药能在给药前期达到较好的抑制作用,并持续维持抑制效果较长时间。对血浆TC在8周内的变化进行曲线下面积(AUC)统计,以便更加直观地比较三种治疗方案的效果,其中Cholesin和瑞舒伐他汀的单独处理均能显著降低血浆中TC水平,二者联合处理的抑制效果更为突出,实现了协同效果。The changes in mouse plasma TC levels are shown in Figure 24A. The mice were able to significantly reduce the TC levels in the plasma of Ldlr -/- mice when they were given rosuvastatin or Cholesin alone. Among them, rosuvastatin was observed to slightly increase the plasma TC level when the drug dosage remained unchanged after 4 weeks of administration, indicating that there was a drug resistance problem in the later stage of rosuvastatin administration. The combined administration of rosuvastatin and Cholesin had the best effect on reducing plasma TC, which was better than the single administration of rosuvastatin or Cholesin. The combined administration could achieve a good inhibitory effect in the early stage of administration and maintain the inhibitory effect for a long time. The area under the curve (AUC) of the changes in plasma TC within 8 weeks was statistically analyzed in order to more intuitively compare the effects of the three treatment regimens. Among them, the single treatment of Cholesin and rosuvastatin could significantly reduce the TC level in plasma, and the inhibitory effect of the combined treatment of the two was more prominent, achieving a synergistic effect.

给药8周后小鼠肝脏中的TC水平如图24B所示。小鼠给药瑞舒伐他汀或Cholesin后肝脏的TC水平均显著降低,瑞舒伐他汀和Cholesin联合给药对肝脏TC的降低效果最好,优于单独的瑞舒伐他汀或Cholesin给药,实现了协同效果。The TC level in the liver of mice after 8 weeks of administration is shown in Figure 24B. The TC level in the liver of mice was significantly reduced after administration of rosuvastatin or Cholesin. The combined administration of rosuvastatin and Cholesin had the best effect on reducing liver TC, which was better than the single administration of rosuvastatin or Cholesin, achieving a synergistic effect.

小鼠血浆和肝脏中的TG水平如图24C和24D所示。单独给药Cholesin和瑞舒伐他汀均能降低血浆或肝脏中的TG水平,而当二者联合使用对血浆或肝脏中的TG的抑制作用最为明显,说明二者联合实现了协同效果。The TG levels in mouse plasma and liver are shown in Figures 24C and 24D. Cholesin and rosuvastatin alone can reduce the TG levels in plasma or liver, and the combined use of the two has the most significant inhibitory effect on TG in plasma or liver, indicating that the combination of the two achieves a synergistic effect.

小鼠肝脏中胆固醇合成相关基因的表达水平Expression levels of cholesterol synthesis-related genes in mouse liver

总胆固醇的减少可能是由于胆固醇的合成被抑制,因此通过qPCR和免疫印迹对小鼠肝脏中胆固醇合成相关基因Hmgcs1、Hmgcr、Mvd、Mvk、Pmvk和Sqle以及调控这些基因表达的转录因子SREBP2的表达水平进行了检测。结果如图25A和25B所示。The reduction in total cholesterol may be due to the inhibition of cholesterol synthesis, so the expression levels of cholesterol synthesis-related genes Hmgcs1, Hmgcr, Mvd, Mvk, Pmvk and Sqle in mouse liver and the transcription factor SREBP2 that regulates the expression of these genes were detected by qPCR and immunoblotting. The results are shown in Figures 25A and 25B.

结果显示,给药8周后瑞舒伐他汀处理的小鼠肝脏中胆固醇合成相关基因及转录因 子SREBP2的表达均明显上调,而注射Cholesin能显著抑制胆固醇合成相关基因及转录因子SREBP2的表达,并在Cholesin和瑞舒伐他汀联合使用时显著缓解他汀药带来的胆固醇合成基因上调的情况,将胆固醇合成相关基因的表达水平抑制到较低水平。The results showed that after 8 weeks of administration, the cholesterol synthesis-related genes and transcription factors in the liver of rosuvastatin-treated mice were significantly upregulated. The expression of SREBP2 was significantly upregulated in both groups, while the injection of Cholesin could significantly inhibit the expression of cholesterol synthesis-related genes and transcription factor SREBP2. When Cholesin and rosuvastatin were used in combination, the upregulation of cholesterol synthesis genes caused by statins was significantly alleviated, and the expression level of cholesterol synthesis-related genes was suppressed to a lower level.

小鼠全主动脉油红染色Oil red staining of whole mouse aorta

为了观察小鼠动脉的整体病变情况,对小鼠全主动脉进行油红染色。油红O是一种强染脂剂,能特异性将中性甘油三酯、脂质及脂蛋白等染成红色。由于动脉粥样硬化斑块中富含脂质,因此斑块部分能被染成红色,统计染色后的斑块面积占整个主动脉的比例从而评估病变情况。In order to observe the overall pathological changes of mouse arteries, the whole aorta of mice was stained with Oil Red O. Oil Red O is a strong lipid staining agent that can specifically stain neutral triglycerides, lipids and lipoproteins red. Since atherosclerotic plaques are rich in lipids, the plaque part can be stained red. The proportion of the stained plaque area to the entire aorta is calculated to evaluate the pathological condition.

具体实验步骤如下:1)麻醉小鼠,将小鼠仰卧固定在泡沫板上,开胸腔暴露心脏,注射器吸取20ml PBS,注射器针头自心尖插入左心室,在右心房剪开一小口,手推PBS进行全身灌流,可见灌流后肝脏颜色由红变黄。留置针头,注射器吸取10ml 4%多聚甲醛/PBS继续灌流,灌入固定液后可见鼠尾轻微摆动,提示灌流固定效果较好;2)移除小鼠内脏,暴露脊柱,在体式显微镜下小心剥离心脏和全主动脉,主动脉剪至髂动脉分叉以下,在靠近心脏位置将升主动脉剪断,全主动脉剥离后还需仔细剔除血管外膜上的脂肪组织,主要采用钝性剥离,避免损伤血管,处理好的全主动脉保存在4%多聚甲醛/PBS中等待染色;3)取出充分固定的主动脉,在体式显微镜下用Vannas剪自升主动脉断口处伸入血管,沿主动脉弓大弯侧向远端将主动脉弓纵向剖开,保留三个分支不做处理,将剖好的主动脉内膜朝上展开,用最小号昆虫针固定在黑底胶板上;4)配置油红O染液:称取油红O粉末0.5g,加入异丙醇100ml,90℃水浴1小时,过滤后得到饱和油红染液,使用时取饱和液和双蒸水按3:2混合,42℃水浴10分钟后再过滤一次,即为油红工作液,饱和液和工作液均需低温避光保存;5)染色时先将样本在60%异丙醇中预处理10分钟,随后在油红工作液中染色15分钟,染色结束后在60%异丙醇中脱色5分钟降低背景,染色后的样本可长期保存在固定液中;6)随后,使用Zeiss Stemi 508立体显微镜拍摄染色后的主动脉图像。使用Image J软件对胸主动脉的病变区域进行量化。用ImageJ测量斑块面积和整个主动脉内膜面积,再用斑块面积除以整个内膜面积×100%,即为斑块相对百分比。除以内膜面积主要是为了消除小鼠体型差异产生的影响。The specific experimental steps are as follows: 1) Anesthetize the mouse, fix it in supine position on a foam board, open the chest cavity to expose the heart, draw 20 ml of PBS into the syringe, insert the syringe needle into the left ventricle from the apex of the heart, cut a small hole in the right atrium, and manually push PBS for systemic perfusion. It can be seen that the color of the liver changes from red to yellow after perfusion. Leave the needle in place and draw 10 ml of 4% paraformaldehyde/PBS into the syringe to continue perfusion. After the fixative is injected, the mouse tail can be seen swinging slightly, indicating that the perfusion fixation effect is good; 2) Remove the mouse viscera, expose the spine, carefully peel off the heart and the entire aorta under a stereo microscope, cut the aorta below the bifurcation of the iliac artery, and cut the ascending aorta near the heart. After the entire aorta is peeled off, the fat tissue on the adventitia of the blood vessels must be carefully removed. Blunt peeling is mainly used to avoid damaging the blood vessels. The processed whole aorta is stored in 4% paraformaldehyde/PBS and wait for staining; 3) Take out the fully fixed aorta, use Vannas scissors to extend into the blood vessels from the fracture of the ascending aorta under a stereo microscope, and longitudinally dissect the aortic arch along the greater curvature of the aortic arch to the distal end, leaving the three branches untreated. The good aortic intima was unfolded upwards and fixed on a black-bottomed plastic plate with the smallest insect pin; 4) Prepare Oil Red O dye solution: weigh 0.5g of Oil Red O powder, add 100ml of isopropanol, and place in a water bath at 90℃ for 1 hour. After filtering, a saturated Oil Red dye solution was obtained. When using, the saturated solution and double distilled water were mixed in a ratio of 3:2, and then filtered again after a water bath at 42℃ for 10 minutes. This is the Oil Red working solution. Both the saturated solution and the working solution need to be stored at low temperature and away from light; 5) When staining, the sample was first pretreated in 60% isopropanol for 10 minutes, and then stained in the Oil Red working solution for 15 minutes. After staining, the sample was decolorized in 60% isopropanol for 5 minutes to reduce the background. The stained sample can be stored in the fixative for a long time; 6) Then, a Zeiss Stemi 508 stereo microscope was used to take images of the stained aorta. Image J software was used to quantify the lesion area of the thoracic aorta. ImageJ was used to measure the plaque area and the entire aortic intima area, and then the plaque area was divided by the entire intima area × 100% to obtain the relative percentage of plaques. Dividing by the intima area was mainly to eliminate the effects of differences in mouse body size.

实验结果如图26所示,载剂对照组中可见明显斑块,瑞舒伐他汀和Cholesin单独给药均能有效抑制动脉病变,而二者联用可以取得更好的治疗效果,实现了协同效果。The experimental results are shown in FIG26 . Obvious plaques can be seen in the vehicle control group. Rosuvastatin and Cholesin can both effectively inhibit arterial lesions when administered alone, and their combined use can achieve better therapeutic effects and realize a synergistic effect.

小鼠体重和进食情况Mouse body weight and food intake

给药过程中的小鼠体重变化和采食量变化分别如图27A和27B所示。结果显示,瑞舒伐他汀组和对照组之间体重没有显著性差异,而Cholesin单独处理和Cholesin与瑞舒 伐他汀联合处理均能抑制小鼠体重增长的幅度(图27A)。此外,在给药过程中小鼠的采食量不受影响,四组之间没有明显差异(图27B),这也表明小鼠的体重变化不是由于小鼠的采食量差异引起的。The changes in body weight and food intake of mice during the administration process are shown in Figures 27A and 27B, respectively. The results showed that there was no significant difference in body weight between the rosuvastatin group and the control group, while Cholesin alone and Cholesin combined with rosuvastatin Vastatin combined treatment can inhibit the increase of mouse body weight (Figure 27A). In addition, the food intake of mice was not affected during the administration process, and there was no significant difference among the four groups (Figure 27B), which also indicates that the weight change of mice is not caused by the difference in food intake of mice.

小鼠肝脏组织HE染色HE staining of mouse liver tissue

通过对肝脏组织进行HE染色来观察肝脏脂质堆积情况,具体操作如下:1)取下一块小鼠肝脏组织放入4%多聚甲醛中固定过夜;2)由低浓度到高浓度酒精作脱水剂,逐渐脱去组织块中的水份,再将组织块置于二甲苯中透明,以二甲苯替换出组织块的中酒精,浸蜡包埋;3)将已透明的组织块置于已溶化的石蜡中,放入溶蜡箱保温,待石蜡完全浸入组织块后进行包埋;4)将包埋好的蜡块固定于切片机上,切成薄片,一般为5μm,贴片后放45℃恒温箱中烘干;5)依次将切片放入二甲苯Ⅰ10min-二甲苯Ⅱ10min-无水乙醇Ⅰ 5min-无水乙醇Ⅱ 5min-95%酒精5min-90%酒精5min-80%酒精5min-70%酒精5min-蒸馏水洗;6)切片入Harris苏木素染3-8min,自来水洗,1%的盐酸酒精分化数秒,自来水冲洗,0.6%氨水返蓝,流水冲洗;7)切片入伊红染液中染色1-3min;8)将切片依次放入95%酒精I 5min-95%酒精II 5min-无水乙醇Ⅰ 5min-无水乙醇II 5min-二甲苯Ⅰ 5min-二甲苯Ⅱ 5min中脱水透明,将切片从二甲苯拿出来稍晾干,中性树胶封片;9)显微镜镜检,图像采集分析。The accumulation of lipids in the liver was observed by HE staining of liver tissue. The specific operation was as follows: 1) A piece of mouse liver tissue was taken and fixed in 4% paraformaldehyde overnight; 2) Alcohol was used as a dehydrating agent from low concentration to high concentration to gradually remove the water in the tissue block, and then the tissue block was placed in xylene to make it transparent, and the alcohol in the tissue block was replaced with xylene, and then immersed in wax and embedded; 3) The transparent tissue block was placed in the melted paraffin, placed in a wax melting box for insulation, and embedded after the paraffin was completely immersed in the tissue block; 4) The embedded wax block was fixed on a microtome and cut into thin slices, generally 5μm, and placed in a 45℃ constant temperature box for drying after pasting; 5) The slices were placed in xylene I for 10min-xylene II for 10min-anhydrous ethanol I for 5min-anhydrous ethanol I for 5min Ethanol II 5min-95% alcohol 5min-90% alcohol 5min-80% alcohol 5min-70% alcohol 5min-wash with distilled water; 6) Stain the slices with Harris hematoxylin for 3-8min, wash with tap water, differentiate with 1% hydrochloric acid alcohol for a few seconds, rinse with tap water, blue with 0.6% ammonia water, and rinse with running water; 7) Stain the slices with eosin staining solution for 1-3min; 8) Dehydrate and make transparent the slices in 95% alcohol I 5min-95% alcohol II 5min-anhydrous ethanol I 5min-anhydrous ethanol II 5min-xylene I 5min-xylene II 5min, take the slices out of xylene and dry them slightly, and seal them with neutral gum; 9) Examine under a microscope, collect and analyze images.

HE染色结果如图28所示,细胞内的白色区域即为脂质堆积,可以观察到对照组胞内白色脂滴最多,实验组均明显减弱,表明Cholesin给药后的小鼠肝脏脂质堆积明显减少。The HE staining results are shown in Figure 28. The white area in the cells is lipid accumulation. It can be observed that the control group has the most white lipid droplets in the cells, while the experimental groups have significantly weakened them, indicating that the lipid accumulation in the liver of mice after Cholesin administration is significantly reduced.

小鼠血浆中的ALT和AST水平ALT and AST levels in mouse plasma

为评估给药后小鼠的肝脏损伤,用丙氨酸氨基转移酶(谷丙转氨酶/ALT/GPT)试剂盒(南京建成)和天冬氨酸氨基转移酶(谷草转氨酶/AST/GOT)试剂盒(南京建成)检测4组小鼠血浆中的AST和ALT水平,结果如图29A和29B所示。To evaluate the liver damage in mice after administration, the AST and ALT levels in the plasma of the four groups of mice were detected using an alanine aminotransferase (alanine aminotransferase/ALT/GPT) kit (Nanjing Jiancheng) and an aspartate aminotransferase (aspartate aminotransferase/AST/GOT) kit (Nanjing Jiancheng). The results are shown in Figures 29A and 29B.

结果显示,Cholesin单独给药或与瑞舒伐他汀联合给药后AST和ALT水平均显著降低,表明Cholesin对肝脏损伤具有缓解作用。The results showed that AST and ALT levels were significantly reduced after Cholesin was administered alone or in combination with rosuvastatin, indicating that Cholesin has an alleviating effect on liver damage.

实施例9的上述结果表明,无论是单独使用Cholesin还是Cholesin与瑞舒伐他汀联合使用,均对高胆固醇血症和动脉粥样硬化具有显著的保护作用。The above results of Example 9 indicate that Cholesin, whether used alone or in combination with rosuvastatin, has a significant protective effect on hypercholesterolemia and atherosclerosis.

本申请参考了各种发行的专利、公开的专利申请、期刊文章和其他出版物,将所有这些引入本申请作为参考。若任何引入的参考文献和本说明书有冲突,则以本说明书为准。此外,落入现有技术范围的本发明的任何具体实施方案可以明确地从任何一个或多个权 利要求中排除。因为所述实施方案被认为是本领域技术人员已知的,它们可以被排除,即使所述排除没有在本申请中明确列出。本发明的任何具体实施方案可从任何权利要求中以任何理由排除,不管是否与现有技术的存在有关。This application refers to various issued patents, published patent applications, journal articles, and other publications, all of which are incorporated herein by reference. If any incorporated reference conflicts with this specification, the present specification shall prevail. In addition, any specific embodiment of the present invention that falls within the scope of the prior art may be specifically identified from any one or more of the claims. Because the embodiments are considered to be known to those skilled in the art, they may be excluded even if the exclusion is not explicitly listed in the present application. Any specific embodiment of the present invention may be excluded from any claim for any reason, whether or not related to the existence of prior art.

虽然已经参考其特定实施方案描述本发明,本领域的技术人员应当理解可以进行各种改变且可以替换等同物而不脱离本发明的真正的精神和范围。另外,可作出许多修改以使特定的情况,材料,组合物,方法,方法步骤适于本发明的目的,精神和范围。所有这些修改都旨在权利要求的范围内。 Although the present invention has been described with reference to specific embodiments thereof, it will be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the present invention. In addition, many modifications may be made to adapt specific situations, materials, compositions, methods, process steps to the purpose, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims.

Claims (33)

一种降低受试者中的血浆胆固醇水平和/或甘油三酯水平的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。A method for reducing plasma cholesterol level and/or triglyceride level in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein. 根据权利要求1所述的方法,其中所述血浆胆固醇选自血浆总胆固醇和血浆LDL-胆固醇。The method according to claim 1, wherein the plasma cholesterol is selected from the group consisting of plasma total cholesterol and plasma LDL-cholesterol. 一种抑制受试者中的肝脏胆固醇合成的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。A method for inhibiting liver cholesterol synthesis in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein. 根据权利要求3所述的方法,所述方法抑制与胆固醇合成或摄取相关基因的表达,并且所述基因选自Hmgcr、Hmgcs1、Mvd、Mvk、Pmvk、Sqle和Ldlr。The method according to claim 3, wherein the method inhibits the expression of genes related to cholesterol synthesis or uptake, and the genes are selected from Hmgcr, Hmgcs1, Mvd, Mvk, Pmvk, Sqle and Ldlr. 根据权利要求3或4所述的方法,所述方法抑制促进胆固醇合成或摄取相关基因表达的转录因子的表达,并且所述转录因子为SREBP2。The method according to claim 3 or 4, wherein the method inhibits the expression of a transcription factor that promotes the expression of a gene related to cholesterol synthesis or uptake, and the transcription factor is SREBP2. 一种抑制受试者中的肝脏VLDL-胆固醇分泌的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。A method for inhibiting hepatic VLDL-cholesterol secretion in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein. 一种治疗或预防受试者中与血浆胆固醇水平升高相关的疾病的方法,其包括向所述受试者施用有效量的Cholesin蛋白、编码所述Cholesin蛋白的核酸、或包含所述核酸的表达载体的步骤,或增强所述受试者的内源性Cholesin蛋白的表达或活性的步骤。A method for treating or preventing a disease associated with elevated plasma cholesterol levels in a subject, comprising the step of administering to the subject an effective amount of a Cholesin protein, a nucleic acid encoding the Cholesin protein, or an expression vector comprising the nucleic acid, or the step of enhancing the expression or activity of the subject's endogenous Cholesin protein. 根据权利要求1-7中任一项所述的方法,其中所述Cholesin蛋白是人Cholesin蛋白。The method according to any one of claims 1 to 7, wherein the Cholesin protein is human Cholesin protein. 根据权利要求1-8中任一项所述的方法,其中所述核酸是DNA或RNA。 The method according to any one of claims 1 to 8, wherein the nucleic acid is DNA or RNA. 根据权利要求1-9中任一项所述的方法,其中所述表达载体选自慢病毒载体、腺病毒载体、腺相关病毒(AAV)载体、逆转录病毒载体、质粒、DNA载体、mRNA载体、基于转座子的载体和人工染色体。The method according to any one of claims 1 to 9, wherein the expression vector is selected from a lentiviral vector, an adenoviral vector, an adeno-associated virus (AAV) vector, a retroviral vector, a plasmid, a DNA vector, an mRNA vector, a transposon-based vector and an artificial chromosome. 根据权利要求7-10中任一项所述的方法,其中所述与血浆胆固醇水平升高相关的疾病选自高脂血症(例如,高胆固醇血症、高甘油三酯血症)、动脉粥样硬化、心血管疾病(例如,心肌梗死、冠心病)、脑血管疾病(例如,脑血栓、脑出血、脑梗死)、高血糖症、糖尿病、肥胖症、高血压、脂肪肝、肝硬化、肾病综合征和胆结石。The method according to any one of claims 7 to 10, wherein the disease associated with elevated plasma cholesterol levels is selected from hyperlipidemia (e.g., hypercholesterolemia, hypertriglyceridemia), atherosclerosis, cardiovascular disease (e.g., myocardial infarction, coronary heart disease), cerebrovascular disease (e.g., cerebral thrombosis, cerebral hemorrhage, cerebral infarction), hyperglycemia, diabetes, obesity, hypertension, fatty liver, cirrhosis, nephrotic syndrome and gallstones. 根据权利要求11所述的方法,其中所述与血浆胆固醇水平升高相关的疾病为高胆固醇血症。The method according to claim 11, wherein the disease associated with elevated plasma cholesterol levels is hypercholesterolemia. 根据权利要求11所述的方法,其中所述与血浆胆固醇水平升高相关的疾病为动脉粥样硬化。The method according to claim 11, wherein the disease associated with elevated plasma cholesterol levels is atherosclerosis. 根据权利要求1-13中任一项所述的方法,其中所述方法还包括施用第二治疗剂的步骤,优选地,所述第二治疗剂选自蛋白质或肽、核酸和小分子药物。The method according to any one of claims 1 to 13, wherein the method further comprises the step of administering a second therapeutic agent, preferably, the second therapeutic agent is selected from proteins or peptides, nucleic acids and small molecule drugs. 根据权利要求14所述的方法,其中所述第二治疗剂为选自以下的降胆固醇药物:HMGCR抑制剂、PCSK9抑制剂和胆固醇吸收抑制剂。The method of claim 14, wherein the second therapeutic agent is a cholesterol-lowering drug selected from the group consisting of an HMGCR inhibitor, a PCSK9 inhibitor, and a cholesterol absorption inhibitor. 根据权利要求15所述的方法,其中所述HMGCR抑制剂为他汀类药物。The method of claim 15, wherein the HMGCR inhibitor is a statin. 根据权利要求16所述的方法,其中所述他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀,优选瑞舒伐他汀。The method according to claim 16, wherein the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin. 根据权利要求15所述的方法,其中所述PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。The method according to claim 15, wherein the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran and Tafolecimab. 根据权利要求15所述的方法,其中所述胆固醇吸收抑制剂为NPC1L1抑制剂,优 选依折麦布或海博麦布。The method according to claim 15, wherein the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably Choose ezetimibe or hebomibe. 根据权利要求14-19中任一项所述的方法,其中所述第二治疗剂在所述Cholesin蛋白、所述核酸或所述表达载体之前、之后或同时施用。The method according to any one of claims 14-19, wherein the second therapeutic agent is administered before, after or simultaneously with the Cholesin protein, the nucleic acid or the expression vector. 根据权利要求1-20中任一项所述的方法,其中所述受试者为人。The method according to any one of claims 1-20, wherein the subject is a human. 根据权利要求1-14和20-21中任一项所述的方法,其中所述受试者对胆固醇吸收抑制剂、HMGCR抑制剂和PCSK9抑制剂中的一种或多种无应答或不耐受,例如所述受试者具有LDL受体(LDLR)缺陷。The method according to any one of claims 1-14 and 20-21, wherein the subject is unresponsive or intolerant to one or more of a cholesterol absorption inhibitor, an HMGCR inhibitor, and a PCSK9 inhibitor, e.g., the subject has an LDL receptor (LDLR) defect. 组合物,其包含Cholesin蛋白、编码所述Cholesin蛋白的核酸、包含所述核酸的表达载体、或增强人内源性Cholesin蛋白的表达或活性的试剂,和药学上可接受的载剂或赋形剂。The composition comprises a Cholesin protein, a nucleic acid encoding the Cholesin protein, an expression vector comprising the nucleic acid, or an agent for enhancing the expression or activity of human endogenous Cholesin protein, and a pharmaceutically acceptable carrier or excipient. 根据权利要求23所述的组合物,其中所述组合物用于以下中的一种或多种:The composition according to claim 23, wherein the composition is used for one or more of the following: 1)降低受试者中的血浆胆固醇水平;1) reducing plasma cholesterol levels in a subject; 2)降低受试者中的血浆总胆固醇或血浆LDL-胆固醇水平;2) reducing plasma total cholesterol or plasma LDL-cholesterol levels in a subject; 3)降低受试者中的血浆甘油三酯水平;3) reducing plasma triglyceride levels in a subject; 4)抑制受试者中的肝脏胆固醇合成;4) inhibiting hepatic cholesterol synthesis in a subject; 5)抑制受试者中的肝脏VLDL-胆固醇分泌;和5) inhibiting hepatic VLDL-cholesterol secretion in a subject; and 6)治疗或预防受试者中与血浆胆固醇水平升高相关的疾病。6) Treating or preventing a disease associated with elevated plasma cholesterol levels in a subject. 根据权利要求23或24所述的组合物,其中所述Cholesin蛋白是人Cholesin蛋白。The composition according to claim 23 or 24, wherein the Cholesin protein is human Cholesin protein. 根据权利要求23-25中任一项所述的组合物,其中所述组合物还包含第二治疗剂,优选地,所述第二治疗剂选自蛋白质或肽、核酸和小分子药物。The composition according to any one of claims 23-25, wherein the composition further comprises a second therapeutic agent, preferably, the second therapeutic agent is selected from proteins or peptides, nucleic acids and small molecule drugs. 根据权利要求26所述的组合物,其中所述第二治疗剂选自降胆固醇药物。The composition of claim 26, wherein the second therapeutic agent is selected from cholesterol-lowering drugs. 根据权利要求27所述的组合物,其中所述降胆固醇药物选自HMGCR抑制剂、 PCSK9抑制剂和胆固醇吸收抑制剂。The composition according to claim 27, wherein the cholesterol-lowering drug is selected from HMGCR inhibitors, PCSK9 inhibitors and cholesterol absorption inhibitors. 根据权利要求28所述的组合物,其中所述HMGCR抑制剂为他汀类药物。The composition of claim 28, wherein the HMGCR inhibitor is a statin. 根据权利要求29所述的组合物,其中所述他汀类药物选自瑞舒伐他汀、洛伐他汀、辛伐他汀、阿托伐他汀、普伐他汀、氟伐他汀、西立伐他汀和匹伐他汀,优选瑞舒伐他汀。The composition according to claim 29, wherein the statin is selected from rosuvastatin, lovastatin, simvastatin, atorvastatin, pravastatin, fluvastatin, cerivastatin and pitavastatin, preferably rosuvastatin. 根据权利要求28所述的组合物,其中所述PCSK9抑制剂选自Evolocumab、Alirocumab、Inclisiran和Tafolecimab。The composition according to claim 28, wherein the PCSK9 inhibitor is selected from Evolocumab, Alirocumab, Inclisiran and Tafolecimab. 根据权利要求28所述的组合物,其中所述胆固醇吸收抑制剂为NPC1L1抑制剂,优选依折麦布或海博麦布。The composition according to claim 28, wherein the cholesterol absorption inhibitor is an NPC1L1 inhibitor, preferably ezetimibe or hebomibe. 根据权利要求23-32中任一项所述的组合物,其中所述组合物被配制为注射用制剂或口服制剂。 The composition according to any one of claims 23 to 32, wherein the composition is formulated as an injection preparation or an oral preparation.
PCT/CN2024/126779 2023-10-24 2024-10-23 Use of cholesin in modulating cholesterol homeostasis Pending WO2025087287A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202311385254.5A CN117180404A (en) 2023-10-24 2023-10-24 Use of Cholesin for regulating cholesterol homeostasis
CN202311385254.5 2023-10-24

Publications (1)

Publication Number Publication Date
WO2025087287A1 true WO2025087287A1 (en) 2025-05-01

Family

ID=89003612

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2024/126779 Pending WO2025087287A1 (en) 2023-10-24 2024-10-23 Use of cholesin in modulating cholesterol homeostasis

Country Status (2)

Country Link
CN (1) CN117180404A (en)
WO (1) WO2025087287A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117180404A (en) * 2023-10-24 2023-12-08 清华大学 Use of Cholesin for regulating cholesterol homeostasis

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1448403A (en) * 2002-04-03 2003-10-15 上海新世界基因技术开发有限公司 Human protein with cancer cell growth suppressing function and its coding sequence
US20090036394A1 (en) * 2005-10-10 2009-02-05 Takeda Pharmaceutical Company Limited GRP 146 Receptor
WO2010009534A1 (en) * 2008-07-24 2010-01-28 Xenon Pharmaceuticals Inc. Methods for the treatment, prevention and diagnosis of lipid metabolism associated diseases
CN114832105A (en) * 2021-02-01 2022-08-02 复旦大学附属中山医院 Application of inhibitor of C7orf50 gene or protein
CN115837071A (en) * 2022-12-13 2023-03-24 清华大学 Use of Feimin in the regulation of sugar homeostasis
CN117180404A (en) * 2023-10-24 2023-12-08 清华大学 Use of Cholesin for regulating cholesterol homeostasis

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1448403A (en) * 2002-04-03 2003-10-15 上海新世界基因技术开发有限公司 Human protein with cancer cell growth suppressing function and its coding sequence
US20090036394A1 (en) * 2005-10-10 2009-02-05 Takeda Pharmaceutical Company Limited GRP 146 Receptor
WO2010009534A1 (en) * 2008-07-24 2010-01-28 Xenon Pharmaceuticals Inc. Methods for the treatment, prevention and diagnosis of lipid metabolism associated diseases
CN114832105A (en) * 2021-02-01 2022-08-02 复旦大学附属中山医院 Application of inhibitor of C7orf50 gene or protein
CN115837071A (en) * 2022-12-13 2023-03-24 清华大学 Use of Feimin in the regulation of sugar homeostasis
CN117180404A (en) * 2023-10-24 2023-12-08 清华大学 Use of Cholesin for regulating cholesterol homeostasis

Also Published As

Publication number Publication date
CN117180404A (en) 2023-12-08

Similar Documents

Publication Publication Date Title
Zhu et al. STAT3 signaling promotes cardiac injury by upregulating NCOA4-mediated ferritinophagy and ferroptosis in high-fat-diet fed mice
Kawarazaki et al. Salt causes aging-associated hypertension via vascular Wnt5a under Klotho deficiency
Shan et al. Overexpression of TRPC3 increases apoptosis but not necrosis in response to ischemia-reperfusion in adult mouse cardiomyocytes
Moreno et al. The inflammatory cytokines TWEAK and TNFα reduce renal klotho expression through NFκB
Jiang et al. Lipoprotein metabolism in nonalcoholic fatty liver disease
Déliot et al. Parathyroid hormone treatment induces dissociation of type IIa Na+-Pi cotransporter-Na+/H+ exchanger regulatory factor-1 complexes
She et al. VDAC2 malonylation participates in sepsis-induced myocardial dysfunction via mitochondrial-related ferroptosis
EP2898898B1 (en) Secreted AP2 and methods of inhibiting same
Huang et al. Protective effects of grape seed proanthocyanidins on cardiovascular remodeling in DOCA-salt hypertension rats
Sorokina et al. Intracellular targeting of peroxiredoxin 6 to lysosomal organelles requires MAPK activity and binding to 14-3-3ε
Tao et al. Melatonin suppresses atherosclerosis by ferroptosis inhibition via activating NRF2 pathway
Zhuang et al. RCAN1 deficiency aggravates sepsis-induced cardiac remodeling and dysfunction by accelerating mitochondrial pathological fission
CN115837071A (en) Use of Feimin in the regulation of sugar homeostasis
Jia et al. CD73 regulates hepatic stellate cells activation and proliferation through Wnt/β-catenin signaling pathway
WO2025087287A1 (en) Use of cholesin in modulating cholesterol homeostasis
Li et al. C1q/TNF-related protein 4 induces signal transducer and activator of transcription 3 pathway and modulates food intake
Zhao et al. DJ-1 activates the AMPK/mTOR pathway by binding RACK1 to induce autophagy and protect the myocardium from ischemia/hypoxia injury
Sheng et al. A pan-PPAR agonist E17241 ameliorates hyperglycemia and diabetic dyslipidemia in KKAy mice via up-regulating ABCA1 in islet, liver, and white adipose tissue
Vachon et al. Downregulation of low-density lipoprotein receptor mRNA in lymphatic endothelial cells impairs lymphatic function through changes in intracellular lipids
Tang et al. GPSM1 in POMC neurons impairs brown adipose tissue thermogenesis and provokes diet-induced obesity
Jiang et al. Protections of transcription factor BACH2 and natural product myricetin against pathological cardiac hypertrophy and dysfunction
Li et al. Counteracting TGM2 by a Fibroin peptide ameliorated Adriamycin-induced nephropathy via regulation of lipid metabolism through PANX1-PPAR α/PANK1 pathway
KR20230172930A (en) Composition for preventing or treating of obesity comprising peptides derived from Neuronal growth regulator 1(NEGR1)
Qian et al. PGC-1α regulates hepatic hepcidin expression and iron homeostasis in response to inflammation
Huang et al. GPR146 regulates pulmonary vascular remodeling by promoting pulmonary artery smooth muscle cell proliferation through 5-lipoxygenase

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24881651

Country of ref document: EP

Kind code of ref document: A1