WO2025165814A1 - Compositions et méthodes de régulation de l'ischémie post-angiogenèse cardiaque - Google Patents
Compositions et méthodes de régulation de l'ischémie post-angiogenèse cardiaqueInfo
- Publication number
- WO2025165814A1 WO2025165814A1 PCT/US2025/013510 US2025013510W WO2025165814A1 WO 2025165814 A1 WO2025165814 A1 WO 2025165814A1 US 2025013510 W US2025013510 W US 2025013510W WO 2025165814 A1 WO2025165814 A1 WO 2025165814A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nucleic acid
- acid molecule
- inhibitory nucleic
- seq
- nucleotides
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering nucleic acids [NA]
- C12N2310/141—MicroRNAs, miRNAs
Definitions
- This disclosure relates to inhibitory nucleic acid molecules useful for targeting Decorin (DCN), and methods of using such inhibitory nucleic acid molecules for (i) treating medical complications resulting from a myocardial infarction (Ml), and/or (ii) promoting angiogenesis in a subject after the subject has suffered an Ml.
- DCN myocardial infarction
- Ml myocardial infarction
- Angiogenesis around the infarct is crucial to limiting damage and recovery of cardiac function.
- Patients with diabetes are more likely to suffer from myocardial injury compared to those without diabetes.
- longevity and quality of life suffer considerably post-MI, in part due to impaired angiogenesis and tissue repair.
- elevated blood glucose level hyperglycemia
- EC endothelial cell
- microRNAs microRNAs
- the invention provides an inhibitory nucleic acid molecule comprising sufficient complementarity to a target nucleic acid molecule, wherein (i) the inhibitory nucleic acid molecule is at least 15 nucleotides in length, and (ii) the target nucleic acid molecule comprises a nucleotide sequence having at least 80% sequence identity to SEQ ID NO: 14.
- the target nucleic acid molecule comprises a nucleotide sequence having 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%, or at least 99% sequence identity to SEQ ID NO: 14.
- the target nucleic acid molecule comprises the nucleotide sequence of
- the inhibitory nucleic acid molecule is 15 to 6,850 nucleotides in length (e.g., 50 to 6,850 nucleotides in length, 100 to 6,850 nucleotides in length, 250 to 6,850 nucleotides in length, 500 to 6,850 nucleotides in length, 750 to 6,850 nucleotides in length, 1 ,000 to 6,850 nucleotides in length, 1 ,500 to 6,850 2,000 to 6,850, 4,000 to 6,850, 5,000 to 6,850, or 6,000 to 6,850 nucleotides in length).
- the inhibitory nucleic acid molecule is 15 to 6,850 nucleotides in length (e.g., 50 to 6,850 nucleotides in length, 100 to 6,850 nucleotides in length, 250 to 6,850 nucleotides in length, 500 to 6,850 nucleotides in length, 750 to 6,850 nucleotides in length, 1 ,000 to 6,850 nucleotides in length, 1 ,500 to 6,850
- the inhibitory nucleic acid molecule is 15 to 49 nucleotides in length (e.g., 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42,
- nucleotides in length 50 to 99 nucleotides in length (e.g., 50, 51 , 52, 53, 54,
- the inhibitory nucleic acid molecule is 20 to 28 nucleotides in length (e.g., 20, 21 , 22, 23, 24, 25, 26, 27, or 28 nucleotides in length).
- the inhibitory nucleic acid molecule is 18 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 19 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 20 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 21 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 22 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 23 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 24 nucleotides in length. In some embodiments, the inhibitory nucleic acid molecule is 25 nucleotides in length.
- the inhibitory nucleic acid molecule is 23 or 25 nucleotides in length.
- the inhibitory nucleic acid molecule comprises at least 85% (e.g., 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) complementarity to the target nucleic acid molecule, or splice variant thereof.
- the inhibitory nucleic acid molecule includes at least 90% (e.g., 90% 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) complementarity to the target nucleic acid molecule, or splice variant thereof.
- the inhibitory nucleic acid molecule includes at least 95% (e.g., 95%, 96%, 97%, 98%, 99%, or 100%) complementarity to the target nucleic acid molecule, or splice variant thereof.
- the inhibitory nucleic acid molecule is complementary to the target nucleic acid molecule, or splice variant thereof.
- the inhibitory nucleic acid molecule further includes a modification.
- the modification includes: (a) a non-natural or modified nucleoside or nucleotide; and/or (b) a covalently or non-covalently conjugated moiety.
- the non-natural or modified nucleoside or nucleotide is selected from the group consisting of: a locked nucleic acid (LNA), a 2'-O-methyl (2'-0-Me) modified nucleoside, a phosphorothioate (PS) bond between nucleosides, and a 2'-fluoro (2’-F) modified nucleoside; and/or (b) the covalently or non-covalently conjugated moiety is selected from the group consisting of: a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer.
- LNA locked nucleic acid
- PS phosphorothioate
- the targeting moiety is vascular cell adhesion protein 1 (VCAM1). In some embodiments, the targeting moiety is arginylglycylaspartic acid (RGD),
- the inhibitory nucleic acid molecule is selected from the group consisting of: a small interfering RNA (siRNA), a double-stranded RNA (dsRNA), a microRNA (miRNA), a short hairpin RNA (shRNA), an anti-sense oligonucleotide (ASO), and a gapmeR.
- siRNA small interfering RNA
- dsRNA double-stranded RNA
- miRNA microRNA
- shRNA short hairpin RNA
- ASO anti-sense oligonucleotide
- the inhibitory nucleic acid molecule is a dsRNA.
- the inhibitory nucleic acid molecule is a miRNA.
- the inhibitory nucleic acid molecule is an shRNA.
- the inhibitory nucleic acid molecule is an ASO.
- the inhibitory nucleic acid molecule is a gapmeR.
- the inhibitory nucleic acid molecule is an siRNA.
- the siRNA comprises an antisense strand comprising at least 88% (e.g., 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to any one of SEQ ID NOs: 1 -6.
- the antisense strand comprises at least 92% (e.g., 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to any one of SEQ ID NOs: 1 -6.
- the antisense strand comprises at least 96% (e.g., 97%, 98%, 99%, or 100%) sequence identity to any one of SEQ ID NOs: 1 -6.
- the antisense strand comprises the nucleotide sequence of any one of SEQ ID NOs: 1 -6.
- the siRNA further comprises a sense strand comprising at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to any one of SEQ ID NOs: 7-12.
- the siRNA includes: (a) the antisense strand of SEQ ID NO: 1 and the sense strand of SEQ ID NO: 7; (b) the antisense strand of SEQ ID NO: 2 and the sense strand of SEQ ID NO: 8; (c) the antisense strand of SEQ ID NO: 3 and the sense strand of SEQ ID NO: 9; (d) the antisense strand of SEQ ID NO: 4 and the sense strand of SEQ ID NO: 10; (e) the antisense strand of SEQ ID NO: 5 and the sense strand of SEQ ID NO: 11 ; or (f) the antisense strand of SEQ ID NO: 6 and the sense strand of SEQ ID NO: 12.
- the siRNA contains 3’ overhangs selected from the group consisting of: (i) a single uracil overhang at one or more 3’ ends of the siRNA; (ii) a double uracil overhang at one or more 3’ ends of the siRNA; (iii) a single thymine overhang at one or more 3’ ends of the siRNA; (iv) a double thymine overhang at one or more 3’ ends of the siRNA; or (v) a single cytosine and single thymine overhang at one or more 3’ ends of the siRNA.
- the siRNA targets the nucleotide sequence of any one of SEQ ID NOs:
- the inhibitory nucleic acid molecule is a miRNA.
- the miRNA comprises a modification selected from: (a) a non-natural or modified nucleoside or nucleotide; and/or (b) a covalently or non-covalently conjugated moiety.
- the non-natural or modified nucleoside or nucleotide is selected from the group consisting of: a locked nucleic acid (LNA), a 2'-O-methyl (2'-0-Me) modified nucleoside, a phosphorothioate (PS) bond between nucleosides, and a 2'-fluoro (2’-F) modified nucleoside; and/or (b) the covalently or non-covalently conjugated moiety is selected from the group consisting of: a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer.
- LNA locked nucleic acid
- PS phosphorothioate
- the targeting moiety is vascular cell adhesion protein 1 (VCAM1). In some embodiments, the targeting moiety is arginylglycylaspartic acid (RGD).
- the miRNA comprises a nucleotide sequence comprising at least 86% (e.g., 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to SEQ ID NO: 13.
- the miRNA comprises a nucleotide sequence comprising at least 91% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to SEQ ID NO: 13.
- the miRNA comprises a nucleotide sequence comprising at least 95% (e.g., 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to SEQ ID NO: 13.
- the miRNA comprises the nucleotide sequence of SEQ ID NO: 13.
- the miRNA is miR-342-3p.
- the inhibitory nucleic acid molecule is formulated in a delivery vehicle.
- the delivery vehicle is selected from the group consisting of: a vector, a plasmid, a micelle, a liposome, an exosome, and a lipid nano particle (LNP).
- a vector a plasmid, a micelle, a liposome, an exosome, and a lipid nano particle (LNP).
- LNP lipid nano particle
- the vector is a viral vector.
- the viral vector is an adeno-associated viral (AAV) vector.
- AAV adeno-associated viral
- the AAV vector is selected from the group consisting of: AAV1 , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11 , and AAV12.
- the inhibitory nucleic acid molecule is formulated as a pharmaceutical composition.
- the pharmaceutical composition includes a pharmaceutically acceptable excipient, diluent, and/or carrier.
- the invention provides a method of treating or reducing the likelihood of a medical complication resulting from a myocardial infarction (Ml) in a subject, the method comprising administering the inhibitory nucleic acid molecule of the first aspect.
- Ml myocardial infarction
- the medical complication is an arrhythmic-related complication, an ischemic-related complication, a mechanical-related complication, an inflammatory-related complication, and/or a systemic complication.
- the arrhythmic-related complication is a heart block, an atrial arrhythmia, and/or a ventricular arrhythmia;
- the ischemic-related complication is reinfarction, periinfarct ischemia, and/or an infarct extension;
- the mechanical-related complication is a mitral valve rupture or tear, a chordae rupture or tear, a ventricular septal defect (VSD), a ventricular free wall rupture, a cardiac tamponade, and/or an aneurysm;
- the inflammatory-related complication is pericarditis and/or Dressier syndrome; and/or
- the systemic complication is cardiogenic shock, cardiomyopathy, heart failure, embolic stroke, systemic embolism, and/or a lower extremity embolism.
- the invention provides a method of promoting angiogenesis in a subject, the method comprising administering the inhibitory nucleic acid molecule of the first aspect.
- the subject has previously experienced a myocardial infarction.
- the subject has an ischemic injury.
- the subject has a cardiovascular disease.
- the cardiovascular disease is coronary artery disease, peripheral artery disease, cardiomyopathy, or stroke.
- the subject has a metabolic disorder, or the subject is at risk of developing the metabolic disorder.
- the metabolic disorder is diabetes.
- the subject at risk of developing diabetes is prediabetic and/or has one or more of the following: (a) hyperglycemia; (b) glucose resistance; (c) insulin resistance; (d) hyperlipidemia; and (e) has a family history of diabetes.
- the inhibitory nucleic acid molecule is administered to the subject intravenously, intraperitoneally, subcutaneously, intraarticularly, or intramuscularly.
- the inhibitory nucleic acid molecule is delivered to the coronary endothelium, remote zone of the heart, and/or border zone of the heart.
- the inhibitory nucleic acid molecule is delivered to an endothelial cell, a cardiomyocyte, a fibroblast, a vascular smooth muscle cell, and/or a leukocyte.
- the additional therapeutic agent is a statin or hepatocyte growth factor (HGF).
- HGF hepatocyte growth factor
- FIG. 1A-FIG. 1G demonstrate that miR-342-3p is a proangiogenic miRNA.
- FIG. 1A displays the expression kinetics of miR-342-3pin cardiac endothelial cells (ECs) post-myocardial infarction (Ml) between chow and high fat sucrose containing (HFSC) diet groups. Dominant phases for inflammation and angiogenesis are indicated.
- FIG. 1B is a representative image of a spheroid sprouting assay following transfection of ECs with miR-342-3p mimics. These data demonstrate that miR-342-3p promoted spheroid sprouting while its inhibition elicited angiostatic responses.
- FIG. 1A displays the expression kinetics of miR-342-3pin cardiac endothelial cells (ECs) post-myocardial infarction (Ml) between chow and high fat sucrose containing (HFSC) diet groups. Dominant phases for inflammation and angiogenesis are indicated.
- FIG. 1B is
- FIG. 1C are graphs quantifying the number of sprouts after the transfection of ECs with miR-342-3p mimics. This data demonstrates that miR-342-3p promoted spheroid sprouting while its inhibition elicited angiostatic responses. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 1D is a representative image of a scratch closure assay following transfection of ECs with miR-342-3p mimics. These data demonstrate that miR-342-3p accelerated scratch closure while its inhibition elicited angiostatic responses.
- FIG. 1E are graphs quantifying the percent scratch area after the transfection of ECs with miR-342-3p mimics.
- FIG. 1F are graphs quantifying the area under the curve of a scratch area assay after the transfection of ECs with miR-342-3p mimics. These data demonstrate that miR-342-3p accelerated scratch closure while its inhibition elicited angiostatic responses. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 1G are graphs quantifying a BrdU incorporation assay following transfection of ECs with miR-342-3p mimics. These data demonstrate that miR-342-3p increased EC proliferation while its inhibition elicited angiostatic responses. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 2A-FIG. 2H show miR-342-3p’s targets and associated signaling pathways.
- FIG. 2A is a Venn diagram illustrating the identification of decorin (DCN) as one of several targets of miR-342-3p.
- FIG. 2B is a graph quantifying DCN transcripts following miR-342-3p overexpression. These data demonstrate that overexpression miR-342-3p suppresses DCN at the transcriptional level in ECs. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 2C are a Western blot (WB) and a graph quantifying DCN protein following miR-342-3p overexpression. These data demonstrate that overexpression miR-342-3p suppresses DCN at the translational level in ECs.
- FIG. 2D shows DCN’s 3’ UTR and the seed sequence for miR-342-3p (upper) (SEQ ID NOs: 21 -23), along with bar graphs (lower) quantifying DCN 3’UTR transcriptional activity, which was reduced significantly in the presence of miR-342-3p vs control. Mutated DCN 3’UTR is not targeted. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 2E is a chord plot showing top up- regulated and top down-regulated genes, as well as their associated pathways in miR-342-3p overexpressing ECs vs control.
- FIG. 2F are graphs showing that miR-342-3p overexpression induces MET mRNA expression (upper graph) and protein expression (lower graph). Two-tailed t-tests, p ⁇ 0.05.
- FIG. 2G is a WB showing that miR-342-3p overexpression induces MET protein expression.
- FIG. 2H is a graph showing that miR-342-3p induces HGF mRNA concordantly.
- HGF is the canonical ligand of the MET receptor. Two-tailed t-tests, p ⁇ 0.05.
- FIG. 3A-FIG. 3F demonstrate the effect of hypoxia and diabetogenic stimuli on miR-342-3p, DCN and MET.
- FIG. 3A is a graph showing that hypoxia induces miR-342-3p expression under control conditions, but high glucose conditions blunted this induction. One-way analysis of variance (ANOVA), p ⁇ 0.05.
- FIG. 3B is a graph showing that DCN is suppressed by hypoxia and is induced by glucose conditions.
- One-way ANOVA p ⁇ 0.05.
- FIG. 3C is a graph showing that MET expression mirrors that of miR-342-3p; it is induced by hypoxia but abrogated by glucose.
- One-way ANOVA p ⁇ 0.05.
- FIG. 3D is a graph showing that hypoxia induces miR-342-3p expression under control conditions, but palmitate treatment blunts this induction.
- One-way ANOVA p ⁇ 0.05.
- FIG. 3E is a graph showing that DCN is suppressed by hypoxia and is induced by palmitate treatment.
- One-way ANOVA p ⁇ 0.05.
- FIG. 3F is a graph showing that MET expression mirrors that of miR-342-3p; it is induced by hypoxia but abrogated by palmitate treatment.
- FIG. 4A-FIG. 41 demonstrate that miR-342-3p rescues cardiac function and promotes angiogenesis in vivo.
- FIG. 4A is a schematic of the experimental procedure in which miR-342-3p or control mimic was injected in the border zone (BZ) of mice following 45 minutes of ischemia.
- FIG. 4B are representative mode images of non-specific control (NS) vs miR-342-3p treated hearts.
- FIG. 4C is a graph quantifying the ejection fraction (EF) and fractional shortening (FS) in miR-342-3p-injected hearts, which had significantly higher EF and FS at day 14 when compared to control. Two-way ANOVA, two- tailed t-tests, p ⁇ 0.05.
- FIG. 4D is a graph of a quantitative pCR (qPCR) validation of miR-342-3p overexpression in the BZ at day 14.
- FIG. 4E is a graph showing that DCN mRNA was suppressed in the miR-342-3p injected hearts vs control. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 4F is a graph showing that MET mRNA was significantly upregulated in the miR-342-3p treated group at day 14. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 4G are a representative immunofluorescence images showing proliferating (ki67+) endothelial cells in BZs of control or miR-342-3p mimic injected hearts.
- FIG. 4H is a graph quantifying capillary density, which was higher in miR-342-3p hearts vs. control, respectively.
- Two-way ANOVA two-tailed t-tests, p ⁇ 0.05.
- FIG. 41 is a graph quantifying ki67+ EC count, which was higher in miR-342-3p hearts vs. control, respectively.
- Two-way ANOVA two-tailed t-tests, p ⁇ 0.05.
- FIG. 5A-FIG. 5H are a series of graphs demonstrating that miR-342-3p induces EC proliferation via HGF-MET signaling. For all graphs: one-way ANOVA, two-way ANOVA. p ⁇ 0.05.
- FIG. 5A is a graph of BrdU incorporation assays showing inhibition of the MET receptor by PHA-665752 attenuates EC proliferation induced by miR-342-3p.
- FIG. 5B is a graph showing that the addition of exogenous recombinant DCN (rDCN) protein neutralized miR-342-3p-induced EC proliferation, mirroring results from FIG. 5A.
- rDCN exogenous recombinant DCN
- FIG. 5C is a graph showing that PHA-665752 blunted EC proliferation, even in the presence of HGF.
- FIG. 5D is a graph showing that rDCN blunted EC proliferation, even in the presence of HGF.
- FIG. 5E is a graph quantifying a BrdU assay performed under mannitol or glucose conditions in the absence of HGF. While miR-342-3p induced EC proliferation, its induction was attenuated by a MET inhibitor.
- FIG. 5F is a graph quantifying a BrdU assay performed under mannitol or glucose conditions in the presence of HGF. While miR-342-3p induced EC proliferation, its induction was attenuated by a MET inhibitor.
- 5G is a graph showing that exogenous rDCN counteracts the proliferative effects of miR- 342-3p in the absence of HGF.
- FIG. 5H is a graph showing that exogenous rDCN counteracts the proliferative effects of miR-342-3p in the presence of HGF.
- FIG. 6A-FIG. 60 demonstrate that DCN knockdown (KD) phenocopied miR-342-3p overexpression (OE).
- FIG. 6A is a WB showing the induction of MET and repression of DCN by miR-342- 3p.
- FIG. 6B is a graph showing the densitometry of relative MET levels from FIG. 6A. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6C is a graph showing the densitometry of relative DCN levels from FIG. 6A. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6D is a WB showing that siRNA-mediated KD of DCN enhanced MET protein while suppressing DCN; these results phenocopy the results from FIG. 6A- FIG. 6C.
- FIG. 6E is a quantification of MET proteins from FIG. 6D. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6F is a quantification of DCN protein from FIG. 6D. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6G is a graph showing that the KD of DCN induced EC proliferation under normal and high glucose conditions. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6H is a panel of representative endothelial spheroid images showing robust sprouting in the miR-342-3p OE group under both high mannitol and glucose conditions.
- FIG. 61 is a panel of representative endothelial spheroid images showing that DCN KD also promoted sprouting under mannitol and glucose conditions.
- FIG. 6J is a panel of representative endothelial spheroid images showing that the addition of exogenous rDCN attenuates miR-342-induced sprouting.
- FIG. 6K is a panel of representative endothelial spheroid images showing that supplementation with rDCN abrogates siDCN-mediated sprouting, echoing results from FIG. 6J.
- FIG. 6L is a quantification of sprout numbers from FIG. 6H. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6M is a quantification of sprout numbers from FIG. 61. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 6N is a quantification of sprout numbers from FIG. 6J. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 60 is a quantification of sprout numbers from FIG. 6K. Two-way ANOVA, two-tailed t-tests, p ⁇ 0.05.
- FIG. 7 is a graphical summary of miR-342-3p-induced angiogenesis.
- hypoxia induces miR-342-3p expression, inhibiting DCN.
- miR-342-3p induced HGF-MET signaling precipitates sprouting angiogenesis.
- Diabetes associated stimuli such as high glucose or fatty acid levels, prevents hypoxia-induced expression of miR-342- 3p, causing an overabundance of DCN protein which can then be secreted and bind to the MET receptor.
- DCN-MET binding leads to internalization and degradation of the MET receptor. This blockade of the HGF-MET pathway impairs angiogenesis.
- FIG. 8A-FIG. 8B demonstrate that DCN protein expression is impacted by diabetes-associated stimuli.
- FIG. 8A is a WB blot and quantification thereof showing the suppression of DCN due to hypoxia in mannitol treated ECs.
- High glucose treatment significantly elevates DCN levels under normoxia and hypoxia vs mannitol control.
- FIG. 8B is a WB and quantification thereof showing that hypoxia suppressed DCN protein expression in a time-dependent manner vs normoxia when treated with BSA. Palmitate treatment significantly upregulates DCN protein and keeps it elevated even under hypoxic conditions Two-way ANOVA p ⁇ 0.05.
- FIG. 9A-FIG. 9E demonstrate that miR-342-3p acts via the HGF-MET signaling pathway in both mice and humans. For all graphs: one-way and two-way ANOVA p ⁇ 0.05.
- FIG. 9A is a quantification of a BrdU assay showing that EC proliferation is induced by HGF (50 ng/mL) supplementation in HUVECs transfected with either control or miR-342 mimic.
- FIG. 9B is a graph showing that the addition of PHA- 665752 blunted EC proliferation caused by miR-342 under normal (mannitol) conditions. HUVECs proliferated less under high glucose conditions even in the absence of the inhibitor. Addition of the inhibitor did not synergistically reduce proliferation.
- FIG. 9C is a graph showing that supplementation with rDCN attenuated proliferation in a manner similar to that of the MET inhibitor.
- FIG. 9D is a quantification of a BrdU assay showing similar results FIG. 9B except with added HGF.
- FIG. 9E is a graph showing that rDCN acts similarly to the conditions set forth in FIG. 9C, even in the presence of added HGF.
- FIG. 10A-FIG. 10D are dose-responses of PHA-665752 and DCN using the scratch closure assay.
- FIG. 10A is a dose curve showing scratch closure rates in response to vehicle (DMSO) or increasing concentrations of PHA-665752 - a MET inhibitor.
- FIG. 10B is an area under curve (AUC) analysis from FIG. 10A showing significantly higher AUC, or slower scratch closure, in ECs treated with 100 nM of the inhibitor. One-way ANOVA p ⁇ 0.05.
- FIG. 10C is a dose curve showing scratch closure rates in response to increasing concentrations of rDCN protein.
- FIG. 10D is an AUC analysis from FIG. 10C showing effective inhibition of scratch closure at 25 nM dose. One-way ANOVA p ⁇ 0.05.
- FIG. 11 A displays the expression kinetics of DCN mRNA in cardiac ECs post-MI.
- FIG. 11B displays the expression kinetics of MET mRNA in cardiac ECs post-MI.
- FIG. 11C is a panel of representative light-sheet images in which infarct size and capillary density was quantified.
- the term "about,” as applied to one or more values of interest, refers to a value that falls within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of a stated reference value, unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
- administration refers to providing or giving a subject a therapeutic agent by any effective route. Exemplary routes of administration are described herein below.
- the term “administered in combination” or “combined administration” means that two or more agents are administered to a subject at the same time or within an interval such that there may be an overlap of an effect of each agent on the patient. In some embodiments, they are administered within about 60, 30, 15, 10, 5, or 1 minute of one another. In some embodiments, the administrations of the agents are spaced sufficiently closely together such that a combinatorial (e.g., a synergistic) effect is achieved.
- auxiliary moiety refers to any moiety, including, but not limited to, a small molecule, a peptide, a carbohydrate, a neutral organic polymer, a positively charged polymer, a therapeutic agent, a targeting moiety, an endosomal escape moiety, and any combination thereof, which can be conjugated to a nucleic acid molecule.
- an "auxiliary moiety" is linked to an inhibitory nucleic acid molecule disclosed herein by forming one or more covalent or non-covalent bonds with one or more conjugating groups attached to a phosphate linkage, a phosphorothioate linkage, a 5' positions of a nucleotide sugar, or any portion of a nucleobase.
- conjugating groups attached to a phosphate linkage, a phosphorothioate linkage, a 5' positions of a nucleotide sugar, or any portion of a nucleobase.
- delivery vehicle refers to any substance (e.g., molecule, peptide, conjugate, and construct) that facilitates, at least in part, the in vivo delivery of a nucleic acid molecule to targeted cells.
- the terms “effective amount,” “therapeutically effective amount,” and a “sufficient amount” of a composition described herein refer to a quantity sufficient to, when administered to the subject, effect beneficial or desired results; as such, an “effective amount” or synonym thereto depends upon the context in which it is being applied. For example, in the context of decreasing decorin (DCN), it is an amount of the composition sufficient to achieve a treatment response as compared to the response obtained without administration of the composition.
- DCN decreasing decorin
- the amount of a given composition described herein that will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical compositions, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, weight) or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art.
- a “formulation” includes at least an inhibitory nucleic acid molecule and a delivery vehicle.
- in vitro refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism (e.g., animal, plant, or microbe).
- in vivo refers to events that occur within an organism (e.g., animal, plant, or microbe or cell or tissue thereof).
- inhibitory nucleic acid molecule refers to a nucleic acid molecule that has sufficient complementarity to bind to a target nucleic acid molecule to inhibit expression of a product (e.g., a mRNA) encoded by the target nucleic acid molecule.
- exemplary inhibitory nucleic acid molecules are anti-sense oligonucleotides (ASOs), small interfering RNA (siRNAs), short hairpin RNA (shRNAs), double stranded RNAs (dsRNAs), and microRNA (miRNAs).
- Inhibitory nucleic acid molecules may reduce the target’s expression by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99% or more).
- the target nucleic acid molecule encodes DCN.
- the term “modified” refers to a changed state or structure of a nucleic acid molecule described herein. Molecules may be modified in many ways including chemically, structurally, and functionally. In one embodiment, the inhibitory nucleic acid molecules of the present invention are modified by the introduction of non-natural nucleosides and/or nucleotides. In other embodiments, the inhibitory nucleic acid molecules of the present invention are modified by conjugation of an auxiliary moiety.
- the term “pharmaceutical composition” refers to a mixture containing a therapeutic agent, optionally in combination with one or more pharmaceutically acceptable excipients, diluents, and/or carriers, to be administered to a subject, such as a mammal, e.g., a human, in order to prevent, treat or control a particular disease or condition affecting or that may affect the subject.
- the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues a subject, such as a mammal (e.g., a human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
- Percent (%) sequence identity with respect to a reference polynucleotide or polypeptide sequence is defined as the percentage of nucleic acids or amino acids in a candidate sequence that are identical to the nucleic acids or amino acids in the reference polynucleotide or polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid or amino acid sequence identity can be achieved in various ways that are within the capabilities of one of skill in the art, for example, using publicly available computer software such as BLAST, BLAST-2, or Megalign software.
- percent sequence identity values may be generated using the sequence comparison computer program BLAST.
- percent sequence identity of a given nucleic acid or amino acid sequence, A, to, with, or against a given nucleic acid or amino acid sequence, B, (which can alternatively be phrased as a given nucleic acid or amino acid sequence, A that has a certain percent sequence identity to, with, or against a given nucleic acid or amino acid sequence, B) is calculated as follows:
- an inhibitory nucleic acid molecule e.g., an siRNA, a dsRNA, a miRNA, a shRNA, an ASO, or a gapmeR
- a target nucleic acid molecule e.g., a target mRNA, e.g., DCN
- the inhibitory nucleic acid molecule includes a nucleotide sequence capable of hybridizing to, and triggering the destruction of, the target nucleic acid molecule (e.g., by RISC-mediated cleavage or Rnase H-mediated cleavage of the target nucleic acid molecule).
- the inhibitory nucleic acid molecule can be designed such that every nucleotide is complementary to a nucleotide in the target nucleic acid molecule. Alternatively, mismatched nucleotides may be introduced so long as there remains hybridization and destruction of the target nucleic acid molecule.
- therapeutic agent refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
- treatment and “treating” in reference to a disease or condition, refer to an approach for obtaining beneficial or desired results, e.g., clinical results.
- beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease or condition; stabilized (i.e., not worsening) state of disease, disorder, or condition; preventing spread of disease or condition; delay or slowing the progress of the disease or condition; amelioration or palliation of the disease or condition; and remission (whether partial or total), whether detectable or undetectable.
- Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
- Those in need of treatment include those already with the condition or disorder, as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
- vector is considered a replicon, such as plasmid, phage, viral construct or cosmid, to which another nucleic acid (e.g., DNA or RNA) segment may be attached.
- vectors are used to transduce and express the nucleic acid segment in cells.
- compositions for reducing expression of a target nucleic acid molecule (e.g., an mRNA molecule encoding decorin (DCN), e.g., SEQ ID NO: 14) and methods thereof for (i) treating a medical condition resulting from a myocardial infarction (Ml), and/or (ii) promoting angiogenesis in a subject.
- a target nucleic acid molecule e.g., an mRNA molecule encoding decorin (DCN), e.g., SEQ ID NO: 14
- DCN mRNA molecule encoding decorin
- the inhibitory nucleic acid molecule may be a small interfering RNA (siRNA), a double-stranded RNA (dsRNA), an anti-sense oligonucleotide (ASO), a microRNA (miRNA), or a short hairpin RNA (shRNA)), or a gapmeR described herein, or a composition (e.g., pharmaceutical composition) thereof.
- the composition described herein provides therapeutic effects (e.g., angiogenesis) for cardiac tissue following myocardial infarction (Ml).
- inhibitory nucleic acid molecules of the disclosure are siRNAs, dsRNAs, ASOs, miRNAs, gapmeRs, and shRNAs; however, any nucleic acid molecule capable of reducing DCN (e.g., SEQ ID NO: 14), or a variant thereof, is envisioned for use of the methods described herein.
- the inhibitory nucleic acid molecules of the disclosure may be referred to as RNA inhibitory (RNAi) molecules.
- the inhibitory nucleic acid molecule contains at least some sequence complementarity to the nucleotide sequence of SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 15 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 16 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 17 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 18 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 19 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 20 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 21 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 22 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 23 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 24 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 25 contiguous nucleotides set forth within SEQ ID NOs: 16. In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 26 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 27 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 28 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 29 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 30 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the inhibitory nucleic acid molecule comprises or consists of a sequence complementary to at least 15 to 6850 contiguous nucleotides (e.g., 15 to 49, 20 to 28, 23-25, 50 to 99, 100 to 200, 150 to 300, 200 to 400, 300 to 700, 500 to 1000, 1000 to 5000, 100 to 6850, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 36 to 30, 27 to 30, 28 to 30, or 29 to 30 contiguous nucleotide) set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- 15 to 6850 contiguous nucleotides e.g., 15 to 49, 20 to 28, 23-25, 50 to 99, 100 to 200, 150 to 300, 200 to 400, 300 to 700, 500 to 1000, 1000 to 5000, 100 to 6850, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to
- the inhibitory nucleic acid is an siRNA targeting DON (e.g., SEQ ID NO: 14), or a variant thereof. In some embodiments, the inhibitory nucleic acid is an dsRNA targeting DON (e.g., SEQ ID NO: 14), or a variant thereof. In some embodiments, the inhibitory nucleic acid is an ASO targeting DON (e.g., SEQ ID NO: 14), or a variant thereof. In some embodiments, the inhibitory nucleic acid is a gapmeR targeting DON (e.g., SEQ ID NO: 14), or a variant thereof.
- the inhibitory nucleic acid is a miRNA targeting DON (e.g., SEQ ID NO: 14), or a variant thereof.
- the inhibitory nucleic acid is an shRNA targeting DON (e.g., SEQ ID NO: 14), or a variant thereof.
- small interfering RNA (siRNA) siRNAs of the disclosure are single-stranded (ss) or double-stranded (ds) nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- RISC RNA-induced silencing complex
- siRNAs of the disclosure may include a nucleotide sequence of about 10 to about 30 nucleotides in length (e.g., 9, about 10, about 11 , about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21 , about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, or 31 nucleotides in length).
- siRNAs of the disclosure may include a nucleotide sequence of 10 to 30 nucleotides in length (e.g., 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 26 to 30, 27 to 30, 28 to 30, or 29 to 30 nucleotides in length, e.g., 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length).
- 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 26 to 30, 27 to 30, 28 to 30, or 29 to 30 nucleotides in length e.g., 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length).
- the siRNA includes a sequence complementary at least 15 to 30 contiguous nucleotides (e.g., 15 to 25, 20 to 28, 23 to 25, 25 to 28, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 36 to 30, 27 to 30, 28 to 30, or 29 to 30 contiguous nucleotides, e.g., 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 contiguous nucleotides) set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- contiguous nucleotides e.g., 15 to 25, 20 to 28, 23 to 25, 25 to 28, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 36 to 30, 27 to 30, 28 to 30, or 29 to 30 contiguous nucleotides, e.g., 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29,
- the siRNA contains an antisense strand.
- lengths for an antisense strand of the siRNA molecules of the present disclosure is between 10 and 30 nucleotides (e.g., 10 nucleotides, 11 nucleotides, 12 nucleotides, 13 nucleotides, 14 nucleotides, 15 nucleotides, 16 nucleotides, 17 nucleotides, 18 nucleotides, 19 nucleotides, 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides, 25 nucleotides, 26 nucleotides, 27 nucleotides, 28 nucleotides, 29 nucleotides, or 30 nucleotides), between 15 and 25 nucleotides (e.g., 15 nucleotides, 16 nucleotides, 17 nucleotides, 18 nucleotides, 19 nucleotides, 20 nucleotides, 21 nu
- the antisense strand is 17 nucleotides. In some embodiments, the antisense strand is 18 nucleotides. In some embodiments, the antisense strand is 19 nucleotides. In some embodiments, the antisense strand is 20 nucleotides. In some embodiments, the antisense strand is 21 nucleotides. In some embodiments, the antisense strand is 22 nucleotides. In some embodiments, the antisense strand is 23 nucleotides. In some embodiments, the antisense strand is 24 nucleotides. In some embodiments, the antisense strand is 25 nucleotides. In some embodiments, the antisense strand is 26 nucleotides.
- the antisense strand is 27 nucleotides. In some embodiments, the antisense strand is 28 nucleotides. In some embodiments, the antisense strand is 29 nucleotides. In some embodiments, the antisense strand is 30 nucleotides.
- the siRNA contains a sense strand.
- the sense strand of the siRNA molecules of the present disclosure is between 10 and 30 nucleotides (e.g., 10 nucleotides, 11 nucleotides, 12 nucleotides, 13 nucleotides, 14 nucleotides, 15 nucleotides, 16 nucleotides, 17 nucleotides, 18 nucleotides, 19 nucleotides, 20 nucleotides, 21 nucleotides, 22 nucleotides, 23 nucleotides, 24 nucleotides, 25 nucleotides, 26 nucleotides, 27 nucleotides, 28 nucleotides, 29 nucleotides, or 30 nucleotides), between 15 and 25 nucleotides (e.g., 15 nucleotides, 16 nucleotides, 17 nucleotides, 18 nucleotides, 19 nucleotides, 20 nucleotides, 21 nucleotides, 22
- the sense strand is 15 nucleotides. In some embodiments, the sense strand is 16 nucleotides. In some embodiments, the sense strand is 17 nucleotides. In some embodiments, the sense strand is 18 nucleotides. In some embodiments, the sense strand is 19 nucleotides. In some embodiments, the sense strand is 20 nucleotides. In some embodiments, the sense strand is 21 nucleotides. In some embodiments, the sense strand is 22 nucleotides. In some embodiments, the sense strand is 23 nucleotides. In some embodiments, the sense strand is 24 nucleotides. In some embodiments, the sense strand is 25 nucleotides.
- the sense strand is 26 nucleotides. In some embodiments, the sense strand is 27 nucleotides. In some embodiments, the sense strand is 28 nucleotides. In some embodiments, the sense strand is 29 nucleotides. In some embodiments, the sense strand is 30 nucleotides.
- the sense and antisense strands of an siRNA molecule of the disclosure are completely complementary. In some embodiments, the sense and antisense strands of an siRNA molecule of the disclosure are completely complementary to the extent that their lengths overlap with one another. Depending on the sequence of the first and second strand, complementarity need not be complete or perfect, which means that the first and second strand are not 100% base-paired due to mismatches. One or more mismatches may be present within the ds siRNA without impacting the siRNA’s ability to reduced expression of a target gene of interest.
- the nucleotide sequence of an siRNA of the disclosure may contain sufficient complementarity to a portion of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3) such that the siRNA can hybridize with the target gene of interest.
- the siRNA is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3), or a portion thereof.
- the siRNA is complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3), or a portion thereof.
- the nucleotide sequence of the siRNA may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the nucleotide sequence of the siRNA may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the siRNA of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DCN (e.g., SEQ ID NO: 14), or a variant thereof.
- the siRNA may hybridize to a target sequence of any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- the target gene of interest may be DCN (e.g., SEQ ID NO: 14, or a splice variant thereof, e.g., see Table 3).
- the siRNAs described herein have 0-7 nucleotide 3’ overhangs or 0-4 nucleotide 5’ overhangs.
- the siRNA molecule has a single uracil (e.g., U) overhang at each 3’ end of the siRNA.
- the siRNA molecule has a double uracil (e.g., UU) overhang at each 3’ end of the siRNA.
- the siRNA molecule has a single thymine (e.g., T) overhang at each 3’ end of the siRNA.
- the siRNA molecule has a double thymine (e.g., TT) overhang at each 3’ end of the siRNA. In some embodiments, the siRNA molecule has a cytosine and thymine (e.g., CT) overhang at each 3’ end of the siRNA.
- TT double thymine
- CT cytosine and thymine
- siRNAs can be combined for decreasing mRNA expression of DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- a combination of two siRNAs may be used in a method of the invention, such as two different siRNAs, three different siRNAs, four different siRNAs, or five different siRNAs targeting the same gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- the siRNA sequence may contain at least 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to any one or more of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA sequence may contain the sequence of any one or more of SEQ ID NOs: 1 -6 (e.g., see Table 1 ).
- the siRNA contains at least 15 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains at least 16 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains at least 17 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains at least 18 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ).
- the siRNA contains at least 19 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains at least 20 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains 21 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains 22 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ).
- the siRNA contains 23 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains 24 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ). In some embodiments, the siRNA contains 25 contiguous nucleotides set forth within any one of SEQ ID NOs: 1 -6 (e.g., see Table 1 ).
- the siRNA further contains the sequence of any one of SEQ ID NOs: 7-12.
- Table 1 below provides the antisense and sense strands of exemplary siRNA sequences of the invention.
- the siRNA of the disclosure may target a nucleotide sequence of any one of SEQ ID NOs: 14-20 (e.g., see Table 2), or a complementary sequence thereof, or variant thereof with at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% thereto.
- the siRNA comprises a sequence complementary to at least 15 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 16 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 17 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the siRNA comprises a sequence complementary to at least 18 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 19 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 20 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the siRNA comprises a sequence complementary to at least 21 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 22 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 23 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the siRNA comprises a sequence complementary to at least 24 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 25 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 26 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the siRNA comprises a sequence complementary to at least 27 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 28 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3). In some embodiments, the siRNA comprises a sequence complementary to at least 29 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the siRNA comprises a sequence complementary to at least 30 contiguous nucleotides set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the nucleotide sequence of SEQ ID NO: 14 is set forth in Table 2.
- Double-stranded RNA (ds RNA) dsRNAs of the disclosure are ds nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- dsRNAs are longer than an siRNA and are processed within a cell to form an siRNA molecule.
- the siRNA is then incorporated into an RNA-induced silencing complex (RISC).
- RISC RNA-induced silencing complex
- dsRNAs of the disclosure may include a sense strand and an antisense strand, each containing a nucleotide sequence of about 25 to about 5000 nucleotides in length, or longer (e.g., 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about
- a sense strand and an antisense strand each containing a nucleotide sequence of about 25 to about 5000 nucleotides in length, or longer (e.g., 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120
- dsRNAs of the disclosure may include a sense strand and an antisense strand, each containing a nucleotide sequence of 25 to 5000 nucleotides in length, or longer (e.g., 25 to 5000, 50 to 5000, 75 to 5000, 100 to 5000, 125 to 5000, 150 to 5000, 175 to 5000, 200 to 5000, 225 to 5000, 250 to 5000, 275 to 5000, 300 to 5000, 325 to 5000, 350 to 5000, 375 to 5000, 400 to 5000, 425 to
- 1800 to 5000 1900 to 5000, 2000 to 5000, 2100 to 5000, 2200 to 5000, 2300 to 5000, 2400 to 5000,
- 2500 to 5000, 3000 to 5000, 3500 to 5000, 4000 to 5000, or 4500 to 5000 nucleotides in length e.g., 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145,
- the dsRNA includes a sequence complementary at least 25 to 6850 contiguous nucleotides (e.g., 25 to 6850, 50 to 6850, 75 to 6850, 100 to 6850, 125 to 6850, 150 to 6850, 175 to 6850, 200 to 6850, 225 to 6850, 250 to 6850, 275 to 6850, 300 to 6850, 325 to 6850, 350 to 6850, 375 to 6850, 400 to 6850, 425 to 6850, 450 to 6850, 475 to 6850, 500 to 6850, 550 to 6850, 600 to 6850, 650 to 6850, 700 to 6850, 750 to 6850, 800 to 6850, 850 to 6850, 900 to 6850, 950 to 6850, 1000 to 6850, 1050 to 6850, 1100 to 6850, 1150 to 6850, 1200 to 6850, 1300 to 6850, 1400 to 6850, 1500 to 6850, 1600 to 6850, 1700 to 6850, 1800 to 6
- the sense and antisense strands of an dsRNA molecule of the disclosure are completely complementary. In some embodiments, the sense and antisense strands of an dsRNA molecule of the disclosure are completely complementary to the extent that their lengths overlap with one another. Depending on the sequence of the first and second strand, complementarity need not be complete or perfect, which means that the first and second strand are not 100% base-paired due to mismatches. One or more mismatches may be present within the ds dsRNA without impacting the dsRNA’s ability to reduced expression of a target gene of interest.
- the nucleotide sequence of an dsRNA of the disclosure may contain sufficient complementarity to a portion of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof) such that the dsRNA can hybridize with the target gene of interest.
- the dsRNA is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the dsRNA is complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the nucleotide sequence of the dsRNA may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3). In some embodiments, the nucleotide sequence of the dsRNA may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3).
- the dsRNA of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3.
- the target sequence may be any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- Different dsRNAs can be combined for decreasing the protein expression of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- a combination of two dsRNAs may be used in a method of the invention, such as two different dsRNAs, three different dsRNAs, four different dsRNAs, or five different dsRNAs targeting the same gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof.
- micro RNA (miRNA) miRNAs of the disclosure are single stranded (ss) nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- ss single stranded nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- RISC RNA-induced silencing complex
- miRNAs of the disclosure may include a nucleotide sequence of about 6 to about 30 nucleotides in length (e.g., 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, or 31 nucleotides in length).
- miRNAs of the disclosure may include a nucleotide sequence of 6 to 30 nucleotides in length (e.g., 6 to 30, 7 to 30, 8 to 30, 9 to 30, 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 26 to 30, 27 to 30, 28 to 30, or 29 to 30 nucleotides in length, e.g., 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length).
- 6 to 30, 7 to 30, 8 to 30, 9 to 30, 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 26 to 30, 27 to 30, 28 to 30, or 29 to 30 nucleotides in length e.g., 6, 7, 8, 9, 10, 11 ,
- the miRNA comprises a sequence complementary at least 6 to 30 contiguous nucleotides (e.g., 6 to 30, 7 to 30, 8 to 30, 9 to 30, 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25 to 30, 26 to 30, 27 to 30, 28 to 30, or 29 to 30 contiguous nucleotides, e.g., 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, or 30 contiguous nucleotides) set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- contiguous nucleotides e.g., 6 to 30, 7 to 30, 8 to 30, 9 to 30, 10 to 30, 11 to 30, 12 to 30, 13 to 30, 14 to 30, 15 to 30, 16 to 30, 17 to 30, 18 to 30, 19 to 30, 20 to 30, 21 to 30, 22 to 30, 23 to 30, 24 to 30, 25
- the nucleotide sequence of the miRNA may contain sufficient complementarity to a portion of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof) such that the miRNA can hybridize with the target gene of interest.
- the miRNA is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the miRNA is complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the nucleotide sequence of the miRNA may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the nucleotide sequence of the miRNA may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the miRNA of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DCN (e.g., SEQ ID NO: 14), or a variant thereof.
- the target sequence may be any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- the nucleotide sequence of the miRNA includes at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to SEQ ID NO: 13.
- the miRNA may further include a modification described herein (e.g., a non-natural or modified nucleoside or nucleotide, and/or a covalently or non- covalently conjugated moiety).
- the miRNA is miR-342-3p (e.g., SEQ ID NO: 13).
- Different miRNAs can be combined for decreasing the protein expression of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- a combination of two or more miRNAs may be used in a method of the invention, such as two different miRNAs, three different miRNAs, four different miRNAs, or five different miRNAs targeting the same target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3).
- At least one of the miRNAs is miR-342-3p (e.g., SEQ ID NO: 13), or a variant thereof having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity thereto.
- short hairpin RNA (shRNA) shRNAs of the disclosure are ss or ds nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- RISC RNA- induced silencing complex
- shRNAs of the disclosure may include a nucleotide sequence of about 50 to about 100 nucleotides in length (e.g., 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, or 110 nucleotides in length).
- shRNAs of the disclosure may include a nucleotide sequence of 50 to 100 nucleotides in length (e.g., 50 to 100, 55 to 100, 60 to 100, 65 to 100, 70 to 100, 75 to 100, 80 to 100, 85 to 100, 90 to 100, or 95 to 100 nucleotides in length, e.g., 50, 51 , 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, or 100 nucleotides in length).
- shRNAs of the disclosure contain a variable hairpin loop structure and a stem sequence.
- the stem sequence may be 10 to 50 nucleotides in length (e.g., 10 to 50, 11 to 50, 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50,
- the hairpin size is between 4 to 50 nucleotides in length (e.g., 4 to 50, 5 to 50, 6 to 50, 7 to 50, 8 to 50, 9 to 50, 10 to 50, 11 to 50, 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50,
- shRNA molecules of the disclosure may contain mismatches, for example G-U mismatches between two strands of the shRNA stem without decreasing potency.
- shRNAs are designed to include one or several G-U pairings in the hairpin stem to stabilize hairpins during propagation in bacteria, for example.
- the shRNA includes a sequence (e.g., a stem sequence) complementary at least 10 to 50 contiguous nucleotides (e.g., 10 to 50, 11 to 50, 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to
- a sequence e.g., a stem sequence
- complementary at least 10 to 50 contiguous nucleotides e.g., 10 to 50, 11 to 50, 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to
- contiguous nucleotides e.g., 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, or 50 contiguous nucleotides) set forth within SEQ ID NO: 14 (or a variant thereof, e.g., see Table 3).
- the nucleotide sequence of the shRNA may contain sufficient complementarity to a portion of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof) such that the shRNA can hybridize with the target gene of interest.
- the shRNA is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the shRNA is complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the nucleotide sequence of the shRNA may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the nucleotide sequence of the shRNA may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the shRNA of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DCN (e.g., SEQ ID NO: 14), or a variant thereof.
- the target sequence may be any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- Different shRNAs can be combined for decreasing the protein expression of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- a combination of two or more shRNAs may be used in a method of the invention, such as two different shRNAs, three different shRNAs, four different shRNAs, or five different shRNAs targeting the same gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- ASOs of the disclosure are single (ss) nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- ss single (ss) nucleic acid molecules made of DNA, RNA, or both DNA and RNA (e.g., a chimeric) that are complementary to a target gene of interest and prevent translation of the target’s mRNA into a protein.
- RNase H Upon hybridization to a target mRNA, RNase H will degrade the mRNA by hydrolyzation, resulting in reduced mRNA and protein levels of the target.
- ASOs of the disclosure may include a nucleotide sequence of about 12 to about 50 nucleotides in length (e.g., 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, or 51 nucleotides in length).
- ASOs of the disclosure may include a nucleotide sequence of 12 to 50 nucleotides in length (e.g., 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to 50, 28 to 50, 29 to 50, 30 to 50, 31 to
- the ASO includes a sequence complementary at least 12 to 50 contiguous nucleotides (e.g., 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to 50, 28 to 50, 29 to 50, 30 to 50,
- the nucleotide sequence of the ASO may contain sufficient complementarity to a portion of a target gene of interest (e.g., DON (e.g., SEQ ID NO: 14), or a variant thereof) such that the ASO can hybridize with the target gene of interest.
- the ASO is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DON (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the ASO is complementary to the target gene of interest (e.g., DON (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the nucleotide sequence of the ASO may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DON (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the nucleotide sequence of the ASO may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DON (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the ASO of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DON (e.g., SEQ ID NO: 14), or a variant thereof.
- the target sequence may be any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- ASOs can be combined for decreasing the protein expression of a target gene of interest (e.g., DON (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3).
- a combination of two ASOs may be used in a method of the invention, such as two different ASOs, three different ASOs, four different ASOs, or five different ASOs targeting the same gene of interest (e.g., DON (e.g., SEQ ID NO: 14), or a variant thereof, e.g., see Table 3).
- GapmeRs of the disclosure are single (ss) nucleic acid molecules made of DNA and RNA with the central 8-10 nucleotide of the gapmeR being DNA that is complementary to a target gene of interest, which prevent translation of the target’s mRNA into a protein.
- RNase H Upon hybridization to a target mRNA, RNase H will degrade the mRNA by hydrolyzation, resulting in reduced mRNA and protein levels of the target.
- gapmeRs of the disclosure may include a nucleotide sequence of about 12 to about 50 nucleotides in length (e.g., 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, or 51 nucleotides in length).
- gapmeRs of the disclosure may include a nucleotide sequence of 12 to 50 nucleotides in length (e.g., 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to 50, 28 to 50, 29 to 50, 30 to 50, 31 to 50, 32 to 50, 33 to 50, 34 to 50, 35 to 50, 36 to 50, 37 to 50, 38 to 50, 39 to 50, 40 to 50, 41 to 50, 42 to 50, 43 to 50, 44 to 50, 45 to 50, 46 to 50, 47 to 50, 48 to 50, or 49 to 50 nucleotides in length, e.g., 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44 to 50, 45
- gapmeRs of the disclosure may include a nucleotide sequence of 8 to 9, 8 to 10, or 9 to 10 (e.g., 8, 9, or 10) internal DNA nucleotides.
- the gapmeR includes a sequence complementary at least 12 to 50 contiguous nucleotides (e.g., 12 to 50, 13 to 50, 14 to 50, 15 to 50, 16 to 50, 17 to 50, 18 to 50, 19 to 50, 20 to 50, 21 to 50, 22 to 50, 23 to 50, 24 to 50, 25 to 50, 26 to 50, 27 to 50, 28 to 50, 29 to 50, 30 to 50,
- the nucleotide sequence of the gapmeR may contain sufficient complementarity to a portion of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof) such that the gapmeR can hybridize with the target gene of interest.
- the gapmeR is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the gapmeR is complementary to the target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof), or a portion thereof.
- the nucleotide sequence of the gapmeR may contain sufficient complementarity to an exon sequence of a target gene of interest (e.g., an exon of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the nucleotide sequence of the gapmeR may contain sufficient complementarity to an intron sequence of a target gene of interest (e.g., an intron of DCN (e.g., SEQ ID NO: 14), or a variant thereof). In some embodiments, the gapmeR of the disclosure may contain sufficient complementarity to a pre-mRNA transcript or an mRNA transcript encoding DCN (e.g., SEQ ID NO: 14), or a variant thereof.
- the target sequence may be any one of SEQ ID NOs: 15-20 (e.g., see Table 2).
- Different gapmeRs can be combined for decreasing the protein expression of a target gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- a combination of two gapmeRs may be used in a method of the invention, such as two different gapmeRs, three different gapmeRs, four different gapmeRs, or five different gapmeRs targeting the same gene of interest (e.g., DCN (e.g., SEQ ID NO: 14), or a variant thereof).
- inhibitory nucleic acid molecules disclosed herein may be used in the methods disclosed herein in an unmodified or in a modified form.
- Unmodified inhibitory nucleic acid molecules contain nucleobases that include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U). Modified nucleic acid molecules are described in more detail below.
- Modifications may be achieved by systematically adding or removing linked nucleosides to generate longer or shorter sequences.
- Modifications may be achieved by incorporating, for example, one or more alternative nucleosides, alternative 2’ sugar moieties, and/or alternative internucleoside linkages, which are described further below.
- these types of modifications are introduced to optimize the molecule’s efficacy or biophysical properties (e.g., increasing serum stability or circulating half-life, increasing thermal stability, enhancing transmembrane delivery, reduce immunogenicity, and/or targeting to a particular location or cell type).
- Modification may further be achieved by covalently or non-covalently conjugating a moiety (e.g., a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer) to the 5’ end and/or 3’ end of the inhibitory nucleic acid molecule, as described in more detail below.
- a moiety e.g., a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer
- VCAM1 vascular ceil adhesion protein 1
- RGD arginylglycylaspartic acid
- the inhibitory nucleic acid molecules may also include nucleobases in which the purine or pyrimidine base is replaced with other heterocycles, for example 7- deaza-adenine, 7-deazaguanosine, 2-aminopyridine, and/or 2-pyridone. Further modification of the inhibitory nucleic acid molecules described herein may include nucleobases disclosed in US 3,687,808; Kroschwitz, J. I., ed. The Concise Encyclopedia of Polymer Science and Engineering, New York, John Wiley & Sons, 1990, pp.
- Modifications of the inhibitory nucleic acid molecules described herein may also include one or more of the following 2’ sugar modifications: 2’-O-methyl (2’-0-Me), 2'-methoxyethoxy (2'-O- CH2CH2OCH3, also known as 2'-O-(2-methoxyethyl) or 2'-MOE), 2'-dimethylaminooxyethoxy, i.e.
- a O(CH2)2ON(CH3)2 group also known as 2'-DMAOE, and/or 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethylamino-ethoxy-ethyl or 2'-DMAEOE), i.e., 2'-O-CH2OCH2N(CH3)2.
- Other possible 2'-modifications that can modify the inhibitory nucleic acid molecules described herein include all possible orientations of OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-O- alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl.
- 2'-sugar substituent groups may be in the arabino (up) position or ribo (down) position.
- the 2'-arabino modification is 2'-F.
- Similar modifications may also be made at other positions on the interfering RNA molecule, particularly the 3' position of the sugar on the 3' terminal nucleoside or in 2'-5' linked oligonucleotides and the 5' position of 5' terminal nucleotide.
- Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
- Modifications of the inhibitory nucleic acid molecules described herein may include one or more of the following internucleoside modifications: phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'- alkylene phosphonates, phosphinates, phosphoramidates including 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates, and boranophosphates having normal 3'-5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage.
- any of the inhibitory nucleic acid molecules described herein may be modified via the addition of an auxiliary moiety, e.g., a cell penetrating peptide (CPP), a polymer, a hydrophobic moiety, or a targeting moiety.
- the auxiliary moiety may be present as a 5’ terminal modification (e.g., covalently bonded to a 5’- terminal nucleoside), a 3’ terminal modification (e.g., covalently bonded to a 3’-terminal nucleoside), or an internucleoside linkage (e.g., covalently bonded to phosphate or phosphorothioate in an internucleoside linkage).
- CPPs are known in the art (e.g., TAT or Arg8) (Snyder and Dowdy, 2005, Expert Opin. Drug Deliv. 2, 43-51 ). Specific examples of CPPs are provided in WO2011157713, which is incorporated herein by reference in its entirety.
- Inhibitory nucleic acid molecules of the disclosure may include covalently attached neutral polymer-based auxiliary moieties.
- Neutral polymers include poly(C1 -6 alkylene oxide), e.g., polyethylene glycol) and polypropylene glycol) and copolymers thereof, e.g., di- and triblock copolymers.
- An inhibitory nucleic acid molecule containing a hydrophobic moiety may exhibit superior cellular uptake, as compared to an inhibitory nucleic acid molecule lacking the hydrophobic moiety.
- a hydrophobic moiety is a monovalent group (e.g., a bile acid (e.g., cholic acid, taurocholic acid, deoxycholic acid, oleyl lithocholic acid, or oleoyl cholenic acid), glycolipid, phospholipid, sphingolipid, isoprenoid, vitamin, saturated fatty acid, unsaturated fatty acid, fatty acid ester, triglyceride, pyrene, porphyrine, texaphyrine, adamantine, acridine, biotin, coumarin, fluorescein, rhodamine, Texas-Red, digoxygenin, dimethoxytrityl, t-butydimethylsilyl, t-butyldiphenylsilyl, cyanine dye (e.g., Cy3 or Cy5), Hoechst 33258 dye, psoralen, or ibuprofen) covalently linked to the nucleic
- a targeting moiety is selected based on its ability to target oligonucleotides of the invention to a desired or selected cell population that expresses the corresponding binding partner (e.g., either the corresponding receptor or ligand) for the selected targeting moiety.
- a binding partner e.g., either the corresponding receptor or ligand
- an oligonucleotide of the invention could be targeted to hepatocytes expressing asialoglycoprotein receptor (ASGP-R) by selecting a targeting moiety containing N-acetylgalactosamine (GalNAc).
- the targeting moiety is vascular cell adhesion protein 1 (VCAM1). In some embodiments, the targeting moiety is arginylglycylaspartic acid (RGD).
- a targeting moiety may include one or more ligands (e.g., 1 to 9 ligands, 1 to 6 ligands, 1 to 3 ligands, 3 ligands, or 1 ligand).
- the ligand may target a cell expressing asialoglycoprotein receptor (ASGP-R), IgA receptor, HDL receptor, LDL receptor, or transferrin receptor.
- ASGP-R asialoglycoprotein receptor
- Non-limiting examples of the ligands include N-acetylgalactosamine (e.g., a triantennary N-acetylgalactosamine), glycyrrhetinic acid, glycyrrhizin, lactobionic acid, lactoferrin, IgA, or a bile acid (e.g., litrocholyltaurine or taurocholic acid).
- N-acetylgalactosamine e.g., a triantennary N-acetylgalactosamine
- glycyrrhetinic acid glycyrrhizin
- lactobionic acid lactoferrin
- IgA lactoferrin
- a bile acid e.g., litrocholyltaurine or taurocholic acid
- the ligand may be a small molecule, e.g., a small molecule targeting a cell expressing asialoglycoprotein receptor (ASGP-R).
- ASGP-R asialoglycoprotein receptor
- a non-limiting example of a small molecule targeting an asialoglycoprotein receptor is N-acetylgalactosamine.
- the ligand can be an antibody or an antigen-binding fragment or an engineered derivative thereof (e.g., Fcab or a fusion protein (e.g., scFv)).
- Inhibitory nucleic acid molecules of the disclosure may be prepared using techniques and methods known in the art for the oligonucleotide synthesis.
- inhibitory nucleic acid molecules of the disclosure may be prepared using a phosphoramidite-based synthesis cycle.
- This synthesis cycle includes the steps of (1 ) de-blocking a 5’-protected nucleotide to produce a 5’-deblocked nucleotide, (2) coupling the 5’-deblocked nucleotide with a 5’-protected nucleoside phosphoramidite to produce nucleosides linked through a phosphite, (3) repeating steps (1 ) and (2) one or more times as needed, (4) capping the 5’-terminus, and (5) oxidation or sulfurization of internucleoside phosphites.
- the reagents and reaction conditions useful for the oligonucleotide synthesis are known in the art.
- the inhibitory nucleic acid molecules disclosed herein may be linked to solid support as a result of solid-phase synthesis.
- Cleavable solid supports that may be used are known in the art.
- Non-limiting examples of the solid support include, e.g., controlled pore glass or macroporous polystyrene bonded to a strand through a cleavable linker (e.g., succinate-based linker) known in the art (e.g., UnyLinkerTM).
- a nucleic acid linked to solid support may be removed from the solid support by cleaving the linker connecting a nucleic acid and solid support.
- the inhibitory nucleic acid molecules described herein may be formulated into various compositions (e.g., a pharmaceutical composition) for administration to a subject in a biologically compatible form suitable for administration in vivo.
- the inhibitory nucleic acid molecules described herein e.g., the siRNA molecules of SEQ ID NOs: 1 -12, or variants thereof
- a suitable diluent, carrier, or excipient may further contain a preservative, e.g., to prevent the growth of microorganisms.
- Conventional procedures and ingredients for the selection and preparation of suitable compositions are described, for example, in Remington, J.P. The Science and Practice of Pharmacy, Easton, PA. Mack Publishers, 2012, 22 nd ed. And in The United States Pharmacopeial Convention, The National Formulary, United States
- compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to any other animal, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of the pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates and mammals.
- compositions containing the inhibitory nucleic acids described herein may further include a second therapeutic agent (e.g., a nucleic acid molecule to be expressed within a cell, a polypeptide, or a drug).
- a second therapeutic agent may be a blood pressure medication, an antiinflammatory medication (e.g., a steroid or colchicine), or immunosuppressive agent.
- the second therapeutic agent is a statin.
- Non-limiting examples of second therapeutic agents are a statin (e.g., atorvastatin), a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor (e.g., an siRNA or monoclonal antibody targeting PCSK9), hepatocyte growth factor (HGF), inclisiran (e.g., LEQVIOTM), or ezetimibe (e.g., ZETIATM).
- a statin e.g., atorvastatin
- PCSK9 inhibitor e.g., an siRNA or monoclonal antibody targeting PCSK9
- HGF hepatocyte growth factor
- inclisiran e.g., LEQVIOTM
- ezetimibe e.g., ZETIATM
- the second therapeutic agent e.g., statin
- the second therapeutic agent is administered in combination with an inhibitory nucleic acid molecule of the disclosure.
- the subject is orally administered a statin.
- the subject is administered a statin daily.
- the disclosure provides methods of (i) treating a medical condition resulting from a myocardial infarction (Ml), and/or (ii) promoting angiogenesis in a subject.
- the method includes the step of administering to a subject an inhibitory nucleic acid molecule described herein, wherein the inhibitory nucleic acid molecule targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- DCN e.g., SEQ ID NO: 14
- a variant thereof e.g., see Table 3
- the method includes the step of administering to a subject an siRNA described herein (e.g., any one or more of SEQ ID NOs: 1 -12, or a variant thereof), wherein the siRNA targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the method includes the step of administering to a subject an dsRNA described herein, wherein the dsRNA targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the method includes the step of administering to a subject an ASO described herein, wherein the ASO targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the method includes the step of administering to a subject a gapmeR described herein, wherein the gapmeR targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the method includes the step of administering to a subject an miRNA described herein, wherein the miRNA targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the method includes the step of administering to a subject an shRNA described herein, wherein the shRNA targets DCN (e.g., SEQ ID NO: 14), or a variant thereof (e.g., see Table 3).
- the medical complication is an arrhythmic-related complication (e.g., a heart block, an atrial arrhythmia, and/or a ventricular arrhythmia), an ischemic-related complication (e.g., reinfarction, peri-infarct ischemia, and/or an infarct extension), a mechanical-related complication (e.g., a mitral valve rupture or tear, a chordae rupture or tear, a ventricular septal defect (VSD), a ventricular free wall rupture, a cardiac tamponade, and/or an aneurysm), an inflammatory-related complication (e.g., pericarditis and/or Dressier syndrome), and/or a systemic complication (e.g., cardiogenic shock, cardiomyopathy, heart failure, an embolic stroke, a systemic embolism, and/or a lower extremity embolism).
- arrhythmic-related complication e.g., a
- any of the methods can administer a composition (e.g., a pharmaceutical composition) or delivery vehicle (e.g., a vector or nanoparticle) that contains or expresses any of the inhibitory nucleic acid molecules described herein (e.g., siRNA, dsRNA, miRNA, shRNA, ASO, or gapmeR).
- a composition e.g., a pharmaceutical composition
- delivery vehicle e.g., a vector or nanoparticle
- any of the inhibitory nucleic acid molecules described herein e.g., siRNA, dsRNA, miRNA, shRNA, ASO, or gapmeR.
- the methods of (i) treating a medical condition resulting from a myocardial infarction (Ml), and/or (ii) promoting angiogenesis in a subject further includes administering a second therapeutic agent (e.g., a nucleic acid molecule to be expressed within a cell, a polypeptide, or a drug).
- a second therapeutic agent e.g., a nucleic acid molecule to be expressed within a cell, a polypeptide, or a drug.
- a second therapeutic agent may be a blood pressure medication, an anti-inflammatory medication (e.g., a steroid or colchicine), or immunosuppressive agent.
- the second therapeutic agent is a statin.
- Non-limiting examples of second therapeutic agents are a statin (e.g., atorvastatin), a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor (e.g., an siRNA or monoclonal antibody targeting PCSK9), hepatocyte growth factor (HGF), inclisiran (e.g., LEQVIOTM), or ezetimibe (e.g., ZETIATM).
- the additional therapeutic agent can be administered prior to, subsequent to, or concurrently with an inhibitory nucleic acid described herein.
- the inhibitory nucleic acid molecule of the disclosure may be delivered to a subject (e.g., a human) using any suitable delivery vehicle.
- a delivery vehicle for any of the inhibitory nucleic acid molecules described herein may be a vector, plasmid, or nano particle, (e.g., a micelle, a liposome, an exosome, or a lipid nano particle (LNP)).
- LNP lipid nano particle
- the inhibitory nucleic acid molecule of the disclosure and compositions thereof may be delivered to a subject via a vector (e.g., a viral vector).
- a viral vector system can be used including, e.g., adenoviruses (e.g., Ad2, Ad5, Ad9, Ad15, Ad17, Ad19, Ad20, Ad22, Ad26, Ad27, Ad28, Ad30, or Ad39), rhabdoviruses (e.g., vesicular stomatitis virus), retroviruses, adeno-associated vectors (AAV), poxviruses, herpes viral vectors, and Sindbis viral vectors.
- the vector may be an AAV vAAV1 , AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11 , or AAV12 vector.
- the inhibitory nucleic acid molecule of the disclosure and compositions thereof may be delivered to a subject via liposomes.
- Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as a delivery vehicle for the administration of the inhibitory nucleic acids described herein, and compositions thereof.
- Liposomes can be of different sizes such as, but not limited to, a multilamellar vesicle (MLV) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, a small unicellular vesicle (SUV) which may be smaller than 50 nm in diameter, and a large unilamellar vesicle (LUV) which may be between 50 and 500 nm in diameter.
- MLV multilamellar vesicle
- SUV small unicellular vesicle
- LUV large unilamellar vesicle
- Liposome design may include, but is not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis.
- Liposomes may contain a low or a high pH in order to improve the delivery of the pharmaceutical composition.
- exosomes produced from cells can be collected from cell culture medium by any suitable method.
- a preparation of exosomes can be prepared from cell culture or tissue supernatant by centrifugation, filtration or combinations of these methods.
- exosomes can be prepared by differential centrifugation, that is low speed ( ⁇ 20000 g) centrifugation to pellet larger particles followed by high speed (>100000 g) centrifugation to pellet exosomes, size filtration with appropriate filters (for example, 0.22 micrometer filter), gradient ultracentrifugation (for example, with sucrose gradient) or a combination of these methods.
- the inhibitory nucleic acid molecules of the disclosure, and compositions thereof, may be delivered to a subject via LNPs.
- the inhibitory nucleic acid molecules e.g., siRNA, dsRNA, miRNA, shRNA, ASO, or gapmeR
- a lipid nanoparticle such as those described in International Publication No. WO2012170930, herein incorporated by reference in its entirety.
- LNP formulations may contain cationic lipids, distearoylphosphatidylcholine (DSPC), cholesterol, polyethylene glycol (PEG), R-3-[(w-methoxy polyethylene glycol)2000)carbamoyl)]-1 ,2- dimyristyloxl-propyl-3-amine (PEG-c-DOMG), distearoyl-rac-glycerol (DSG) and/or dimethylaminobutanoate (DMA).
- DSPC distearoylphosphatidylcholine
- DMA dimethylaminobutanoate
- the PEG-c-DOMG may be replaced with a PEG lipid such as, but not limited to, PEG-DSG (1 ,2-Distearoyl-sn-glycerol, methoxypoly ethylene glycol) or PEG-DPG (1 ,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol).
- PEG-DSG 1,2-Distearoyl-sn-glycerol, methoxypoly ethylene glycol
- PEG-DPG 1,2-Dipalmitoyl-sn-glycerol, methoxypolyethylene glycol
- the cationic lipid may be selected from any lipid known in the art such as, but not limited to, (6Z,9Z,28Z,31Z)- heptatriacont-6,9,28,31 -tetraene-19-yl 4-(dimethylamino)butanoate (DLin-MC3-DMA), 1 ,2-dil inoleyloxy- n,n-dimethyl-3-aminopropane (DLin-DMA), C 12-200, and N,N-dimethyl-2,2-di-(9Z,12Z)-9,12- octadecadien-1 -yl-1 ,3-dioxolane-4-ethanamine (DLin-KC2-DMA).
- Exemplary commercial reagents useful for lipid-based delivery of inhibitory nucleic acid molecules including, but not limited to, TransIT-TKOTM (Mirus, Catalog No. MIR 2150), Trans messengergerTM (Qiagen, Catalog No. 301525), OligofectamineTM and LipofectamineTM (Invitrogen, Catalog No. MIR 12252-011 and Catalog No. 13778-075), siPORTTM (Ambion, Catalog No. 1631 ), and DharmaFECTTM (Fisher Scientific, Catalog No. T-2001 -01 ). Subject
- the subject to be treated may have previously experienced a myocardial infarction (Ml).
- the subject to be treated may have an ischemic injury, which may have occurred as a result of the Ml.
- the subject to be treated may have a cardiovascular disease, including, but not limited to, coronary artery disease, peripheral artery disease.
- the subject may also have, or be at risk of developing, a stroke.
- the subject to be treated may have a metabolic disorder, or is at risk of developing a metabolic disorder, such as diabetes.
- Subjects at risk of developing diabetes may be prediabetic and/or have experienced one or more of the following risk factors: hyperglycemia, glucose resistance, insulin resistance, hyperlipidemia, or has a family history of diabetes.
- the inhibitory nucleic acid molecules of the disclosure, and compositions thereof may be delivered to the subject’s coronary endothelium, remote zone of the heart, and/or border zone of the heart.
- the inhibitory nucleic acid molecules of the disclosure, and compositions thereof may be delivered to an endothelial cell in the subject (e.g., an endothelial cell, a cardiomyocyte, a fibroblast, a vascular smooth muscle cell, and/or a leukocyte in the subject’s coronary endothelium, remote zone, and/or border zone).
- Such delivery can promote angiogenesis in the subject, e.g., by stimulating endothelial cell proliferation.
- the actual dosage amount of a composition of the present disclosure administered to a subject can be determined by physical and physiological factors such as body weight, severity of condition, previous or concurrent therapeutic interventions, idiopathy of the patient and on the route of administration. Depending upon the dosage (e.g., mg/kg) and the route of administration, the number of administrations of a preferred dosage and/or an effective amount may vary according to the response of the subject. The practitioner responsible for administration will, in any event, determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. Administration may occur any suitable number of times per day, and for as long as necessary. Subjects may be adult or pediatric humans, with or without comorbid diseases.
- compositions utilized in the methods described herein can be administered to a subject by any suitable route of administration.
- a composition containing an inhibitory nucleic acid of the disclosure may be administered intramuscularly, intravenously, intradermally, percutaneously, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, by catheter, by lavage, in cremes, or in lipid compositions.
- compositions utilized in the methods described herein can be administered to the subject intravenously. In some embodiments, the compositions utilized in the methods described herein can be administered to the subject subcutaneously. In some embodiments, the compositions utilized in the methods described herein can be administered to the subject intraarticularly. In some embodiments, the compositions utilized in the methods described herein can be administered to the subject intramuscularly.
- Example 1 MicroRNA-342-3p Rescues Post-Ischemic Cardiac Function in Diabetic Mice via Activation of the HGF-MET Pathway
- This example describes the discovery of miR-342-3p’s role in regulating angiogenesis under physiologic and diabetic conditions. Additionally, this example describes the discovery of decorin (DCN) as a therapeutic target for promoting post-ischemic angiogenesis, which can be used to treat or reduce the likelihood of a medical complication resulting from myocardial infarction (Ml).
- DCN decorin
- miRNAs were sequenced from cardiac endothelial cells (ECs) obtained from Ldlr / - mice on either chow or high fat sucrose containing (HFSC) diet at 0-, 3-, 7- and 14-days post-MI. Among the top differentially expressed miRNAs at day 14 was miR-342-3p (FIG. 11 A). In vitro, overexpression of miR-342-3p promotes angiogenic sprouting and migration in endothelial cells (ECs) by activating the hepatocyte growth factor (HGF)-mesenchymal-epithelial transition factor (MET) signaling pathway.
- HGF hepatocyte growth factor
- MET meenchymal-epithelial transition factor
- miR-342-3p Delivery of miR-342-3p mimics to the border zone increased capillary density, EC proliferation, and cardiac function post-MI.
- DCN was identified as a direct target of miR-342-3p and validated by Western blot (WB) and luciferase reporter assays.
- RNA sequencing and subsequent WBs implicated HGF-MET signaling as one of the top upregulated pathways in miR-342-3p overexpressing ECs (FIG. 11 B).
- both high glucose (25 mM) and palmitate (100 pM) treatments had the opposite effect.
- DCN and MET binding was confirmed by co-immunoprecipitation assay.
- RNA from the EC fraction was sequenced and the raw data analyzed for differentially expressed (fold change (FC) > 1 .5, adjusted p value ⁇ 0.05) miRNAs between chow and HFSC groups.
- the resulting list of miRNAs had unique expression kinetics across different time points, indicating the diverse roles played by miRNAs during various phases following ischemic insult, e.g., inflammation, angiogenesis, and vascular remodeling.
- ischemic insult e.g., inflammation, angiogenesis, and vascular remodeling.
- day- 14 the peak of the angiogenesis phase, was interrogated.
- miR-342-3p was among the top dysregulated miRNAs from this list. From the miRNA-seq data, miR-342-3p was higher in the chow group vs the HFSC group at day 14 (FIG.
- rniR-342 m human umbilical vein endothelial cells
- spheroid sprouting assay scratch wound closure assay
- BrdU incorporation proliferation
- spheroids composed of miR-342 inhibitor (miR-342i) transfected HUVECs showed much lower number of sprouts as well as cumulative sprout length vs non-specific (NS) control (NSi) (FIG. 1 B and FIG. 1 C).
- scratch closure assays were performed. Scratches created in a monolayer of rniR-342 m transfected HUVECs closed significantly faster compared to those in the NSm group (FIG. 1 D and FIG. 1 E). This was confirmed by area under curve (AUC) analysis (FIG. 1 F). In contrast, the miR-342i scratches failed to close completely even after 24 hours (FIG. 1 D and FIG. 1 E).
- HUVECs transfected with either control or miR-342 mimic were incubated with the nucleoside analogue, BrdU (5-Bromo-2'- deoxyuridine).
- BrdU 5-Bromo-2'- deoxyuridine
- Cells in the synthesis (s) phase of cell division incorporated this thymidine analog in their DNA.
- a colorimetric test was used to quantify the BrdU levels in both groups. rniR-342 m HUVECs had significantly higher BrdU incorporation vs control, while inhibition of miR-342 reduced endothelial proliferation dramatically (FIG. 1 G)
- Predicted miR-342-3p targets were cross-referenced with the top downregulated mRNAs (day 0 vs day 14) in the chow group (FIG. 2A) and arrived at four potential targets. Of these, only DCN was significantly downregulated at the protein level (FIG. 2B and FIG. 2C). DCN is a small leucine rich protein which stabilizes collagen fibers and is a component of the extracellular matrix. Soluble DCN is secreted and has been shown to bind to various extracellular receptors by competing with their canonical ligands. While miR-342 overexpression repressed DCN protein, its inhibition promoted DCN expression, suggesting that DCN is a novel target of miR-342.
- DCN mRNA was confirmed as a target of miR-342 using a 3’-UTR luciferase reporter assay (FIG. 2D). Luciferase activity was significantly reduced in wildtype DCN 3’ UTR transfected cells when co-transfected with miR-342 mimic vs scramble control (FIG. 2D). Mutation of the 3’ UTR seed sequence abolished transcriptional repression by miR-342 (FIG. 2D). miR-342 overexpression promotes HGF-MET signaling in ECs
- RNA from miR-342 overexpressing HUVECs was sequenced and analyzed for differentially expressed (DE) genes (DEGs).
- DE differentially expressed
- RNA-seq results showed that one of the top upregulated genes was MET, which codes for the c-MET receptor.
- HGF is the natural ligand of the MET receptor.
- HGF-MET signaling is a well-studied pathway involved in cell proliferation[26- 28]. It has been implicated in angiogenesis in diverse tissue types and in different disease contexts[29- 31].
- IPA Ingenuity Pathway Analysis revealed HGF-MET signaling as one of the top upregulated pathways (FIG. 2E).
- MET receptor mRNA and protein was upregulated in ECs transfected with miR-342 mimic vs control at the transcript and protein level (FIG. 2F and FIG. 2G), validating this studies RNA-seq results.
- miR-342 also induced HGF mRNA expression (FIG. 2H). This result is also mirrored in the RNA-seq data from the initial Ml study, where MET mRNA levels were higher in the chow group at day 14 vs HFSC (data not shown).
- miR-342-3p expression is responsive to hypoxia, high glucose, and palmitate treatments
- ECs can sense and respond to diverse stimuli such as oxygen concentration, circulating glucose, fatty acid levels, or paracrine factors.
- ischeemia hypoxia/dyslipidemia
- ECs were pre-treated with either 25 mM mannitol or 25 mM glucose for 48 hours under normoxic conditions, followed by 0, 2, 16 or 24 hours of hypoxia (2% O2).
- ECs were treated with 100 uM Palmitate or 100 uM BSA and 2% O2 for 0, 2, 16 or 24 hours.
- Ejection fraction (EF) and fractional shortening (FS) was significantly improved in the miR-342 injected group vs control at day 14 (FIG. 4C and FIG. 4D).
- miR-342-3p overexpression was confirmed in the border zone by qPCR (FIG. 4D).
- DCN expression was significantly reduced in the miR-342 group vs control, while MET mRNA was upregulated (FIG. 4E and FIG. 4F).
- Immunofluorescence on heart sections revealed increased capillary density and EC proliferation in miR- 342 mimic vs control group (FIGS. 4G-I and FIG. 11 C).
- ECs were transfected with scramble or miR-342 mimic and added a selective small molecule MET inhibitor, PHA-665752 or its vehicle (DMSO) and performed a proliferation assay using BrdU pulse-chase approach. While miR-342 overexpression in the vehicle group dramatically increased proliferation, the addition of the MET inhibitor significantly blunted this effect (FIG. 5A). This experiment was repeated under high glucose or mannitol conditions. Results show that the inhibitor reduced proliferation under basal conditions; but in the high glucose group ECs proliferated less, even in the absence of this inhibitor.
- DMSO selective small molecule MET inhibitor
- rDCN recombinant DCN protein
- ECs were transfected with either scramble control or miR-342 mimic with or without rDCN (10 nM).
- rDCN significantly reduced the proliferative effect of miR-342, while its addition had no effect on the scramble control group (FIG. 5B).
- Supplementation with HGF significantly increased proliferation in NSm and rniR-342 m treated groups, while addition of MET inhibitor or rDCN reduced it in the rniR-342 m treated group only (FIG. 5C and FIG. 5D).
- rDCN reduced proliferation (FIG. 5G and FIG. 5H).
- High glucose treated NSm cells were significantly less proliferative vs rnannitol-NSm.
- miR-342 rescued proliferation under high glucose conditions.
- Addition of rDCN did not inhibit proliferation in either NSm or rniR-342 m groups in the high glucose treated group (FIG. 5G and FIG. 5H).
- These results demonstrate the attenuation of miR-342 induced proliferation by DCN.
- inhibition of MET signaling abolishes miR-342-induced endothelial proliferation.
- the effects of the MET inhibitor (PHA-665752) and rDCN on EC proliferation are strikingly similar across the experiments, supporting DCN as an inhibitor of the MET receptor and counters the pro-proliferative effect of miR-342.
- DCN Having established DCN as a target of miR-342, the effect of its silencing on endothelial proliferation and angiogenesis was investigated, along with whether siRNA mediated knockdown (KD) of DCN phenocopied the effects of miR-342 overexpression.
- miR-342 overexpression suppressed DCN protein while boosting MET receptor expression (FIGS. 6A-6C).
- the increase in MET expression in siDCN treated cells phenocopies the effect of miR-342 overexpression.
- mannitol or glucose treated ECs were transfected with siDCN (ThermoFisher, USA) or a non-specific/scramble control (siNS) followed by a BrdU incorporation assay.
- siDCN ThermoFisher, USA
- siNS non-specific/scramble control
- BrdU incorporation assay As observed in similar experiments described above, high glucose treatment significantly reduced endothelial proliferation vs control.
- DCN KD significantly upregulated MET protein under basal conditions, mirroring the effect of miR-342 overexpression (FIG. 6D and FIG. 6E).
- spheroid sprouting assays were performed on ECs transfected with either siNS or siDCN and cultured in either mannitol or high glucose containing media.
- siDCN spheroids had significantly more sprouts vs siNS spheroids (FIG. 6I and FIG. 6M).
- DCN KD also significantly increased the number of sprouts under high glucose conditions.
- the cumulative sprout length was also higher in siDCN group vs siNS under both mannitol and glucose conditions.
- Parallel experiments tested the effects of miR-342 overexpression (rniR-342 m or NSm) on spheroid sprouting under similar conditions.
- rniR-342 m spheroids exhibited enhanced sprouting vs NSm under both conditions (FIG. 6H and FIG. 6L). Further, supplementing the culture medium with rDCN abrogated the effects of miR-342 overexpression (FIG. 6J and FIG. 6N). Similarly, the addition of rDCN also suppressed siDCN mediated sprouting (FIG. 6K and FIG. 60). Taken together, these results demonstrate that DCN deficiency phenocopies the effects of miR-342 overexpression on angiogenesis and that DCN supplementation antagonizes these effects. miR-342-3p acts via the HGF-MET signaling pathway in both mice and humans
- a BrdU proliferation assay demonstrated that EC proliferation was induced by HGF (50 ng/mL) supplementation in HUVECs transfected with a miR-342 mimic, relative to control (FIG. 9A).
- HGF 50 ng/mL
- PHA-665752 blunted EC proliferation observed by the miR-342 mimic under normal (mannitol) conditions.
- HUVECs proliferated less under high glucose conditions even in the absence of the inhibitor, which did not synergistically reduce proliferation (FIG. 9B).
- mice Male Ldlr-/- mice were placed on a high fat sucrose containing (HFSC) diet for 4 weeks, followed by temporary left anterior descending (LAD) coronary artery ligation surgery. Mice were subjected to 45 minutes of myocardial ischemia followed by reperfusion. Immediately before reperfusion, either lipid encapsulated miR-342-3p mimic or non-specific mimic (40 ng per animal) was injected into the border zone of the heart. Mimics were encapsulated with 20% (v/v) Lipofectamine 2000 reagent and delivered as a 20 pL volume (2 injections of 10 pL each) on the left and right margins of the border zone. Echocardiography was used to assess cardiac function at days 3, 7 and 14 post-surgery. Mice continued their special diet during this period. Mimics were encapsulated in Lipofectamine 2000 as described below:
- UCUCACACAGAAAUCGCACCCGU-3’ SEQ ID NO: 13
- mouse miR-342-3p mimic or non-specific control was prepared by dissolving 250 nmol or 1867 ug of powder in 250 pL of nuclease-free water.
- One pL of this stock solution was added to 99 pL of sterile 1x PBS to get a 74.70 ng/pL working solution.
- the negative control or scramble mimic was prepared in a similar fashion.
- miR-342-3p sequences (same human and mouse):
- HGF receptor up-regulation contributes to the angiogenic phenotype of human endothelial cells and promotes angiogenesis in vitro. Blood 101 (12):4816-4822.
- Hepatocyte growth factor is a survival factor for endothelial cells and is expressed in human atherosclerotic plaques. Atherosclerosis 164 (1 ):79-87.
- An inhibitory nucleic acid molecule comprising sufficient complementarity to a target nucleic acid molecule, wherein (i) the inhibitory nucleic acid molecule is at least 15 nucleotides in length, and (ii) the target nucleic acid molecule comprises a nucleotide sequence having at least 80% sequence identity to SEQ ID NO: 14. 2.
- the inhibitory nucleic acid molecule of embodiment 1 wherein the target nucleic acid molecule comprises a nucleotide sequence having at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to SEQ ID NO: 14.
- inhibitory nucleic acid molecule of embodiment 1 or 2 wherein the target nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 14.
- inhibitory nucleic acid molecule of embodiment 4 wherein the inhibitory nucleic acid molecule is 15 to 49 nucleotides in length, 50 to 99 nucleotides in length, or 100 to 6,850 nucleotides in length.
- inhibitory nucleic acid molecule of embodiment 5 wherein the inhibitory nucleic acid molecule is 20 to 28 nucleotides in length.
- inhibitory nucleic acid molecule of embodiment 8 wherein the inhibitory nucleic acid molecule comprises at least 90% complementarity to the target nucleic acid molecule.
- inhibitory nucleic acid molecule of embodiment 9 wherein the inhibitory nucleic acid molecule comprises at least 95% complementarity to the target nucleic acid molecule.
- inhibitory nucleic acid molecule of embodiment 10 wherein the inhibitory nucleic acid molecule is complementary to the target nucleic acid molecule.
- inhibitory nucleic acid molecule of any one of embodiments 1 -11 , further comprising a modification.
- inhibitory nucleic acid molecule of embodiment 12, wherein the modification comprises:
- the non-natural or modified nucleoside or nucleotide is selected from the group consisting of: a locked nucleic acid (LN A), a 2'-O-methyl (2'-O-Me) modified nucleoside, a phosphorothioate (PS) bond between nucleosides, and a 2'-fluoro (2’-F) modified nucleoside; and/or
- the covalently or non-covalently conjugated moiety is selected from the group consisting of: a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer.
- siRNA small interfering RNA
- dsRNA doublestranded RNA
- miRNA microRNA
- shRNA short hairpin RNA
- ASO anti-sense oligonucleotide
- inhibitory nucleic acid molecule of embodiment 16 wherein the siRNA comprises an antisense strand comprising at least 88% sequence identity to any one of SEQ ID NOs: 1 -6. 18.
- antisense strand comprises at least 92% sequence identity to any one of SEQ ID NOs: 1 -6.
- inhibitory nucleic acid molecule of embodiment 18, wherein the antisense strand comprises at least 96% sequence identity to any one of SEQ ID NOs: 1 -6.
- inhibitory nucleic acid molecule of embodiment 19, wherein the antisense strand comprises the nucleotide sequence of any one of SEQ ID NOs: 1 -6.
- the non-natural or modified nucleoside or nucleotide is selected from the group consisting of: a locked nucleic acid (LNA), a 2'-O-methyl (2'-O-Me) modified nucleoside, a phosphorothioate (PS) bond between nucleosides, and a 2’-fluoro (2'-F) modified nucleoside; and/or
- LNA locked nucleic acid
- PS phosphorothioate
- the covalently or non-covalently conjugated moiety is selected from the group consisting of: a targeting moiety, a hydrophobic moiety, a cell penetrating peptide, or a polymer.
- inhibitory nucleic acid molecule of embodiment 26 or 27, wherein the miRNA comprises a nucleotide sequence comprising at least 86% sequence identity to SEQ ID NO: 13.
- inhibitory nucleic acid molecule of embodiment 30, wherein the miRNA comprises the nucleotide sequence of SEQ ID NO: 13.
- inhibitory nucleic acid molecule of embodiment 31 wherein the miRNA is miR-342-3p.
- the inhibitory nucleic acid molecule of embodiment 33 wherein the delivery vehicle is selected from the group consisting of: a vector, a plasmid, a micelle, a liposome, an exosome, and a lipid nano particle (LNP).
- the delivery vehicle is selected from the group consisting of: a vector, a plasmid, a micelle, a liposome, an exosome, and a lipid nano particle (LNP).
- inhibitory nucleic acid molecule of embodiment 34 wherein the vector is a viral vector.
- inhibitory nucleic acid molecule of embodiment 36 wherein the pharmaceutical composition comprises a pharmaceutically acceptable excipient, diluent, and/or carrier.
- a method of treating or reducing the likelihood of a medical complication resulting from a myocardial infarction (Ml) in a subject comprising administering the inhibitory nucleic acid molecule of any one of embodiments 1 -37.
- the medical complication is an arrhythmic-related complication, an ischemic-related complication, a mechanical-related complication, an inflammatory- related complication, and/or a systemic complication.
- the arrhythmic-related complication is a heart block, an atrial arrhythmia, and/or a ventricular arrhythmia;
- the ischemic-related complication is reinfarction, peri-infarct ischemia, and/or an infarct extension;
- the mechanical-related complication is a mitral valve rupture or tear, a chordae rupture or tear, a ventricular septal defect (VSD), a ventricular free wall rupture, a cardiac tamponade, and/or an aneurysm;
- VSD ventricular septal defect
- the inflammatory-related complication is pericarditis and/or Dressier syndrome;
- the systemic complication is cardiogenic shock, cardiomyopathy, heart failure, embolic stroke, systemic embolism, and/or a lower extremity embolism.
- a method of promoting angiogenesis in a subject comprising administering the inhibitory nucleic acid molecule of any one of embodiments 1 -37.
- inhibitory nucleic acid molecule is delivered to an endothelial cell, a cardiomyocyte, a fibroblast, a vascular smooth muscle cell, and/or a leukocyte.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Biomedical Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Wood Science & Technology (AREA)
- Biophysics (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Cardiology (AREA)
- Heart & Thoracic Surgery (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
L'invention concerne des compositions (par exemple, une molécule d'acide nucléique inhibitrice) pour réduire l'expression de la décorine (DCN) et des méthodes associées pour (i) traiter une affection médicale résultant d'un infarctus du myocarde (IDM), et/ou (II) favoriser l'angiogenèse chez un sujet. La molécule d'acide nucléique inhibitrice peut être un petit ARN interférent (ARNsi), un ARN double brin (ARNdb), un oligonucléotide antisens (ASO), un microARN (miARN), ou un ARN court en épingle à cheveux (ARNsh)), ou un gapmeR décrit ici, ou une composition (par exemple, une composition pharmaceutique) associée. De manière avantageuse, la composition décrite ici assure des effets thérapeutiques (par exemple, l'angiogenèse) pour un tissu cardiaque suite à un infarctus du myocarde (IDM).
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202463626184P | 2024-01-29 | 2024-01-29 | |
| US63/626,184 | 2024-01-29 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2025165814A1 true WO2025165814A1 (fr) | 2025-08-07 |
| WO2025165814A8 WO2025165814A8 (fr) | 2025-09-18 |
Family
ID=96591424
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2025/013510 Pending WO2025165814A1 (fr) | 2024-01-29 | 2025-01-29 | Compositions et méthodes de régulation de l'ischémie post-angiogenèse cardiaque |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025165814A1 (fr) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20030198983A1 (en) * | 2002-02-01 | 2003-10-23 | Affymetrix, Inc. | Methods of genetic analysis of human genes |
| US20050244851A1 (en) * | 2004-01-13 | 2005-11-03 | Affymetrix, Inc. | Methods of analysis of alternative splicing in human |
| US20100273180A1 (en) * | 2009-03-20 | 2010-10-28 | Banerjee Abhijit G | Decorin polypeptide and methods and compositions of use thereof |
-
2025
- 2025-01-29 WO PCT/US2025/013510 patent/WO2025165814A1/fr active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20030198983A1 (en) * | 2002-02-01 | 2003-10-23 | Affymetrix, Inc. | Methods of genetic analysis of human genes |
| US20050244851A1 (en) * | 2004-01-13 | 2005-11-03 | Affymetrix, Inc. | Methods of analysis of alternative splicing in human |
| US20100273180A1 (en) * | 2009-03-20 | 2010-10-28 | Banerjee Abhijit G | Decorin polypeptide and methods and compositions of use thereof |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2025165814A8 (fr) | 2025-09-18 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP5697988B2 (ja) | 干渉rnaを使用したポロ様キナーゼ発現のサイレンシング方法 | |
| JP2022008404A (ja) | C/EBPα小分子活性化RNA組成物 | |
| JP5873168B2 (ja) | Hsp47発現の調節を増強するレチノイド−リポソーム | |
| US11464873B2 (en) | RNA-modulating agents | |
| KR20170136542A (ko) | C/EBP 알파 saRNA 조성물 및 사용 방법 | |
| WO2023051822A1 (fr) | Oligonucléotide de ciblage pour le traitement de maladies associées à pcsk9 | |
| JP7707065B2 (ja) | Angptl8を阻害するための新規のrna組成物および方法 | |
| WO2016134146A2 (fr) | Agents thérapeutiques à interférence arn dirigés contre le virus ebola | |
| JP2025061882A (ja) | Apoc3関連疾患および障害の処置のための方法 | |
| JP2024532019A (ja) | アルファ-1アンチトリプシン発現を阻害するための組成物及び方法 | |
| JP2020537653A (ja) | アシアロ糖タンパク質受容体1の発現を阻害するためのRNAi剤および組成物 | |
| WO2025165814A1 (fr) | Compositions et méthodes de régulation de l'ischémie post-angiogenèse cardiaque | |
| WO2013007766A1 (fr) | Moyens et procédés pour le traitement d'une angiogenèse pathologique | |
| US20170362590A1 (en) | Pharmaceutical compositions comprising microrna | |
| WO2025165840A1 (fr) | Compositions et méthodes de traitement d'une maladie cardiovasculaire | |
| WO2025208204A1 (fr) | Compositions et procédés pour inhiber l'expression ou la fonction d'une protéine d'échange activée directement par camp (epac) | |
| WO2024026565A1 (fr) | Compositions et procédés d'inhibition de l'adénylate cyclase 9 (ac9) | |
| JP7525578B2 (ja) | C/EBPα小分子活性化RNA組成物 | |
| US20240150768A1 (en) | Methods for the treatment of nash with advanced fibrosis and/or cirrhosis | |
| EP4162046B1 (fr) | Thérapie pour empêcher un remodelage cardiaque indésirable après un infarctus aigu du myocarde | |
| Mainkar et al. | The Potential of RNA Therapeutics in Treating Cardiovascular Disease | |
| WO2025162482A1 (fr) | Aso pour la prévention et le traitement de métastases cancéreuses et son utilisation | |
| TW202538051A (zh) | 寡核苷酸的遞送載體及其使用方法 | |
| JP2024026903A (ja) | siRNA及び医薬組成物並びに予防及び/又は治療剤 | |
| HK40091624A (en) | Treatment method of left ventricular dysfunction following an acute myocardial infarction |
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: 25748811 Country of ref document: EP Kind code of ref document: A1 |