WO2025077898A1 - A therapeutic nucleic acid-encased nanoworm for gene delivery and endosomal escape - Google Patents
A therapeutic nucleic acid-encased nanoworm for gene delivery and endosomal escape Download PDFInfo
- Publication number
- WO2025077898A1 WO2025077898A1 PCT/CN2024/124523 CN2024124523W WO2025077898A1 WO 2025077898 A1 WO2025077898 A1 WO 2025077898A1 CN 2024124523 W CN2024124523 W CN 2024124523W WO 2025077898 A1 WO2025077898 A1 WO 2025077898A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- nanoworm
- pda
- nws
- cells
- composition
- 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
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7105—Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/711—Natural deoxyribonucleic acids, i.e. containing only 2'-deoxyriboses attached to adenine, guanine, cytosine or thymine and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/51—Nanocapsules; Nanoparticles
- A61K9/5107—Excipients; Inactive ingredients
- A61K9/5115—Inorganic compounds
-
- 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]
Definitions
- NPs nanoparticles
- nucleic acid-based NPs Despite the clinical application of nucleic acid-based NPs, obstacles remain that hinder their efficient intracellular delivery, as the nucleic acid-based NPs are readily degraded by nucleases and possess unfavorable physicochemical properties that impede their uptake by cells.
- the genes delivered by transfection agents often face pronounced endolysosomal entrapment, leading to degradation within lysosomes and therefore inefficient gene release into cytosol. Therefore, their subsequent escape from the acidic organelles is crucial for applications in gene therapy.
- the subject polymer NW can enter cells and escape the endosomes and/or lysosomes and effectively regulate genes.
- the NWs can exit the late endosomes by increasing chloride ion accumulation in the intracellular vesicles, resulting in vesicle swelling and subsequent membrane rupture.
- the platform of the subject invention compared with the widely used transfection agent Lipofectamine 3000, the platform of the subject invention has more effective gene regulation in vitro and in vivo, thereby enhancing cell-based therapy through genetic engineering.
- Figure 2H shows subcellular location of ClC3 (green) relative to intracellular vesicles (red) .
- Scale bar 10 ⁇ m.
- Figure 2I shows RNAi-mediated knockdown of ClC3 significantly disrupted the escape of Au@PDA@T 21 NWs (red) to acidic vesicles (green) .
- Figure 2J shows pharmacological inhibition of general chloride ion channels by niflumic acid (NFA) impaired the escape of Au@PDA@T 21 NWs (red) to acidic vesicles (green) .
- Figures 3A-3H illustrate ion concentration and membrane rupture of endosomes in A549 cells.
- Figure 3A shows Cl - concentration of intracellular vesicles containing Cy5-labeled Au@PDA@T 21 NWs in cells without (upper row) or with siClC3 treatment (lower row) .
- MQAE staining signals are inversely correlated to Cl - concentration.
- White arrows indicate the intracellular vesicles containing Cy5-labeled Au@PDA@T 21 NWs in WT A549 cells have relatively high concentration of Cl - when compared to the A549 cells treated with siClC3 (indicated by magenta arrows) .
- Figures 5A-5K illustrate ex vivo lipid NW-enabled transfection of mRNA in hMSCs for cell-based therapy and alleviating kidney fibrosis.
- Figure 5A shows schematic illustration of the synthesis of mRNA-encased, lipid-coated Au@PDA NWs.
- Figure 5B shows Cy5-labeled Au@PDA@mCXCR4/BMP7 NWs did not accumulate in acidic organelles (green) 24 h post-incubation.
- White number indicates PCC between Cy5-labeled Au@PDA@mCXCR4/mBMP7 NWs (red) and Lysotracker (green) .
- Blue nuclei.
- Figure 5C shows confocal image shows hMSCs that were co-transfected by mRNA expressing CXCR4-GFP (green) and mRNA expressing BMP7-OFP (red) using Au@PDA@lipid NWs 24 h post-incubation.
- Blue nuclei
- Figure 5D shows schematic illustration of hMSC-based therapy of kidney fibrosis in mice. After ligating the left ureter twice with sutures, UUO mice received a single i.v. injection of saline, or hMSCs under different treatments on Day 3 and were sacrificed on Day 14 post-UUO surgery.
- Figure 5E shows quantification of CXCR4 and BMP7 expression in vitro.
- Figure 7D illustrates organ-level distribution of DiR-labeled hMSC, Lipo-hMSC HGF , and LNW-hMSC HGF in the STZ-induced diabetic mice 24 h post-injection.
- Figure 7E shows representative IHC images showing expression of type I collagen and ⁇ -SMA of the UUO kidneys.
- Quantification of Figure 7F shows Col-I-positive and
- Figure 7G shows ⁇ -SMA-positive area from the IHC-stained sections.
- Figures 8A-8C illustrate representative TEM images of citrate-capped 40 nm Au NPs (Figure 8A) , Au 40 @PDA NPs ( Figure 8B) , Au 40 @PDA NWs ( Figure 8C) .
- Figure 24 illustrates validation of siClC5 knockdown in A549 cells.
- Figure 25 illustrates experimental design of pH measurement of intracellular vesicles.
- A549 cells were incubated with a mixture of dextran 10K pHrodo green, dextran 10K AF594, and Cy5-labeled Au@PDA@T 21 NW for 8 h.
- Some vesicles will contain dextran 10K pHrodo green only, dextran 10K AF594 only, both dyes, Cy5-labeled Au@PDA@T 21 NW only, dextran 10K pHrodo green with Cy5-labeled Au@PDA@T 21 NW, dextran 10K AF594 with Cy5-labeled Au@PDA@T 21 NW.
- Only those intracellular vesicles that were positive for both dextran 10K AF594 and dextran 10K pHrodo green signals were considered for pH measurements.
- SEQ ID NO: 3 antisense DNA against enhanced green fluorescence protein (asEGFP)
- SEQ ID NO: 4 Mouse GAPDH Forward sequence 5’ to 3’
- SEQ ID NO: 14 Mouse CD206 Forward sequence 5’ to 3’
- SEQ ID NO: 16 Mouse TNF- ⁇ Forward sequence 5’ to 3’
- SEQ ID NO: 20 Mouse INF- ⁇ Forward sequence 5’ to 3’
- SEQ ID NO: 21 Mouse INF- ⁇ Reverse sequence 3’ to 5’
- SEQ ID NO: 22 Mouse Arg1 Forward sequence 5’ to 3’
- SEQ ID NO: 29 Human RUNX2 Reverse sequence 3’ to 5’
- SEQ ID NO: 31 Human C1C3 Reverse sequence 3’ to 5’
- SEQ ID NO: 33 Human Col-1 Reverse sequence 3’ to 5’
- SEQ ID NO: 35 Mouse ⁇ -SMA Reverse sequence 3’ to 5’
- the subject invention provides effective, safe compositions comprising nanoworms that can be used in methods of delivering nucleic acids to a subject. More specifically, the subject invention provides a polymer-coated nanoworm (NW) . Further, the subject invention provides a therapeutic nucleic acid-encased nanoworm for effective cellular delivery with limited localization inside acidic organelles.
- NW polymer-coated nanoworm
- compositions containing amounts of ingredients where the terms “about” are used these compositions contain the stated amount of the ingredient with a variation (error range) of 0-10%around the value (X ⁇ 10%) .
- the term “about” is providing a variation (error range) of 0-10%around a given value (X ⁇ 10%) .
- this variation represents a range that is up to 10%above or below a given value, for example, X ⁇ 1%, X ⁇ 2%, X ⁇ 3%, X ⁇ 4%, X ⁇ 5%, X ⁇ 6%, X ⁇ 7%, X ⁇ 8%, X ⁇ 9%, or X ⁇ 10%.
- the terms “therapeutically-effective amount, ” “therapeutically-effective dose, ” “effective amount, ” and “effective dose” are used to refer to an amount or dose of a compound or composition that, when administered to a subject, is capable of treating, preventing, or improving a condition, disease, or disorder in a subject. In other words, when administered to a subject, the amount is “therapeutically effective. ” The actual amount will vary depending on a number of factors including, but not limited to, the particular condition, disease, or disorder being treated, prevented, or improved; the severity of the condition; the weight, height, age, and health of the patient; and the route of administration.
- the method comprises administration of multiple doses of the nanoworm of the subject invention.
- the method may comprise administration of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100 or more therapeutically effective doses of a composition comprising the compounds of the subject invention as described herein.
- doses are administered over the course of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 14 days, 21 days, 30 days, 2 months, 3 months, 6 months, 9 months, 1 year, 1.5 years, 2 years, 2.5 years, 5 years, or more than 10 years.
- the frequency and duration of administration of multiple doses of the compositions is such as prevent or treat a disease, such as, for example kidney disease.
- treatment of a subject with a therapeutically effective amount of the compounds of the invention can include a single treatment or can include a series of treatments.
- the effective dosage of a compound used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of testing for kidney disease, such as, for example, glomerular filtration rate.
- the method comprises administration of the compounds at several times per day, including but not limiting to 2 times per day, 3 times per day, and 4 times per day.
- subject refers to an animal, such as a mammal, for example a human.
- the methods described herein can be useful in both humans and non-human animals.
- the subject is a mammal (such as an animal model of disease)
- the subject is a human.
- the terms “subject” and “patient” can be used interchangeably.
- the animal may be for example, humans, pigs, horses, goats, cats, mice, rats, dogs, apes, fish, chimpanzees, orangutans, guinea pigs, hamsters, cows, sheep, birds, chickens, as well as any other vertebrate or invertebrate.
- the subject is a cell line, such as, for example, A549 human lung cells, bEnd. 3 endothelial cells, bone-marrow-derive macrophage (BMDM) cells, and human mesenchymal stem cells (hMSCs) .
- BMDM bone-marrow-derive macrophage
- hMSCs human mesenchymal stem cells
- an “isolated” or “purified” compound is substantially free of other compounds.
- purified compounds are at least 60%by weight (dry weight) of the compound of interest.
- the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight of the compound of interest.
- a purified compound is one that is at least 90%, 91%, 92%, 93%, 94%, 95%, 98%, 99%, or 100% (w/w) of the desired compound by weight. Purity is measured by any appropriate standard method, for example, by column chromatography, thin layer chromatography, or high-performance liquid chromatography (HPLC) analysis.
- an isolated nucleic acid molecule is generally introduced into a vector (e.g., a cloning vector or an expression vector) for convenience of manipulation or to generate a fusion nucleic acid molecule.
- an isolated nucleic acid molecule can include an engineered nucleic acid molecule such as a recombinant or a synthetic nucleic acid molecule.
- oligos As used herein, the terms “oligos” , “oligonucleotides” are used interchangeably to describe short single strands of synthetic DNA or RNA, such as, for example, about a 5 nucleic acid base sequence to about a 500 nucleic acid base sequence. “Oligo delivery” and “oligonucleotide delivery” are also used interchangeably to describe the method by which oligonucleotides are delivered into cells or targeted to specific cell types, tissues, or organs.
- gene means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) involved in the transcription/translation of the gene product and the regulation of the transcription/translation, as well as intervening sequences (introns) between individual coding segments (exons) .
- a “pharmacokinetics” refers to the branch of pharmacology which specializes in the movement of drugs within the body.
- the subject invention provides a polymer-coated nanoworm, such as, for example a polydopamine-coated nanoworm (PDA NW) , with a 1-dimensional assembly, which can reduce endosomal and/or lysosomal entrapment of nucleic acids when administering nucleic acids to a subject.
- a polymer-coated nanoworm such as, for example a polydopamine-coated nanoworm (PDA NW)
- PDA NW polydopamine-coated nanoworm
- the NW is an integrated core-shell nanoparticle with a metal nanochain, such as, for example, a gold (Au) nanochain, serving as the skeleton and a polymer shell, such as, for example, a polydopamine shell for adsorbing nucleic acids, such as, for example, oligonucleotides or an additional lipid coating to complex with a nucleic acid molecule, such as, for example, mRNA.
- the oligonucleotides can be about 15 to about 50 bases in length.
- the mRNA can be about 700 to about 5000 bases in length.
- the metal nanochain is gold (Au) , copper, silver, iron oxide, ceria, zinc oxide, titanium oxide, rhodium, platinum, iron oxide, or any combination thereof.
- the metal nanoparticle can be capped with citrate.
- the mechanism for endosomal escape is distinct from the proton sponge effect due to the involvement of ClC3.
- the anionic gold-polydopamine core-shell nanoworm of the subject invention can be used for programming cellular responses, for example in primary macrophage polarization and stem cell differentiation, and for treating diseases, including, but not limited to, kidney fibrosis and acute liver injury.
- an oligonucleotide can be adsorbed onto the polymer-coated nanoworm by electrostatic interaction in acidic conditions, such as, for example, at a pH of less than or equal to about 3.0 or about 2.5.
- the oligonucleotide can be labeled with a dye, such as, for example, cyanine (Cy) .
- a dye such as, for example, cyanine (Cy) .
- Cy 5-labeled 21 thymines Cy5-T 21
- the mRNA can encode hepatocyte growth factor (HGF) , chemokine receptor type 4 (CXCR4) , bone morphogenetic protein 7 (BMP-7) , enhanced green fluorescence protein (EGFP; SEQ ID NO: 3: UAC CAC UCG UUC CCG CUC CUC GAC AAG UGG CCC CAC CAC GGG UAG_GAC CAG CUC GAC CUG CCG CUG CAU UUG CCG GUG UUC AAG UCG CAC AGG CCG CUC CCG CUA CGG UGG AUG CCG UUC GAC UGG GAC UUC AAG UAG_ACG UGG UGG CCG UUC GAC GGG CAC GGG ACC GGG UGG GAG CAC UGG UGG GAC UGG AUG CCG CAC GUC ACG AAG UCG GCG AUG GGG CUG GUG UAC UGG CGG UGG CGG GAC UGG AUG CCG CAC GUC ACG AAG UCG GCG AUG GGG CUG G
- the DOTAP is at a concentration of about 33 mol%to about 37 mol%or about 35 mol%.
- the cholesterol is at a concentration of about 47 mol%to about 51 mol%or about 49 mol%.
- the DOPE is at a concentration of about 14 mol%to about 18 mol%or about 16 mol%.
- the mol%of each lipid refers to the percentage in the whole lipid mixture (e.g., a mixture of cholesterol, DOTAP, and DOPE) initially added to the polymer-coated nanoworm.
- the lipid and polymer-coated nanoworm has an average hydrodynamic size of about 220 nm to about 275 nm or about 252 nm and a zeta potential of about +30 mV to about +45 mV or about +40 mV.
- the lipid and polymer-coated nanoworm can be formed by mixing a lipid with the polymer-coated nanoworm.
- lipid and polymer-coated nanoworm can further comprise an oligonucleotide, such as, for example, mRNA, which can be prepared by mixing the lipid and polymer-coated nanoworm with an oligonucleotide.
- an oligonucleotide can be adsorbed onto the lipid and polymer-coated nanoworm by electrostatic interaction in acidic conditions, such as, for example, at a pH of less than or equal to about 3.0 or about 2.5.
- the positive charge of the lipid coating on the nanoworms allows for the complexation of large, macromolecular nucleic acids, such as, for example, mRNA, with the lipid nanoworms in nuclease-free water.
- the shorter oligonucleotides can be adsorbed onto the polydopamine shell of the nanoworms via electrostatic interactions in an acidic citrate buffer, whereby the polymer shell becomes protonated and acidic.
- the lipid and polymer-coated nanoworm with an therapeutic mRNA has an average hydrodynamic size of about 225 nm to about 280 nm or about 258 nm and an overall zeta potential of about -30 mV to -50 mV about -32 mV.
- the nanoworm can have overall anionic charge and can readily load different types of therapeutic nucleic acids, including, for example, antisense oligonucleotides (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , and messenger RNA (mRNA) .
- ASO antisense oligonucleotides
- siRNA short interfering RNA
- miRNA microRNA
- mRNA messenger RNA
- the zeta potential of the nanoworm is less than about -10 mV (e.g., -20 mV) .
- the negative charge on the surface of nanoworms is evenly dispersed.
- the nanoworm has a limited colocalization with the endosome and/or lysosomes, with a colocalization coefficient of about 0.1 to about 0.2.
- the therapeutic nucleic acid can be used to treat a variety of diseases and disorders, such as, for example, chronic kidney disease, acute liver disease, cancers, cell-based immunotherapy, or any combination thereof.
- the nanoworm enters epithelial cells, liver cells, kidney cells, endothelial cells, primary macrophages, mesenchymal stem cells, neural cells, or any combination thereof in the subject.
- compositions of the invention are administered at specific time intervals.
- the subject nanoworm can be used for in vitro transfection, ex vivo transfection followed by injection into a subject as cell-based therapy, or direct in vivo transfection via injection of nanoworm into the subject.
- the cells can be dosed once with specific nucleic acid-encased nanoworms for about 2 to about 3 days for the cellular process to occur (e.g., macrophage polarization, stem cell differentiation, gene knockdown) .
- stem cells can be incubated with therapeutic nucleic acid-encased lipid nanoworms once for about 12 hours; subsequently, the transfected stem cells can be injected into a subject, such as, for example, a subject with kidney disease.
- a subject such as, for example, a subject with kidney disease.
- at least about 1 day to about 6 months, or about 11 days to about 4 weeks can pass before efficacy assessment.
- At least 1, 2, 3, 4, 5, 6, 7, 8, 9 10, about 20, about 30, about 40, about 50, about 100 or more dose of therapeutic nucleic acid-encased lipid nanoworms can be administered to a subject, such as, for example, a subject with acute liver injury.
- at least about 6 hours to about 168 hours or about 48 hours can pass before efficacy assessment.
- the therapeutically effective amount of a nanoworm composition of the invention can be administered through intravenous, oral, rectal, bronchial, nasal, topical, buccal, sub-lingual, transdermal, vaginal, intramuscular, intraperitoneal, intra-arterial, intracerebral, intraocular administration or in a form suitable for administration by inhalation or insufflation, including powders and liquid aerosol administration, or by sustained release systems such as semipermeable matrices of solid hydrophobic polymers containing the nanoworms of the invention.
- Administration may be also by way of other carriers or vehicles such as patches, micelles, liposomes, vesicles, implants (e.g. microimplants) , synthetic polymers, microspheres, nanoparticles, and the like.
- the nanoworm compositions of the instant invention may be formulated for parenteral administration e.g., by injection, for example, bolus injection or continuous infusion.
- the nanoparticle compositions may be presented in unit dose form in ampoules, pre-filled syringes, and small volume infusion or in multi-dose containers with or without an added preservative.
- the nanoparticle compositions may be in forms of suspensions, solutions, or emulsions in oily or aqueous vehicles.
- the composition may further contain formulation agents such as suspending, stabilizing and/or dispersing agents.
- nanoworms do not have huge concerns over genotoxicity.
- the nanoworms of the subject invention do not exert any voltage on the cells and can overcome the delivery barriers of deep tissue through various routes of administration, such as, articular injections, intrathecal injections, and retrobulbar injections.
- confocal images of Lysotracker staining can show that the effect of a treatment on the colocalization of organelles.
- the methods of the subject invention found that the treatment of Bafilomycin A1 did not alter the colocalization of the acidic organelles and the NWs, indicating that the endosomal escape of NWs is not based on the proton sponge effect ( Figure 2A) .
- Figure 4A and Figure 4D demonstrates the results of IF staining using red, green, and blue dyes.
- IF staining can be used to evaluate polarization.
- intense CD80-positive (M1-specific maker) signals were found in the PBS, pristine NW, and free miR223 groups while cells pretreated with Au@PDA@miR-223 NW showed the weakest signals for CD80 but stronger signals for CD206 (M2-specific marker) .
- qRT-PCR analysis also verified the markedly decreased expression of M1-associated genes (e.g., IL-12, CD80, TNF- ⁇ , and IFN- ⁇ ) in Au@PDA@miR-223 NW group (downregulated by at least 50%) , and Lipo + miR-223 group showed modest depressed expression (downregulated by ⁇ 25%) .
- M1-associated genes e.g., IL-12, CD80, TNF- ⁇ , and IFN- ⁇
- the cells were stained with 2 ⁇ g/mL Alexa Fluor 488-labeled, goat secondary antibody against rabbit (Invitrogen, A-11008) in 2%BSA for 1 h at RT, stained by 1 ⁇ g/mL DAPI (Invitrogen, D1306) in PBS for 10 min at RT, and rinsed with PBS twice. Coverslips were mounted with Antifade Mountant (Thermo Scientific; P36980) for confocal laser scanning microscopy.
- the excitation wavelengths of DAPI, Alexa Fluor 488, and Cy5 are 405 nm, 488 nm and 650 nm, respectively.
- the emission wavelengths of DAPI, Alexa Fluor 488, and Cy5 are 410–470 nm, 495–600 nm, and 655–750 nm, respectively.
- the polarized BMDMs were stained with primary antibodies against CD206 (1: 150; Abcam, ab64693) and CD80 antibodies (1: 100; Abcam, ab254579) in 2.5%BSA overnight at 4°C. After three PBS rinses, the cells were stained by 1 ⁇ g/mL Alexa Fluor 488-labeled, goat secondary antibody against rabbit for 1 h at RT, stained by 1 ⁇ g/mL DAPI in PBS for 10 min at RT, and rinsed with PBS twice.
- 2.5%horse serum Vector Laboratories, S-2012-50
- Western blotting is a laboratory technique used to detect a specific protein in a blood or tissue sample.
- the method uses gel electrophoresis to separate the sample’s proteins.
- the western blotting technique begins with an antigen sample that is placed in separation gel. Then the separated proteins are transferred to a nitrocellulose sheet (blot) in a blotting tank. Following the blotting tank, the antibodies are labeled, and the blot is immune-stained. The sample then goes through autoradiography and is developed into a fixed autoradiograph where the antigen bands can be visualized.
- Oligo delivery or oligonucleotide delivery describes a method by with oligonucleotides are delivered into cells or targeted to specific cell types, tissues, or organs. In some embodiments, this method can be used to silence the expression of a gene. Oligos/oligonucleotides are short single strands of synthetic DNA or RNA that serve as the starting point for many biological methods. Oligos can be used in genetic testing, forensic research, and sequencing. In some embodiments, oligonucleotides can be administrated via parenteral routes, which comprise either intravenous infusion or subcutaneous injection.
- oligonucleotides oligos
- oligos oligonucleotides
- Cy5-T 21 cyanine 5-labeled 21 thymines
- DOTAP 1-dioleoyl-3-trimethylammonium propane; Cayman, 15110) , cholesterol (Sigma, C8667) , and DOPE (dioleoylphosphatidylethanolamine; Cayman, 15091) were added to chloroform ( ⁇ 99.8%, Fisher Chemical) at a molar ratio of 35: 49: 16, keeping the total lipid concentration at 1 mg/mL.
- chloroform ⁇ 99.8%, Fisher Chemical
- the 1.2%native agarose gel made in diethyl pyrocarbonate (DEPC; Macklin, D6079 X) -treated TBE (Tris-borate-ethylenediaminetetraacetic acid) buffer, contains 1X SYBR gold nucleic acid gel stain (1: 10000 dilution; Thermo Scientific, S11494) .
- DEPC diethyl pyrocarbonate
- TBE Tris-borate-ethylenediaminetetraacetic acid
- the concentration of NWs and Au@PDA NPs was determined by inductive coupled-plasma mass spectrometry (ICP-MS) (Agilent 7900) with reference to a standard curve of known gold concentration (Au 197 isotope) in parts per billion (ppb) .
- ICP-MS inductive coupled-plasma mass spectrometry
- the hydrodynamic diameters and zeta potential of NWs and NPs were measured by the DelsaMax PRO dynamic light scattering (DLS) analyzer (Beckman Coulter) .
- DLS data represent the values from three independent measurements. The DLS data quality was interpreted by analyzing the autocorrelation function and its fit, where the sum of square (SOS) threshold was set to be under 100. (Only measurements with an SOS value ⁇ 100 were counted) . As independent samples were used for different assays or experiments, a slight batch-to-batch variation was observed.
- the NW solutions were dispersed in DMEM containing 10 %fetal bovine serum (FBS; Gibco, 10270106) . After 24 h of incubation at 37 °C, the NW solutions were analyzed using UV-Vis spectrometry and DLS. Reported DLS values represent mean ⁇ SD from three independent measurements.
- the loading of each type of nucleic acid on the NWs was calculated based on a fluorescence-based assay, following literature precedent.
- the concentration of Cy5-labeled oligos before and after their adsorption onto NWs or Au@PDA NPs was measured by microplate reader (MULTISKAN GO, Thermo Scientific) , followed by dividing the difference in nucleic acid concentration by the concentration of NWs and NPs to obtain the loading of nucleic acid.
- DMEM Modified Eagle Medium
- P/S penicillin-streptomycin
- Human mesenchymal stromal cells (hMSCs, Lonza) were cultured in ⁇ -minimum essential medium ( ⁇ -MEM; Gibco, 11900073) supplemented with 16.7%FBS, 1%P/Sand 1%L-glutamine (Gibco, 25030081) .
- Bone marrow-derived macrophages were isolated from Balb/c mice of 9–11 wk old. Briefly, hind legs were dislocated from the hip bone and sterilized in three changes of 75%ethanol for 1 min. After removing the bones below the knee joint by cutting the ligaments, the separated femurs and tibias were rinsed with precooled phosphate buffered saline (PBS; Gibco, 21600010) . The ends of the femur and tibia were cut, and each marrow cavity was irrigated with 5 mL of RPMI 1640 medium (Gibco, 11875093) containing 10%FBS.
- PBS phosphate buffered saline
- Cells were plated on 96-well plates and cultured till ⁇ 80%confluence. Then, cells were incubated with Au@PDA@oligonucleotide NWs (0.2 nM) formulated in 0.1 mL of full culture medium or mRNA-LNWs (10 ⁇ g) in 0.1 mL full ⁇ -MEM for 24 h. After two PBS rinses, cell viability was measured by the alamarBlue assay (Invitrogen, DAL1025) according to the manufacturer’s protocol.
- the cells were fixed with cold methanol for 10 min and stained by DAPI (4', 6-diamidino-2-phenylindole; Thermo Scientific, D1306) for confocal microscopy.
- DAPI 4, 6-diamidino-2-phenylindole
- Thermo Scientific, D1306 for confocal microscopy.
- the excitation wavelengths for DAPI and Gal8-GFP are 405 and 488 nm, respectively, and the emission wavelengths are 410–480 nm and 495–600 nm, respectively.
- the blocker-containing medium was removed, and 0.4 mL of fresh full DMEM that contains the same blocker at the original concentration and 0.2 nM T 21 @NWs was added to the cells.
- the cells were rinsed with PBS twice and trypsinized (0.25%Trypsin-EDTA, Gibco) for cell counting by a hemocytometer. The cells were further centrifuged at 1500 rpm for 5 min to form a pellet.
- Ultrathin sections of ⁇ 70 nm in thickness were deposited onto 200-mesh copper grids (Electron Microscopy Sciences, G200-Cu) and stained with 4%uranyl acetate in 50%methanol/water (EMS, 541-09-3) and Reynold’s lead citrate for observation under TEM at a beam voltage of 100 kV (Hitachi H7700) .
- PVDF polyvinylidene difluoride
- TBST Tris-buffered saline with Tween-20
- HRP horseradish peroxidase
- BMDMs Day 7 BMDMs were stimulated by 1 mL of full DMEM containing 100 ng/mL LPS and 25 ng/mL IFN- ⁇ (Sinobiological, 11725-H08H) . After 24 h, the LPS/IFN- ⁇ -containing medium was removed and rinsed by PBS twice, and the cells were repolarized by incubating with 1 mL of 0.2 nM Au@PDA@miR-223 NWs in full DMEM for 48 h.
- hMSCs were seeded in a 24-well plate until ⁇ 90 %confluence. Next, hMSCs were treated with 0.4mL of osteogenic medium (OM) (0.05 mM ascorbic acid, 10 mM glycerophosphate, and 0.1 ⁇ M dexamethasone in full ⁇ -MEM) containing 0.2 nM Au@PDA@siNog NWs for 7 of 14 d, with medium changed every 3 days.
- OM osteogenic medium
- Apoptotic and necrotic cells were visualized and quantified using the Click-iT TM Plus TUNEL Assay for In Situ Apoptosis Detection (Invitrogen, C10617; Alexa Fluor 488) .
- Paraffin liver sections of 5 ⁇ m thick were stained per the manufacturer's instructions, co- stained with DAPI for 10 min at RT, and mounted with Antifade Mountant for confocal laser scanning microscopy.
- the excitation wavelengths of DAPI and Alexa Fluor 488 are 405 nm and 488 nm, respectively.
- the emission wavelength ranges of DAPI and Alexa Fluor 488 are 415–480 nm and 495–650 nm, respectively.
- RNA-Seq Whole-transcriptome analysis with total RNA sequencing
- DEG Differential expressed gene
- GO gene ontology
- Q values corrected p values
- qRT-PCR was performed on the StepOnePlus Real-Time PCR System (Applied Biosystems) using TB SYBR Green Premix Ex Taq kit (Takara, RR82WR) following the manufacturer’s instructions.
- Transcript levels were analyzed using the ⁇ CT method and normalized to the house-keeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH) .
- Gene expression was quantified using pre-designed primers purchased from Shanghai Rui Mian Bio-Tech (see the sequences in Table 3) .
- APAP acetaminophen
- ALI acute liver injury
- the staining solution was washed off, and the hMSCs were trypsinized for cell counting by a hemocytometer.
- the hMSCs were further centrifuged at 1500 rpm for 5 min and resuspended in 200 ⁇ L PBS for injection.
- the injected mice were sacrificed 24 hour post injection and perfused with PBS before imaging.
- Ex vivo fluorescence imaging of the excised internal organs liver, lung, heart, spleen, and kidneys was performed using an Odyssey infrared imaging system (excitation wavelength: 700 nm, emission wavelength: ⁇ 700nm) .
- Au 40 @PDA NWs by sonicating citrate-capped 40-nm gold NPs with monomeric dopamine at alkaline pH for 1 h ( Figure 1A) .
- Au 40 @PDA NWs bear 4–5 gold cores, a PDA shell of ⁇ 20 nm thick, a length of ⁇ 224 nm, and a zeta potential of –25.7 mV ( Figure 1B and Figure 8C) .
- BMDMs primary bone marrow-derived macrophages
- hMSCs human mesenchymal stromal cells 24 h post incubation
- Treatment with amiloride reduced the association of T 21 -encased NWs to all four cell types most pronouncedly of all pharmacological blockers tested, indicating macropinocytosis as a key uptake pathway (similar to PDA-coated NPs) .
- ClC1, ClC2, ClC-Ka, and ClC-Kb are plasma membrane Cl - channels
- ClC3 through ClC7 are H + /2Cl - exchanger in the endosomes or lysosomes for the regulation of luminal acidification and Cl - accumulation 20, 21 .
- ClC5 TTYH1 (Tweety Family Member 1, a Cl - channel) and TMEM206 (aproton-activated Cl - channel) are also upregulated.
- ClC3 is an ion exchanger
- MQAE N- (ethoxycarbonylmethyl) -6-methoxyquinolinium bromide
- MQAE is not a dye of Cl - specific for late endosomes or lysosomes
- TRITC tetramethylrhodamine
- the red-to-green (R/G) ratio fluorescence should indicate the pH of vesicles containing NWs.
- the mean pH of the organelles was 5.44, characteristic of late endosomes.
- treatment of wildtype cells with NWs lowered the mean pH to 5.20, implying the acidification of organelles.
- the organellar pH before and after NW treatment were 5.70 and 5.55, respectively ( Figures 3B and 3D) , both less acidic than those on wildtype cells.
- Gal8-GFP is a cytosolic protein and will recruit to the inner leaflet of ruptured endosomal membrane, yielding strong GFP fluorescence.
- Treatment of Gal8-GFP-expressing cells with Au@PDA@T 21 NWs yielded punctate spots ( ⁇ 15 dots per cell) , proof of endosomal rupture ( Figures. 3G and 26) .
- TEM imaging also captured the impaired endosomal membrane and escape of Au@PDA@T 21 NWs to the cytosol, with rupture sites on membrane segments near NWs but not the entire membrane ( Figures 3H and 27) .
- miR-223 (aM2 macrophage stimulator [27] ) to primary BMDMs that were pre-induced to M1 phenotype.
- the M1 BMDMs were treated with miR-223-encased NW, free miR-223, pristine Au@PDA NW, or Lipo+miR-223 for 48 h for M2 repolarization.
- miR-223-encased NWs activated M2-related genes (IL-10, CD206, TGF- ⁇ , and arginase-I) and inhibited M1-related genes (IL-12, CD80, TNF- ⁇ , and IFN- ⁇ ) most effectively (Figure 4C and 31) .
- Au@PDA@lipid NW was larger than Au@PDA NW (278.5 nm) and positively charged (+30.5 mV) , but upon adsorption with mEGFP at w/w ratio of 1: 50 ( Figures 35 and 36) , the resultant Au@PDA@mEGFP NW were larger (280.3 nm) and negatively charged (-35.2 mV) , with ⁇ 68 mEGFP strands (Table 8 and 9) . Confocal imaging verified NW escape from late endosomes 10 h post-incubation ( Figure 37) .
- Embodiment 2 The nanoworm composition of embodiment 1, wherein the at least two metal nanoparticle cores are at least two gold nanoparticle cores.
- Embodiment 3 The nanoworm composition of any preceding embodiment, wherein each of the at least two metal nanoparticle cores has a diameter of about 40 nm.
- Embodiment 6 The nanoworm composition of any preceding embodiment, wherein the polymer coating comprises polydopamine (PDA) , polyethylene glycol (PEG) , polyethylenimine (PEI) , or silica.
- PDA polydopamine
- PEG polyethylene glycol
- PEI polyethylenimine
- Embodiment 10 The nanoworm composition of any preceding embodiment, wherein the therapeutic nucleic acid is an antisense oligonucleotide (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , messenger RNA (mRNA) , or any combination thereof.
- ASO antisense oligonucleotide
- siRNA short interfering RNA
- miRNA microRNA
- mRNA messenger RNA
- mRNA messenger RNA
- Embodiment 11 The nanoworm composition of embodiment 8, wherein the therapeutic oligonucleotide nucleic acid is adsorbed onto the polymer coating.
- Embodiment 13 The nanoworm composition of any preceding embodiment, comprising
- lipid coating over the polymer coating, wherein the therapeutic nucleic acid is bound to the lipid coating.
- Embodiment 14 The nanoworm composition of embodiment 13, wherein the lipid coating comprises 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) , cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DOPE) , (1, 1′- ( (2- (4- (2- ( (2- (bis (2-hydroxydodecyl) amino) ethyl) (2-hydroxydodecyl) amino) ethyl) piperazin-1-yl) ethyl) azanediyl) bis (dodecan-2-ol) ) (C12-200) , (6Z, 9Z, 28Z, 31Z) -Heptatriaconta-6, 9, 28, 31-tetraen-19-yl 4- (dimethylamino) butanoate (DLin-MC3-DMA) , ( (4-hydroxybutyl) azanediyl] di (hexan
- Embodiment 16 The nanoworm composition of any preceding embodiment, wherein the nanoworm comprises an Au x @PDA NW, wherein x is about 20, about 40, or about 60 nm in diameter.
- Embodiment 17 The nanoworm composition of any preceding embodiment, wherein the nanoworm comprises an Au@PDA NR (NanoRod) , wherein the size of the NR is about 45 nm x about 180 nm.
- Au@PDA NR NanoRod
- Embodiment 19 The method of embodiment 18, wherein the therapeutic nucleic acid regulates gene expression in the subject.
- Embodiment 24 The composition of embodiment 18, wherein the nanoworm is administered by intra-an articular injection, an intrathecal injection, or a retrobulbar injection.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- General Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Physics & Mathematics (AREA)
- Wood Science & Technology (AREA)
- Biotechnology (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Biophysics (AREA)
- Dermatology (AREA)
- Inorganic Chemistry (AREA)
- Nanotechnology (AREA)
- Optics & Photonics (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
An effective and safe composition comprises an anionic gold-polydopamine core-shell nanoworm as an alternative gene carrier for bypassing the bottleneck of endosomal entrapment. The nanoworm can be used in methods of delivering therapeutic oligonucleotides to a subject. A polydopamine shell supports the surface adsorption of nucleic acids. The anionic nucleic acid-encased nanoworm can then enter cells without transfection agents and activate the ClC3 H +/Cl - exchanger in late endosomes to mediate vesicular accumulation of H + and Cl -, which causes membrane rupture, and finally escape to cytosol without cell-penetrating peptides or mechanical stimuli. The nanoworm can be further used for programming cellular responses, i. e., primary macrophage polarization and stem cell differentiation, for the treatment of diseases, such as kidney fibrosis and acute liver injury.
Description
CROSS-REFERENCE TO RELATED APPLICATION
This application claims the benefit of U.S. Provisional Application Serial No. 63/590,241, filed October 13, 2023, which is hereby incorporated by reference in its entirety including any tables, figures, or drawings.
In recent decades, there has been a resurgence of interest in gene therapy. Currently, there are several approved and marketed gene therapies, such as (Alnylam Pharmaceuticals, Cambridge, MA) , (Akcea Therapeutics, Carlsbad, CA) , and (Akcea Therapeutics) .
Effective intracellular delivery of nucleic acid forms the basis of important biomedical applications, such as gene regulation, expression, and editing. To date, there are several methods for gene delivery, including viral transduction, electroporation, and nanoparticle-based transfection. Viral transduction has a high transduction efficiency but raises safety concerns over genotoxicity and insertional mutagenesis. Electroporation suffers from cytotoxicity due to high voltage and limited penetration depth in vivo. A significant advancement in delivering nucleic acids is the utilization of nanoparticles (NPs) . NPs are the most promising non-viral, non-physical methods for gene therapy. Despite the clinical application of nucleic acid-based NPs, obstacles remain that hinder their efficient intracellular delivery, as the nucleic acid-based NPs are readily degraded by nucleases and possess unfavorable physicochemical properties that impede their uptake by cells. Unfortunately, upon initial cellular entry, the genes delivered by transfection agents often face pronounced endolysosomal entrapment, leading to degradation within lysosomes and therefore inefficient gene release into cytosol. Therefore, their subsequent escape from the acidic organelles is crucial for applications in gene therapy.
The field of nanoparticle (NP) -based gene delivery boomed with the global deployment of mRNA-based COVID-19 vaccines, but there were only 11 out of more than 33000 publications that reported correlation coefficients of ≤0.2 (denoting little or no spatial correlation) between NPs and endosomes since 2019 (Table 1) . Rather, most studies focused
on materials design and downstream transfection efficiency, often lacking details on the efficiency of endosomal escape or the biological mechanism for endosomal escape (Table 2) .
NPs offer a customized gene delivery vehicle with an adjustable size and shape, as well as versatile functionalization. Many nanocarriers enable efficient cell uptake and packaging of gene cargos, but endosomal/lysosomal entrapment can hinder efficacy by causing degradation, leading to an ineffective gene release into the cytosol. Currently, there are several frequently employed carriers that aim to overcome the obstacle of endosomal escape. These include (1) cationic or pH-responsive polymer-based nanoparticles, such as polyethyleneimine (PEI) , poly (l-lysine) , and poly (amidoamine) (PAMAM) , through the proton sponge effect; (2) lipid-based nanoparticles (e.g., liposomes, lipid nanoparticles) through the membrane fusion or proton sponge effect (ionizable lipids) ; and (3) cell penetration peptides through membrane disruption. However, the probability of endosomal escape for the aforementioned carriers remains low; only approximately 1%of the ionizable lipid NPs have reported successful endosomal escape [1] . Additionally, positively charged cationic NPs are frequently linked with high cytotoxicity [2] .
Cationic lipids [3] and polymers [4] are classical gene carriers. Cationic, lipid-based transfection agents electrostatically complex with the therapeutic gene cargo and form a near-neutral nanoparticle for easier cellular entry. They condense nucleic acids into nanoparticle (NP) -based complexes to overcome the electrostatic repulsion from the anionic cell membrane and facilitate cellular entry. Also, based on the celebrated “proton sponge effect” , cationic carriers have amine groups with sufficient pH buffering capacity to trigger the endosomal accumulation of protons by V-ATPase and balance chloride ions, ultimately elevating osmotic pressure to cause membrane rupture. [5] Yet, cationic carriers induce cytotoxicity or inflammatory responses, [6, 7] and their endosomal escape is inefficient. [1] Recently, some anionic bionanomaterials have become alternative gene carriers for they abundantly enter cells via cell-surface receptors [8] without inducing cytotoxicity. Still, anionic carriers suffer from endosomal entrapment [9] and often require pH buffering groups, [10] cell-penetration peptides, [11] or mechanical stimuli [12] for escape. A gene carrier that marries the advantages of cationic and anionic carriers (biocompatibility, robust cell entry, and endosomal escape driven by its physicochemical nanostructure) is rare. However, efficient gene delivery remains essential for effective gene regulation. Therefore, a safe and robust gene delivery nanocarrier is urgently needed for more effective gene regulation to enhance cell-based therapy.
BRIEF SUMMARY OF THE INVENTION
The subject invention relates to a novel polymer-coated nanoworm (NW) . In certain embodiments, the polymer is polydopamine (PDA) , polyethylene glycol (PEG) , polyethylenimine (PEI) , or silica. More specifically, the NW is an integrated core-shell nanoparticle with a metal nanochain serving as the skeleton and a polymer shell for adsorbing molecules, such as, for example, oligonucleotides or additional lipid coating to complex with mRNA. In certain embodiments, the metal is gold (Au) , copper, silver, iron oxide, ceria, zinc oxide, titanium oxide, rhodium, platinum, and iron oxide. In preferred embodiments, the NW comprise citrate-capped Au NPs (Cit-Au NPs) . In certain embodiments, the diameter of the Cit-Au NPs ranges from about 20 to about 60 nm. In preferred embodiments, the diameter of the Cit-Au NPs is about 40 nm. In certain embodiments, the NPs of the subject invention comprise Aux@PDA NWs, where x is about 20, about 40, or about 60 nm in diameter. In certain embodiments, the NPs of the subject invention comprise Au@PDA NRs (Nanorods) , which size is about 45 nm x about 180 nm.
In certain embodiments, the subject polymer NW can enter cells and escape the endosomes and/or lysosomes and effectively regulate genes. In certain embodiments, the NWs can exit the late endosomes by increasing chloride ion accumulation in the intracellular vesicles, resulting in vesicle swelling and subsequent membrane rupture. In certain embodiments, compared with the widely used transfection agent Lipofectamine 3000, the platform of the subject invention has more effective gene regulation in vitro and in vivo, thereby enhancing cell-based therapy through genetic engineering.
In certain embodiments, the subject therapeutic nucleic acid-encased nanoworm is effective at cellular delivery with limited localization inside acidic organelles, such as, for example, late endosomes and lysosomes. In certain embodiments, the subject nanoworm has an overall anionic charge. In certain embodiments, the nanoworm can be loaded with different types of therapeutic nucleic acids, such as, for example, antisense DNA, antisense oligonucleotides (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , and messenger RNA (mRNA) ; enter multiple cell types in vitro, such as, for example, epithelial cells, endothelial cells, primary macrophages, and human mesenchymal stromal cells, and in vivo, such as for example, in liver cells; and have limited colocalization with the endosomes and/or lysosomes, with colocalization coefficients in the range of about 0.1 to about 0.2.
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing (s) will be provided by the Office upon request and payment of the necessary fee.
Figures 1A-1E illustrate oligonucleotide-encased, gold@polydopamine nanoworms (Au@PDA@oligonucleotide NWs) for intracellular delivery and endosomal escape. Figure 1A shows preparation route of oligonucleotides-encased NWs for delivering multiple types of oligonucleotides. Figure 1B shows representative TEM image of Au@PDA@T21 NW shows the core-shell Au@PDA NW structure. Scale bar = 50 nm. Figure 1C shows confocal images of four different cell types show limited colocalization of Cy5-labeled Au@PDA@T21 NW with intracellular acidic compartments 24 h post-incubation. Blue = nuclei. White number indicates Pearson colocalization coefficient (PCC) between Cy5-labeled Au@PDA@T21 NW (red) and Lysotracker (green) . Figure 1D shows representative TEM images of four different cell types show cytosolic accumulation of Au@PDA@T21 NW 24 h post incubation. Nu = nucleus; Cy = Cytosol. Scale bar = 500 nm. Figure 1E shows trafficking of Cy5-labeled Au@PDA@T21 NWs (red) in A549 cells as a function of incubation time. EEA1, Rab9, and LAMP1 are markers of early endosome, late endosome, and lysosome, respectively. Blue = Nuclei. White number indicates PCC between Cy5-labeled Au@PDA@T21 NWs (red) and different organelle markers (green) . Scale bar =20 μm.
Figures 2A-2L illustrate ClC3 mediates the endosomal escape of Au@PDA@T21 NWs in A549 cells. Figure 2A shows pharmacological inhibition of V-ATPase by bafilomycin A1 or chloroquine did not affect the endosomal escape of Au@PDA@T21 NWs. White number indicates PCC between Cy5-labeled Au@PDA@T21 NWs (red) and Lysotracker (green) . Scale bar = 20 μm. Figure 2B shows titration curves of Au@PDA@T21 NW. Saline and branched polyethylenimine (bPEI) are served as controls. Figures 2C and 2D show key enriched biological processes (Q < 0.05) found by comparing (c) “Au@PDA@T21 NW; 24 h” to “Au@PDA@T21 NP 24 h” groups and (d) “Au@PDA@T21 NW; 24 h” to “Au@PDA@T21 NW; 8 h” groups. n = 3 for all groups. Figures 2E and 2F show volcano plot shows the distribution of DETs found by comparing (e) “Au@PDA@T21 NW; 24 h” to “Au@PDA@T21 NP; 24 h” groups and (f) “Au@PDA@T21 NW; 24 h” to “Au@PDA@T21 NW; 8 h” groups. n = 3 for all groups. Figure 2G shows western blot analysis revealed upregulation of ClC3 in the “Au@PDA@T21 NW; 24 h” group relative to the “Au@PDA@T21 NP; 24 h” (top row) and the “Au@PDA@T21 NW; 8 h” (bottom row) groups. n = 3, across one experiment. Figure 2H shows subcellular location of ClC3 (green)
relative to intracellular vesicles (red) . Scale bar = 10 μm. Figure 2I shows RNAi-mediated knockdown of ClC3 significantly disrupted the escape of Au@PDA@T21 NWs (red) to acidic vesicles (green) . Figure 2J shows pharmacological inhibition of general chloride ion channels by niflumic acid (NFA) impaired the escape of Au@PDA@T21 NWs (red) to acidic vesicles (green) . Figure 2K shows activation of ClC3 accelerates endosomal escape of Au@PDA@T21 NWs 8 h post-incubation. Blue = nuclei. Figures 2A, 2I, 2J, and 2K, show white number indicates PCCs between Cy5-labeled Au@PDA@T21 NWs (red) and Lysotracker (green) . Blue = nucleus. Figure 2L shows summary of PCC values under different treatment conditions. Data are presented as mean ± SEM, n = 10 frames per group (~150 cells were calculated, and 1 mean value was calculated per frame) .
Figures 3A-3H illustrate ion concentration and membrane rupture of endosomes in A549 cells. Figure 3A shows Cl-concentration of intracellular vesicles containing Cy5-labeled Au@PDA@T21 NWs in cells without (upper row) or with siClC3 treatment (lower row) . MQAE staining signals are inversely correlated to Cl-concentration. White arrows indicate the intracellular vesicles containing Cy5-labeled Au@PDA@T21 NWs in WT A549 cells have relatively high concentration of Cl-when compared to the A549 cells treated with siClC3 (indicated by magenta arrows) . Figure 3B shows pH measurement of intracellular vesicles without (upper row) or with siClC3 treatment (lower row) 8 h post co-incubation with Cy5-labeled Au@PDA@T21 NWs (magenta) , dextran 10K pHrodo (green) , and dextran 10K AF594 (red) . White arrows indicate intracellular vesicles that contain Cy5-labeled Au@PDA@T21 NWs while yellow arrows indicate intracellular vesicles that do not contain Cy5-labeled Au@PDA@T21 NWs. The R/G ratio is the ratio of fluorescence intensity of dextran 10K pHrodo green to the fluorescence intensity of dextran 10k AF594 and is presented in heatmap. Scale bar = 10 μm. Figure 3C shows quantification of the fluorescence intensity of MQAE staining based on (a) . Figure 3D shows quantification of the pH value of individual vesicles in WT cells and siClC3-treated cells by measuring the R/G ratio of intracellular vesicles that contain Cy5-labeled Au@PDA@T21 NWs and do not contain Cy5-labeled Au@PDA@T21 NWs. n = 300 intracellular vesicles. Figure 3E shows evaluation of V-ATPase activity before and after Au@PDA@T21 NWs incubation. The activity of V-ATPase was measured by the colocalization of subunit V0 and V1. White number indicates PCCs between anti-V1 (red) and anti-V0 (green) . Blue = nucleus. Figure 3F shows proposed mechanism of the endosomal escape of Au@PDA@T21 NWs. Figure 3G shows green clusters indicate the recruitment of Gal8-GFP to the ruptured organelle membrane 24 h post-incubation with Au@PDA@T21 NWs but not Au@PDA@T21 NPs. Blue
= nuclei. Figure 3H shows representative TEM image shows the escape of Au@PDA@T21 NWs (orange arrows) from the ruptured membrane of intracellular vesicles. Cy = cytosol, ex = extracellular space.
Figures 4A-4H in vitro NW-enabled transfection of oligonucleotides for inducing the polarization of BMDMs and osteogenic differentiation of hMSCs. Figure 4A shows Cy5-labeled Au@PDA@miR-223 NWs (red) did not accumulate in acidic organelles (green) in BMDMs 24 h post-incubation. Figure 4B shows representative TEM image of endosomal escape of Au@PDA@miR-223 NWs in BMDMs 24 h post-incubation. Figure 4C shows qRT-PCR measurements of M2 phenotype markers in BMDMs. n = 6 per group, across 2 experiments. Figure 4D shows confocal images showed the repolarization of BMDMs from M1 to M2, as evidenced by stronger M2 markers (CD206; green) and weaker M1 markers (CD80; red) . Figure 4E shows Cy5-labeled Au@PDA@siNog NWs (red) did not accumulate in acidic organelles (green) 24 h post -incubation in hMSCs. White number indicates PCC between Cy5-labeled Au@PDA@siNog NWs (red) and Lysotracker (green) . Figures 4A, 4D, and 4E blue = nucleus. Figure 4F show representative TEM images of endosomal escape of Au@PDA@siNog NWs in hMSCs 24 h post-incubation. Figure 4G show qRT-PCR measurements of markers of osteogenesis in hMSCs 14 d post-induction of differentiation. n = 6 per group, across 2 experiments. Figure 4H shows calcium deposit of hMSCs 14 d post-osteogenic differentiation stained by Alizarin Red. Scale bar = 100 μm. n = 3 per group, across 1 experiment. Figure 4C and 4G show data are presented as means ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey’s Test for post-hoc analysis. ns = not significant (P > 0.05) .
Figures 5A-5K illustrate ex vivo lipid NW-enabled transfection of mRNA in hMSCs for cell-based therapy and alleviating kidney fibrosis. Figure 5A shows schematic illustration of the synthesis of mRNA-encased, lipid-coated Au@PDA NWs. Figure 5B shows Cy5-labeled Au@PDA@mCXCR4/BMP7 NWs did not accumulate in acidic organelles (green) 24 h post-incubation. White number indicates PCC between Cy5-labeled Au@PDA@mCXCR4/mBMP7 NWs (red) and Lysotracker (green) . Blue = nuclei. Figure 5C shows confocal image shows hMSCs that were co-transfected by mRNA expressing CXCR4-GFP (green) and mRNA expressing BMP7-OFP (red) using Au@PDA@lipid NWs 24 h post-incubation. Blue = nuclei Figure 5D shows schematic illustration of hMSC-based therapy of kidney fibrosis in mice. After ligating the left ureter twice with sutures, UUO mice received a single i.v. injection of saline, or hMSCs under different treatments on Day 3 and were sacrificed on Day 14 post-UUO surgery. Figure 5E
shows quantification of CXCR4 and BMP7 expression in vitro. The level of therapeutic protein of hMSCs was evaluated 24 h-post transfection by ELISA. Figure 5F shows ex vivo fluorescence images of UUO kidneys showed the strongest fluorescence for the DiR-labeled Au@PDA@mCXCR4/mBMP7-transfected hMSC (NWs-hMSCCXCR4/BMP7) group 24 h post-injection. Lipo-hMSCCXCR4/BMP7: Lipo+mCXCR4/mBMP7-transfected hMSC. Figures 5G shows IHC images showing the expression of Col-I and α-SMA in the UUO kidney sections. Figures 5H and 5I show quantification of Col-I-positive and α-SMA-positive areas in the UUO kidney sections, based on an average of 6 images per mouse. n = 7 per group, across 2 experiments. Figures 5J and 5K show mRNA expression of Col-I and α-SMA quantified by qRT-PCR. Figure 5L shows quantification of BMP7 level in the UUO kidney on Day 14 by ELISA. Data are presented as mean ± SEM. Statistical significance was calculated by two-way ANOVA with Tukey’s multiple comparisons test. ns = not significant (P > 0.05) . n = 7 per group, across 2 experiments.
Figures 6A-6I illustrate in vivo NW-enabled transfection of mRNA in liver cells for reducing acute liver injury (ALI) . Figure 6A shows disease development and treatment plan of ALI. After i. p. injecting APAP to induce ALI, mice received a single i.v. injection of saline, Au@PDA@mHGF NWs, or mixture of Lipofectamine and mHGF on Day 1 and were sacrificed on Day 2. Figure 6B shows cellular-level distribution of Cy5-labeled Au@PDA@mHGF NWs (red) in the liver 24 h post-injection into ALI mice. Blue = nuclei. Green = hepatocytes (labeled with anti-HNF4α antibody) . Figure 6C shows TEM images of the ALI liver showed that Au@PDA@mHGF NWs accumulated inside intracellular vesicles 5 h post-injection (green arrow) , but they resided in the cytosol (Cy) 10 h post-injection (blue arrow) . Orange arrows indicate the Au@PDA@mHGF NWs were escaping from the ruptured membrane. Nu = Nucleus. Cy = cytosol. Figure 6D shows confocal images of the ALI liver revealed limited colocalization of Cy5-labeled Au@PDA@mHGF NWs with organelles (late endosomes labeled by anti-Rab9 antibody and lysosomes labeled by anti-LAMP1 antibody, respectively) 10 h-post injection. White number indicates PCC between Cy5-labeled Au@PDA@mHGF NWs (red) and organelles (green) . Blue = nuclei. Figure 6E, 6F, and 6I show Au@PDA@mHGF NWs most effectively reduced the necrosis area (dotted yellow line) by histological staining and apoptotic cells (TUNEL-positive; green) in the ALI liver sections, and serum ALT level 24 h post-treatment. Blue = nuclei. Figures 6G and 6H, show quantification of necrotic area and TUNEL-positive area. n = 7 per group, across 2 experiments. Data are presented as mean ± SEM. All statistics are two-way ANOVA with Tukey’s multiple comparisons test. ns = not significant (P > 0.05) .
Figures 7A-7G illustrate mRNA-transfected MSC ameliorating streptozotocin (STZ) -induced diabetic nephropathy. Figure 7A shows schematic of disease development and treatment. Figure 7B shows secreted HGF concentration measured by ELISA in conditioned media of Lipo+mHGF transfected MSCs, Au@PDA@mHGF NWs-transfected hMSCs, and hMSCs. Data are presented as mean ± SEM. All statistics are two-way ANOVA with Tukey’s multiple comparisons test. ns = not significant (P > 0.05) . Figure 7C shows ex vivo images of biodistribution of injected hMSCs in kidneys after 24 h by NIRF imaging. Figure 7D illustrates organ-level distribution of DiR-labeledhMSC, Lipo-hMSCHGF, and LNW-hMSCHGF in the STZ-induced diabetic mice 24 h post-injection. Representative ex vivo NIRF images of STZ-induced diabetic mice after intravenous injection of free DiR dye molecules, DiR-labeledhMSC, Lipo-hMSCHGF, and LNW-hMSCHGF. Figure 7E shows representative IHC images showing expression of type I collagen and α-SMA of the UUO kidneys. Quantification of Figure 7F shows Col-I-positive and Figure 7G shows α-SMA-positive area from the IHC-stained sections. For Figure 7E and 7F, data of kidney sections were from an average of six images per mouse, from n = 7 mice per group (across two independent experiments) . Data are presented as means ± s. e. m.
Figures 8A-8C illustrate representative TEM images of citrate-capped 40 nm Au NPs (Figure 8A) , Au40@PDA NPs (Figure 8B) , Au40@PDA NWs (Figure 8C) .
Figure 9A-9F illustrate representative TEM images of citrate-capped 60 nm Au NPs (Figure 9A) , Au60@PDA NWs (Figure 9B) , citrate-capped 20 nm Au NPs (Figure 9C) , Au20@PDA NWs (Figure 9D) , citrate-capped Au45 NRs (45 × 200 nm) (Figure 9E) , Au45@PDA NRs (Figure 9F) ,
Figures 10A-10D illustrate cytotoxicity of oligos-NWs in vitro. (Figure 10A) shows Au40@PDA@T21 NWs, (Figure 10B) shows Au40@PDA@asEGFP NWs, (Figure 10C) shows Au40@PDA@miR223 NWs, and (Figure 10D) shows Au40@PDA@siNog NWs incubated with A549 cells, bEnd. 3 cells, BMDMs, and hMSCs for 24 h, respectively. By the alamarBlue assay, the cells remained largely viable after incubation with all concentration of oligos-encased NWs tested. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns: not significant (P > 0.05) . n = 3 per group, across 1 experiment.
Figures 11A-11D illustrate cytotoxicity of pharmacological inhibitors. A549 cells (Figure 11A) , bEnd. 3 cells (Figure 11B) , BMDMs (Figure 11C) , and hMSCs (Figure 11D) were incubated with various inhibitors of cellular uptake pathways for 4 h, including 250 μg/mL amiloride, 25 μg/mL filipin III, 50 mM sodium azide, and 50 μg/mL fucoidan. By the
alamarBlue assay, the cells remained largely viable after incubation with all the inhibitors tested. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns: not significant (P > 0.05) . n = 6 per group, across 1 experiment.
Figures 12A-12D illustrate cellular uptake pathway of oligo-NWs. A549 cells (Figure 12A) , bEnd. 3 cells (Figure 12B) , BMDMs (Figure 12C) , and hMSCs (Figure 12D) were pre-incubated with various inhibitors of cellular uptake pathways for 1 h and then co-incubated with T21-, asEGFP-, miR223-, and siNog-encased NWs for another 4 h, respectively. The level of cellular uptake was measured by ICP-MS. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns: not significant (P > 0.05) . n = 6 per group, across 1 experiment.
Figures 13A-13B illustrate representative confocal and TEM image of Au40@PDA@T21 NPs in A549 cells. Figure 13A shows Au40@PDA@T21 NPs were highly colocalized with Lysotracker 24 h post-incubation. Blue = DAPI (nuclei) . White number indicates Pearson colocalization coefficients (PCCs) between Cy5-labeled Au40@PDA@T21 NPs (red) and Lysotracker (green) . Figure 13B shows TEM images confirmed Au40@PDA@T21 NPs were entrapped in the intracellular vesicles. Nu = nucleus. Cy =cytosol. Ex = Extracellular space. Yellow asterisks denote vesicles. The two smaller images on the right show the enlargement of the boxed areas of the larger image on the left.
Figures 14A-14B illustrate representative TEM images of Au40@PDA@T21 NWs in A549 cells. Most Au40@PDA@T21 NWs were (Figure 14A) entrapped in late endosomes 8 h post-incubation and (Figure 14B) accumulated freely in the cytosol (or outside of vesicles) 24 h post-incubation. Nu = Nucleus. Cy = cytosol. Ex = Extracellular space. Yellow asterisks denote vesicles, and blue arrows denote free c in the cytosol. In Figures 14A and 14B the two smaller images on the right show the enlargement of the boxed areas (blue or yellow) of the larger image on the left.
Figures 15A-15B illustrate Au20@PDA@T21 NWs (Figure 15A) and Au60@PDA@T21 NWs (red) (Figure 15B) entrapped inside lysosomes (green) 24 h post-incubation, unlike Au40@PDA@T21 NWs that escaped endosomes. Blue = nuclei. White number indicates PCCs between Cy5-labeled NWs (red) and Lysotracker (green) .
Figures 16A-16B illustrate endosomal escape and cellular uptake of Au45@PDA@T21 NRs in A549 cells. Figure 16A shows colocalization of Au45@PDA@T21 NRs in A549 cells 24 h post-incubation. Blue = nuclei. White number indicates PCCs between Cy5-labeled Au45@PDA@T21 NRs (red) and Lysotracker (green) . Figure 16B
illustrates ICP-MS measurement showing that the uptake of Au45@PDA@T21 NRs is ~ 65%less than the Au40@PDA@T21 NWs. Data are presented as mean ± SEM. Statistical significance was calculated by Student’s t-test. n = 3 per group, across one experiment.
Figure 17 illustrates zeta potential measurement of Au40@PDA@T21 NWs in buffer. Data showed that Au@PDA@T21 NWs maintained negatively charged from pH 4.5 to pH 7.5. Data are presented as mean ± SEM. n = 3 per group, across one experiment.
Figure 18 illustrates influence of Na+, K+, and Ca2+ channels on the endosomal escape of T21-NWs in A549 cells. For all pharmacological inhibitors tested, there was no significant escape of Au40@PDA@T21 NWs (red) from lysosomes (green) 24 h post-incubation. White number indicates PCC between Au@PDA@T21 NWs (red) and Lysotracker (green) . Blue = nucleus.
Figure 19 illustrates influence of Na+, K+, Ca2+, Cl-channel and V-ATPase on the endosomal escape of Au40@PDA@T21 NWs in bEnd. 3 cells, BMDMs and hMSCs. White number indicates PCCs between Au@PDA@T21 NWs (red) and Lysotracker (green) . For bEnd. 3 cells, NFA (atypical general Cl-inhibitor) showed minor effect on the endosomal escape while inhibition of Ca2+ channel and V-ATPase have limited effects. For BMDMs and hMSCs, all the inhibitors tested showed minor or no significant influence on the endosomal escape of Au@PDA@T21 NWs, respectively.
Figures 20A-20B illustrate quantification of western blot data of ClC3 expression under different comparison, including (Figure 20A) time-dependent comparison and (Figure 20B) shape-dependent comparison. Data are presented as mean ± SEM. Statistical significance was calculated by Student’s t-test. n = 3 per group, across one experiment.
Figure 21 illustrates subcellular location of ClC3 relative to intracellular vesicles in bEnd. 3 cells, BMDMs, and hMSCs. ClC3 was strongly colocalized with lysosomes (LAMP1) and, to a lesser extent, late endosomes (Rab9) . White number indicates PCCs between ClC3 (green) and intracellular vesicles (red) . Scale bar = 10μm.
Figure 22 illustrates validation of siClC3 knockdown in A549 cells. Cells were incubated with two different siClC3 sequences at three different concentrations using Lipofectamine 3000 and their relative mRNA expression of ClC3 was assessed 48 h post-incubation. A scrambled siRNA sequence (siNC) served as a negative control. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 per group, across 1 experiment.
Figures 23A-23B illustrates validation of ClC3 activation by bufalin in A549 cells. Figure 23A shows relative mRNA expression of ClC3 and Figure 23B shows cell viability after incubation with different concentrations of bufalin for 8 h. We chose 0.5 μM bufalin for our subsequent studies due to its effective upregulation of ClC3 and low cytotoxicity. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Dunnett’s Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 per group, across 1 experiment.
Figure 24 illustrates validation of siClC5 knockdown in A549 cells. Cells were incubated with siClC5 sequences at three different concentrations using Lipofectamine 3000 and their relative mRNA expression of ClC3 was assessed 48 h post-incubation. Data are presented as means ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 per group, across 1 experiment.
Figure 25 illustrates experimental design of pH measurement of intracellular vesicles. A549 cells were incubated with a mixture of dextran 10K pHrodo green, dextran 10K AF594, and Cy5-labeled Au@PDA@T21 NW for 8 h. Some vesicles will contain dextran 10K pHrodo green only, dextran 10K AF594 only, both dyes, Cy5-labeled Au@PDA@T21 NW only, dextran 10K pHrodo green with Cy5-labeled Au@PDA@T21 NW, dextran 10K AF594 with Cy5-labeled Au@PDA@T21 NW. Only those intracellular vesicles that were positive for both dextran 10K AF594 and dextran 10K pHrodo green signals were considered for pH measurements.
Figures 26A-26D illustrate endosomal rupture due to treatment with Au@PDA@T21 NW. Additional representative confocal images of Gal8-GFP transfected A549 cells that were (Figure 26A) untreated, (Figure 26B) treated with Au@PDA@T21 NW for 24 h, and (Figure 26C) treated with Au@PDA@T21 NP for 24 h. Green clusters indicate the recruitment of Gal8-GFP to the ruptured organelle membrane 24 h post-incubation with T21-NWs. Blue = nuclei. Scale bar = 10 μm. Figure 26D shows quantification of Gal8 clusters. Data are presented as mean ± SEM. n = 20 per group, across 3 experiments. Statistical significance was calculated by one-way ANOVA with Tukey’s test for post-hoc analysis. ns: not significant (P > 0.05) .
Figures 27A-27B illustrate membrane rupture of intracellular vesicles. Representative TEM images showing the vesicular membrane rupture in (Figure 27A) A549 cells and (Figure 27B) hMSCs 8 h after Au@PDA@T21 NW and Au@PDA@siNog NW
incubation, respectively. Orange arrows denote the NWs were escaping from the ruptured vesicular membrane. Ex = extracelular space. Cy = cytosol.
Figures 28A-28D illustrate EGFP knockdown in bEnd. 3 cells mediated by Au@PDA@asEGFP NWs. Figure 28A shows confocal images of EGFP-expressing bEnd. 3 cells show limited colocalization of Cy5-labeled Au@PDA@asEGFP NWs with intracellular acidic compartments 24 h post-incubation. Blue = nuclei. White number indicates Pearson colocalization coefficients (PCCs) between Cy5-labeled Au@PDA@asEGFP NWs (red) and Lysotracker (green) . Figure 28B shows representative confocal images of EGFP-expressing bEnd. 3 cells upon various treatments. Green = EGFP; Blue = nuclei. Figure 28C shows percentage of EGFP-positive cells and Figure 28D shows mean fluorescence intensity (MFI) of EGFP by flow cytometry. Data are presented as mean ± SEM n = 3 per group, across 1 experiment. Statistical significance was calculated by one-way ANOVA with Tukey’s test for post-hoc analysis. ns: not significant (P > 0.05) .
Figure 29 illustrates representative TEM images of bEnd. 3 cells show the cytosolic accumulation of Au@PDA@asEGFP NWs (blue arrows) 24 h post-incubation. Nu = nucleus; Cy = cytosol; Ex = extracellular space. The two smaller images on the right show the enlargement of the boxed areas (blue or yellow) of the larger image on the left.
Figure 30 illustrates representative TEM images of BMDMs show cytosolic accumulation of Au@PDA@miR-223 NWs (blue arrows) 24 h post-incubation. Nu = nucleus; Cy = cytosol; Ex = extracellular space. The two smaller images on the right show the enlargement of the boxed areas (blue or yellow) of the larger image on the left.
Figure 31 illustrate in vitro NW-enabled transfection of miR-223 for inducing M2 polarization of BMDMs. qRT-PCR measurements of M1 phenotype markers. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey’s Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 6 per group, across 2 experiments.
Figures 32A-32C illustrate in vitro NW-enabled transection of miR-223 for inducing the depolarization of BMDMs. Figure 32A shows confocal images showed M1 depolarization of BMDMs, as evidenced by stronger M2 markers (CD206; green) and weaker M1 markers (CD80; red) . Blue = nucleus. Scale bar = 20 μm. qRT-PCR measurements of M1 phenotype markers (Figure 32B) and M2 phenotype markers (Figure 32C) . Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 6 per group, across 2 experiments.
Figure 33 illustrates representative TEM images of hMSCs show cytosolic accumulation of Au@PDA@siNog NWs (blue arrows) 24 h post-incubation. Nu = nucleus; Cy = cytosol; Ex = extracellular space. The two smaller images on the right show the enlargement of the boxed areas (blue or yellow) of the larger image on the left.
Figures 34A-34C illustrate in vitro NW-enabled transfection of siNog for inducing the osteogenic differentiation of hMSCs. Figure 34A shows qRT-PCR measurements confirming the knockdown of Noggin 7 d post-incubation. Quantification of calcium deposits (red) (Figure 34B) stained by Alizarin Red (Figure 34C) 7 d post-osteogenic differentiation. Scale bar = 100 μm. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 or 6 per group, across 1–2 experiments. OM: osteogenesis medium; pristine NWs: Au@PDA NWs without siNog.
Figure 35 illustrates native agarose gel electrophoresis of mRNA as synthesized by in vitro transfection (IVT) . From left to right, the samples are an RNA ladder (500–9000 nt) , mEGFP, mBMP-7-OFP, mHGF, and mCXCR4-GFP. The expected sizes of mEGFP, mBMP-7-OFP, mHGF, and mCXCR4-GFP is ~ 1100 nt, 2200 nt, 2500 nt, and 2100 nt, respectively.
Figure 36 illustrates gel retardation analysis of mRNA loading on Au@PDA@lipid NWs. Model mRNA (mEGFP) was complexed with Au@PDA@lipid NWs at varying weight ratio (mEGFP: Au@PDA@lipid) from 1: 20 to 1: 100. The mRNA loading was saturated from the ratio of 1: 50.
Figure 37 Trafficking of Cy5-labeled mRNA-encased NWs (red) in hMCSs as a function of incubation time. Time-lapse confocal images showed that the mRNA-encased NWs were highly colocalized with acidic organelles (stained by Lysotracker) 5 h-post incubation and escaped from those vesicles 10 h-post incubation. Blue = Nuclei. White number indicates PCCs between Cy5-labled mRNA-encased NWs (red) and Lysotracker (green) .
Figures 38A-38B illustrate Cl-accumulation in hMSCs. Figure 38A shows Cl-accumulation of intracellular vesicles containing mRNA-encased NWs in hMSCs without (upper row) or with siClC3 treatment (lower row) . MQAE staining signals are inversely correlated to Cl-concentration. White arrows indicate the vesicles containing Cy5-labeled mRNA-encased NWs in WT hMSCs have relatively high concentration of Cl-when compared to those in the siClC3-treated hMSCs. Figure 38B shows quantification of the fluorescence intensity of MQAE staining based on Figure 38A. Data are presented as mean
± SEM. Statistical significance was calculated by Student’s t-test. n = 300 intracellular vesicles per group. ns = not significant (P > 0.05) .
Figures 39A-39C illustrate pH measurement of the intracellular vesicles in hMSCs. Figure 39A shows pH measurement of intracellular vesicles without (upper row) or with siClC3 (lower row) 5 h post co-incubation with mRNA-encased NWs (magneta) , dextran 10K pHrodo (green) , and dextran 10K AF594 (red) . White arrows indicate intracellular vesicles that contain Cy5-labeled mRNA-encased NWs. Scale Bar = 10 μm. Figure 39B shows activity of V-ATPase of hMSCs upon incubation with mRNA-encased NWs as a function of time. The activity of V-ATPase was enhanced 5 h-post incubation of mRNA-encased NWs. Blue = nuclei. White number indicates PCCs between V1 (green) and V0 (red) . Figure 39C shows quantification of the pH value of individual vesicles in WT hMSCs and siClC3-treated hMSCs by measuring the R/G ratio of intracellular vesicles that contain Cy5-labeled mRNA-encased NWs and those do not contain Cy5-labeled mRNA-encased NWs. Data are presented as mean ± SEM. Statistical significance was calculated by Student’s t-test. n = 300 intracellular vesicles per group. ns = not significant (P > 0.05) .
Figures 40A-40C illustrate in vitro lipid-coated NW (Au@PDA@lipid NW) -enabled transfection of mRNA for expressing of EGFP in hMSCs. Figure 40A shows representative fluorescence images confirming the expression of EGFP (green) due to transfection by Lipofectamine3000 and LNWs 24 h and 48 h post-incubation. Scale bar = 50 μm. Figure 40B shows transfection efficiency of mEGFP (as measured by flow cytometry) . Figure 40C shows cell viability 24 and 48 h post-incubation. Data are presented as mean ±SEM. Statistical significance was calculated by one-way ANOVA with Tukey's Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 6 per group, across 2 experiments.
Figures 41A-41C illustrate ex vivo lipid NW-enabled transfection of mRNA expressing CXCR4 and BMP7 in hMSCs. Figure 41A shows in vitro transfection efficiency of mCXCR4-GFP and mBMP-7-OFP 24 h post-transfection of hMSCs was evaluated by flow cytometry. Figures 41B-41C show relative expression of CXCR4 (Figure 41B) , and BMP-7 (Figure 41C) in hMSCs under different treatments. Measured by ELIZA, data suggested that LNWs enabled the highest expression of both mRNA compared to the hMSCs and Lipofection. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey’s Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 per group, across 1 experiment.
Figure 42 illustrates establishment of renal fibrosis 3 days post UUO surgery. The kidneys were collected from UUO mice 3 days post-surgery and were stained by IHC to
evaluate the expression of Col-I and α-SMA in the UUO kidney and contralateral kidney (CL kidney) .
Figure 43 illustrates accumulation of injected hMSCs in major internal organs. Figure 43 shows representative ex vivo near-infrared fluorescence imaging (NIRF) 24 h post-i.v. injection of DiR-labeled hMSCs in UUO mice. The stronger fluorescence of kidneys from the Au@PDA@mCXCR4/BMP7 NW-transfected hMSC (NWs-hMSCCXCR4/BMP7) group thanhMSCs and the Lipofectamine-transfected hMSCs (Lipo-hMSCCXCR4/BMP7) group suggests an enhanced homing ability of hMSCs to the UUO kidney due to the robust transfection of mCXCR4 mediated by Au@PDA@mCXCR4/mBMP7 NWs.
Figures 44A-44B illustrate quantification of Col-I-positive (Figure 44A) and α-SMA-positive (Figure 44B) area in the UUO kidney sections, based on an average of 6 images per mouse. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with Tukey’s Test for post-hoc analysis. n=7 per group, across 2 experiments.
Figures 45A-45B illustrate in vivo toxicity of NW-hMSCmCXCR4/BMP7 in UUO mice 7 d post-i.v. injection (or 14 d post-UUO surgery) . Injection of Au@PDA@mCXCR4/BMP7 NWs-transfected hMSC did not change (Figure 45A) blood chemistry, cell count, and (Figure 45B) liver function. Grey shaded area represents the normal range of markers or cell counts for mice. WBC: white blood cell. RBC: red blood cell. Absolute neutrophils, lymphocytes, or monocytes (103/μL) : cell number per volume of blood. Neutrophils%, lymphocytes%, monocytes%: percentage of cells out of total WBC. RBC (106/μL) : cell number per volume of blood. AST: aspartate aminotransferase. ALT: alanine transaminase. Data are from n = 3, across 1 experiment.
Figure 46 illustrates in vivo toxicity of NW-hMSCCXCR4/BMP7 in UUO mice. Histological examination of the heart, liver, lung, and spleen showed NW-hMSCCXCR4/BMP7 led to no appreciable change in tissue morphology 11 d post-injection. Representative images were chosen from n = 3 mice per group, across 1 experiment.
Figures 47A-47C illustrate in vivo distribution of Au@PDA@mHGF NW in ALI mice. Gold contents were measured by ICP-MS. Figure 47A shows blood pharmacokinetics. Figure 47B shows organ-level distribution data showed that Au@PDA@mHGF NWs accumulate most abundantly in the liver and spleen (>60 %ID within 24 h post-injection) . Figure 47C shows amount of HGF in the liver tissue 24 h post-injection by ELISA. Data are presented as mean ± SEM. Statistical significance was calculated by one-way ANOVA with
Tukey’s Test for post-hoc analysis. ns = not significant (P > 0.05) . n = 3 or 6 per group, across 1 experiment.
Figure 48 illustrates in vivo toxicity of Au@PDA@mHGF NW in ALI mice. Histological examination of the heart, kidney, lung, and spleen showed Au@PDA@mHGF NW led to no appreciable change in tissue morphology 24 h post-injection. Representative images were chosen from n = 3 mice per group, across 1 experiment. Scale bar = 100 μm.
Figure 49 illustrates in vivo toxicity of Au@PDA@mHGF NW in ALI mice 24 h post-i.v. injection (or 48 h post-APAP disease induction) . Au@PDA@mHGF NW did not change blood chemistry or cell count. Grey shaded area represents the normal range of markers or cell counts for mice. WBC: white blood cell. RBC: red blood cell. Absolute neutrophils, lymphocytes, or monocytes (103/μL) : cell number per volume of blood. Neutrophils%, lymphocytes%, monocytes%: percentage cells among total WBC. RBC (106/μL) : cell number per volume of blood. Data are from n = 3, across 1 experiment.
Table 1. Endosomal escape of NPs for gene delivery with colocalization coefficient (< 0.5) in the literature from 2019 to 2024.
Table 2. Endosomal escape of NPs for gene delivery without reporting colocalization coefficient in the literature from 2019 to 2024.
Table 3. Primer sequences used in this work.
Table 4. Hydrodynamic diameter and zeta potential were measured by dynamic light scattering (DLS) at room temperature (RT) in water. For the serum stability test, different types of NWs were incubated in DMEM with 10%FBS for 24 h at 37 ℃.
Table 5. Loading of oligonucleotides on Au40@PDA NWs as determined by fluorescence assay.
Table 6. Top enriched GO terms identified from DEGs in A549 cells based on the pairwise comparison of “Au@PDA@T21 NW 24 h” group to “Au@PDA@T21 NP 24 h” groups. Data are from n = 3, across 1 experiment. Q < 0.05. Bolded GO terms are relevant to intracellular vesicles and ion homeostasis.
Table 7. Top enriched GO terms identified from DEGs in A549 cells based on the pairwise comparison of “Au@PDA@T21 NW 24 h” group to “Au@PDA@T21 NW 8 h” groups. Data are from n = 3, across 1 experiment. Q < 0.05. Bolded GO terms are relevant to intracellular vesicles and ion homeostasis.
Table 8. Physicochemical characterization of different types of mRNA-encased NWs by DLS. PDI = polydispersity index.
Supplementary Table 9. Loading of mRNA on Au@PDA@lipid NWs.
BRIEF DESCRIPTION OF THE SEQUENCES
SEQ ID NO: 1: miR223
SEQ ID NO: 2: siNog
SEQ ID NO: 3: antisense DNA against enhanced green fluorescence protein (asEGFP)
SEQ ID NO: 4: Mouse GAPDH Forward sequence 5’ to 3’
SEQ ID NO: 5: Mouse GAPDH Reverse sequence 3’ to 5’
SEQ ID NO: 6: Human GADPH Forward sequence 5’ to 3’
SEQ ID NO: 7: Human GADPH Reverse sequence 3’ to 5’
SEQ ID NO: 8: Mouse IL-12 Forward sequence 5’ to 3’
SEQ ID NO: 9: Mouse IL-12 Reverse sequence 3’ to 5’
SEQ ID NO: 10: Mouse IL-10 Forward sequence 5’ to 3’
SEQ ID NO: 11: Mouse IL-10 Reverse sequence 3’ to 5’
SEQ ID NO: 12: Mouse CD80 Forward sequence 5’ to 3’
SEQ ID NO: 13: Mouse CD80 Reverse sequence 3’ to 5’
SEQ ID NO: 14: Mouse CD206 Forward sequence 5’ to 3’
SEQ ID NO: 15: Mouse CD206 Reverse sequence 3’ to 5’
SEQ ID NO: 16: Mouse TNF-α Forward sequence 5’ to 3’
SEQ ID NO: 17: Mouse TNF-α Reverse sequence 3’ to 5’
SEQ ID NO: 18: Mouse TGF-β Forward sequence 5’ to 3’
SEQ ID NO: 19: Mouse TGF-β Reverse sequence 3’ to 5’
SEQ ID NO: 20: Mouse INF-γ Forward sequence 5’ to 3’
SEQ ID NO: 21: Mouse INF-γ Reverse sequence 3’ to 5’
SEQ ID NO: 22: Mouse Arg1 Forward sequence 5’ to 3’
SEQ ID NO: 23: Mouse Arg1 Reverse sequence 3’ to 5’
SEQ ID NO: 24: Human Noggin Forward sequence 5’ to 3’
SEQ ID NO: 25: Human Noggin Reverse sequence 3’ to 5’
SEQ ID NO: 26: Human BMP-2 Forward sequence 5’ to 3’
SEQ ID NO: 27: Human BMP-2 Reverse sequence 3’ to 5’
SEQ ID NO: 28: Human RUNX2 Forward sequence 5’ to 3’
SEQ ID NO: 29: Human RUNX2 Reverse sequence 3’ to 5’
SEQ ID NO: 30: Human C1C3 Forward sequence 5’ to 3’
SEQ ID NO: 31: Human C1C3 Reverse sequence 3’ to 5’
SEQ ID NO: 32: Human Col-1 Forward sequence 5’ to 3’
SEQ ID NO: 33: Human Col-1 Reverse sequence 3’ to 5’
SEQ ID NO: 34: Mouse α-SMA Forward sequence 5’ to 3’
SEQ ID NO: 35: Mouse α-SMA Reverse sequence 3’ to 5’
DETAILED DISCLOSURE OF THE INVENTION
The subject invention provides effective, safe compositions comprising nanoworms that can be used in methods of delivering nucleic acids to a subject. More specifically, the subject invention provides a polymer-coated nanoworm (NW) . Further, the subject invention provides a therapeutic nucleic acid-encased nanoworm for effective cellular delivery with limited localization inside acidic organelles.
Selected Definitions
As used herein, the singular forms “a” , “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms “including” , “includes” , “having” , “has” , “with” , or variants thereof are used in either the detailed description and/or the claims, such terms are intended to be inclusive in a manner similar to the term “comprising” . The transitional terms/phrases (and any grammatical variations thereof) “comprising” , “comprises” , “comprise” , “consisting essentially of” , “consists essentially of” , “consisting” and “consists” can be used interchangeably.
The phrases “consisting essentially of” or “consists essentially of” indicate that the claim encompasses embodiments containing the specified materials or steps and those that do not materially affect the basic and novel characteristic (s) of the claim.
The term “about” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured, i.e., the limitations of the measurement system. In the context of compositions containing amounts of ingredients where the terms “about” are used, these compositions contain the stated amount of the ingredient with a variation (error range) of 0-10%around the value (X ± 10%) . In other contexts, the term “about” is providing a variation (error range) of 0-10%around a given value (X ± 10%) . As is apparent, this variation represents a range that is up to 10%above or below a given value, for example, X ± 1%, X ± 2%, X ± 3%, X ± 4%, X ± 5%, X ± 6%, X ± 7%, X ± 8%, X ± 9%, or X ± 10%.
In the present disclosure, ranges are stated in shorthand to avoid having to set out at length and describe each and every value within the range. Any appropriate value within the range can be selected, where appropriate, as the upper value, lower value, or the terminus of the range. For example, a range of 0.1-1.0 represents the terminal values of 0.1 and 1.0, as well as the intermediate values of 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, and all intermediate ranges encompassed within 0.1-1.0, such as 0.2-0.5, 0.2-0.8, 0.7-1.0, etc. Values having at
least two significant digits within a range are envisioned, for example, a range of 5-10 indicates all the values between 5.0 and 10.0 as well as between 5.00 and 10.00 including the terminal values. When ranges are used herein, combinations and subcombinations of ranges (e.g., subranges within the disclosed range) and specific embodiments therein are explicitly included.
As used herein, the terms “therapeutically-effective amount, ” “therapeutically-effective dose, ” “effective amount, ” and “effective dose” are used to refer to an amount or dose of a compound or composition that, when administered to a subject, is capable of treating, preventing, or improving a condition, disease, or disorder in a subject. In other words, when administered to a subject, the amount is “therapeutically effective. ” The actual amount will vary depending on a number of factors including, but not limited to, the particular condition, disease, or disorder being treated, prevented, or improved; the severity of the condition; the weight, height, age, and health of the patient; and the route of administration.
As used herein, the term “treatment” refers to eradicating; reducing; ameliorating; abatement; remission; diminishing of symptoms or delaying the onset of symptoms; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; and/or improving a subject's physical or mental well-being or reversing a sign or symptom of a health condition, disease or disorder to any extent, and includes, but does not require, a complete cure of the condition, disease, or disorder. Treating can be curing, improving, or partially ameliorating a disorder. “Treatment” can also include improving or enhancing a condition or characteristic, for example, bringing the function of a particular system in the body to a heightened state of health or homeostasis.
As used herein, “preventing” a health condition, disease, or disorder refers to avoiding, delaying, forestalling, or minimizing the onset of a particular sign or symptom of the condition, disease, or disorder. Prevention can, but is not required, to be absolute or complete; meaning, the sign or symptom may still develop at a later time. Prevention can include reducing the severity of the onset of such a condition, disease, or disorder, and/or inhibiting the progression of the condition, disease, or disorder to a more severe condition, disease, or disorder.
In some embodiments of the invention, the method comprises administration of multiple doses of the nanoworm of the subject invention. The method may comprise administration of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100 or more therapeutically effective doses of a composition comprising the compounds of the
subject invention as described herein. In some embodiments, doses are administered over the course of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 14 days, 21 days, 30 days, 2 months, 3 months, 6 months, 9 months, 1 year, 1.5 years, 2 years, 2.5 years, 5 years, or more than 10 years. The frequency and duration of administration of multiple doses of the compositions is such as prevent or treat a disease, such as, for example kidney disease. Moreover, treatment of a subject with a therapeutically effective amount of the compounds of the invention can include a single treatment or can include a series of treatments. It will also be appreciated that the effective dosage of a compound used for treatment may increase or decrease over the course of a particular treatment. Changes in dosage may result and become apparent from the results of testing for kidney disease, such as, for example, glomerular filtration rate. In some embodiments of the invention, the method comprises administration of the compounds at several times per day, including but not limiting to 2 times per day, 3 times per day, and 4 times per day.
As used herein, “subject” refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both humans and non-human animals. In some embodiments, the subject is a mammal (such as an animal model of disease) , and in some embodiments, the subject is a human. The terms “subject” and “patient” can be used interchangeably. The animal may be for example, humans, pigs, horses, goats, cats, mice, rats, dogs, apes, fish, chimpanzees, orangutans, guinea pigs, hamsters, cows, sheep, birds, chickens, as well as any other vertebrate or invertebrate. In some embodiments, the subject is a cell line, such as, for example, A549 human lung cells, bEnd. 3 endothelial cells, bone-marrow-derive macrophage (BMDM) cells, and human mesenchymal stem cells (hMSCs) .
As used herein, an “isolated” or “purified” compound is substantially free of other compounds. In certain embodiments, purified compounds are at least 60%by weight (dry weight) of the compound of interest. Preferably, the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight of the compound of interest. For example, a purified compound is one that is at least 90%, 91%, 92%, 93%, 94%, 95%, 98%, 99%, or 100% (w/w) of the desired compound by weight. Purity is measured by any appropriate standard method, for example, by column chromatography, thin layer chromatography, or high-performance liquid chromatography (HPLC) analysis.
In this disclosure, the term “isolated nucleic acid” molecule means a nucleic acid molecule that is separated from other nucleic acid molecules that are usually associated with the isolated nucleic acid molecule. Thus, an “isolated nucleic acid molecule” includes, without limitation, a nucleic acid molecule that is free of nucleotide sequences that naturally
flank one or both ends of the nucleic acid in the genome of the organism from which the isolated nucleic acid is derived (e.g., a cDNA or genomic DNA fragment produced by PCR or restriction endonuclease digestion) . Such an isolated nucleic acid molecule is generally introduced into a vector (e.g., a cloning vector or an expression vector) for convenience of manipulation or to generate a fusion nucleic acid molecule. In addition, an isolated nucleic acid molecule can include an engineered nucleic acid molecule such as a recombinant or a synthetic nucleic acid molecule. A nucleic acid molecule existing among hundreds to millions of other nucleic acid molecules within, for example, a nucleic acid library (e.g., a cDNA or genomic library) or a gel (e.g., agarose, or polyacrylamide) containing restriction-digested genomic DNA, is not an “isolated nucleic acid” .
The term “nucleic acid” or “polynucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single-or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogs of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) , alleles, orthologs, single nucleotide polymorphisms (SNPs) , and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19: 5081 (1991) ; Ohtsuka et al., J. Biol. Chem. 260: 2605-2608 (1985) ; and Rossolini et al., Mol. Cell. Probes 8: 91-98 (1994) ) . The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.
As used herein, the terms “oligos” , “oligonucleotides” are used interchangeably to describe short single strands of synthetic DNA or RNA, such as, for example, about a 5 nucleic acid base sequence to about a 500 nucleic acid base sequence. “Oligo delivery” and “oligonucleotide delivery” are also used interchangeably to describe the method by which oligonucleotides are delivered into cells or targeted to specific cell types, tissues, or organs.
The term “gene” means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) involved in the transcription/translation of the gene product and the regulation of the transcription/translation, as well as intervening sequences (introns) between individual coding segments (exons) .
In certain embodiments, treatments are administered to a subject that “significantly reduce or abolish the expression of a gene or functioning of a protein encoded by the gene. ” This phrase refers to a reduction of gene expression in an amount of at least (or at least about) 30%as compared to a non-treated subject. Thus, the treated subjects exhibit significantly reduced or abolished expression of a gene exhibit a reduction in gene expression or expression of an active protein that can range from about 30%to about 99.99%about 40%to about 99.99%, about 50%to about 99.99%, about 60%to about 99.99%, about 70%to about 99.99%, about 80%to about 99.99%, about 90%to about 99.99% or are devoid of expression (expression is abolished) of the gene or an active protein encoded by the gene.
By “reduces” is meant a negative alteration of at least 1%, 5%, 10%, 25%, 50%, 75%, or 100%.
By “increases” is meant as a positive alteration of at least 1%, 5%, 10%, 25%, 50%, 75%, or 100%.
As used herein, a “pharmacokinetics” refers to the branch of pharmacology which specializes in the movement of drugs within the body.
As used herein, the terms “determining, ” “measuring, ” and “assessing, ” and “assaying” are used interchangeably and include both quantitative and qualitative determinations.
As used herein, “stromal cell differentiation” refers to the process in which a stromal cell changes to a differentiated type of cell. Stromal cell transplants occur when stromal cells replace cells damaged by chemotherapy or disease.
As used herein, “macrophage polarization” refers to the process by which macrophages produce distinct functional phenotypes as a reaction to specific microenvironmental stimuli and signals.
Nanoworm Compositions
The subject invention provides a polymer-coated nanoworm, such as, for example a polydopamine-coated nanoworm (PDA NW) , with a 1-dimensional assembly, which can reduce endosomal and/or lysosomal entrapment of nucleic acids when administering nucleic acids to a subject. In certain embodiments, the NW is an integrated core-shell nanoparticle with a metal nanochain, such as, for example, a gold (Au) nanochain, serving as the skeleton and a polymer shell, such as, for example, a polydopamine shell for adsorbing nucleic acids, such as, for example, oligonucleotides or an additional lipid coating to complex with a nucleic acid molecule, such as, for example, mRNA. In certain embodiments, the oligonucleotides can be about 15 to about 50 bases in length. In certain embodiments, the
mRNA can be about 700 to about 5000 bases in length. In certain embodiments, the metal nanochain is gold (Au) , copper, silver, iron oxide, ceria, zinc oxide, titanium oxide, rhodium, platinum, iron oxide, or any combination thereof. In certain embodiments, the metal nanoparticle can be capped with citrate.
In certain embodiments, the subject invention comprises an anionic gold-polydopamine core-shell nanoworm (Au@PDA NW) , which is used as a dual agent for gene delivery and for activating endosomal escape. In certain embodiments, the gold cores enable the tracking of endosomal escape by transmission electron microscopy (TEM) , and the PDA shell supports the surface adsorption of DNA, siRNA, miRNA and, with extra lipids, mRNA. In certain embodiments, the nucleic acid-encased NW is overall anionic, can enter cells, including, but not limited to, A549, bEnd3 endothelial cells, primary bone marrow-derived macrophages (BMDM) , and human mesenchymal stromal cells (hMSCs) , without transfection agents. Without being bound to any theory, the nucleic acid-encased NW enter the cells naturally activating the ClC3 H+/Cl-ion exchanger to mediate endosomal escape and accumulating H+ and Cl-for membrane rupture, and robustly escaping from late endosomes, where the PCC is ≤0.2, without cell-penetrating peptides or mechanical stimuli. In preferred embodiments, the mechanism for endosomal escape is distinct from the proton sponge effect due to the involvement of ClC3. In more preferred embodiments, the anionic gold-polydopamine core-shell nanoworm of the subject invention can be used for programming cellular responses, for example in primary macrophage polarization and stem cell differentiation, and for treating diseases, including, but not limited to, kidney fibrosis and acute liver injury.
In certain embodiments, to prepare the polymer-coated nanoworm, a metal nanoparticle can be sonicated with a polymer to produce a core-shell nanochain. In certain embodiments, the duration of the sonication is about 1 hour at a frequency of about 40 kHz. In certain embodiments, each metal nanoparticle has a diameter of about 10 nm to about 100 nm or about 40 nm. In certain embodiments, the nanoworm has 2, 3, 4, 5, 6, or 7 metal nanoparticle cores in a chain. In preferred embodiments, the nanoworm has 4 or 5 metal nanoparticle cores per chain. In certain embodiments, the polymer shell has thickness of about 7 nm to about 35 nm or about 20 nm. In certain embodiments, the polymer comprises polydopamine, poly (ethylene glycol) , poly (ethylene oxide) , poly (lactic acid) , poly (glycolic acid) , poly (ethylene oxide) -poly (lactic acid) , poly (ethylene oxide) -poly (glycolic acid) , poly (glycolic acid) -poly (lactic acid) , sodium alginate, dextran, polyethylenimine (PEI) , silica, or any combination thereof.
In certain embodiments, an oligonucleotide can be adsorbed onto the polymer-coated nanoworm by electrostatic interaction in acidic conditions, such as, for example, at a pH of less than or equal to about 3.0 or about 2.5. In certain embodiments, the oligonucleotide can be labeled with a dye, such as, for example, cyanine (Cy) . In certain embodiments, to generate an oligonucleotide (oligo) -containing nanoworm, the negatively charged oligo, Cy 5-labeled 21 thymines (Cy5-T21) , can be adsorbed onto the polymer-coated nanoworm by electrostatic interaction in acidic conditions. In certain embodiments, the oligonucleotides have a length of about 15 to about 50 bases. In certain embodiments, the oligonucleotides can be single stranded oligonucleotides, such as, for example, microRNA, and/or double stranded oligonucleotides, such as, for example, siRNA. In certain embodiments, to measure the loading of oligonucleotides on the nanoworms, the nucleic acids can be labeled, such as, for example, by using Cy5-labeled oligonucleotides or mRNA, and then the concentration of the oligonucleotides in the supernatant can be subtracted after purification. In certain embodiments, for the adsorption of oligonucleotides, the oligonucleotides can be mixed with the nanoworms in an acidic buffer, such as, for example, in an acidic citrate buffer at a pH of less than or about 3. In certain embodiments, for the adsorption of mRNA, the mRNA can be mixed with the nanoworms in nuclease-free water at a pH of about 7. In certain embodiments, exemplary oligonucleotides include miR223 (3'-CCCAUAAACUGUUUGACUG-5'; SEQ ID NO: 1) and siNog (5′-AAC ACU UAC ACU CGG AAA UGA UGG G-3′; SEQ ID NO: 2) . In certain embodiments, the mRNA can encode hepatocyte growth factor (HGF) , chemokine receptor type 4 (CXCR4) , bone morphogenetic protein 7 (BMP-7) , enhanced green fluorescence protein (EGFP; SEQ ID NO: 3: UAC CAC UCG UUC CCG CUC CUC GAC AAG UGG CCC CAC CAC GGG UAG_GAC CAG CUC GAC CUG CCG CUG CAU UUG CCG GUG UUC AAG UCG CAC AGG CCG CUC CCG CUC CCG CUA CGG UGG AUG CCG UUC GAC UGG GAC UUC AAG UAG_ACG UGG UGG CCG UUC GAC GGG CAC GGG ACC GGG UGG GAG CAC UGG UGG GAC UGG AUG CCG CAC GUC ACG AAG UCG GCG AUG GGG CUG GUG UAC UUC GUC GUG CUG AAG AAG UUC AGG CGG UAC GGG CUU CCG AUG CAG GUC CUC GCG UGG UAG_AAG AAG UUC CUG CUG CCG UUG AUG UUC UGG GCG CGG CUC CAC UUC AAG CUC CCG CUG UGG GAC CAC UUG GCG UAG_CUC GAC UUC CCG UAG_CUG AAG UUC CUC CUG CCG UUG UAG_GAC CCC GUG UUC GAC CUC AUG UUG AUG UUG UCG GUG UUG CAG AUA UAG_UAC CGG CUG UUC GUC UUC UUG CCG UAG_UUC CAC UUG AAG UUC UAG_GCG GUG UUG UAG_CUC CUG CCG UCG CAC GUC GAG CGG CUG GUG AUG
GUC GUC UUG UGG GGG UAG_CCG CUG CCG GGG CAC GAC GAC GGG CUG UUG GUG AUG GAC UCG UGG GUC AGG CGG GAC UCG UUU CUG GGG UUG CUC UUC GCG CUA GUG UAC CAG GAC GAC CUC AAG CAC UGG CGG CGG CCC UAG_UGA_GAG CCG UAC CUG CUC GAC AUG UUC AUU) , red fluorescence protein (RFP) , or any combination thereof, the miRNA is miR223, or the siRNA is siNog.
In certain embodiments, the polymer-coated nanoworm can further comprise a lipid coating. In certain embodiments, the lipid coating comprises a lipid, such as, for example, 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) , cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DOPE) , 1, 1′- ( (2- (4- (2- ( (2- (bis (2-hydroxydodecyl) amino) ethyl) (2-hydroxydodecyl) amino) ethyl) piperazin-1-yl) ethyl) azanediyl) bis (dodecan-2-ol) (C12-200) , (6Z, 9Z, 28Z, 31Z) -Heptatriaconta-6, 9, 28, 31-tetraen-19-yl 4- (dimethylamino) butanoate (DLin-MC3-DMA) , ( (4-hydroxybutyl) azanediyl] di (hexane-6, 1-diyl) bis (2-hexyldecanoate) (ALC-0315) , (dimethyldioctadecylammonium bromide) (DDA) , (1, 2-distearoyl-sn-glycero-3-phosphocholine) (DSPC) , (1, 2-dioleoyl-3-dimethylammonium-propane) DODAP, (1, 2-dioleyloxy-3-dimethylaminopropane) (DODMA) , (Dipalmitoylphosphatidylcholine) (DPPC) , or any combination thereof. In certain embodiments, the DOTAP is at a concentration of about 33 mol%to about 37 mol%or about 35 mol%. In certain embodiments, the cholesterol is at a concentration of about 47 mol%to about 51 mol%or about 49 mol%. In certain embodiments, the DOPE is at a concentration of about 14 mol%to about 18 mol%or about 16 mol%. In certain embodiments, the mol%of each lipid refers to the percentage in the whole lipid mixture (e.g., a mixture of cholesterol, DOTAP, and DOPE) initially added to the polymer-coated nanoworm. In certain embodiments, the lipid and polymer-coated nanoworm has an average hydrodynamic size of about 220 nm to about 275 nm or about 252 nm and a zeta potential of about +30 mV to about +45 mV or about +40 mV. In certain embodiments, the lipid and polymer-coated nanoworm can be formed by mixing a lipid with the polymer-coated nanoworm.
In certain embodiments, lipid and polymer-coated nanoworm can further comprise an oligonucleotide, such as, for example, mRNA, which can be prepared by mixing the lipid and polymer-coated nanoworm with an oligonucleotide. In certain embodiments, an oligonucleotide can be adsorbed onto the lipid and polymer-coated nanoworm by electrostatic interaction in acidic conditions, such as, for example, at a pH of less than or equal to about 3.0 or about 2.5. In certain embodiments, the positive charge of the lipid coating on the nanoworms allows for the complexation of large, macromolecular nucleic acids, such as, for example, mRNA, with the lipid nanoworms in nuclease-free water. In certain embodiments,
for the oligo-encased nanoworms, the shorter oligonucleotides can be adsorbed onto the polydopamine shell of the nanoworms via electrostatic interactions in an acidic citrate buffer, whereby the polymer shell becomes protonated and acidic. In certain embodiments, the lipid and polymer-coated nanoworm with an therapeutic mRNA has an average hydrodynamic size of about 225 nm to about 280 nm or about 258 nm and an overall zeta potential of about -30 mV to -50 mV about -32 mV.
In certain embodiments, the nanoworm can have overall anionic charge and can readily load different types of therapeutic nucleic acids, including, for example, antisense oligonucleotides (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , and messenger RNA (mRNA) . In certain embodiments, the zeta potential of the nanoworm is less than about -10 mV (e.g., -20 mV) . In certain embodiments, the negative charge on the surface of nanoworms is evenly dispersed. In certain embodiments, the nanoworm has a limited colocalization with the endosome and/or lysosomes, with a colocalization coefficient of about 0.1 to about 0.2. When compared to a widely used transfection agent, such as, for example, Lipofectamine3000, the subject nanoworm exhibits more effective gene regulation to support stem cell differentiation and macrophage polarization in vitro, gene expression ex vivo to support stem cell-based treatment of kidney fibrosis in vivo, and, finally, gene expression in vivo to rescue liver injury.
Methods of Administering Nanoworm Compositions
In certain embodiments, the nanoworm of the subject invention can deliver various types of therapeutic nucleic acids, such as, for example ASO, siRNA, miRNA, and mRNA, with low cytotoxicity and high endosomal escape efficiency in vitro and in vivo. In certain embodiments, the escape efficiency can be quantified using, for example, confocal imaging of the labeled nanoworm inside the cell, such as, for example, fluorescently labeled nucleic acids of the nanoworms. In certain embodiments, acidic organelles, such as, for example, late endosomes and lysosomes, can be stained utilizing a dye, such as, for example, LysoTracker. In certain embodiments, the lack of overlap between stained organelles and labeled nanoworms, as indicated by a Pearson correlation coefficient (PCC) <0.4 through ImageJ to measure the colocalization of the two signals, suggests escape. In certain embodiments, a PCC >0.6 indicates strong overlap, indicating entrapment of the nanoworms inside the acidic compartments. In preferred embodiments, the PCC is ≤0.2.
In certain embodiments, the therapeutic nucleic acid can be used to treat a variety of diseases and disorders, such as, for example, chronic kidney disease, acute liver disease,
cancers, cell-based immunotherapy, or any combination thereof. In certain embodiments, the nanoworm enters epithelial cells, liver cells, kidney cells, endothelial cells, primary macrophages, mesenchymal stem cells, neural cells, or any combination thereof in the subject.
In specific embodiments, several applications of the compositions of the invention are administered at specific time intervals. In certain embodiments, the subject nanoworm can be used for in vitro transfection, ex vivo transfection followed by injection into a subject as cell-based therapy, or direct in vivo transfection via injection of nanoworm into the subject.
In certain embodiments, for in vitro transfection the cells can be dosed once with specific nucleic acid-encased nanoworms for about 2 to about 3 days for the cellular process to occur (e.g., macrophage polarization, stem cell differentiation, gene knockdown) . In certain embodiments, for ex vivo transfection stem cells can be incubated with therapeutic nucleic acid-encased lipid nanoworms once for about 12 hours; subsequently, the transfected stem cells can be injected into a subject, such as, for example, a subject with kidney disease. In certain embodiments, at least about 1 day to about 6 months, or about 11 days to about 4 weeks can pass before efficacy assessment. In certain embodiments, for direct in vivo transfection, at least 1, 2, 3, 4, 5, 6, 7, 8, 9 10, about 20, about 30, about 40, about 50, about 100 or more dose of therapeutic nucleic acid-encased lipid nanoworms can be administered to a subject, such as, for example, a subject with acute liver injury. In certain embodiments, at least about 6 hours to about 168 hours or about 48 hours can pass before efficacy assessment.
The skilled artisan will understand that the dosage of the compositions of the instant invention varies, depending upon, for example, the route of administration, the particular nanoworm to be used in the composition, other drugs being administered, and the age, condition, gender and seriousness of the disease in the subject as described above. An effective dose of a nanoworm composition of the invention generally ranges between about 1 μg/kg of body weight and 100 mg/kg of body weight. Examples of such dosage ranges include, but are not limited to, about 1.5 μg/kg to about 90 mg/kg; about 2 μg/kg to about 80 mg/kg; about 5 μg/kg to about 70 mg/kg; about 7.5 μg/kg to about 65 mg/kg; about 10 μg/kg to about 60 mg/kg; about 12.5 μg/kg to about 55 mg/kg; about 15 μg/kg to about 50 mg/kg; about 17.5 μg/kg to about 45 mg/kg; about 20 μg/kg to about 40 mg/kg; about 22.5 μg/kg to about 35 mg/kg; about 25 μg/kg to about 30 mg/kg; about 27.5 μg/kg to about 25 mg/kg; about 30 μg/kg to about 20 mg/kg; about 32.5 μg/kg to about 18 mg/kg; about 35 μg/kg to about 17 mg/kg; about 37.5 μg/kg to about 16 mg/kg; about 40 μg/kg to about 15 mg/kg; about 42.5 μg/kg to about 14 mg/kg; about 45 μg/kg to about 13 mg/kg; about 47.5 μg/kg to about 12 mg/kg; about 50 μg/kg to about 11 mg/kg; about 52.5 μg/kg to about 10 mg/kg;
about 55 μg/kg to about 9 mg/kg; about 57.5 μg/kg to about 8 mg/kg; about 60 μg/kg to about 7 mg/kg; about 62.5 μg/kg to about 6 mg/kg; about 65 μg/kg to about 5 mg/kg; about 67.5 μg/kg to about 4 mg/kg; about 70 μg/kg to about 3 mg/kg; about 72.5 μg/kg to about 2 mg/kg; about 75 μg/kg to about 1 mg/kg; about 77.5 μg/kg to about 800 μg/kg; about 80 μg/kg to about 700 μg/kg; about 82.5 μg/kg to about 600 μg/kg; about 85 μg/kg to about 500 μg/kg; about 87.5 μg/kg to about 400 μg/kg; about 90 μg/kg to about 300 μg/kg; about 92.5 μg/kg to about 200 μg/kg; about 95 μg/kg to about 100 μg/kg.
In certain embodiments, the nanoworms of the invention are administered at a dose of about 50 μg/kg to about 200 μg/kg, preferably, about 75 μg/kg to about 150 μg/kg. or, most preferably, about 100 μg/kg to about 120 μg/kg.
In some embodiments, the therapeutically effective amount of a nanoworm composition of the invention can be administered through intravenous, oral, rectal, bronchial, nasal, topical, buccal, sub-lingual, transdermal, vaginal, intramuscular, intraperitoneal, intra-arterial, intracerebral, intraocular administration or in a form suitable for administration by inhalation or insufflation, including powders and liquid aerosol administration, or by sustained release systems such as semipermeable matrices of solid hydrophobic polymers containing the nanoworms of the invention. Administration may be also by way of other carriers or vehicles such as patches, micelles, liposomes, vesicles, implants (e.g. microimplants) , synthetic polymers, microspheres, nanoparticles, and the like.
In certain embodiments, the nanoworm compositions of the instant invention may be formulated for parenteral administration e.g., by injection, for example, bolus injection or continuous infusion. In addition, the nanoparticle compositions may be presented in unit dose form in ampoules, pre-filled syringes, and small volume infusion or in multi-dose containers with or without an added preservative. The nanoparticle compositions may be in forms of suspensions, solutions, or emulsions in oily or aqueous vehicles. The composition may further contain formulation agents such as suspending, stabilizing and/or dispersing agents. In further embodiments, the active ingredients of the compositions according to the instant invention may be in powder form, obtained by aseptic isolation of sterile solid or by lyophilization from solution for constitution with a suitable vehicle, e.g. sterile, pyrogen-free water, before use.
Advantageously, our nanoworms do not have huge concerns over genotoxicity. The nanoworms of the subject invention do not exert any voltage on the cells and can overcome the delivery barriers of deep tissue through various routes of administration, such as, articular injections, intrathecal injections, and retrobulbar injections.
In certain embodiments, the nucleic acid-encased nanoworm effectively regulate genes by efficiently entering multiple cell lines and escaping the endosome and/or lysosomes. In certain embodiments, the nanoworm can increase or decrease the transcription and/or translation rate by at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 98%, about 99%, or 100%. The subject invention provides a nanoworm for effective cellular delivery with limited localization inside acidic organelles (e.g., late endosomes and lysosomes) . In some embodiments, the nanoworm can exit the late endosomes by increasing chloride ion accumulation in the intracellular vesicles, resulting in vesicle swelling and subsequent membrane rupture.
MATERIALS AND METHODS
Lysotracker Staining
Lysotracker staining uses lysotracker probes to label lysosomes in live cells. Lysotracker staining dyes mammalian cells and yeast with fluorescent dyes that can be blue, green, visible red, and far-red fluorescence. In some embodiments, lysotracker staining can be done under confocal imagining. In one embodiment, the subject invention discovered the colocalization of Cy5-T21@NW and Lysotracker signals are consistently low across all tested cell lines, with low Pearson’s correlation coefficient (PCC) value at 0.1~ 0.2 (Figure 1D) . This was done by lysotracker staining.
In some embodiments, confocal images of Lysotracker staining can show that the effect of a treatment on the colocalization of organelles. For example, the methods of the subject invention found that the treatment of Bafilomycin A1 did not alter the colocalization of the acidic organelles and the NWs, indicating that the endosomal escape of NWs is not based on the proton sponge effect (Figure 2A) .
In other embodiments, lysotracker staining can be used to test the endosomal escape efficiency of nanoworms. For example, testing the endosomal escape efficiency of asEGFP-encased NWs by lysotracker staining. This showed that the PCC is 0.134 after 24 h incubation (Figure 28A) .
Immunofluorescence (IF) Staining
IF staining is a method that permits visualization of many components in a given tissue or cell type. IF staining is completed through combinations of specific antibodies tagged with fluorophores, fluorescent chemical compounds that can re-emit light upon light excitation.
Figure 4A and Figure 4D demonstrates the results of IF staining using red, green, and blue dyes. In some embodiments, IF staining can be used to evaluate polarization. Such as is shown in Figure 4D, intense CD80-positive (M1-specific maker) signals were found in the PBS, pristine NW, and free miR223 groups while cells pretreated with Au@PDA@miR-223 NW showed the weakest signals for CD80 but stronger signals for CD206 (M2-specific marker) . qRT-PCR analysis also verified the markedly decreased expression of M1-associated genes (e.g., IL-12, CD80, TNF-α, and IFN-γ) in Au@PDA@miR-223 NW group (downregulated by at least 50%) , and Lipo + miR-223 group showed modest depressed expression (downregulated by ~ 25%) .
Intracellular trafficking of A549 cells
Seeded in 35-mm confocal dishes, cells were incubated with 0.2 nM Cy5-labeled Au@PDA@T21 NWs, formulated in 1 mL of full DMEM, for various time points. After two PBS rinses and fixation with cold methanol for 10 min and ice-cold acetone for 1 min, the cells were blocked by 2%BSA for 1 h at RT, stained with primary antibodies against EEA1 (1: 100; Abcam, ab2900) , Rab 9 (1: 100 dilution; Abcam, ab179815) , or LAMP1 (1: 150 dilution; Abcam, ab24170) in 2%BSA overnight at 4℃. After two PBS rinses, the cells were stained with 2 μg/mL Alexa Fluor 488-labeled, goat secondary antibody against rabbit (Invitrogen, A-11008) in 2%BSA for 1 h at RT, stained by 1 μg/mL DAPI (Invitrogen, D1306) in PBS for 10 min at RT, and rinsed with PBS twice. Coverslips were mounted with Antifade Mountant (Thermo Scientific; P36980) for confocal laser scanning microscopy. The excitation wavelengths of DAPI, Alexa Fluor 488, and Cy5 are 405 nm, 488 nm and 650 nm, respectively. The emission wavelengths of DAPI, Alexa Fluor 488, and Cy5 are 410–470 nm, 495–600 nm, and 655–750 nm, respectively.
ClC3 colocalization staining of A549 cells
After fixation and BSA blocking, cells were stained with primary antibodies against CCl3 (1: 100; Invitrogen, #MA5-45554) After two PBS rinses, the cells were stained with 2 μg/mL Alexa Fluor 488-labeled, goat secondary antibody against rabbit and Cy5-labeled goat secondary antibody against mouse (Invitrogen, A10524) in 2%BSA for 1 h at RT, stained by 1 μg/mL DAPI in PBS for 10 min at RT, and rinsed with PBS twice.
BMDM polarization
After fixation and BSA blocking, the polarized BMDMs were stained with primary antibodies against CD206 (1: 150; Abcam, ab64693) and CD80 antibodies (1: 100; Abcam, ab254579) in 2.5%BSA overnight at 4℃. After three PBS rinses, the cells were stained by 1 μg/mL Alexa Fluor 488-labeled, goat secondary antibody against rabbit for 1 h at RT, stained by 1 μg/mL DAPI in PBS for 10 min at RT, and rinsed with PBS twice.
Tissue sections
Fresh tissues were embedded in OCT and stored at –80℃. Tissue cryosections of 8μm thick were dried for 20 min and fixed with 4%PFA for 15 min at RT. Next, the cryosections were blocked with 2.5%horse serum (Vector Laboratories, S-2012-50) at RT for 1 h and stained with primary antibodies against HNF4α (1: 100; Thermo Scientific, MA1199) in 2.5%horse serum at 4℃ overnight. After PBS rinses, the cryosections were stained by Alexa Fluor 532-labeled, goat secondary antibody against mouse (1: 1000; Invitrogen, A11002) at RT for 1 h, stained by 1 μg/mL DAPI in PBS for 10 min at RT, washed, and mounted with Antifade Mountant for confocal laser scanning microscopy. The excitation and emission wavelengths of Alexa Fluor 532 are 532 nm and 542–640 nm, respectively.
Western Blotting
Western blotting is a laboratory technique used to detect a specific protein in a blood or tissue sample. The method uses gel electrophoresis to separate the sample’s proteins. The western blotting technique begins with an antigen sample that is placed in separation gel. Then the separated proteins are transferred to a nitrocellulose sheet (blot) in a blotting tank. Following the blotting tank, the antibodies are labeled, and the blot is immune-stained. The sample then goes through autoradiography and is developed into a fixed autoradiograph where the antigen bands can be visualized.
In some embodiments, RNA sequencing results can be verified by western blotting. Figure 2G demonstrates a western blot analysis of ClC3 expression in A549 cells treated with NWs and Au@PDA NPs, and A549 cells treated with NWs for “8 h” and “24 h” .
Oligo Delivery
Oligo delivery or oligonucleotide delivery describes a method by with oligonucleotides are delivered into cells or targeted to specific cell types, tissues, or organs. In some embodiments, this method can be used to silence the expression of a gene.
Oligos/oligonucleotides are short single strands of synthetic DNA or RNA that serve as the starting point for many biological methods. Oligos can be used in genetic testing, forensic research, and sequencing. In some embodiments, oligonucleotides can be administrated via parenteral routes, which comprise either intravenous infusion or subcutaneous injection.
In one embodiment, to generate oligonucleotides (oligos) -containing NWs, we adsorbed the negatively charged oligos, cyanine (Cy) 5-labeled 21 thymines (Cy5-T21) onto the NWs by electrostatic interaction at acidic condition for proof of concept.
The methods of the subject invention further provide applications for nanoworms to deliver oligos into cells.
Synthesis of Au@PDA NWs and Au@PDA NRs
Citrate-capped Au NPs (Cit-Au NPs) of ~40 nm in diameter were synthesized according to an established seed-mediated growth method. Ci-Au NPs of ~20 nm and ~60 nm in diameter and Cit-Au NRs (45 nm × 180 nm) were purchased from Nanopartz. Aux@PDA NWs and Au@PDA NRs were prepared using our published method. Briefly, 1 mL of 0.1 nM Au NPs (x = 20, 40, or 60 nm) and Au NRs was diluted by adding 1 mL of Tris buffer (10 mM, pH=8.5) . Then, 2 mL of freshly prepared dopamine (DA) solution (1 mg/mL) in Tris buffer was quickly injected into the diluted gold NP solution under sonication. After 60 min, the resultant NWs and NRs were collected and purified by centrifugation at 5000 rpm (for Au40@PDA NWs, Au60@PDA NWs, and Au40@PDA NRs) or 10000 rpm (for Au20@PDA NWs) for 15 min twice. The purified nanoparticles were resuspended in Nanopure water and stored at 4℃ for future use.
Synthesis of unassembled Au@PDA NPs
Briefly, 1 mL of Cit-Au NPs of 40 nm stock solution (0.1 nM) was injected into 1 mL of HS-PEG5000-COOH (Jenkem; 0.1 mg/mL) in Nanopure water. After 1 h of sonication, the resultant Au@PEG NPs were collected by centrifugation at 10,000 rpm for 15 min twice. Next, 1 mL of Au@PEG NPs was diluted to 0.05 nM by adding 1 mL of Tris buffer (5 mM, pH=8.5) , followed by quickly injecting 2 mL of freshly prepared 0.5 mg/mL DA in Tris buffer. Upon 1 h of sonication, the resultant unassembled Au@PDA NPs were collected and purified by repeated centrifugation at 5000 rpm for 15 min twice. Purified Au@PDA NPs were resuspended in Nanopure water and stored at 4 ℃.
Synthesis of oligonucleotide-encased NWs (Au@PDA@oligo NWs)
Unless otherwise specified, oligonucleotide-encased NWs were synthesized by using Au@PDA NWs with 40-nm Au cores as starting materials. Typically, concentrated Au@PDA NWs were resuspended into 5mL of citrate buffer (20 mM, pH=3, made with nuclease-free water) to a final concentration of 1 nM. After 30 min of sonication to tune the negative PDA shell of NW to a positive surface charge, the same volume of oligonucleotide solution (500 nM, nuclease-free water) was added to the NWs; the oligonucleotides were either T21 DNA, ASO against enhanced green fluorescent protein (asEGFP) , miR-223, or siRNA against Noggin (Idobio) . After 1 h of incubation, the resultant oligonucleotide-encased Au@PDA NWs were collected by centrifugation at 10,000 rpm for 15 min at 4 ℃twice and resuspended in nuclease-free water and stored at 4 ℃. Loading oligonucleotides on Au@PDA NPs would follow the same protocol as Au@PDA NWs, keeping the total concentration of Au cores the same.
Preparation of mRNA-encased, lipid-coated Au@PDA NWs
Step 1. Synthesis of lipid-coated NWs
DOTAP (1, 2-dioleoyl-3-trimethylammonium propane; Cayman, 15110) , cholesterol (Sigma, C8667) , and DOPE (dioleoylphosphatidylethanolamine; Cayman, 15091) were added to chloroform (≥99.8%, Fisher Chemical) at a molar ratio of 35: 49: 16, keeping the total lipid concentration at 1 mg/mL. Upon rotary evaporation of the lipid mixture at 32 ℃ to remove the chloroform, 10 mL of Au@PDA NWs (0.5 mg/mL in Nanopure water) was added to the dried lipid film dropwise. After sonicating the mixture for 1 h, the excess lipids were removed by centrifugation at 5000 rpm for 15 min twice, and the purified LNWs were dispersed in Nanopure water and stored at 4 ℃.
Step 2. Synthesis of mRNA by in vitro transcription (IVT)
Circular plasmid DNA template of pEGFP (Addgene, BPK1098) dilluted in 16 μL nuclease-free waterwere linearized by adding 1 μL of FastDigest BshTI (Thermo Scientific, FD1464) and 2 μL diggestion buffer per μg of DNA at 37 ℃ for 1 h. Likewise, circular plasmid DNA templates of pHGF, pBMP7-OFP, and pCXCR4-GFP (Sino Biological) were linearized by adding FastDigest XbaI (Thermo Scientific, FD0684) . After column purification by thePCR &DNA cleanup kit (NEB, T1030S) , the linearized DNA template underwent IVT by the T7 HiScribe ARCA mRNA Transcription Kit with tailing (NEB, E2060S) . Briefly, 2 μL (0.5 μg/μL) of linearized DNA was mixed with 10 μL of 2X ARCA/NTP Mix and 2 μL T7 RNA polymerase Mix, diluted with nuclease-free water to 20
μL, and incubated at 37 ℃ for 30 min. After removing the unreacted DNA template by adding 2 μL DNase I for incubation at 37 ℃ for 15 min, the mRNA was tailed with poly (A) using 5 μL poly (A) polymerase for incubation at 37 ℃ for 30 min. Upon purification by the RNA Cleanup kit (NEB, T2040L) , the mRNA product was characterized by Nanodrop (Thermo Scientific) for its concentration and gel electrophoresis for its molecular weight. The 1.2%native agarose gel, made in diethyl pyrocarbonate (DEPC; Macklin, D6079 X) -treated TBE (Tris-borate-ethylenediaminetetraacetic acid) buffer, contains 1X SYBR gold nucleic acid gel stain (1: 10000 dilution; Thermo Scientific, S11494) . For each gel lane, 2 μL of single-strand RNA ladder (NEB, N0362S) or mRNA product (200 ng) was mixed with 8 μL of RNA loading dye, followed by heating at 90 ℃ for 2 min and cooling down on ice for 2 min before loading. Upon electrophoresis at 80 mV for 1 h, the gel was imaged by the ChemiDocTM Touch imaging system (Bio-Rad) . The mRNA was product stored in -70 ℃.
Step 3. Preparation of mRNA-encased, lipid-coated Au@PDA NWs
Typically, 1 mL of mRNA was added to 1 mL of lipid-coated Au@PDA NWs (in nuclease-free water) at a mass ratio of 1: 100. Upon incubation for 30 min, the excess mRNA was removed by centrifugation at 5000 rpm for 15 min twice at 4 ℃.
Characterization of Au@PDA NWs and Au@PDA NPs
The concentration of NWs and Au@PDA NPs was determined by inductive coupled-plasma mass spectrometry (ICP-MS) (Agilent 7900) with reference to a standard curve of known gold concentration (Au 197 isotope) in parts per billion (ppb) . To quantify the gold content in NWs solutions, Au@PDA NPs solutions, tissues, and cells for the efficacy study, we first converted ICP-MS raw data from ppb to μg/L using the equation 1ppb = 1 μg/L, followed by multiplying the volume of dilute 2%nitric acid that contains the digested samples.
The hydrodynamic diameters and zeta potential of NWs and NPs were measured by the DelsaMax PRO dynamic light scattering (DLS) analyzer (Beckman Coulter) . Reported DLS data represent the values from three independent measurements. The DLS data quality was interpreted by analyzing the autocorrelation function and its fit, where the sum of square (SOS) threshold was set to be under 100. (Only measurements with an SOS value <100 were counted) . As independent samples were used for different assays or experiments, a slight batch-to-batch variation was observed. To test the colloidal stability, the NW solutions were dispersed in DMEM containing 10 %fetal bovine serum (FBS; Gibco, 10270106) . After 24 h
of incubation at 37 ℃, the NW solutions were analyzed using UV-Vis spectrometry and DLS. Reported DLS values represent mean ± SD from three independent measurements.
The loading of each type of nucleic acid on the NWs was calculated based on a fluorescence-based assay, following literature precedent. The concentration of Cy5-labeled oligos before and after their adsorption onto NWs or Au@PDA NPs was measured by microplate reader (MULTISKAN GO, Thermo Scientific) , followed by dividing the difference in nucleic acid concentration by the concentration of NWs and NPs to obtain the loading of nucleic acid.
Cell culture
All cell types were maintained at 37℃ and 5%CO2. A549 lung epithelial cells (ATCC) and bEnd. 3 brain endothelial cells (ATCC were cultured in Dulbecco’s Modified Eagle Medium (DMEM; Gibco, 12100046) , supplemented with 10%feFBSadde) and 1%penicillin-streptomycin (P/S; Gibco, 15140122) . Human mesenchymal stromal cells (hMSCs, Lonza) were cultured in α-minimum essential medium (α-MEM; Gibco, 11900073) supplemented with 16.7%FBS, 1%P/Sand 1%L-glutamine (Gibco, 25030081) . Bone marrow-derived macrophages (BMDMs) were isolated from Balb/c mice of 9–11 wk old. Briefly, hind legs were dislocated from the hip bone and sterilized in three changes of 75%ethanol for 1 min. After removing the bones below the knee joint by cutting the ligaments, the separated femurs and tibias were rinsed with precooled phosphate buffered saline (PBS; Gibco, 21600010) . The ends of the femur and tibia were cut, and each marrow cavity was irrigated with 5 mL of RPMI 1640 medium (Gibco, 11875093) containing 10%FBS. Cells were pelleted by centrifugation at 1500 rpm for 5 min, treated with iced red blood cell lysis buffer (BioLegend, 420301) for 2 min, pelleted again, and cultured in DMEM containing 10%FBS, 1%P/S, and 50 ng/mL colony stimulating factor-1 (CSF-1; SinoBiological, 51112-MNAH) for 7 d.
Cytotoxicity
Cells were plated on 96-well plates and cultured till ~80%confluence. Then, cells were incubated with Au@PDA@oligonucleotide NWs (0.2 nM) formulated in 0.1 mL of full culture medium or mRNA-LNWs (10 μg) in 0.1 mL full α-MEM for 24 h. After two PBS rinses, cell viability was measured by the alamarBlue assay (Invitrogen, DAL1025) according to the manufacturer’s protocol.
Staining of intracellular chloride ions
Cells were seeded in a 35-mm confocal dish (SPL Life Sciences) and cultured till 80–90%confluence. Next, 1 mL of 10 mM MQAE (N- (ethoxycarbonylmethyl) -6-methoxyquinolinium bromide; Invitrogen, E3101) , formulated in OptiMEM (Gibco, 31985070) , was added. After incubation at 37 ℃ for 30 min, the staining solution was removed, and cells were rinsed with PBS twice and replenished with phenol red-free DMEM (Gibco, 31053028) . Live cell images were acquired by a confocal laser scanning microscope (TCS SP8, Leica) at identical imaging settings. The excitation and emission wavelengths for MQAE are 405 and 410~480 nm, respectively.
Ratiometric imaging for intracellular pH measurement
Cells were seeded in a 35-mm confocal dish and cultured till 80–90%confluence. The pH of intracellular acidic vesicles was analyzed by ratiometric analysis of the fluorescence intensities of the pH-sensitive dye pHrodo green (10 kDa dextran, Invitrogen, P10361) and the pH-insensitive dye Alexa Fluor 594 (10 kDa dextran; Invitrogen, D22913) on the same day. After coincubation with both dyes at 100 μg/mL for 8 h, the cells were fixed with 4%formaldehyde for 10 min and then incubated in calibration buffers (Invitrogen, P35379) at a range of pH values (4.5, 5.5, 6.5, and 7.5) for 2 h before confocal imaging. In each image, individual integral intensities of both dyes in each acidic vesicle were calculated at different pH values in three independent experiments with three replicates per experiment (n= 300 vesicles) . Typically, the fluorescence intensity of pHrodo green increases with lowering pH, but that of Alexa 594 remains consistent. Then, the R/G fluorescence ratio of Alexa 594 to pHrodo green was calculated and plotted. To determine the pH of NW-containing intracellular vesicles, cells were co-incubated with Cy5-labeled Au@PDA T21 NWs (0.2 nM) , pHrodo green (100 μg/mL) , and Alexa Fluor 594 (100 μg/mL) for 8 h and observed live under confocal microscopy. For each group, ~ 300 vesicles were calculated.
Gal8 recruitment assay
Cells were plated on 35-mm confocal dishes and cultured till ~80%confluence. Next, 500 ng of Gal8-GFP pDNA (addgene, 127191) and 1.5 μL of Lipofectamine3000 (Invitrogen, L3000008) was added to 50 μL of OptiMEM; after 10 min, the mixture was diluted by adding 100 μL of full culture medium. After incubating cells with the transfection medium for 6 h, cells were replenished by fresh full culture medium and incubated for another 36 h. Next, cells were incubated with 0.2 nM Au@PDA T21 NWs or Au@PDA T21 NPs in DMEM
(supplemented with 1%FBS) for another 24 h. The cells were fixed with cold methanol for 10 min and stained by DAPI (4', 6-diamidino-2-phenylindole; Thermo Scientific, D1306) for confocal microscopy. The excitation wavelengths for DAPI and Gal8-GFP are 405 and 488 nm, respectively, and the emission wavelengths are 410–480 nm and 495–600 nm, respectively.
Pharmacological Blocking
Cells were plated on 24-well plates and cultured till ~80%confluence. Cells were pretreated with 0.3 mL of full DMEM that contains chemical blockers for 1 h prior to incubation with NWs. These blockers include filipin III (2.5 μg/mL; MCE, HY-N6718) , dynasore (120 μM; Cayman Chemical, #HY-N6718) , methyl-β-cyclodextrin (mβCD) (10 mM;Tokyo Chemical Industry, M1356) , sodium azide (0.1%; Sigma, S2002) , and amiloride (0.5 mg/mL; Sigma, BP008) . Next, the blocker-containing medium was removed, and 0.4 mL of fresh full DMEM that contains the same blocker at the original concentration and 0.2 nM T21@NWs was added to the cells. After 2 h of incubation, the cells were rinsed with PBS twice and trypsinized (0.25%Trypsin-EDTA, Gibco) for cell counting by a hemocytometer. The cells were further centrifuged at 1500 rpm for 5 min to form a pellet. Cell pellets were digested in 0.25 mL of aqua regia (3: 1 v/v ratio of 38%HCl and 68%HNO3) overnight and diluted to 5 mL by the matrix solution (2%HCl, 2%HNO3) for ICP-MS measurements (Agilent 7900) of the Au content associated with the cells. Data are shown as the mean ± SD from four biological replicates (i.e., 3 wells per treatment group) .
Live cell imaging for intracellular trafficking
After two PBS rinses, NW-treated cells were stained with 100 nM LysoTracker Green DND-26 (Thermo Scientific, L7526) , formulated in 1 mL of phenol red-free full DMEM, for 30 min. Next, the cells were rinsed, stained with Hoechst 33342 (Thermo Scientific; #62249) for 10 min, rinsed again, and replenished with 1 mL of phenol red-free full DMEM for confocal microscopy. The excitation wavelengths for Hoechst 33342, LysoTracker Green DND-26, and Cy5-labeled Au@PDA@T21 NWs are 405, 488, and 650 nm, respectively, and their corresponding emission wavelengths are 410–480 nm, 495–580 nm, and 655–750 nm, respectively.
Bio-TEM imaging
Freshly harvested cell pellets or tissues were fixed with glutaraldehyde (2.5%in PB; EMS, 16000) for 2 h and stained by osmium tetroxide (1%in PB; EMS, 19100) for another 2 h. Samples were dehydrated in increasing ethanol gradients (30%, 50%, 70%, 80%90%, 100%) and propylene oxide, embedded in Epon 812 resins (EMS) , and polymerized at 55 ℃for 48 h. Ultrathin sections of ~70 nm in thickness were deposited onto 200-mesh copper grids (Electron Microscopy Sciences, G200-Cu) and stained with 4%uranyl acetate in 50%methanol/water (EMS, 541-09-3) and Reynold’s lead citrate for observation under TEM at a beam voltage of 100 kV (Hitachi H7700) .
siRNA Transfection
Cells were plated on 24-well plates and cultured till ~80%confluence. The transfection solution, formulated in 50 μL of OptiMEM, contains 1.2 μL of Lipo and 40 pmoles of small-interfering RNA (siRNA) that specifically targets either human or mouse ClC3 (Dharmacon SMARTpool; Santa Cruz, sc-42381) , human ClC5 (Santa Cruz, sc-42385) , or nontargeting siRNA control (siCtrl) (Idobio) . After incubation for 10 min and dilution by adding 350 μL of full DMEM, the transfection mixture was added to each well of 24-well plate. The cells were incubated for 48 h before harvesting the RNA for q-PCR analysis or confocal imaging.
Immunoblotting
Cells were lysed with 0.2 mL of ice-cold RIPA buffer (Themo Scientific, 89900) with Pierce Protease Inhibitors (Thermo Scientific, A32963) . After gentle swirling on ice for 5 min, the lysates were centrifuged at 14000×g at 4 ℃ to pellet the cell debris for removal, and the supernatant was subject to the bicinchoninic acid (BCA) assay (Thermo Scientific, #23225) for quantifying the protein concentration before loading to a 10%gel [with sodium dodecyl sulfate (SDS) ] for reducing polyacrylamide gel electrophoresis (PAGE) . After transferring the gel proteins to polyvinylidene difluoride (PVDF) membranes by a Power Blotter Station (Invitrogen) , the membranes were blocked by 3%bovine serum albumin (BSA; Rocland immunochemicals, BSA-50) in TBST (Tris-buffered saline with Tween-20) at RT for 1 h, incubated with primary antibodies against ClC3 (1: 1000 in 3%BSA/TBST; Thermo Scientific, MA5-27709) and against β-tubulin (1: 1000 in 3%BSA/TBST; Abcam, ab108342) at 4℃ overnight, and incubated with horseradish peroxidase (HRP) -conjugated secondary antibodies (1: 1000 anti-rabbit, Bio-Rad 1706515; 1: 3000, anti-mouse, Invitrogen 31430) in 5%milk (Bio-Rad) /TBST for 1 h at RT. After 5 washes with TBST, Clarity Max
ECL Western Blotting Substrate (Bio-Rad, 1705062) was used to detect the proteins on the blots using the ChemiDocTM Touch imaging system.
BMDM polarization
For M1 depolarization, Day 7 BMDMs were incubated with 1 mL of 0.2 nM Au@PDA@miR-223 NWs in 1 mL full DMEM for 12 h. After two PBS rinses, the cells were stimulated by 0.4 mL of full culture medium containing 100 ng/mL LPS (lipopolysaccharides; Sigma, L7895) . After 24 h, the LPS-containing medium was removed, and the cells were cultured in fresh full culture medium or lysed by RNAiso Plus (Takara, #9108) for RNA isolation. For M2 repolarization, Day 7 BMDMs were stimulated by 1 mL of full DMEM containing 100 ng/mL LPS and 25 ng/mL IFN-γ (Sinobiological, 11725-H08H) . After 24 h, the LPS/IFN-γ-containing medium was removed and rinsed by PBS twice, and the cells were repolarized by incubating with 1 mL of 0.2 nM Au@PDA@miR-223 NWs in full DMEM for 48 h.
hMSC osteogenic differentiation
hMSCs were seeded in a 24-well plate until ~90 %confluence. Next, hMSCs were treated with 0.4mL of osteogenic medium (OM) (0.05 mM ascorbic acid, 10 mM glycerophosphate, and 0.1 μM dexamethasone in full α-MEM) containing 0.2 nM Au@PDA@siNog NWs for 7 of 14 d, with medium changed every 3 days. After fixation in 95%ethanol at RT for 10 min and 3 PBS rinses, the cells were 400 μL of Alizarin Red S staining solution (Abcam, ab146374) at 37℃ for 45 min, washed 3 times by Nanopure water, and observed under a light microscope (10× objective) to confirm stem cell differentiation. To quantify the calcium deposit, the cells were treated with 200 μL of 10%acetic acid at RT for 30 min with shaking to detach the cell monolayer, transferred to a microcentrifuge tube, heated at 85℃ for 10 min, and cooled on ice for 5 min. Upon centrifugation at 10,000 rpm for 15 min and removal of the supernatant, 75 μL of 10%ammonium hydroxide was added to dissolve the pellet. Finally, the absorbance of the pellet sample at 405 nm, as measured by the microplate reader, indicates the level of Alizarin Red (or calcium deposit) .
TUNEL assay staining
Apoptotic and necrotic cells were visualized and quantified using the Click-iTTM Plus TUNEL Assay for In Situ Apoptosis Detection (Invitrogen, C10617; Alexa Fluor 488) . Paraffin liver sections of 5 μm thick were stained per the manufacturer's instructions, co-
stained with DAPI for 10 min at RT, and mounted with Antifade Mountant for confocal laser scanning microscopy. The excitation wavelengths of DAPI and Alexa Fluor 488 are 405 nm and 488 nm, respectively. The emission wavelength ranges of DAPI and Alexa Fluor 488 are 415–480 nm and 495–650 nm, respectively.
Whole-transcriptome analysis with total RNA sequencing (RNA-Seq)
Cells of 80–90%confluence were seeded in 6-well plates with complete DMEM overnight, followed by splitting the cells into 4 groups (3 wells per group) to study the effects of NP shape and incubation time. Upon incubation with Au@PDA@T21 NWs, cells were sent to the Beijing Genomics Institute (BGI) Group for cDNA preparation and whole-transcriptome sequencing. The process followed a standardized procedure monitored by BGI’s Quality Control System. mRNA was isolated from total RNA using oligo (dT) magnetic beads following the manufacturer’s instructions for cDNA library construction. Double-stranded cDNA was sequenced using the DNBseq platform. At least 20 million clean reads per sample on the DNBseq platform were generated for data analysis. Differential expressed gene (DEG) detection, gene ontology (GO) analysis of DEG, and other analyses based on gene expression were performed by BGI. GO terms with corrected p values (Q values) of 0.05 or less were considered significantly enriched among the DEGs.
RNA isolation and qRT-PCR analysis
RNA was isolated using RNAiso Plus and reverse-transcribed using the RevertAid First Strand cDNA Synthesis Kit (Thermo Scientific, K1622) to generate cDNA. qRT-PCR was performed on the StepOnePlus Real-Time PCR System (Applied Biosystems) using TB SYBR Green Premix Ex Taq kit (Takara, RR82WR) following the manufacturer’s instructions. Transcript levels were analyzed using the ΔΔCT method and normalized to the house-keeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH) . Gene expression was quantified using pre-designed primers purchased from Shanghai Rui Mian Bio-Tech (see the sequences in Table 3) .
Animals
All procedures followed the guidelines stipulated by the Animal Experimentation Ethics Committee at The Chinese University of Hong Kong (CUHK) . Male Balb/c and C57 mice between 9 and 11 weeks of age were used and randomly divided into various treatment groups. All mice were housed in a temperature-and humidity-controlled environment with a
12-h light/dark cycle. For all biodistribution and efficacy studies, the sample size (n) indicates biological replicates.
Mouse model of unilateral ureteral obstruction (UUO)
Male Balb/c mice of 9–11 wk old were anesthetized by an intraperitoneal (i. p. ) injection of 0.2 mL of saline containing ketamine (100 mg/kg; Alfasan International B. V. ) and xylazine (10 mg/kg; Alfasan International) . The peritoneum was cut along the midline, and the left ureter was isolated and ligated twice using a 5-0 suture (NingBo Cheng-He Microsurgical Instruments) . The bowel was laid back and the peritoneum was closed with suture. Mice were placed under a heating lamp to maintain body temperature until recovery from anesthesia. For analgesia, three i. p. injections of 0.1 mL of saline containing buprenorphine (Temgesic, 0.05 mg/kg) were given to the mice subcutaneously every 12 h post-surgery. To verify the development of kidney tubulointerstitial fibrosis, mice were sacrificed 7 dpost-surgery, and the kidneys were weighted and processed for histological analysis.
Mouse model of acetaminophen (APAP) -induced acute liver injury (ALI)
Male C57BL/6 mice of 10–12 wk old were fast for 12 h prior to APAP (Solarbio, IA0030) injection for inducing ALI. After receiving a single i. p. injection of 0.3 mL of 500 mg/kg APAP (body weight) in saline, mice were allowed free access to food. To verify the establishment of ALI, mice were sacrificed 24 h post-APAP injection, serum was collected to measure the alanine transaminase (ALT) level (an indicator of liver function) , and the liver was harvested for histological analysis.
Ex vivo fluorescence imaging of biodistribution of hMSCs in UUO mice
Three days post ligation surgery, UUO mice received a single i.v injection with either free DiR dye (10 mg/kg body weight; Invitrogen, D12731) or DiR-labeled 5×105 cells/kg body weight hMSCs (hMSCs, LipomCXCR4/BMP7-treated hMSCs and LNWmCXCR4/BMP7-treated hMSCs) . To label the hMSCs with DiR, the 10 μM DiR staining solution formulated in full α-MEM was added to the cells and incubated for 30 min at 37℃. The staining solution was washed off, and the hMSCs were trypsinized for cell counting by a hemocytometer. The hMSCs were further centrifuged at 1500 rpm for 5 min and resuspended in 200 μL PBS for injection. The injected mice were sacrificed 24 hour post injection and perfused with PBS before imaging. Ex vivo fluorescence imaging of the
excised internal organs (liver, lung, heart, spleen, and kidneys) was performed using an Odyssey infrared imaging system (excitation wavelength: 700 nm, emission wavelength: ≥700nm) .
Blood pharmacokinetics
Mice received an intravenous (i.v. ) injection of 0.1 mL of LNWs containing 10 μg mHGF via the tail vein. At different time points, 0.5 mL of blood was withdrawn by terminal intracardiac puncture (using a 25-Gauge needle) and then stored inside EDTA-coated tubes (Becton Dickinson) . After centrifugation at 1500 ×g for 10 min, 0.2 mL of the blood plasma in the supernatant was collected, lysed by adding 0.5 mL of aqua regia, and further diluted to 5 mL by the matrix solution (2%HCl, 2%HNO3) for ICP-MS measurements of the Au content. Data were fitted to a mono-exponential decay model by using the Prism Graphpad software.
In vivo toxicity of Au@PDA@mHGF NWs
ALI mice received a single i.v injection of 0.15 mL of 0.5 mg Au@PDA@mHGF NWs containing 10 μg mHGF in PBS. 24 h later, 0.5 mL of whole blood was collected from the heart via an intracardiac puncture under anesthesia, left to sit at RT for 30 min, and centrifuged at 2,000 ×g at 4℃ for 10 min. About 200 μL of blood serum were collected from the supernatant and sent to the PathLab Medical Laboratories (Hong Kong) on the same day for blood biochemistry analysis. The heart, liver, spleen, kidneys, and lung were trimmed and fixed in 10%neutral buffered formalin for 48 h, embedded in paraffin, and cut into sections of 5 μm thick. Paraffin sections were deparaffinized in xylene (5 min × 3 times) , and rehydrated through a series of ethanol (100%, 95%, 70%) as well as deionized water and counterstained by hematoxylin and eosin (H&E; Sigma) for 2 min and 1 min, respectively. The stained sections were dehydrated in ethanol, cleared in xylene, and mounted with DPX mountant (Sigma) for visualization under a Ti-E motorized inverted fluorescence microscope (Nikon) in bright-field mode.
Protein purification for proteomics analysis
Desorbed proteins were mixed with 5× loading buffer (Beyotime) and boiled for 5 min. After loading the prepared samples and protein marker into the wells of a 12%separating gel, the bands were resolved by PAGE at 80 kV for 15 min and then at 120 kV for another 15 min, the gel was stained with Coomassie blue dye (Beyotime) . Three gel slices
from each gel were cut into 1 mm3 cubes, transferred to a 1.5 mL microcentrifuge tube, and washed with 500 μL of 50 mM ammonium bicarbonate/acetonitrile (ACN) (1: 1, v/v) solution until Coomassie blue disappeared. After discarding the supernatant, 500 μL of ACN was added and the mixture was incubated for 10 min until the gel pieces became opaque and stuck together. After removing the ACN, the gel slices were rehydrated in 10 mM dithiothreitol (DTT) /50 mM ammonium bicarbonate, incubated at 56℃ for 1 h, and the supernatant was discarded. For the rinsing step, 500 μL of ACN was added, the mixture was incubated for another 10 min, and ACN was removed. Next, the gel slices were incubated in 50 mM iodoacetamide (IAA) /50 mM ammonium bicarbonate at RT in the dark for 1 h, and the IAA/ammonium bicarbonate was discarded. After repeating the rinsing step, the gel pieces were incubated in proteomics grade trypsin (Promega) digestion solution on ice for 45 min. Another 5–20 μL of enzyme digestion solution was added, followed by overnight digestion at 37 ℃. After adding 100 μL of extraction solution (5%trifluoroacetic acid (TFA) -50%ACN-45%ddH2O) to each sample, the mixture was incubated in a 37℃ water bath for 1 h and sonicated for another 5 min. The extract was transferred to a fresh microcentrifuge tube and extraction was repeated.
Proteomics analysis with LC-MS/MS
All reagents were chromatography grade and bought from Thermo Fisher Scientific. Extracted peptides were lyophilized and resuspended in 10 μL of 0.1%formic acid for separation using an Orbitrap Eclipse Mass Spectrometer (Thermo Fisher Scientific) , equipped with a 150 μm×15 cm in-house made column packed with Acclaim PepMap RPLC C18 (1.9 μm,Dr. Maisch GmbH) . The organic gradient was driven by the Nanoflow UPLC system over 120 min using Buffer A (0.1%formic acid in water) and Buffer B (20%0.1%formic acid in water -80%acetonitrile) at a flow rate of 600 nL/min. The gradient was held from 4%to 8 %B for 3 min, from 8 %to 28%B for 86 min, from 28 %to 40%B for 20 min, from 40%to 95%B for 1 min and from 95%to 95%B for 10 min. Eluted peptides were directly sprayed into the mass spectrometer. Ten MS/MS data-dependent scans were acquired simultaneously with one high-resolution (60000 at 400 m/z) full-scan mass spectrum to provide the amino acid sequence and mass-to-charge ratio for the selected peptide ions.
Data processing and statistical analysis
The Prism (GraphPad) and Excel softwares were used for data analysis and graph
construction. To determine the statistical significance in the comparison of two groups, an unpaired two-tail t test was performed. To determine the statistical significance in the comparison of multiple groups, an unpaired one-way analysis of variance (ANOVA) was performed with Tukey's Test for post hoc analysis. Normality of sampling distribution of means was validated by Shapiro-Wilk test. Homogeneity of variance was validated by Bartlett’s test. Results are considered significant at P < 0.05.
All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety, including all figures and tables, to the extent they are not inconsistent with the explicit teachings of this specification.
Following are examples that illustrate procedures for practicing the invention. These examples should not be construed as limiting. All percentages are by weight and all solvent mixture proportions are by volume unless otherwise noted.
EXAMPLE 1-PREPARATION OF OLIGONUCLEOTIDE-ENCASED NANOWORMS
We followed our previous work to prepare Au40@PDA NWs by sonicating citrate-capped 40-nm gold NPs with monomeric dopamine at alkaline pH for 1 h (Figure 1A) . [20] Au40@PDA NWs bear 4–5 gold cores, a PDA shell of ~20 nm thick, a length of ~224 nm, and a zeta potential of –25.7 mV (Figure 1B and Figure 8C) . Then, we constructed oligonucleotide-encased NW (Au40@PDA@T21 NW) by flipping its surface charge to positive at pH 3 and electrostatically adsorbing polythymidine with 21 Ts (T21) , a model noncoding DNA. Each Au40@PDA@T21 NW had ~1024 strands, was stable in 10%fetal bovine serum (FBS) at 37 ℃ for 24 h (Tables 4 and 5) , and were not toxic on A549 lung epithelial cells, bEnd. 3 brain endothelial cells, primary bone marrow-derived macrophages (BMDMs) , or human mesenchymal stromal cells (hMSCs) 24 h post incubation (Figures 10A-10D) . Treatment with amiloride reduced the association of T21-encased NWs to all four cell types most pronouncedly of all pharmacological blockers tested, indicating macropinocytosis as a key uptake pathway (similar to PDA-coated NPs) . [21] Treatment with fucoidan also attenuated cellular association, suggesting a role of scavenger receptor in uptake (similar to oligonucleotide-conjugated NPs [8] ) (Figures 11A-12D) .
EXAMPLE 2-ENDOSOMAL ESCAPE OF OLIGONUCLEOTIDE-ENCASED NWS
We treated A549 cells with Cyanine (Cy) 5-labeled Au40@PDA@T21 NWs and monitored their cellular trafficking by confocal imaging. Confirmed by Lysotracker staining
under confocal imaging, we found the colocalization of Cy5-T21@NW and Lysotracker signals are consistently low across all tested cell lines, with low Pearson’s correlation coefficient (PCC) value at 0.1~ 0.2 (Figure 1C) . In addition to the fluoresce imaging, we also applied bio-TEM imaging to locate T21@NW upon cellular entry. Consistent with imaging data, bio-TEM images showed most T21@NW were exposed in the cytosol after 24 h incubation, implying that the ability of endosomal escape was generalizable across different cell types (Figure 1D) . We then specifically labeled the intracellular vesicles with anti-Rab 9 (late endosomes) and anti-LAPM1 (lysosomes) antibodies and traced the distribution by time-lapse confocal imaging. Abundant Cy5-T21@NWs entered the cells 4 h post incubation and significantly accumulated in late endosomes at 8 h, with PCC value of 0.781. We found that the PCC of Rab 9 and Cy5-T21@NWs dropped to 0.407 at 12 h, indicating that more than half of Cy5-T21@NWs escaped from the late endosomes. Most T21@NWs escaped from late endosomes at 24 h, with PPC value of 0.305. In contrast, the colocalization with lysosomes maintained at ~0.3 across all the time points we tested, respectively (Figure 1E) . So, the NWs escaped from late endosomes before reaching lysosomes.
To clarify the effect of nanostructure shape on endosomal escape, we prepared a control NP “Au40@PDA@T21 NP” by adsorbing the same density of T21 strands (~380 per NP) to an unassembled Au@PDA NP (Figure 8B) with the same gold core size and PDA shell thickness as Au40@PDA@T21 NW. Au40@PDA-T21 NPs were predominantly inside acidic organelles 24 h post-incubation (PCC ~0.755; Figure 13) , matching our reported data on PDA NPs [21] and DNA-coated NPs. [8] We prepared elongated Au@PDA nanostructures of varying sizes and shapes, keeping the NP concentrations added to cells similar. We made Au20@PDA@T21 and Au60@PDA@T21 NWs using gold cores of 20 and 60 nm (Figure 9A-9D and Table 4) ; NWs composed of larger cores contained less cores (~6 for Au20@PDA@T21 NW and ~2 Au60@PDA@T21 NW) . Both NWs were strongly localized to acidic organelles (PCC >0.7) instead (Figures 15A and 15B) . Yet, when we prepared unassembled T21-encased Au@PDA nanorods (core size: 45 × 200 nm) with a similar size, PDA thickness, and aspect ratio to Au40@PDA@T21 NW (Figures 9E-9F) , the nanorods barely colocalized with acidic organelles (PCC =0.137) but have reduced cellular uptake compared to the Au40@PDA@T21 NW (Figures 16A and 16B) . These data suggest that an aspect ratio of ~3.5 and a length of ~240 nm are suitable parameters for endosomal escape.
EXAMPLE 3-ROLE OF CLC3 CHLORIDE ION EXCHANGER IN THE ENDOSOMAL ESCAPE OF NWS
Proton sponge effect is a mainstream mechanism for endosomal escape. Here, pretreatment of A549 cells by bafilomycin A1 or chloroquine (both inhibitors of endosomal acidification) did not impair the endosomal escape of Au40@PDA@T21 NWs (Figure 2A) . Also, the NWs did not exhibit pH buffering capacity between 4.5 (lysosome) and 7.5 (extracellular space) (Figure 2B) , with zeta potentials more negative than –25 mV (Figure 17) . Instead of proton sponge effect, we searched for other plausible mechanism for endosomal escape. Pretreatment with niflumic acid (ageneral pharmacological blocker of Cl-channels) disrupted the endosomal escape of NWs, but pretreatment with lidocaine (ablocker of Na+ channels) , amiodarone (ablocker of K+ channels) , or nifedipine (ablocker of Ca2+channels) did not (Figures 18 and 19) . Therefore, we hypothesized a role of Cl-in endosomal escape, but which specific Cl-channel was involved is questionable.
To probe the mechanism for endosomal escape, we used unbiased RNA-sequencing (RNA-seq) to analyze the changes in gene expression upon incubating NW with of A549 cells, screening for the differentially expressed mRNA transcripts (DETs) with a statistical significance of Q < 0.05. Initially, we addressed the shape effect by comparing gene changes upon treatment with Au40@PDA@T21 NW versus Au40@PDA@T21 NP for 24 h. This comparison saw an enrichment in gene ontology (GO) terms related to late endosome, cytoplasm, membrane, and cytosol (Figure 2C and Table 6) . Then, we addressed the time-dependent effect by comparing gene changes upon incubation of Au40@PDA@T21 NW for 24 h (when NWs were in the cytosol) versus 8 h (when NWs were in late endosomes) . This comparison saw an enrichment in GO terms related to transport vesicles, and similarly late endosomes and endosome membranes (Figure 2D and Table 7) . Still, it was unclear which DETs were crucial.
From our RNA-seq data, we screened the DETs related to Cl-with a log2 fold change >2 (Q < 0.05) and detected drastic upregulation of ClC3 (aH+/2Cl-exchanger) in both comparisons (Figures 2E and 2F) , with log2 fold changes of 20.12 for “Au40@PDA@T21 NW 24 h vs. Au40@PDA@T21 NP 24 h” (Q = 5.69 × 10-5) and 9.93 for “Au40@PDA@T21 NW 24 h vs. Au40@PDA@T21 NW 8 h” (Q = 0.029) . In mammals, there are nine ClC isoforms. Among them, ClC1, ClC2, ClC-Ka, and ClC-Kb are plasma membrane Cl-channels, whereas ClC3 through ClC7 are H+/2Cl-exchanger in the endosomes or lysosomes for the regulation of luminal acidification and Cl-accumulation20, 21. In DET analysis, we found that ClC5, TTYH1 (Tweety Family Member 1, a Cl-channel) and TMEM206 (aproton-activated Cl-channel) are also upregulated. Western blot analysis revealed pronouncedly higher expression of ClC3 in A549 cells in the “NW 24 h” group than “NP 24
h” or “NW 8 h” group (Figures 2G, 20A, and 20B) . Confocal immunofluorescence confirmed the expression of ClC3 largely in lysosomes (PCC ~0.662) and moderately in late endosomes (PCC =0.530) (Figures 2H and 21) . Thus, we hypothesized that ClC3 mediates the endosomal escape of NWs.
As supporting evidence, genetic knockdown by RNA interference, using two different sequences of siRNA against ClC3 (Figure 22) , increased the accumulation of NWs in acidic organelles of A549 cells 24h post-incubation (PCC >0.7; Figure 2I) , but treatment with a nonspecific control siRNA sequence did not. Activation of ClC3 using bufalin [22] [as verified by quantitative reverse transcription polymerase chain reaction (qRT-PCR) ; Figures 23A and 23B yielded limited overlap of NWs with acidic organelles 8 h post-incubation (PCC =0.249; Figure 2K) , while NWs were entrapped in late endosomes without bufalin treatment (Figure 1E) . These data underscore a pivotal role of CCl3 in endosomal escape. Meanwhile, siRNA knockdown of ClC5 led to moderate accumulation in acidic organelles (PCC =0.442; Figures 2L and 24) , suggesting a minor role of ClC5 in endosomal escape.
EXAMPLE 4-IONIC ACCUMULATION AND ENDOSOMAL MEMBRANE RUPTURE
As ClC3 is an ion exchanger, we tracked the changes in the concentrations of H+ and Cl-in acidic organelles due to NW treatment. We used a Cl-indicator fluorescent dye called MQAE (N- (ethoxycarbonylmethyl) -6-methoxyquinolinium bromide) to qualitatively assess the amount of Cl-in wildtype A549 cells; MQAE fluorescence drops with increasing Cl-concentrations. Because MQAE is not a dye of Cl-specific for late endosomes or lysosomes, we co-incubated the cells with Cy5-labeled NWs and tetramethylrhodamine (TRITC) -labeled dextran, a tracer NP that enter cells via macropinocytosis, the same pathway adopted by the NW; [21] intriguingly, there was limited colocalization of MQAE and Cy5 fluorescence 8 h post-incubation, indicating the elevated concentration of Cl-inside those vesicles that contained NWs than those did not. On ClC3-silenced A549 cells, however, there was stronger overlap between MQAE and Cy5 fluorescence, indicating a reduced Cl-concentration in the NW-containing organelles when compared to wildtype cells (Figures 3A and 3C) . Next, we probed the pH of NW-containing vesicles using a ratiometric method, [24] based on the ratio of fluorescence of the pH-insensitive dye Alexa Fluor 594-conjugated dextran (red) and the pH-sensitive dye Rodo-Green-conjugated dextran (green) (Figure 25) ; the fluorescence of Rodo-Green drops with increasing pH. When both dyes colocalized with Cy5-labeled NWs, the red-to-green (R/G) ratio fluorescence should indicate the pH of
vesicles containing NWs. After incubating wildtype A549 cells with both pH dyes without NWs for 8 h, the mean pH of the organelles was 5.44, characteristic of late endosomes. By contrast, treatment of wildtype cells with NWs lowered the mean pH to 5.20, implying the acidification of organelles. On ClC3-silenced cells, the organellar pH before and after NW treatment were 5.70 and 5.55, respectively (Figures 3B and 3D) , both less acidic than those on wildtype cells. To probe into the function of V-ATPase, we evaluated the activity of V-ATPase by analyzing the interaction of the major subunits of V-ATPase, V0 and V1. Confocal images suggested that A549 cells treated with NW for 8 h significantly enhanced colocalization of two major subunits of V-ATPase compared to cells without NW treatment (Figure 3E) , indicating more intense activity of V-ATPase after NW’s treatment. These data revealed the enhanced concentrations of H+ and Cl-in organelles due to NW treatment, a cellular event regulated by ClC3. Therefore, we postulated that the internalized NWs trafficked to the late endosomes, where they upregulated ClC3, caused the accumulation of H+ and Cl-, and ultimately membrane rupture and endosomal escape of NWs (Figure 3F) .
To verify this claim, we prepared A549 cells that express galectin-8 (Gal8) fused with the green fluorescent protein (GFP) . Gal8-GFP is a cytosolic protein and will recruit to the inner leaflet of ruptured endosomal membrane, yielding strong GFP fluorescence. [25, 26] Treatment of Gal8-GFP-expressing cells with Au@PDA@T21 NWs yielded punctate spots (~15 dots per cell) , proof of endosomal rupture (Figures. 3G and 26) . There were no obvious Gal8 spots in untreated or Au@PDA@T21 NP-treated cells. TEM imaging also captured the impaired endosomal membrane and escape of Au@PDA@T21 NWs to the cytosol, with rupture sites on membrane segments near NWs but not the entire membrane (Figures 3H and 27) .
EXAMPLE 5-IN VITRO DELIVERY OF DIVERSE OLIGONUCLEOTIDE TYPES TO MULTIPLE CELL TYPES
We utilized Au40@PDA NWs to deliver antisense DNA, miRNA, or siRNA to different cell types. Derived from the same method for attaching T21 to Au40@PDA NW, these oligonucleotide-loaded NWs shared a similar size, surface charge, and oligonucleotide loading to Au40@PDA@T21 NW, were stable in FBS (Tables 4 and 5) , entered cells without transfection agents, were not cytotoxic (Figures 10A-10D) , and escaped endosomes 24 h post-incubation (Figure 28A; Figures 4A, 4B, 4E, and 4F; Figures 29, 30, and 33) . In each application, we added the same amounts of oligonucleotide to cells for all treatment groups.
Our first application was antisense gene regulation, where we delivered antisense DNA against enhanced GFP (asEGFP) to EGFP-expressing bEnd. 3 cells. Besides asEGFP-loaded NWs, cells were also treated with free asEGFP, Lipofectamine 3000 (a commercial transfection agent) , and a mixture of Lipofectamine 3000 and asEGFP (Lipo+asEGFP) . Confocal images revealed that asEGFP-encased NW reduced EGFP fluorescence and flow cytometry data showed 60%less EGFP-positive cells than other groups 72 h post-incubation (Figures 28B and 28C) . Lipofectamine-aided delivery showed ~34%lower EGFP-positive cells (Figure 28D) .
Our second application was macrophage polarization, where we delivered miR-223 (aM2 macrophage stimulator [27] ) to primary BMDMs that were pre-induced to M1 phenotype. The M1 BMDMs were treated with miR-223-encased NW, free miR-223, pristine Au@PDA NW, or Lipo+miR-223 for 48 h for M2 repolarization. miR-223-encased NWs activated M2-related genes (IL-10, CD206, TGF-β, and arginase-I) and inhibited M1-related genes (IL-12, CD80, TNF-α, and IFN-γ) most effectively (Figure 4C and 31) . miR-223-encased NWs yielded an elongated cell shape (M2 phenotype) , but other treatments caused flattened, round cell shapes (M1 phenotype) . miR-223-encased NWs yielded intense signals of C206 but weak signals of CD80, yet pristine Au@PDA NW or free miR-223 yielded strong CD206 signals but weak CD80 signals. Lipofectamine-aided delivery yielded less M2-like and CD206-positive cells (Figure 4D) .
Our third application was stem cell differentiation, where we delivered siRNA against Noggin (anegative regulator of osteogenic differentiation [17] ) to hMSCs. The treatments, including siNog-encased NWs, free siNog, pristine Au@PDA NWs, and Lipo+siNog, lasted for 14 d. siNog-encased NW inhibited Noggin and activated osteogenesis-related genes [bone morphogenetic protein (BMP) -2 and Runt-related transcription factor 2] most effectively and yielded the highest amounts of calcium deposit (Figure 4G, 34A, 34B, and 34C) . Lipofectamine-aided delivery resulted in modest inhibition of Noggin and increase in calcium deposits.
EXAMPLE 6-PREPARATION OF MRNA-ENCASED NWS
We further leveraged Au@PDA NWs to deliver mRNA, a larger gene cargo. Unlike oligonucleotides, direct adsorption of mRNA encoding EGFP (mEGFP) to Au@PDA NW did not yield sufficient mRNA loading or protein expression, so we coated Au@PDA NW with cationic lipids [1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) , 1, 2-distearoyl-sn-glycero-3-phosphocholine (DOPE) ] and cholesterol for adsorbing mRNA (Figure 5A) .
Au@PDA@lipid NW was larger than Au@PDA NW (278.5 nm) and positively charged (+30.5 mV) , but upon adsorption with mEGFP at w/w ratio of 1: 50 (Figures 35 and 36) , the resultant Au@PDA@mEGFP NW were larger (280.3 nm) and negatively charged (-35.2 mV) , with ~68 mEGFP strands (Table 8 and 9) . Confocal imaging verified NW escape from late endosomes 10 h post-incubation (Figure 37) . Similar to the Au@PDA@T21 NWs, Au@PDA@mEGFP NW also enhanced ClC3 expression, V-ATPase activity, and vesicular concentrations of Cl-and H+ (Figures 38A-39C) . Au@PDA@mEGFP NW transfected hMSCs more effectively than Lipofectamine, as evidenced by a higher fraction of EGFP-positive cells and cell viability 48 h post-incubation (Figures 40A-40C) .
EXAMPLE 7-DELIVERY OF MRNA TO HMSCS AS EX VIVO CELL-BASED THERAPY OF KIDNEY FIBROSIS
We utilized NW to deliver mRNA to hMSCs for cellular programming and then injected the genetically engineered hMSCs into mice with unilateral urethral obstruction (UUO) , an established model of kidney fibrosis, as ex vivo cell-based therapy. To the lipid-coated NW, we adsorbed equal portions of mRNA encoding BMP-7 (an anti-fibrosis protein [28] ) -fused with orange fluorescence protein (OFP) and mRNA encoding C-X-C chemokine receptor type 4 (CXCR4; for homing to fibrotic sites [29] ) -fused with GFP. Au@PDA@mCXCR4/mBMP7 NW had similar physicochemical parameters and serum stability as mEGFP-encased LNW (288.1 nm, -33.7 mV) , entered hMSCs, did not induce toxicity, were barely colocalized with acidic organelles (PCC ~0.091) (Figure 5B) , and supported the expression of both fluorescently tagged therapeutic proteins in 85%of hMSCs tested (Figure 5C) , more effective than Lipofectamine at the same amount of mRNA added (Figures 41A-41C) .
We intravenously (i.v. ) injected a single dose of saline, hMSC, hMSCs transfected with mCXCR4/mBMP7-encased NW, or hMSCs transfected with both mRNAs by Lipofectamine into UUO mice. 3 days post-UUO surgery is required for developing fibrosis in the right UUO kidney; the left contralateral kidney remains healthy (Figure 42) . As in vitro validation, Au@PDA@mCXCR4/mBMP7 NW more effectively transfected hMSCs with both mRNA than Lipofectamine (Figure 5E) . By ex vivo fluorescence imaging, NW-transfected hMSCs most abundantly accumulated in the UUO kidney but least abundantly in other key organs (liver, spleen, and lungs) 24 h post-injection, proof of their superior homing ability (Figure 5F and 43) . At sacrifice (14 d post-UUO surgery or 11 d post-treatment) , NW-transfected hMSCs group yielded the sharpest inhibition of type I
collagen and α-smooth muscle actin (both markers of kidney fibrosis) by immunohistochemistry (IHC; Figure 5G-5I, 44A, and 44B) and qRT-PCR (Figures 5J and 5K) and highest expression of BMP7 by the enzyme-linked immunosorbent assay (ELISA; Figure 5L) and in the UUO kidney, evidence of enhanced efficacy of hMSCs due to NW-facilitated genetic engineering. Blood tests and histological examination of major organs did not reveal in vivo toxicity following mCXCR4/mBMP7-encased NW treatment (Figures 45A, 45B, and 46) .
EXAMPLE 8-IN VIVO DELIVERY OF MRNA TO HEPATOCYTES FOR ALLEVIATING ACUTE LIVER INJURY
We utilized NW to deliver mRNA to liver cells in vivo for reducing acetaminophen (APAP) -induced acute liver injury (ALI) in mice (Figure 6A) . To the lipid-coated NW, we adsorbed mRNA encoding hepatocyte growth factor (mHGF) to accelerate hepatocyte recovery and improve liver function30. mHGF-encased NW shared similar properties as mEGFP-encased NW (Table 8 and 9) , had a blood half-life of ~1 h, and accumulated in the liver (30%of injected dose) 24 h post-injection by ICP-MS measurements (Figures 47A and 47B) . Cy5-labeled mHGF-encased NW entered hepatocytes (Figure 6B) and did not overlap with late endosomes (PCC ~0.202) or lysosomes (PCC ~0.153) (Figure 6D) . TEM imaging of the liver captured NW entrapment in organelles 5 h post-injection and cytosolic localization (with ruptured vesicle membranes) 10 h post-injection (Figure 6C) , proof of endosomal escape.
Upon animal recovery (24 h post-APAP induction) , we i.v. injected saline, pristine Au@PDA@lipid NW, Au@PDA@mHGF NW, or Lipo+mHGF into ALI mice, followed by sacrifice 24 h post-treatment. mHGF-encased NW most effectively elevated HGF expression in the liver by ELISA (Figure 47C) , reduced necrotic area (Figures 6E and 6G) and apoptotic cells (Figures 6F and 6H) , and reduced serum alanine transaminase (ALT; indicator of liver function) by ~50% (Figure 6I) , proof of in vivo NW-aided transfection and anti-ALI efficacy. mHGF-encased NWs did not induce toxicity or accumulate in major internal organs 24 h post-injection (Figures 48 and 49) , evidence of in vivo safety.
It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of
this application. In addition, any elements or limitations of any invention or embodiment thereof disclosed herein can be combined with any and/or all other elements or limitations (individually or in any combination) or any other invention or embodiment thereof disclosed herein, and all such combinations are contemplated with the scope of the invention without limitation thereto.
EXEMPLARY EMBODIMENTS
Embodiment 1. A nanoworm composition comprising:
at least two metal nanoparticle cores;
a polymer coating over the at least two metal nanoparticle cores;
a therapeutic nucleic acid; and,
optionally, a lipid coating over the polymer coating.
Embodiment 2. The nanoworm composition of embodiment 1, wherein the at least two metal nanoparticle cores are at least two gold nanoparticle cores.
Embodiment 3. The nanoworm composition of any preceding embodiment, wherein each of the at least two metal nanoparticle cores has a diameter of about 40 nm.
Embodiment 4. The nanoworm composition of any preceding embodiment, comprising 4 or 5 metal nanoparticle cores.
Embodiment 5. The nanoworm composition of any preceding embodiment, wherein the polymer coating has a thickness of about 7 nm to about 35 nm.
Embodiment 6. The nanoworm composition of any preceding embodiment, wherein the polymer coating comprises polydopamine (PDA) , polyethylene glycol (PEG) , polyethylenimine (PEI) , or silica.
Embodiment 7. The nanoworm composition of any preceding embodiment, wherein the nanoworm composition has an anionic charge.
Embodiment 8. The nanoworm composition of any preceding embodiment, wherein the therapeutic nucleic acid is an oligonucleotide or mRNA.
Embodiment 9. The nanoworm composition of any preceding embodiment, wherein the therapeutic nucleic acid is about 15 to about 5000 nucleotides in length.
Embodiment 10. The nanoworm composition of any preceding embodiment, wherein the therapeutic nucleic acid is an antisense oligonucleotide (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , messenger RNA (mRNA) , or any combination thereof.
Embodiment 11. The nanoworm composition of embodiment 8, wherein the therapeutic oligonucleotide nucleic acid is adsorbed onto the polymer coating.
Embodiment 12. The nanoworm composition of embodiment 10, wherein the mRNA encodes hepatocyte growth factor (HGF) , chemokine receptor type 4 (CXCR4) , bone morphogenetic protein 7 (BMP-7) , enhanced green fluorescence protein (EGFP) , red fluorescence protein (RFP) , or any combination thereof; the miRNA is miR223; or the siRNA is siNog.
Embodiment 13. The nanoworm composition of any preceding embodiment, comprising
at least two metal nanoparticle cores;
a polymer coating over the at least two metal nanoparticle cores; and
a lipid coating over the polymer coating, wherein the therapeutic nucleic acid is bound to the lipid coating.
Embodiment 14. The nanoworm composition of embodiment 13, wherein the lipid coating comprises 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) , cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DOPE) , (1, 1′- ( (2- (4- (2- ( (2- (bis (2-hydroxydodecyl) amino) ethyl) (2-hydroxydodecyl) amino) ethyl) piperazin-1-yl) ethyl) azanediyl) bis (dodecan-2-ol) ) (C12-200) , (6Z, 9Z, 28Z, 31Z) -Heptatriaconta-6, 9, 28, 31-tetraen-19-yl 4- (dimethylamino) butanoate (DLin-MC3-DMA) , ( (4-hydroxybutyl) azanediyl] di (hexane-6, 1-diyl) bis (2-hexyldecanoate) (ALC-0315) , dimethyldioctadecylammonium bromide (DDA) , 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC) , 1, 2-dioleoyl-3-dimethylammonium-propane (DODAP) , 1, 2-dioleyloxy-3-dimethylaminopropane (DODMA) , Dipalmitoylphosphatidylcholine (DPPC) , or any combination thereof.
Embodiment 15. The nanoworm composition of any preceding embodiment, wherein the nanoworm comprises a citrate-capped Au NP (Cit-Au NP) having a diameter ranging from about 20 to about 60 nm.
Embodiment 16. The nanoworm composition of any preceding embodiment, wherein the nanoworm comprises an Aux@PDA NW, wherein x is about 20, about 40, or about 60 nm in diameter.
Embodiment 17. The nanoworm composition of any preceding embodiment, wherein the nanoworm comprises an Au@PDA NR (NanoRod) , wherein the size of the NR is about 45 nm x about 180 nm.
Embodiment 18. A method of nucleic acid delivery comprising administering the nanoworm composition of embodiment 1 to a subject.
Embodiment 19. The method of embodiment 18, wherein the therapeutic nucleic acid regulates gene expression in the subject.
Embodiment 20. The method of embodiment 18, wherein the therapeutic nucleic acid escapes endosomal and/or lysosomal degradation.
Embodiment 21. The method of embodiment 18, wherein the therapeutic nucleic acid enhances stromal cell differentiation.
Embodiment 22. The method of embodiment 19, wherein the regulation of gene expression in the subject treats acute liver injury, chronic kidney disease, or a combination thereof.
Embodiment 23. The method of embodiment 18, wherein the nanoworm enters epithelial cells, liver cells, kidney cells, endothelial cells, primary macrophages, mesenchymal stem cells, neural cells, or any combination thereof in the subject.
Embodiment 24. The composition of embodiment 18, wherein the nanoworm is administered by intra-an articular injection, an intrathecal injection, or a retrobulbar injection.
REFERENCES
1. J. Gilleron, W. Querbes, A. Zeigerer, A. Borodovsky, G. Marsico, U. Schubert, K. Manygoats, S. Seifert, C. Andree, M. H. Epstein-Barash, L. Zhang, V. Koteliansky, K. Fitzgerald, E. Fava, M. Bickle, Y. Kalaidzidis, A. Akinc, M. Maier, M. Zerial, Nat Biotechnol 2013, 31, 638.
2. S. Cui, et al. Toxicol Res (Camb) 7, 473–479 (2018) .
3. P.L. Felgner, T.R. Gadek, M. Holm, R. Roman, H.W. Chan, M. Wenz, J.P. Northrop, G.M. Ringold, M. Danielsen, Proc. Natl. Acad. Sci. USA 1987, 84, 7413.
4. O. Boussif, F. LezoualC’H, M.A. Zanta, M.D. Mergny, D. Scherman, B. Demeneix, J.P. Behr, Proc. Natl. Acad. Sci. USA 1995, 92, 7297.
5. J.P. Behr, Chimia (Aarau) 1997, 51, 34.
6. H. Lv, S. Zhang, B. Wang, S. Cui, J. Yan, J. Control. Release 2006, 114, 100.
7. X. Wei, B. Shao, Z. He, T. Ye, M. Luo, Y. Sang, X. Liang, W. Wang, S. Luo, S. Yang, S. Zhang, C. Gong, M. Gou, H. Deng, Y. Zhao, H. Yang, S. Deng, C. Zhao, L. Yang, Z. Qian, J. Li, X. Sun, J. Han, C. Jiang, M. Wu, Z. Zhang, Cell Research 2015 25: 2 2015, 25, 237.
8. Q. Bai, Y. Xiao, H. Hong, X. Cao, L. Zhang, R. Han, L.K.C. Lee, E.Y. Xue, X.Y.Tian, C.H.J. Choi, Proc Natl Acad Sci U S A 2022, 119, 39, e2201443119.
9. H. Yang, Z. Chen, L. Zhang, W.Y. Yung, K.C.F. Leung, H.Y.E. Chan, C.H.J. Choi, Small 2016, 12, 5178.
10. W. Sun, W. Ji, J.M. Hall, Q. Hu, C. Wang, C.L. Beisel, Z. Gu, Angew. Chem. Int. Ed. 2015, 54, 12029.
11. J. Liu, T. Wu, X. Lu, X. Wu, S. Liu, S. Zhao, X. Xu, B. Ding, J Am Chem Soc 2019, 141, 19032.
12. Y. Chen, R. Zhao, L. Li, Y. Zhao, Angew. Chem. 2022, 61, e202206485.
13. C.K.K. Choi, Y.T.E. Chiu, X. Zhuo, Y. Liu, C.Y. Pak, X. Liu, Y.L.S. Tse, J. Wang, C.H.J. Choi, ACS Nano 2019, 13, 5864.
14. S.A. Smith, L.I. Selby, A.P.R. Johnston, & G.K. Such, Bioconjug Chem 2019, 30, 263.
15. A. Shapouri-Moghaddam, et al. J Cell Physiol 2018, 233, 6425.
16. Tran, T. -H., Krishnan, S. & Amiji, M.M. MicroRNA-223 PLoS One 2016, 11, e0152024.
17. A.C.F. Bassit, P. Moffatt, M. -H. Gaumond, R.C. Hamdy, J Nanomed Nanotechnol 2015, 6, 1.
18. Romagnani, P. et al. Chronic kidney disease. Nat Rev Dis Primers 2017, 3, 17088.
19. Wagoner, Z.W. & Zhao, W. Nat Biomed Eng 2021, 5, 379.
20. F. Rizvi, et al. Nat Commun 2021, 12, 613.
21. Y. Liu, C.K.K. Choi, H. Hong, Y. Xiao, M.L. Kwok, H. Liu, X.Y. Tian, C.H.J. Choi, ACS Nano 2021, 15, 13871.
22. J. Liu, D. Zhang, Y. Li, W. Chen, Z. Ruan, L. Deng, L. Wang, H. Tian, A. Yiu, C. Fan, H. Luo, S. Liu, Y. Wang, G. Xiao, L. Chen, W. Ye, J Med Chem 2013, 56, 5734.
23. T.J. Jentsch, Crit Rev Biochem Mol Biol 2008, 43, 3.
24. P. Paramasivam, C. Franke, M. A. S. Bartesaghi, A. Sabirsh, L. Lindfors, M. Yanez Arteta, A. Dahlén, A. Bak, S. Andersson, Y. Kalaidzidis, M. Bickle, M. Zerial, J. Cell Biol. 2022, 221, 2, e202110137.
25. Y. Rui, D.R. Wilson, S.Y. Tzeng, H.M. Yamagata, D. Sudhakar, M. Conge, C.A. Berlinicke, D.J. Zack, A. Tuesca, J.J. Green, Sci Adv 2022, 8, 1, eabk2855.
26. K.V. Kilchrist, S.C. Dimobi, M.A. Jackson, B.C. Evans, T.A. Werfel, E.A. Dailing, S.K. Bedingfield, I.B. Kelly, C.L. Duvall, ACS Nano 2019, 13, 1136.
27. G. Zhuang, C. Meng, X. Guo, P.S. Cheruku, L. Shi, H. Xu, H. Li, G. Wang, A.R. Evans, S. Safe, C. Wu, B. Zhou, Circulation 2012, 125, 2892.
28. M. Zeisberg, J.I. Hanai, H. Sugimoto, T. Mammoto, D. Charytan, F. Strutz, R. Kalluri, Nat. Med. 2003 9: 7 2003, 9, 964.
29. R.J. Miller, G. Banisadr, B.J. Bhattacharyya, J Neuroimmunol 2008, 198, 31.
30. M.Z.C. Hatit, C.N. Dobrowolski, M.P. Lokugamage, D. Loughrey, H. Ni, C. Zurla, A.J. Da Silva Sanchez, A. Radmand, S.G. Huayamares, R. Zenhausern, K. Paunovska, H.E. Peck, J. Kim, M. Sato, J.I. Feldman, M.A. Rivera, A. Cristian, Y.T. Kim, P.J. Santangelo, J.E. Dahlman, Nat. Chem. 2023, 15, 508.
31. M. Hara-Chikuma, B. Yang, N.D. Sonawane, S. Sasaki, S. Uchida, A.S. Verkman, J. Biol. Chem. 2005, 280, 1241.
32. N.D. Sonawane, F.C. Szoka, A.S. Verkman, J. Biol. Chem. 2003, 278, 44826.
33. S.K. Bhangu, L. Mummolo, S. Fernandes, A. Amodio, A. Radziwon, B. Dyett, M. Savioli, N. Mantri, C. Cortez-Jugo, F. Caruso, F. Cavalieri, Adv Funct Mater 2024, 34, 2311240.
34. H. Wang, Y. Yuan, L. Qin, M. Yue, J. Xue, Z. Cui, X. Zhan, J. Gai, X. Zhang, J. Guan, S. Mao, J. Control. Release 2024, 366, 746.
35. S. Narum, B. Deal, H. Ogasawara, J.N. Mancuso, J. Zhang, K. Salaita, ACS Nano 2024, 18, 6186.
36. S. Guo, C. Li, C. Wang, X. Cao, X. Liu, X.J. Liang, Y. Huang, Y. Weng, Acta Pharm Sin B 2024, 14, 781.
37. Y. Ma, O.S. Fenton, Adv. Therap. 2023, 6, 2200305.
38. T.T. Pham, H. Chen, P.H.D. Nguyen, M.K. Jayasinghe, A.H. Le, M.T. Le, Pharmacol Res 2023, 188, 106665.
39. X. Tang, J. Zhang, D. Sui, Q. Yang, T. Wang, Z. Xu, X. Li, X. Gao, X. Yan, X. Liu, Y. Song, Y. Deng, J. Control. Release 2023, 364, 529.
40. B. Li, M. Zhao, W. Lai, X. Zhang, B. Yang, X. Chen, Q. Ni, Angew. Chem. Int. Ed. 2023, 62, e202302676.
41. Y. Zeng, M. Shen, R. Pattipeiluhu, X. Zhou, Y. Zhang, T. Bakkum, T.H. Sharp, A.L. Boyle, A. Kros, Nanoscale 2023, 15, 15206.
42. X. Shen, A. Dirisala, M. Toyoda, Y. Xiao, H. Guo, Y. Honda, T. Nomoto, H. Takemoto, Y. Miura, N. Nishiyama, J. Control. Release 2023, 360, 928.
43. A.P. Pontes, S. van der Wal, S.R. Ranamalla, K. Roelofs, I. Tomuta, L.B. Creemers, J. Rip, Front Bioeng. Biotechnol. 2023, 11, 1290871.
44. Y. Cao, J. Long, H. Sun, Y. Miao, Y. Sang, H. Lu, C. Yu, Z. Zhang, L. Wang, J. Yang, S. Wang, Adv. Sci. 2023, 10, 2302423.
45. P. Roy, N.W. Kreofsky, M.E. Brown, C. Van Bruggen, T.M. Reineke, JACS Au 2023, 3, 1876.
46. X. Feng, B. Liu, Z. Zhou, W. Li, J. Zhao, L. Li, Y. Zhao, Nano Res 2023, 16, 12633.
47. X. Chen, S. Wang, Y. Chen, H. Xin, S. Zhang, D. Wu, Y. Xue, M. Zha, H. Li, K. Li, Z. Gu, W. Wei, Y. Ping, Nat Nanotechnol 2023, 18, 933.
48. Y. Mo, M.H.Y. Cheng, A. D’Elia, K. Doran, L. Ding, J. Chen, P.R. Cullis, G. Zheng, ACS Nano 2023, 17, 4688.
49. Z. Ma, S.W. Wong, H. Forgham, L. Esser, M. Lai, M.N. Leiske, K. Kempe, G. Sharbeen, J. Youkhana, F. Mansfeld, J.F. Quinn, P.A. Phillips, T.P. Davis, M. Kavallaris, J.A. McCarroll, Biomaterials 2022, 285, 121539.
50. S. Liu, J. Liu, H. Li, K. Mao, H. Wang, X. Meng, J. Wang, C. Wu, H. Chen, X. Wang, X. Cong, Y. Hou, Y. Wang, M. Wang, Y.G. Yang, T. Sun, Biomaterials 2022, 287, 121645.
51. Y. Chen, R. Zhao, L. Li, Y. Zhao, Angew. Chem. Int. Ed. 2022, 61, e202206485.
52. Y.J. Sung, H. Guo, A. Ghasemizadeh, X. Shen, W. Chintrakulchai, M. Kobayashi, M. Toyoda, K. Ogi, J. Michinishi, T. Ohtake, M. Matsui, Y. Honda, T. Nomoto, H. Takemoto, Y. Miura, N. Nishiyama, Cancer Sci 2022, 113, 4339.
53. J. Lee, I. Sands, W. Zhang, L. Zhou, Y. Chen, Proc. Natl. Acad. Sci. USA 2021, 118, e2104511118.
54. C. Li, J. Zhou, Y. Wu, Y. Dong, L. Du, T. Yang, Y. Wang, S. Guo, M. Zhang, A. Hussain, H. Xiao, Y. Weng, Y. Huang, X. Wang, Z. Liang, H. Cao, Y. Zhao, X.J. Liang, A. Dong, Y. Huang, Nano Lett. 2021, 21, 3680.
55. Y. Chen, X. Chen, D. Wu, H. Xin, D. Chen, D. Li, H. Pan, C. Zhou, Y. Ping, Chemistry of Materials 2021, 33, 81.
56. Y. Wang, Y. Xie, K. V Kilchrist, J. Li, C.L. Duvall, D. ACS Appl Mater Interfaces 2020, 12, 4308.
57. T. Cristofolini, M. Dalmina, J.A. Sierra, A.H. Silva, A.A. Pasa, F. Pittella, T.B. Creczynski-Pasa, Materials Science and Engineering: C 2020, 109, 110555.
58. W. Zai, W. Chen, Z. Wu, X. Jin, J. Fan, X. Zhang, J. Luan, S. Tang, X. Mei, Q. Hao, H. Liu, D. Ju, ACS Appl Mater Interfaces 2019, 11, 4842.
59. J. Blersch, V. Francisco, C. Rebelo, A. Jiménez‐Balsa, H. Antunes, C. Gonzato, S. Pinto, S. K. Liedl, K. Haupt, L. Ferreira, Angew. Chem. Int. Ed. 2020, 132, 2001.
60. Z. Wang, J. Zhang, Y. Wang, J. Zhou, X. Jiao, M. Han, X. Zhang, H. Hu, R. Su, Y. Zhang, W. Qi, ACS Nano 2024, 18, 10324.
61. M. Gao, M. Tang, W. Ho, Y. Teng, Q. Chen, L. Bu, X. Xu, X. Q. Zhang, ACS Nano 2023, 17, 17721.
62. A. Chakraborty, S. Dharmaraj, N. Truong, R.M. Pearson, ACS Appl Mater Interfaces 2022, 14, 56440.
63. H. Zhang, F. Ding, Z. Zhu, Q. Sun, C. Yang, Int J Pharm 2023, 631, 122489.
64. N. Kong, R. Zhang, G. Wu, X. Sui, J. Wang, N.Y. Kim, S. Blake, D. De, T. Xie, Y. Cao, W. Tao, Proc. Natl. Acad. Sci. USA 2022, 119, e2112696119.
65. A.F. Rodrigues, C. Rebelo, S. C. Paulo, S. Pinho, V. Francisco, L. Ferreira, Adv. Sci. 2023, 10, e2205475.
66. W. Cai, T. Luo, X. Chen, L. Mao, M. Wang, Adv Funct Mater 2022, 32, 2204947.
67. J. Duan, C. Bao, Y. Xie, H. Guo, Y. Liu, J. Li, R. Liu, P. Li, J. Bai, Y. Yan, L. Mu, X. Li, G. Wang, W. Lu, Bioact Mater 2022, 11, 1.
68. X. Zhao, Y. Wang, W. Jiang, Q. Wang, J. Li, Z. Wen, A. Li, K. Zhang, Z. Zhang, J. Shi, J. Liu, Adv. Mater. 2022, 37, 2204585.
69. Y. Wang, P.K. Shahi, X. Wang, R. Xie, Y. Zhao, M. Wu, S. Roge, B.R. Pattnaik, S. Gong, J. Control. Release 2021, 336, 296.
70. P. Zhang, X. Li, Q. Xu, Y. Wang, J. Ji, Colloids Surf. B Biointerfaces 2021, 208, 112125.
71. P. Norouzi, H. Motasadizadeh, F. Atyabi, R. Dinarvand, M. Gholami, M. Farokhi, M.A. Shokrgozar, F. Mottaghitalab, ACS Biomater Sci Eng 2021, 7, 1074.
72. J.H. Park, A. Mohapatra, J. Zhou, M. Holay, N. Krishnan, W. Gao, R.H. Fang, L. Zhang, Angew. Chem. Int. Ed. 2022, 61, e202113671.
73. S. Lu, X. Bao, W. Hai, S. Shi, Y. Chen, Q. Yu, M. Zhang, Y. Xu, J. Peng, Int J Pharm 2020, 575, 118898.
74. Y. Wang, P.K. Shahi, R. Xie, H. Zhang, A.A. Abdeen, N. Yodsanit, Z. Ma, K. Saha, B.R. Pattnaik, S. Gong, J. Control. Release 2020, 324, 194.
75. H. Tanaka, T. Takahashi, M. Konishi, N. Takata, M. Gomi, D. Shirane, R. Miyama, S. Hagiwara, Y. Yamasaki, Y. Sakurai, K. Ueda, K. Higashi, K. Moribe, E. Shinsho, R. Nishida, K. Fukuzawa, E. Yonemochi, K. Okuwaki, Y. Mochizuki, Y. Nakai, K. Tange, H. Yoshioka, S. Tamagawa, H. Akita, Adv. Funct. Mater. 2020, 30, 1910575.
76. W. Tao, A. Yurdagul Jr, N. Kong, W. Li, X. Wang, A.C. Doran, C. Feng, J. Wang, M. Ariful Islam, O.C. Farokhzad, I. Tabas, J. Shi, Sci. Transl. Med. 2020, 12, eaay1063.
77. J. Zhuang, H. Gong, J. Zhou, Q. Zhang, W. Gao, R.H. Fang, L. Zhang, Sci. Adv. 2020, 6, eaaz6108.
78. X. Hou, X. Zhang, W. Zhao, C. Zeng, B. Deng, D.W. McComb, S. Du, C. Zhang, W. Li, Y. Dong, Nat Nanotechnol. 2020, 15, 41.
79. X. Zhang, W. Zhao, G.N. Nguyen, C. Zhang, C. Zeng, J. Yan, S. Du, X. Hou, W. Li, J. Jiang, B. Deng, D.W. McComb, R. Dorkin, A. Shah, L. Barrera, F. Gregoire, M. Singh, D. Chen, D.E. Sabatino, Y. Dong, Sci. adv. 2020.6, eabc2315.
80. P. Zhang, Q. Xu, X. Li, Y. Wang, Mater. Sci. Eng. C 2020, 108, 110396.
81. I.B. Kelly, R.B. Fletcher, J.R. McBride, S.M. Weiss, C.L. Duvall, ACS Appl. Mater. Interfaces 2020, 12, 39602.
82. G. Li, Y. Gao, C. Gong, Z. Han, L. Qiang, Z. Tai, J. Tian, S. Gao, ACS Appl. Mater. Interfaces 2019, 11, 39.
Claims (24)
- A nanoworm composition comprising:at least two metal nanoparticle or nanorod cores;a polymer coating over the at least two metal nanoparticle or nanorod cores;a therapeutic nucleic acid; and,optionally, a lipid coating over the polymer coating.
- The nanoworm composition of claim 1, wherein the at least two metal nanoparticle or nanorod cores are at least two gold nanoparticle cores.
- The nanoworm composition of claim 1, wherein each of the at least two metal nanoparticle or nanorod cores has a diameter of about 40 nm.
- The nanoworm composition of claim 1, comprising 4 or 5 metal nanoparticle or nanorod cores.
- The nanoworm composition of claim 1, wherein the polymer coating has a thickness of about 7 nm to about 35 nm.
- The nanoworm composition of claim 1, wherein the polymer coating comprises polydopamine (PDA) , polyethylene glycol (PEG) , polyethylenimine (PEI) , or silica.
- The nanoworm composition of claim 1, wherein the nanoworm composition has an anionic charge.
- The nanoworm composition of claim 1, wherein the therapeutic nucleic acid is an oligonucleotide or mRNA.
- The nanoworm composition of claim 1, wherein the therapeutic nucleic acid is about 15 to about 5000 nucleotides in length.
- The nanoworm composition of claim 1, wherein the therapeutic nucleic acid is an antisense DNA, an antisense oligonucleotide (ASO) , short interfering RNA (siRNA) , microRNA (miRNA) , messenger RNA (mRNA) , or any combination thereof.
- The nanoworm composition of claim 8, wherein the therapeutic oligonucleotide nucleic acid is adsorbed onto the polymer coating.
- The nanoworm composition of claim 10, wherein the mRNA encodes hepatocyte growth factor (HGF) , chemokine receptor type 4 (CXCR4) , bone morphogenetic protein 7 (BMP-7) , enhanced green fluorescence protein (EGFP) , red fluorescence protein (RFP) , or any combination thereof; the miRNA is miR223; or the siRNA is siNog.
- The nanoworm composition of claim 1, further comprisinga lipid coating over the polymer coating, wherein the therapeutic nucleic acid is bound to the lipid coating.
- The nanoworm composition of claim 13, wherein the lipid coating comprises 1, 2-dioleoyl-3-trimethylammonium-propane (DOTAP) , cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DOPE) , (1, 1′- ( (2- (4- (2- ( (2- (bis (2-hydroxydodecyl) amino) ethyl) (2-hydroxydodecyl) amino) ethyl) piperazin-1-yl) ethyl) azanediyl) bis (dodecan-2-ol) ) (C12-200) , (6Z, 9Z, 28Z, 31Z) -Heptatriaconta-6, 9, 28, 31-tetraen-19-yl 4- (dimethylamino) butanoate (DLin-MC3-DMA) , ( (4-hydroxybutyl) azanediyl] di (hexane-6, 1-diyl) bis (2-hexyldecanoate) (ALC-0315) , dimethyldioctadecylammonium bromide (DDA) , 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC) , 1, 2-dioleoyl-3-dimethylammonium-propane (DODAP) , 1, 2-dioleyloxy-3-dimethylaminopropane (DODMA) , Dipalmitoylphosphatidylcholine (DPPC) , or any combination thereof.
- The nanoworm composition of claim 1, wherein the nanoworm comprises a citrate-capped Au NP (Cit-Au NP) having a diameter ranging from about 20 to about 60 nm.
- The nanoworm composition of claim 15, wherein the nanoworm comprises an Aux@PDA NW, wherein x is about 20, about 40, or about 60 nm in diameter.
- The nanoworm composition of claim 1, wherein the nanoworm comprises an Au@PDA NR (NanoRod) , wherein the size of the NR is about 45 nm x about 180 nm.
- A method of nucleic acid delivery comprising administering the nanoworm composition of claim 1 to a subject.
- The method of claim 18, wherein the therapeutic nucleic acid regulates gene expression in the subject.
- The method of claim 18, wherein the therapeutic nucleic acid escapes endosomal and/or lysosomal degradation.
- The method of claim 18, wherein the therapeutic nucleic acid enhances stem cell differentiation.
- The method of claim 19, wherein the regulation of gene expression in the subject treats acute liver injury, chronic kidney disease, or a combination thereof.
- The method of claim 18, wherein the nanoworm enters epithelial cells, liver cells, kidney cells, endothelial cells, primary macrophages, mesenchymal stem cells, neural cells, or any combination thereof in the subject.
- The composition of claim 18, wherein the nanoworm is administered by intra-an articular injection, an intrathecal injection, or a retrobulbar injection.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363590241P | 2023-10-13 | 2023-10-13 | |
| US63/590,241 | 2023-10-13 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025077898A1 true WO2025077898A1 (en) | 2025-04-17 |
Family
ID=95396666
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2024/124523 Pending WO2025077898A1 (en) | 2023-10-13 | 2024-10-12 | A therapeutic nucleic acid-encased nanoworm for gene delivery and endosomal escape |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025077898A1 (en) |
Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080226562A1 (en) * | 2005-12-22 | 2008-09-18 | Kevin Groves | Biocompatible fluorescent metal oxide nanoparticles |
| CN101512387A (en) * | 2006-09-06 | 2009-08-19 | 帝斯曼知识产权资产管理有限公司 | Core-shell nanoparticles |
| US20110183140A1 (en) * | 2010-01-22 | 2011-07-28 | University Of Maryland, College Park | Method for Polymer Coating and Functionalization of Metal Nanorods |
| US20130034599A1 (en) * | 2010-01-19 | 2013-02-07 | Northwestern University | Synthetic nanostructures including nucleic acids and/or other entities |
| US20130095187A1 (en) * | 2011-10-14 | 2013-04-18 | Postech Academy-Industry Foundation | Composition for nucleic acid delivery using metal nanoparticles and preparing method thereof |
| US20140234210A1 (en) * | 2011-07-08 | 2014-08-21 | The University Of North Carolina At Chapel Hill | Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders |
| CN109310702A (en) * | 2016-05-20 | 2019-02-05 | 芝加哥大学 | Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy and any combination thereof |
| US20190105271A1 (en) * | 2017-03-31 | 2019-04-11 | Rhode Island Council On Postsecondary Education | Nanoparticle-Induced Fusogenicity between Liposome And Endosome Membranes For Targeted Delivery Through Endosomal Escape |
-
2024
- 2024-10-12 WO PCT/CN2024/124523 patent/WO2025077898A1/en active Pending
Patent Citations (8)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080226562A1 (en) * | 2005-12-22 | 2008-09-18 | Kevin Groves | Biocompatible fluorescent metal oxide nanoparticles |
| CN101512387A (en) * | 2006-09-06 | 2009-08-19 | 帝斯曼知识产权资产管理有限公司 | Core-shell nanoparticles |
| US20130034599A1 (en) * | 2010-01-19 | 2013-02-07 | Northwestern University | Synthetic nanostructures including nucleic acids and/or other entities |
| US20110183140A1 (en) * | 2010-01-22 | 2011-07-28 | University Of Maryland, College Park | Method for Polymer Coating and Functionalization of Metal Nanorods |
| US20140234210A1 (en) * | 2011-07-08 | 2014-08-21 | The University Of North Carolina At Chapel Hill | Metal bisphosphonate nanoparticles for anti-cancer therapy and imaging and for treating bone disorders |
| US20130095187A1 (en) * | 2011-10-14 | 2013-04-18 | Postech Academy-Industry Foundation | Composition for nucleic acid delivery using metal nanoparticles and preparing method thereof |
| CN109310702A (en) * | 2016-05-20 | 2019-02-05 | 芝加哥大学 | Nanoparticles for chemotherapy, targeted therapy, photodynamic therapy, immunotherapy and any combination thereof |
| US20190105271A1 (en) * | 2017-03-31 | 2019-04-11 | Rhode Island Council On Postsecondary Education | Nanoparticle-Induced Fusogenicity between Liposome And Endosome Membranes For Targeted Delivery Through Endosomal Escape |
Non-Patent Citations (2)
| Title |
|---|
| "CNKI, Doctoral Dissertation", 1 May 2022, article NIE QING: "PEN coated CLIO-NH2 with siSnail2 combined with cisplatin to treat hepatic carcinoma through Ferroptosis", XP093304036, DOI: 10.27162/d.cnki.gjlin.2022.000393 * |
| HU YUE, NIE QING, CONG XIANLING, WU WEN, WU QIONG, LIU QIHUI, LI YUANYUAN, LIU HAIYAN, GE JINGYAN, CHEN FANGFANG: "PEN-coated superparamagnetic iron-mediated delivery of siSnail2 to inhibit metastasis and promote ferroptosis in the treatment of cancer", INTERNATIONAL JOURNAL OF PHARMACEUTICS, ELSEVIER, AMSTERDAM, NL, vol. 650, 1 January 2024 (2024-01-01), AMSTERDAM, NL, pages 123728, XP093304038, ISSN: 0378-5173, DOI: 10.1016/j.ijpharm.2023.123728 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20240173258A1 (en) | Liposome compositions and methods of use thereof | |
| US11464870B2 (en) | Lipid nanoparticles for in-vivo drug delivery, and uses thereof | |
| US20190292549A1 (en) | Poly(ethylene glycol) brushes for efficient rna delivery | |
| US11560575B2 (en) | High efficient delivery of plasmid DNA into human and vertebrate primary cells in vitro and in vivo by nanocomplexes | |
| CN116744979A (en) | Lipid nanoparticles containing mannose or uses thereof | |
| WO2019110067A1 (en) | Hybrid nanoparticle | |
| US11905338B2 (en) | Peptides for use as cell-penetrating peptides | |
| US9572895B2 (en) | Multiplexed supramolecular assemblies for non-viral delivery of genetic material | |
| Tao et al. | Brain-targeting gene delivery using a rabies virus glycoprotein peptide modulated hollow liposome: bio-behavioral study | |
| WO2011011631A2 (en) | Nucleic acid delivery vehicles | |
| Gandhi et al. | Bioreducible poly (amino ethers) based mTOR siRNA delivery for lung cancer | |
| WO2025077898A1 (en) | A therapeutic nucleic acid-encased nanoworm for gene delivery and endosomal escape | |
| AU2017242657B2 (en) | Synthetic compound for improving efficiency of transfection | |
| CN117241836A (en) | Composition for preventing or treating cancer comprising lipid nanoparticles | |
| JP2023039115A (en) | Substance delivery carrier and composition | |
| US20250367319A1 (en) | Targeted Nanomedicine for Treating Fibrotic Lung Disorders | |
| WO2025054668A1 (en) | Nanoparticle compositions | |
| US10232050B1 (en) | Multi-functional particles and methods of using the same | |
| US20250275921A1 (en) | Composition for preventing or treating cancer, containing lipid nanoparticles | |
| WO2025255152A1 (en) | Polymeric compounds and lipid compositions made using the same | |
| 조계수 | Receptor meditated endocytosis of hyper osmotic folate conjugated poly-sorbitol mediated transporter with siOPA1 for lung cancer therapy | |
| WO2016174430A1 (en) | Gene delivery |
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: 24876703 Country of ref document: EP Kind code of ref document: A1 |