WO2022081245A1 - Compositions and methods for increasing lymphangiogenesis - Google Patents

Compositions and methods for increasing lymphangiogenesis Download PDF

Info

Publication number
WO2022081245A1
WO2022081245A1 PCT/US2021/045918 US2021045918W WO2022081245A1 WO 2022081245 A1 WO2022081245 A1 WO 2022081245A1 US 2021045918 W US2021045918 W US 2021045918W WO 2022081245 A1 WO2022081245 A1 WO 2022081245A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
aibp
cav
composition
seq
Prior art date
Application number
PCT/US2021/045918
Other languages
French (fr)
Inventor
Longhou FANG
Jun-Dae Kim
Original Assignee
The Methodist Hospital System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Methodist Hospital System filed Critical The Methodist Hospital System
Publication of WO2022081245A1 publication Critical patent/WO2022081245A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/775Apolipopeptides
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/072Animals genetically altered by homologous recombination maintaining or altering function, i.e. knock in
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/40Fish
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0312Animal model for Alzheimer's disease
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0318Animal model for neurodegenerative disease, e.g. non- Alzheimer's
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • Lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, dietary lipid transport and immune surveillance.
  • the Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including lymphedema, Alzheimer’s disease, tumor metastasis, cardiovascular disease, obesity, and diabetic mellitus. What are needed are new compositions and methods for increasing lymphangiogenesis and treating the related diseases and disorders.
  • compositions and uses thereof for improving lymphangiogenesis comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • AIBP APOA1 binding protein
  • CAV-1 caveolin-1
  • the polynucleotide can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector).
  • the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
  • the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
  • the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR- Casl3, or CRISPR-Casl3d that ablate either DNA or mRNA. 5.
  • the composition disclosed herein further comprises a stimulator for VEGFR3.
  • the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)).
  • the stimulator comprises Pioglitazone.
  • composition disclosed herein can be contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
  • the composition is contained in a nanoparticle.
  • the composition is conjugated to docosahexaenoic acid (DHA).
  • DHA docosahexaenoic acid
  • compositions disclosed herein comprising administering to the subject a therapeutically effective amount of the composition disclosed herein, wherein the composition comprises i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • AIBP APOA1 binding protein
  • CAV-1 caveolin-1
  • the composition can be administered to the subject intrathecally or intracranially.
  • the neurodegenerative disease comprises Alzheimer’s disease, Parkinson's disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, or Multiple Sclerosis.
  • the composition increases lymphangiogenesis in brain.
  • a method for treating a neurodegenerative disease comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor disclose herein.
  • CAV-1 caveolin-1
  • a method for treating a neurodegenerative disease comprising administering to a subject a composition that is capable of crossing blood-brain barrier, wherein the composition comprises a caveolin-1 (CAV-1) inhibitor.
  • CAV-1 caveolin-1
  • a method for treating lymphedema in a subject comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • AIBP APOA1 binding protein
  • CAV-1 caveolin-1
  • Figures 1A-1D show effect of Aibp2 knockout on lymphatic vessel development.
  • Figure 1A shows lymphatic developmental defects in apoalbp ⁇ zebrafish. Maxiprojection confocal images of TD formation in the flilcc.egfp and apocilbp2 ⁇ / ⁇ ,' flilcc.egfp null zebrafish at 5 dpf. Arrows show the TD, and stars denote absent TD.
  • Figures 2A-2G show AIBP effect on LEC lineage commitment.
  • Figure 2A shows impaired LEC specification in apocilbp2 ⁇ 'f flilcc. egfp zebrafish. Control and cipocilbp2 knockout zebrafish were fixed with 4% PFA at 36 hpf, whole mount immunostaining was performed using Proxl and EGFP antibodies, and images were captured using confocal microscopy.
  • Figure 2B shows enumeration of LEC progenitors in Figure 2A. Arrows show specified LEC progenitors.
  • Figure 2C shows scheme illustration of mESC to LEC differentiation.
  • Figures 2D to 2G show that AIBP augments LEC differentiation from mESCs.
  • the embryoid bodies (EBs) of mESCs were prepared and cultured in the EC differentiation medium containing 2 ng/ml BMP4 and 10 ng/ml bFGF for 3 days.
  • Recombinant VEGFA (50 ng/ml) and VEGFC (50 ng/ml) in combination or AIBP (100 ng/ml) alone were supplemented at day 3 and kept in culture for additional 4 days.
  • the resulting cells were harvested for qPCR analysis of endothelial cell (EC) marker Pecaml expression (Figure 2D) and LEC-associated Lyvel expression (Figure 2E) at the indicated time points.
  • Figures 3A-3E Effect of cholesterol reduction on lymphatic vessel development.
  • Figure 3A shows free cholesterol (FC) content in control and Aibp2 null zebrafish.
  • FC free cholesterol
  • the 5 dpf zebrafish trunk of control or Aibp2 null zebrafish were dissected, total lipids extracted, and free cholesterol levels measured.
  • the FC content were normalized to the protein levels. Fifty embryos were pooled for each measurement. Mean ⁇ SE; *, p ⁇ 0.05.
  • Figure 3B shows that inhibition of cholesterol synthesis rescues TD formation in Aibp2 null animals. TD formation in control zebrafish, Aibp2 null zebrafish treated with 1 pM atorvastatin or control vehicle ethanol at 5 dpf.
  • Figure 3D shows that APOA1 -mediated cholesterol efflux restores TD formation. Confocal images of TD formation in control zebrafish, Aibp2 null zebrafish, or Aibp2 knockout zebrafish with 100 ng human APOA1 mRNA overexpression.
  • Figures 4A-4F show effect of cholesterol efflux on VEGFR3 signaling.
  • Figures 4A and 4B show cholesterol removal potentiates VEGFR3 signaling.
  • Figure 4A shows that hLECs were serum-starved, and treated with 10 mM MpCD for 30 min, and cells were further stimulated with 100 ng/ml VEGFC. The resulting cells were lysed and blotted with Cav-1 or GAPDH antibodies.
  • Figure 4B shows that hLECs were treated as in Figure 4A and cell lysates were immunoprecipitated using VEGFR3 antibody. Immunoblotting was performed using anti- phosphotyrosine (4G10) and VEGFR3 antibodies.
  • FIG. 4C shows that AIBP-mediated cholesterol efflux disrupts caveolae and reduces CAV-1 levels in the caveolar fractions.
  • hLECs were treated with recombinant 200 ng/ml AIBP, 100 pg/ml HDL3, or both in serum-free EBM2 for 6 hours, and the cells were subjected to sucrose-mediated ultracentrifugation. The resulting fractions were collected for Western blot analysis as indicated.
  • Figure 4D shows that AIBP- mediated cholesterol efflux increases VEGFR3 signaling.
  • hLECs were serum-starved and treated as in Figure 4C, and further stimulated with 100 ng/ml VEGFC. The resulting cells were lyzed and immunoblotted as indicated.
  • Figures 5A-5F show effect of CAV-1 on VEGFR3 signaling.
  • Figure 5A shows conserved CAV-1 binding site on VEGFR3 in human (Hu), mouse (Ms), and zebrafish (Zf).
  • Figure 5B shows that VEGFR3 AAA loses its binding to CAV-1.
  • hLECs were transfected with control EGFP (Ctrl), VEGFR3-EGFP (R3), or VEGFR3 AAA -EGFP (RS ⁇ ) using lentivirus- mediated gene transfer. After 72 hours, the resulting cells were lyzed and immunoprecipitated with EGFP antibody coupled to the magnetic Dynabeads and immunoblotted using VEGFR3 and CAV-1 antibodies.
  • VEGFR3-EGFP and VEGFR3 AAA - EGFP were detected using VEGFR3 antibody.
  • the input lysates were shown on the right.
  • Figure 5C shows that VEGFR3 AAA increases VEGFR3 signaling.
  • hLECs were transduced as in Figure 5B, and the resulting cells were serum starved and treated with 100 ng/ml VEGFC for 20 min, cells were then lysed and immunoblotted as indicated.
  • Quantitative data of VEGFR3 activation Figure 5D
  • AKT activation Figure 5E
  • ERK activation Figure 5F
  • Figures 6A-6D show effect of CAV-1 on Aibp2-regulated LEC specification.
  • Figure 6A shows that Cav-1 deficiency rescues LEC specification in Aibp2 knockdown animals.
  • Figure 6B shows quantitative data of LEC progenitors in (Figure 6A).
  • Figure 6C shows that Cav-1 deficiency corrects lymphatic defects in Aibp2 knockdown animals. Confocal imaging of TD formation in the indicated genetically modified flilcc.egfp zebrafish at 5 dpf. Arrows show TD, and stars denote absent TD.
  • Figure 7A-7C show AIBP effect on comeal lymphangiogenesis and LEC specification.
  • Figure 7A shows illustration of murine cornea lymphangiogenesis assay.
  • Figure 7B shows representative images of murine comeal lymphangiogenesis by implantation of pellets containing the indicated recombinant proteins and immunostained using LYVE1 & CD31 antibodies. Enlarged images of the boxed regions (scale bar, 1000 pm) are shown in the lower panels (scale bar, 500 pm).
  • Figure 7C shows quantification of LYVE1+ lymphatic vessel area per cornea.
  • Ctrl control
  • VC VEGFC
  • AA AIBP+APOA1
  • CSD CAV-1 scaffolding domain peptide. **, p ⁇ 0.01; ****, p ⁇ 0.0001.
  • Figures 8A-8D show AIBP expression in cutaneous lymphedema.
  • Figures 8B to 8D show the AIBP signal in the epidermis (Figure 8B), sweat glands (Figure 8C), and dermis (Figure 8D) and were quantified using ImageJ. The enlarged areas of green and yellow boxes are shown on the right. The images were taken separately and stitched together using the EVOS microscope. NL: normal leg; LY: lymphedema. **p ⁇ 0.0I.
  • Figures 9A-9E show AIBP regulates lymphatic vessel development in zebrafish.
  • Figure 9A shows lymphatic developmental defects in Aibp2 knockdown zebrafish. Confocal images of PL at 48 hpf and TD formation at 5 dpf in the control and Aibp2 null zebrafish. Arrows show PL or TD, and stars denote absent PL or TD.
  • Figures 9D and 9E show TD development in apoalbp2-/- and control (CT) flilcc.egfp; lyvel.DsRed zebrafish (Figure 9D) and the quantification ( Figure 9E). Scale bar: 100 pm. **, p ⁇ 0.0I. 21.
  • Figure 10A- 1 OB show that AIBP regulates lymphatic vessel development in zebrafish.
  • Figure 10A shows that Aibp2 ablation disrupts LEC specification.
  • Figure 10B shows quantitative data of LEC progenitors in Figure 10A.
  • Control or apoalbp2 morphants were fixed at 36 hpf, immunostained using Proxl and EGFP antibodies, and images were captured using confocal microscopy. Arrows indicate LEC progenitors. Scale: 25 pm. **p ⁇ 0.01.
  • Figures 11A-1 IB show FACS analysis of murine ESC-derived CD31 + LYVE1 + LECs.
  • Figure 11A shows that AIBP induces LEC lineage commitment from mESCs.
  • the murine ESCs were subjected to mesoderm and then endothelial differentiation as described in Figure 2C. At day 1 and day 7, the resulting cells were dissociated, immunostained with CD31 and LYVE1 antibodies, fixed with 4% PFA, and used for FACS analysis. The percentage of CD31 + and LYVE1 + cells were shown.
  • Figure 1 IB shows quantitative data of FACS-sorted CD31 + and LYVEl + cells in Figure 11A. **p ⁇ 0.01.
  • Figures 12A-12E show generation of Cav-1 knockout zebrafish.
  • Figure 12A depicts diagram showing position of the target site and its sequence (underline) in zebrafish apoalbp2 locus. PAM sequence (GGG) is shown in red.
  • Figure 12B shows Sanger sequencing result of heterozygous mutants revealed an 8-bp genomic DNA fragment insertion from the target site. The PCR amplicons that span the mutated apoalbp2 region were ligated into a T- vector and subsequently transformed into competent cells. Single positive colonies were selected for sequencing.
  • Figure 12C shows that the 8-bp insertion resulted in a frame shift that generates a mutated protein.
  • Figure 12E shows no gross phenotypic defect observed in Cav- 1 knockout zebrafish. Zebrafish embryos at the indicated developmental stages were collected and images of live zebrafish embryos captured, hpf: hour(s) post fertilization, dpf: days postfertilization. WT: wild type.
  • the sequences in Figure 12 include GACGTGATCGCCGAGCCTGCCGG (SEQ ID NO: 40), GATCGCCGAGCCTGCCGGCACCTACAGCTTCGACG (SEQ ID NO: 41), GATCGCCGAGCACCTACAGCTTCGACGGCGTGTGG (SEQ ID NO: 42), MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP KHLNDDVVKVDFEDVIAEPAGTYSFDGVWKASFTTFTVTKYWCYRLLTALVGIPLALV WGIFFAILSFIHIWAVVPCVKSYLIEIHCISRVYSICVHTFCDPLFEAMGKCFSNVRVTATK VV (SEQ ID NO: 43), MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP KHLNDDVVKVDFEDVIAEHLQLRRRVEGELHHLHSNQILV
  • Figures 13A-13B show quantitative PCR analysis of genes regulating lymphatic development in zebrafish.
  • Figure 13A shows that loss of Cav-1 increases LEC gene expression.
  • Figure 13B shows that Cav-1 overexpression reduces LEC gene expression.
  • the zebrafish embryo at one cell stage was injected with Cav-1 mRNA, and the resulting animals or control animals were harvested at 96 hpf, and total RNA extracted for reverse transcription.
  • FIGS 14A-14B show that CAV-1 knockout increases tail lymphangiogenesis in neonatal mice.
  • Figure 14A shows that the CAV-1 knockout mice were purchased from JAX (stock No. 007083). The tail epidermis of CAV-1 knockout mice and control littermates were dissected from the similar anatomical locations and immunostained using LYVE-1 antibodies.
  • Figure 14B shows that quantification of lymphatic vessel length in Figure 14A were performed using ImageJ. *, p ⁇ 0.05.
  • Figures 15A-15D show that AIBP/APOA1 or CSD per se has no effect on adult lymphangiogenesis.
  • Figures 15A and 15C show that PEG pellets containing recombinant AIBP and APOA1 were prepared and implanted into the corneas of B6 mice, control was implanted with control pellets.
  • Figures 15B and 15D show quantification of lymphatic vessel area in Figure 15A and 15C.
  • FIGS 16A-16B depict graphical Abstract.
  • the lymphatics facilitate the drainage the cerebrospinal fluid (CSF) and interstitial fluid (ISF) from the brain.
  • Figure 16B shows the underlying cause of AD is the formation of extracellular Ap aggregation and Tau neurofibrillary tangles. Impaired lymphatic drainage contributes to the A aggregation.
  • Figure 16C shows that nanoparticle mediated delivery enables sustained release of AIBP/VEGFC, which can augment lymphatic vessel growth and lymphatic function, thereby improving AD.
  • ISF interstitial fluid.
  • Figures 17A-17C show that AIBP-CAV1 axis promotes comeal lymphangiogenesis.
  • Figure 17A shows illustration of murine cornea lymphangiogenesis assay.
  • Figure 17B shows representative whole mount images of murine comeal lymphangiogenesis by implantation of PEG pellets containing the indicated recombinant proteins and immunostained using LYVE1 & CD31 antibodies. Enlarged images of the boxed regions (scale bar, 1000 pm) are shown in the lower panels (scale bar, 500 pm).
  • Figure 17C shows quantification of LYVE1+ lymphatic vessel area per cornea.
  • VC VEGFC
  • CSD Cavl scaffolding domain peptide. ***p ⁇ 0.001; *p ⁇ 0.05.
  • FIGS 18A-18C show that AIBP increases LEC lineage specification.
  • Figure 18A shows scheme of mESC to LEC differentiation.
  • Figures 18B-18C show that AIBP augments LEC differentiation from mESCs.
  • the embryoid bodies of mESCs were prepared and cultured in the EC differentiation medium containing BMP4 and bFGF for 3 days.
  • Recombinant VEGFA and VEGFC in combination or AIBP alone were supplemented at day 3 and kept in culture for additional 4 days.
  • Western blot analyses of EC-associated PECAM, LEC marker LYVE1 and PROXI expression Figure 18B
  • Figure 18C Western blot analyses of EC-associated PECAM, LEC marker LYVE1 and PROXI expression
  • C control
  • V VEGFA + VEGFC
  • A AIBP.
  • Figures 19A-19C show that AIBP protects vascular integrity.
  • Figure 19A shows that confluent HRMECs were stimulated for 1 hour with 100 ng/ml VEGF, in the presence or absence of 200 ng/ml recombinant AIBP, followed by immunostaining with anti-VE-Cad antibody. Images are representative of 5 different fields. Stars show paracellular gaps. Scale: 25 pm.
  • Figure 19B shows scheme of permeability assay.
  • Figure 19C shows measurements of leaked FITC-dextran (70 kDa) in the bottom chamber with the indicated treatments. *p ⁇ 0.05.
  • Figure 20 illustrates the effect of three CAV 1 modifying peptides on comeal lymphangiogenesis.
  • Figure 20 shows representative images of murine comeal lymphangiogenesis by implantation of pellets containing the indicated peptide and immunostained using LYVE1 & CD31 antibodies, and quantification of LYVE1 + lymphatic vessel area per cornea.
  • VC VEGFC
  • CSD CAV-1 scaffolding domain peptide
  • CAV1-M CAV1 modulator
  • CAVl-i CAV1 inhibitor.
  • Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed.
  • Activate means to increase an activity, response, condition, or other biological parameter. This may also include, for example, a 10% increase in the activity, response, "or condition, as compared to the native or control level. Thus, the increase can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
  • Activators or “inhibitors” of expression or of activity are used to refer to inhibitory or activating molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., ligands, agonists, antagonists, and their homologs and mimetics.
  • Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or protease activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein, e.g., antagonists.
  • Activators are agents that, e.g., induce or activate the expression of a described target protein or bind to, stimulate, increase, open, activate, facilitate, enhance activation or protease inhibitor activity, sensitize or up regulate the activity of described target protein (or encoding polynucleotide), e.g., agonists.
  • Samples or assays comprising described target protein that are treated with a potential activator or inhibitor are compared to control samples without the inhibitor or activator to examine the extent of effect. Control samples are assigned a relative activity value of 100% inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%.
  • Activation of the described target protein is achieved when the activity value relative to the control is 110%, optionally 150%, optionally 200, 300%, 400%, 500%, or 1000-3000% or more.
  • administering to a subject includes any route of introducing or delivering to a subject an agent. Administration can be carried out by any suitable route, including oral, topical, intravenous, subcutaneous, transcutaneous, transdermal, intramuscular, intra-joint, parenteral, intra-arteriole, intradermal, intraventricular, intracranial, intraperitoneal, intralesional, intranasal, rectal, vaginal, by inhalation, via an implanted reservoir, or via a transdermal patch, and the like. Administration includes self-administration and the administration by another. In some embodiments, the composition disclosed herein is administered intrathecally or intracranially. In some embodiments, the composition disclosed herein is administered topically (e.g., using a skin ointment).
  • agonist refers to a composition that binds to a receptor and activates the receptor to produce a biological response.
  • the terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of agonists specifically mentioned herein, including, but not limited to, salts, esters, amides, reagents, active metabolites, isomers, fragments, analogs, and the like.
  • agonist refers to a composition that binds to a receptor and activates the receptor to produce a biological response.
  • the terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of agonists specifically mentioned herein, including, but not limited to, salts, esters, amides, reagents, active metabolites, isomers, fragments, analogs, and the like.
  • the term “agonist” includes the agent per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, reagents, conjugates, active metabolites, isomers, fragments, analogs, etc.
  • biocompatible generally refers to a material and any metabolites or degradation products thereof that are generally non-toxic to the recipient and do not cause significant adverse effects to the subject.
  • Biological sample refers to a sample of biological material obtained from a subject. Biological samples include all clinical samples useful for detection of disease or disorder in subjects. Appropriate samples include any conventional biological samples, including clinical samples obtained from a human or veterinary subject. Exemplary samples include, without limitation, cells, cell lysates, blood smears, cytocentrifiige preparations, cytology smears, bodily fluids (e.g., blood, plasma, serum, saliva, sputum, urine, bronchial alveolar lavage, semen, cerebrospinal fluid (CSF), etc.), tissue biopsies or autopsies, fine-needle aspirates, and/or tissue sections.
  • CSF cerebrospinal fluid
  • “Complementary” or “substantially complementary” refers to the hybridization or base pairing or the formation of a duplex between nucleotides or nucleic acids, such as, for instance, between the two strands of a double stranded DNA molecule or between an oligonucleotide primer and a primer binding site on a single stranded nucleic acid.
  • Complementary nucleotides are, generally, A and T/U, or C and G.
  • Two single-stranded RNA or DNA molecules are said to be substantially complementary when the nucleotides of one strand, optimally aligned and compared and with appropriate nucleotide insertions or deletions, pair with at least about 80% of the nucleotides of the other strand, usually at least about 90% to 95%, and more preferably from about 98 to 100%.
  • substantial complementarity exists when an RNA or DNA strand will hybridize under selective hybridization conditions to its complement.
  • selective hybridization will occur when there is at least about 65% complementary over a stretch of at least 14 to 25 nucleotides, at least about 75%, or at least about 90% complementary. See Kanehisa (1984) Nucl. Acids Res. 12:203.
  • compositions and methods include the recited elements, but not excluding others.
  • Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. “Consisting of’ shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention. Embodiments defined by each of these transition terms are within the scope of this invention. 45. “Composition” refers to any agent that has a beneficial biological effect.
  • Beneficial biological effects include both therapeutic effects, e.g., treatment of a disorder or other undesirable physiological condition, and prophylactic effects, e.g., prevention of a disorder or other undesirable physiological condition (e.g., a neurodegenerative disorder or lymphadema).
  • the terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of beneficial agents specifically mentioned herein, including, but not limited to, a vector, polynucleotide, cells, salts, esters, amides, reagents, active metabolites, isomers, fragments, analogs, and the like.
  • composition when used, then, or when a particular composition is specifically identified, it is to be understood that the term includes the composition per se as well as pharmaceutically acceptable, pharmacologically active vector, polynucleotide, salts, esters, amides, reagents, conjugates, active metabolites, isomers, fragments, analogs, etc.
  • control is an alternative subject or sample used in an experiment for comparison purposes.
  • Diagnosis refers to the process of identifying a disease by its signs, symptoms and results of various tests. The conclusion reached through that process is also called "a diagnosis.”
  • Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom, Thus, a gene encodes a protein if transcription and translation of mRNA.
  • “Expression vector”, or “vector”, comprises a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed.
  • An expression vector comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system.
  • Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno- associated viruses) that incorporate the recombinant polynucleotide.)
  • fragments can include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the nonmodified peptide or protein. These modifications can provide for some additional property, such as to remove or add amino acids capable of disulfide bonding, to increase its bio-longevity, to alter its secretory characteristics, etc. In any case, the fragment must possess a bioactive property, such as regulating the transcription of the target gene.
  • the term “gene” or “gene sequence” refers to the coding sequence or control sequence, or fragments thereof.
  • a gene may include any combination of coding sequence and control sequence, or fragments thereof.
  • a “gene” as referred to herein may be all or part of a native gene.
  • a polynucleotide sequence as referred to herein may be used interchangeably with the term “gene”, or may include any coding sequence, non-coding sequence or control sequence, fragments thereof, and combinations thereof.
  • the term “gene” or “gene sequence” includes, for example, control sequences upstream of the coding sequence (for example, the ribosome binding site).
  • “Inhibit”, “inhibiting,” and “inhibition” mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
  • Nucleic acid A deoxyribonucleotide or ribonucleotide polymer, which can include analogues of natural nucleotides that hybridize to nucleic acid molecules in a manner similar to naturally occurring nucleotides.
  • a nucleic acid molecule is a single stranded (ss) DNA or RNA molecule, such as a probe or primer.
  • a nucleic acid molecule is a double stranded (ds) nucleic acid, such as a target nucleic acid.
  • modified nucleic acids are those with altered sugar moieties, such as a locked nucleic acid (LN A).
  • Nucleotide The fundamental unit of nucleic acid molecules.
  • a nucleotide includes a nitrogen-containing base attached to a pentose monosaccharide with one, two, or three phosphate groups attached by ester linkages to the saccharide moiety.
  • the major nucleotides of DNA are deoxyadenosine 5 '-triphosphate (dATP or A), deoxyguanosine 5'- triphosphate (dGTP or G), deoxycytidine 5 '-triphosphate (dCTP or C) and deoxythymidine 5'- triphosphate (dTTP or T).
  • RNA The major nucleotides of RNA are adenosine 5 '-triphosphate (ATP or A), guanosine 5 '-triphosphate (GTP or G), cytidine 5 '-triphosphate (CTP or C) and uridine 5'- triphosphate (UTP or U).
  • ATP adenosine 5 '-triphosphate
  • GTP guanosine 5 '-triphosphate
  • CTP cytidine 5 '-triphosphate
  • UDP uridine 5'- triphosphate
  • “Pharmaceutically acceptable” component can refer to a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical formulation of the invention and administered to a subject as described herein without causing significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation in which it is contained.
  • the term When used in reference to administration to a human, the term generally implies the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
  • “Pharmaceutically acceptable carrier” means a carrier or excipient that is useful in preparing a pharmaceutical or therapeutic composition that is generally safe and non-toxic, and includes a carrier that is acceptable for veterinary and/or human pharmaceutical or therapeutic use.
  • carrier or “pharmaceutically acceptable carrier” can include, but are not limited to, phosphate buffered saline solution, water, emulsions (such as an oil/water or water/oil emulsion) and/or various types of wetting agents.
  • carrier encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, lipid, stabilizer, or other material well known in the art for use in pharmaceutical formulations.
  • a carrier for use in a composition will depend upon the intended route of administration for the composition.
  • the preparation of pharmaceutically acceptable carriers and formulations containing these materials is described in, e.g., Remington's Pharmaceutical Sciences, 21st Edition, ed. University of the Sciences in Philadelphia, Eippincott, Williams & Wilkins, Philadelphia, PA, 2005.
  • physiologically acceptable carriers include saline, glycerol, DMSO, buffers such as phosphate buffers, citrate buffer, and buffers with other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEENTM (ICI, Inc.; Bridgewater, New Jersey), polyethylene glycol (PEG), and PLURONICSTM (BASF; Florham Park, NJ).
  • buffers such as phosphate buffer
  • polynucleotide refers to a single or double stranded polymer composed of nucleotide monomers (DNA or RNA).
  • polypeptide refers to a compound made up of a single chain of D- or L-amino acids or a mixture of D- and L-amino acids joined by peptide bonds.
  • peptide As used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another.
  • promoter as used herein is defined as a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence.
  • promoter/regulatory sequence means a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
  • the promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
  • “Recombinant” used in reference to a gene refers herein to a sequence of nucleic acids that are not naturally occurring in the genome of the bacterium.
  • the non-naturally occurring sequence may include a recombination, substitution, deletion, or addition of one or more bases with respect to the nucleic acid sequence originally present in the natural genome of the bacterium.
  • the term “increased” or “increase” as used herein generally means an increase by a statically significant amount; for the avoidance of any doubt, “increased” means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3 -fold, or at least about a 4-fold, or at least about a 5 -fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • reduced generally means a decrease by a statistically significant amount.
  • reduced means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (i.e. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.
  • Sequence identity The similarity between two nucleic acid sequences is expressed in terms of the similarity between the sequences, otherwise referred to as sequence identity.
  • Sequence identity is frequently measured in terms of percentage identity, similarity, or homology; a higher percentage identity indicates a higher degree of sequence similarity.
  • NCBI Basic Local Alignment Search Tool (BLAST), Altschul et al, J. Mol. Biol. 215:403-10, 1990, is available from several sources, including the National Center for Biotechnology Information (NCBI, Bethesda, MD), for use in connection with the sequence analysis programs blastp, blastn, blastx, tblastn and tblastx. It can be accessed through the NCBI website. A description of how to determine sequence identity using this program is also available on the website.
  • homologs When less than the entire sequence is being compared for sequence identity, homologs will typically possess at least 75% sequence identity over short windows of 10-20 amino acids, and can possess sequence identities of at least 85% or at least 90% or 95% depending on their similarity to the reference sequence. Methods for determining sequence identity over such short windows are described on the NCBI website.
  • Subject Any mammal, such as humans, non-human primates, pigs, sheep, horses, dogs, cats, cows, rodents and the like. In two non-limiting examples, a subject is a human subject or a murine subject. Thus, the term "subject" includes both human and veterinary subjects.
  • Treat,” “treating,” “treatment,” and grammatical variations thereof as used herein, include partially or completely delaying, alleviating, mitigating or reducing the intensity of one or more attendant symptoms of a disorder or condition and/or alleviating, mitigating or impeding one or more causes of a disorder or condition.
  • Treatments according to the invention may be applied preventively, prophy tactically, pallatively or remedially.
  • Prophylactic treatments are administered to a subject prior to onset (e.g., before obvious signs of a neurodegenerative disorder or lymphedema), during early onset (e.g. , upon initial signs and symptoms of a neurodegenerative disorder or lymphedema), or after an established development of a neurodegenerative disorder or lymphedema.
  • Prophylactic administration can occur for several days to years prior to the manifestation of symptoms of a disorder.
  • “Therapeutically effective amount” or “therapeutically effective dose” of a composition refers to an amount that is effective to achieve a desired therapeutic result.
  • a desired therapeutic result is the control of a neurodegenerative disorder or lymphedema.
  • a desired therapeutic result is the control of a neurodegenerative disorder or lymphedema, or a symptom of a neurodegenerative disorder or lymphedema.
  • Therapeutically effective amounts of a given therapeutic agent will typically vary with respect to factors such as the type and severity of the disorder or disease being treated and the age, gender, and weight of the subject.
  • the term can also refer to an amount of a therapeutic agent, or a rate of delivery of a therapeutic agent (e.g., amount over time), effective to facilitate a desired therapeutic effect.
  • a therapeutic agent e.g., amount over time
  • the precise desired therapeutic effect will vary according to the condition to be treated, the tolerance of the subject, the agent and/or agent formulation to be administered (e.g., the potency of the therapeutic agent, the concentration of agent in the formulation, and the like), and a variety of other factors that are appreciated by those of ordinary skill in the art.
  • a desired biological or medical response is achieved following administration of multiple dosages of the composition to the subject over a period of days, weeks, or years.
  • Lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, dietary lipid transport and immune surveillance.
  • the Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including lymphedema, tumor metastasis, cardiovascular disease, obesity, Alzheimer’s disease, and diabetic mellitus.
  • Therapeutic augmentation of lymphangiogenesis has been documented to improve lymphatic structure and function.
  • the lymphatic system is derived from the embryonic cardinal vein (CV), where lymphatic endothelial cells (LEC) progenitors are specified and subsequently migrate and establish the lymphatic network.
  • CV embryonic cardinal vein
  • LEC lymphatic endothelial cells
  • Recent studies indicate that a subset of lymphatics can be developed by non-canonical mechanisms.
  • lymphatic vascular network During murine development, the assembly of the lymphatic vascular network is initiated at approximately embryonic day 9.5 (E9.5) under the control of VEGFC signaling through its cognate receptor VEGFR3.
  • LEC fate commitment occurs through SOX18-induced expression of PROX-1, which, in concert with the orphan nuclear factor NR2F2 (also known as COUP-TFII), dictates LEC differentiation from the CV.
  • NR2F2 also known as COUP-TFII
  • GATA2, HHEX, and PROXI itself have recently been shown to upregulate PROXI expression.
  • the newly specified LECs subsequently bud from the CV and migrate (E10.0- E11.5) in a dorsolateral fashion to form lymph sacs and, thereby establish the entire lymphatic vessels.
  • VEGFC-induced VEGFR3 signaling is the major driver of lymphangiogenesis in vertebrates.
  • VEGFR3 signaling also controls LEC progenitor homeostasis by maintaining PROXI expression levels in a positive feedback loop.
  • Vegfc/Vegfr3 signaling increases Proxl + LEC progenitors in the axis vasculature.
  • Mice deficient either in CCBE1, a critical matrix protein regulating VEGFC bioavailability, in VEGFC, or in VEGFR3 show fewer PROXI -positive LECs in the CV.
  • PLs subsequently migrate ventrally and dorsally along the arterial intersegmental vessels (ISVs) at ⁇ 60 hpf, to form the thoracic duct (TD), intersegmental lymphatic vessels and dorsal longitudinal lymphatic vessels (DLLV).
  • ISVs arterial intersegmental vessels
  • DLLV dorsal longitudinal lymphatic vessels
  • AIBP angiogenesis protein binds ECs, accelerates cholesterol efflux from ECs to high-density lipoprotein (HDL) and reduces lipid raft/caveola abundance, which in turn disrupts VEGFR2 signaling, thereby restricting angiogenesis.
  • Lymphatic vessels are structurally and functionally related to blood vessels.
  • Many genes that are required for angiogenesis such as VEGFR3, FGF, D114-Notch, angiopoietin-Tie2, Ephrin B2, and TGFp family member ALK1, Epsin, and others, also function in lymphangiogenesis.
  • APOA1 binding protein (AIBP) polypeptide and/or polynucleotide can used be treat neurodegenerative disorders.
  • Dudau M et al. Arguments for Caveolin-1 Knockout Mice as an Alzheimer’s Disease Model, Austin Alzheimer s J Parkinsons Dis 3(1): idl028 (2016) indicates that caveolin-1 (CAV-1) knockout mice exhibit Alzheimer’s disease-like symptoms and can be used as Alzheimer’s disease model. But this is a genetic model and the mice lose CAV-1 from one cell stage in development, i.e., this cannot not considered as a normal mouse from the beginning of conception.
  • the present disclosure shows that administration of CAV-1 inhibitor and/or AIBP, in the presence of a VEGFR3 stimulator (e.g., VEGFC), surprisingly improves meningeal lymphatics functions and treats a neurodegenerative disease (e.g., Alzheimer’s disease).
  • a VEGFR3 stimulator e.g., VEGFC
  • composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • AIBP APOA1 binding protein
  • CAV-1 caveolin-1
  • the AIBP polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 1 or a fragment thereof.
  • the AIBP polypeptide comprises a sequence of SEQ ID NO: 1.
  • the polynucleotide can be a DNA or an RNA.
  • the polynucleotide encoding the AIBP polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 2 or a fragment thereof.
  • the polynucleotide encoding the AIBP polypeptide comprises a sequence of SEQ ID NO: 2.
  • the polynucleotide is an RNA encoded by a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 2 or a fragment thereof.
  • Caveolin-1 is an oncogenic membrane protein associated with endocytosis, extracellular matrix organization, cholesterol distribution, cell migration and signaling. The present disclosure shows that CAV-1 suppresses VEGFR3 activation in lymphatic endothelial cells.
  • CAV-1 refers herein to a polypeptide that, in humans, is encoded by the C4F7 gene. In some embodiments, the CAV-1 polypeptide is that identified in one or more publicly available databases as follows: HGNC: 1527, Entrez Gene: 857, Ensembl: ENSG00000105974, OMIM: 601047, UniProtKB: Q03135.
  • the CAV-1 polypeptide comprises the sequence of SEQ ID NO: 7, or a polypeptide sequence having at or greater than about 80%, about 85%, about 90%, about 95%, or about 98% homology with SEQ ID NO: 7, or a polypeptide comprising a portion of SEQ ID NO: 7.
  • the CAV-1 polypeptide of SEQ ID NO: 7 may represent an immature or pre-processed form of mature CAV-1, and accordingly, included herein are mature or processed portions of the CAV-1 polypeptide in SEQ ID NO: 7.
  • the composition disclose herein further comprises a CAV-1 inhibitor, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
  • CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
  • Inhibitors of expression or of activity are used to refer to inhibitory molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., antagonists and their homologs and mimetics. Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein. Control samples (untreated with inhibitors) are assigned a relative activity value of 100%. Inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%.
  • CAV-1 is a scaffolding protein that provides a spatially restricted platform for proper signaling of cell surface receptors.
  • CAV-1 contains a signaling motif that interacts with a variety of membrane receptors and modulates their activities (e.g., the activities of VEGFR3).
  • a CAV-1 inhibitor can be an inhibitor for one or more other factors (e.g., one or more genes, proteins, mRNA) involved in the CAV-1 -involving pathway or signaling platform.
  • the CAV-1 inhibitor can include any one or more agents which upon administration to a subject, can inhibit CAV-1.
  • the CAV-1 inhibitor can directly affect CAV-1, for example, by binding to the CAV-1 protein or preventing the transcription or translation of a CAV-1 gene.
  • the CAV-1 inhibitor can inhibit one or more other factors (e.g., one or more genes, proteins, mRNA) involved in the CAV-1 pathway.
  • the CAV-1 inhibitor can be a peptide that competes with the CAV- 1 such that to inhibit the interaction of CAV-1 and a bind protein thereof (including, for example, VEGFR3).
  • the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux. In some embodiments, the CAV-1 inhibitor comprises methyl beta cyclodextrin.
  • the CAV-1 inhibitor is an antibody (e.g., an antagonizing/inhibitory antibody of CAV-1).
  • antibody and “antibodies” are used herein in a broad sense and include polyclonal antibodies, monoclonal antibodies, and bi-specific antibodies. In addition to intact immunoglobulin molecules, also included in the term “antibodies” are fragments or polymers of those immunoglobulin molecules, and human or humanized versions of immunoglobulin molecules or fragments thereof. Antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end.
  • VH variable domain
  • VL variable domain at one end
  • the antibodies can be tested for their desired activity using the in vitro assays described herein, or by analogous methods, after which, their in vivo therapeutic and/or prophylactic activities are tested according to known clinical testing methods.
  • IgA-1, IgG-2, IgG-3, and IgG-4 immunoglobulin-1
  • IgA-1 and IgA -2 One skilled in the art would recognize the comparable classes for mouse.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • antibody encompasses chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab’)2, Fab’, Fab, Fv, scFv, a single-chain variable fragment (scFv) antibody, or a VHH antibody and the like, including hybrid fragments.
  • fragments of the antibodies that retain the ability to bind their specific antigens are provided.
  • the composition disclosed herein comprises a CAV-1 inhibitor, wherein the CAV-1 inhibitor is an antibody, and wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, and/or a VHH antibody.
  • the CAV-1 inhibitor comprises a CAV-1 gene editing tool, including, for example, a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, CRISPR-Casl3, or CRISPR-Casl3d.
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • CRISPR-Cas9 CRISPR-Casl3d.
  • the CAV-1 inhibition method described herein comprises using CRISPR-Cas9 that targets a CAV-1 polynucleotide sequence.
  • CAV-1 polynucleotide sequence is at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 8 or a fragment thereof.
  • the CAV-1 inhibitor comprises a siRNA targeting CAV-1, wherein the siRNA comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 9, 10, or 11 or a fragment thereof.
  • the siRNA comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 9, 10, or 11 or a fragment thereof.
  • the CAV-1 inhibitor comprises a Caveolin-l(Cav-l) scaffolding domain (CSD) (CSD) peptide that can compete with the plasma membrane CAV-1, inhibit the interaction of the proteins and CAV-1, and re -store the functions of CAV-1 binding proteins.
  • the CSD peptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 12 or a fragment thereof.
  • the CAV-1 inhibitor can be a polynucleotide sequence encoding the CSD peptide disclosed herein. Accordingly, in one example, the composition disclosed herein comprises a first polynucleotide encoding an AIBP polypeptide and a second polynucleotide encoding a CSD polypeptide. In another example, the composition disclosed herein comprises a polynucleotide that encodes an AIBP polypeptide and a CSD polypeptide.
  • VEGFR3 refers herein to a polypeptide that, in humans, is encoded by the FLT4 gene.
  • the VEGFR3 polypeptide is that identified in one or more publicly available databases as follows: HGNC: 3767; Entrez Gene: 2324; Ensembl: ENSG00000037280; OMIM: 136352; UniProtKB: P35916.
  • the VEGFR3 tyrosine kinase is expressed mainly in lymphatic vessels.
  • VEGFR3 transmits signals for lymphatic endothelial migration, survival, and proliferation and is involved in the biology and pathology of the lymphatic vasculature.
  • the composition disclosed herein further comprises a stimulator, activator, or agonist of VEGFR3.
  • the stimulator, activator, and/or agonist of VEGFR3 comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
  • the VEGFA polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 3 or a fragment thereof.
  • the VEGFC polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 5 or a fragment thereof.
  • the stimulator, activator, or agonist of VEGFR3 is VEGFC(C156S).
  • VEGFC(C156S) is encoded by a polynucleotide sequence at least about at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 13 or a fragment thereof.
  • the stimulator, activator, or agonist of VEGFR3 is a small molecule (e.g., Pioglitazone).
  • composition disclosed herein can further comprises at least about one polynucleotide encoding a VEGFA polypeptide and/or a VEGFC polypeptide. Accordingly, in some embodiments, the composition disclosed herein comprises a first polynucleotide encoding an AIBP polypeptide, a second polynucleotide encoding a CSD polypeptide, a third polynucleotide encoding a VEGFA polypeptide, and/or a fourth polynucleotide encoding a VEGFC polypeptide, or any combination thereof.
  • the composition disclosed herein comprises a polynucleotide that encodes an AIBP polypeptide, a CSD polypeptide, a VEGFA polypeptide, and/or a VEGFC polypeptide, or any combination thereof.
  • the polynucleotide encoding VEGFA polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 4 or a fragment thereof.
  • the polynucleotide encoding VEGFC polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 6 or a fragment thereof.
  • the polynucleotide disclosed herein can be contained in a vector that can be used to deliver the polynucleotide to cells, either in vitro or in vivo.
  • the vectors and the delivery methods can largely be broken down into two classes: viral based delivery systems and non-viral based delivery systems.
  • the nucleic acids can be delivered through a number of direct delivery systems such as, electroporation, lipofection, calcium phosphate precipitation, plasmids, viral vectors, viral nucleic acids, phage nucleic acids, phages, cosmids, or via transfer of genetic material in cells or carriers such as cationic liposomes.
  • Transfer vectors can be any nucleotide construction used to deliver genes into cells (e.g., a plasmid), or as part of a general strategy to deliver genes, e.g., as part of recombinant retrovirus or adenovirus (Ram et al. Cancer Res. 53:83-88, (1993)).
  • plasmid or viral vectors are agents that transport the disclosed polynucleotides (e.g., a polynucleotide encoding an AIBP polypeptide, a CSD peptide, a VEGFA polypeptide, and/or a VEGFC polypeptide, or a combination thereof) into the cell without degradation and include a promoter yielding expression of the gene in the cells into which it is delivered.
  • the polypeptides are derived from either a virus or a retrovirus.
  • Viral vectors can be, for example, Adenovirus, Adeno-associated virus, Herpes virus, Vaccinia virus, Polio virus, AIDS virus, neuronal trophic virus, Sindbis and other RNA viruses, including these viruses with the HIV backbone. Also preferred are any viral families which share the properties of these viruses which make them suitable for use as vectors.
  • a preferred embodiment is a viral vector which has been engineered so as to suppress the immune response of the host organism, elicited by the viral antigens.
  • Viral vectors can have higher transaction (ability to introduce genes) abilities than chemical or physical methods to introduce genes into cells.
  • viral vectors contain, nonstructural early genes, structural late genes, an RNA polymerase III transcript, inverted terminal repeats necessary for replication and encapsulation, and promoters to control the transcription and replication of the viral genome.
  • viruses When engineered as vectors, viruses typically have one or more of the early genes removed and a gene or gene/promotor cassette is inserted into the viral genome in place of the removed viral DNA.
  • the polynucleotide disclosed herein is contained in an adeno-associated virus (AAV) vector.
  • AAV adeno-associated virus
  • This defective parvovirus is a preferred vector because it can infect many cell types and is nonpathogenic to humans.
  • AAV type vectors can transport about 4 to 5 kb and wild type AAV is known to stably insert into chromosome 19. Vectors which contain this site-specific integration property are preferred.
  • the AAV vector can further comprise the herpes simplex virus thymidine kinase gene, HSV-tk, and/or a marker gene, such as the gene encoding the green fluorescent protein, GFP.
  • the AAV contains a pair of inverted terminal repeats (ITRs) which flank at least one cassette containing a promoter which directs cell-specific expression operably linked to a heterologous gene.
  • ITRs inverted terminal repeats
  • Heterologous refers to any nucleotide sequence or gene which is not native to the AAV or B19 parvovirus.
  • the AAV and B 19 coding regions have been deleted, resulting in a safe, noncytotoxic vector.
  • the AAV ITRs, or modifications thereof confer infectivity and site-specific integration, but not cytotoxicity, and the promoter directs cell-specific expression.
  • US Patent No. 6,261,834 is herein incorporated by reference for material related to the AAV vector.
  • an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide.
  • an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide.
  • an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide.
  • an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide.
  • the disclosed vectors thus provide DNA molecules which are capable of integration into a mammalian chromosome without substantial toxicity.
  • the inserted genes in viral and retroviral can contain promoters, and/or enhancers to help control the expression of the desired gene product.
  • the AAV used herein can be an AAV serotype AAV-5, AAV-6, AAV-8 or AAV-9; a rhesus-derived AAV, or the rhesus-derived AAV AAVrh.l0hCLN2; an organ-tropic AAV, or a neurotropic AAV; and/or an AAV capsid mutant or AAV hybrid serotype.
  • the AAV is engineered to increase efficiency in targeting a specific cell type that is non-permissive to a wild type (wt) AAV and/or to improve efficacy in infecting only a cell type of interest.
  • AAV adeno-associated virus
  • serotypes AAV-8, AAV-9, AAV-DJ or AAV-DJ/8TM are used to deliver the polynucleotide disclosed herein for expression in the CNS.
  • the composition disclosed herein is contained in or conjugated to a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier can be used to deliver the compositions to a CNS or a brain in vivo.
  • the carrier can be the ones described in U.S. Patent Publication No. 20060083737, incorporated by reference herein in its entirety.
  • the pharmaceutically acceptable carrier can be used to deliver the compositions to lymphatic endothelial cells.
  • composition disclosed herein is conjugated to docosahexaenoic acid (DHA).
  • DHA docosahexaenoic acid
  • Liposomes can be made using any method, e.g., as described in Park, et al., U.S. Application Publication No. 20070042031, including method of producing a liposome by encapsulating a composition disclosed herein, the method comprising providing an aqueous solution in a first reservoir; providing an organic lipid solution in a second reservoir, and then mixing the aqueous solution with the organic lipid solution in a first mixing region to produce a liposome solution, where the organic lipid solution mixes with the aqueous solution to substantially instantaneously produce a liposome encapsulating the active agent; and immediately then mixing the liposome solution with a buffer solution to produce a diluted liposome solution.
  • 109 is e.g., as described in Park, et al., U.S. Application Publication No. 20070042031, including method of producing a liposome by encapsulating a composition disclosed herein, the method comprising providing an aqueous solution in a first reservoir
  • the nanoparticle used herein can be any nanoparticle useful for the delivery of nucleic acids and/or polypeptides.
  • nanoparticle refers to a particle or structure which is biocompatible with and sufficiently resistant to chemical and/or physical destruction by the environment of such use so that a sufficient number of the nanoparticles remain substantially intact after delivery to the site of application or treatment and whose size is in the nanometer range.
  • the nanoparticle comprises a lipid-like nanoparticle. See, for example, WO/2017/187531A1, WO/2017/ 176974, WO/2019/027999, or Li, B et al. An Orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo.
  • the nanoparticle is a porous silica nanoparticle (pSi).
  • the nanoparticle comprises poly (lactide-co-glycolide) (PLGA).
  • Porous silica nanoparticles are well known in the art. See, for example, US Patent No. 10,143,660; US Application Publication No. 2013/0216807; International Publication No. 2013/056132; which are incorporated herein by reference in their entireties.
  • Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers.
  • a contemplated nanoparticle may include about 10 to about 99 weight percent of a one or more block co-polymers that include a biodegradable polymer and polyethylene glycol, and about 0 to about 50 weight percent of a biodegradable homopolymer.
  • Polymers can include, for example, both biostable and biodegradable polymers, such as microcrystalline cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyalkylene oxides such as polyethylene oxide (PEG), polyanhydrides, poly(ester anhydrides), polyhydroxy acids such as polylactide (PLA), polyglycolide (PGA), poly(lactide-co-glycolide) (PLGA), poly-3 -hydroxybutyrate (PHB) and copolymers thereof, poly-4-hydroxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof, and combinations thereof.
  • biostable and biodegradable polymers such as microcrystalline cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyalkylene oxides such as polyethylene oxide (PEG), polyanhydrides, poly(ester anhydrides), polyhydroxy acids such as polylactide (PLA), polyglycolide (PGA), poly(lactide
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle.
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
  • the nanoparticle has a diameter from about 1 nm to about 1000 nm.
  • the nanoparticle has a diameter less than, for example, about 1000 nm, about 950 nm, about 900 nm, about 850 nm, about 800 nm, about 750 nm, about 700 nm, about 650 nm, about 600 nm, about 550 nm, about 500 nm, about 450 nm, about 400 nm, about 350 nm, about 300 nm, about 290 nm, about 280 nm, about 270 nm, about 260 nm , about 250 nm, about 240 nm, about 230 nm, about 220 nm, about 210 nm, about 200 nm, about 190 nm, about 180 nm, about 170 nm, about 160 nm, about 150 nm, about 140 nm, about 130 nm, about 120 nm, about 110 nm, about 100 nm, about 90 nm, about 80
  • the nanoparticle has a diameter, for example, from about 20 nm to about 1000 nm, from about 20 nm to about 800 nm, from about 20 nm to about 700 nm, from about 30 nm to about 600 nm, from about 30 nm to about 500 nm, from about 40 nm to about 400 nm, from about 40 nm to about 300 nm, from about 40 nm to about 250 nm, from about 50 nm to about 250 nm, from about 50 nm to about 200 nm, from about 50 nm to about 150 nm, from about 60 nm to about 150 nm, from about 70 nm to about 150 nm, from about 80 nm to about 150 nm, from about 90 nm to about 150 nm, from about 100 nm to about 150 nm, from about 110 nm to about 150 nm, from about 120 nm to about 150 nm, from about 90 nm to about
  • the nanoparticle has a pore size from about 0. 1 nm to about 50 nm (including, for example, about 0. 1 nm, about 0.2 nm, about 0.3 nm, about 0.4 nm, about 0.5 nm, about 0.6 nm, about 0.7 nm, about 0.8 nm, about 0.9 nm, about 1.0 nm, about 1.2 nm, about 1.4 nm, about 1.6 nm, about 1.8 nm, about 2.0 nm, about 2.2 nm, about 2.4 nm, about 2.6 nm, about 2.8 nm, about 3.0 nm, about 3.5 nm, about 4.0 nm, about 4.5 nm, about 5 nm, about 5.5 nm, about 6 nm, about 6.5 nm, about 7 nm, about 7.5 nm, about 8 nm, about 8.5 nm, about 9 nm,
  • nanoparticles or liposomes may be able to efficiently bind to or otherwise associate with a biological entity, for example, a particular membrane component or cell surface receptor on a target cell (e.g., a receptor that facilitates delivery into the CNS or brain, or a receptor on lymphatic endothelial cells).
  • a biological entity for example, a particular membrane component or cell surface receptor on a target cell (e.g., a receptor that facilitates delivery into the CNS or brain, or a receptor on lymphatic endothelial cells).
  • the disclosed nanoparticles or liposomes may be engineered to a ligand binding to a receptor ubiquitously expression on a cell of CNS or brain (e.g., a ligand binding to low density lipoprotein receptor-related protein (LPR)- 1 and/or LPR-2) or binding to receptor or markers expressed on lymphatic endothelial cells, such as CD34, Proxl, podoplanin, LYVE-1, and/or VEGFR-3.
  • LPR low density lipoprotein receptor-related protein
  • a method of increasing, enhancing, improving, and/or stimulating lymphangiogenesis in subject comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor.
  • the extent of effect of increasing, enhancing, improving, and/or stimulating lymphangiogenesis is relative to a control (e.g., a healthy subject or a subject not being administered with the composition).
  • a method of preventing, inhibiting, mitigating, and/or treating a neurodegenerative disease in a subject comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • AIBP APOA1 binding protein
  • CAV-1 caveolin-1
  • the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor.
  • AIBP APOA1 binding protein
  • the extent of effect of preventing, inhibiting, mitigating, and/or treating a neurodegenerative disease is relative to a control (e.g., a subject not being administered with the composition).
  • the polynucleotide disclosed herein can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector).
  • the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
  • the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
  • the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR-Casl3.
  • the composition disclosed herein further comprises a stimulator for VEGFR3.
  • the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)).
  • the stimulator comprises Pioglitazone.
  • VEGFR3 stimulator for examples, VEGFC
  • VEGFC can improve cognition in aged subjects, it also elicits deleterious consequence such as vascular leakage and angiogenesis.
  • AIBP protect against vascular leakage caused by VEGFR3 activation.
  • the composition disclosed herein that comprises i) a stimulator for VEGFR3 and ii) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide.
  • the compositions disclosed herein that comprises a stimulator for VEGFR3 and a CAV-1 inhibitor.
  • the CAV-1 inhibitor is a CSD peptide.
  • composition disclosed herein can be contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
  • the composition is contained in a nanoparticle.
  • the composition is conjugated to docosahexaenoic acid (DHA).
  • DHA docosahexaenoic acid
  • the nanoparticle is a porous silica nanoparticle (pSi).
  • the nanoparticle comprises poly(lactide-co-glycolide) (PLGA).
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle.
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
  • neurodegenerative disease refers to a varied assortment of central nervous system disorders characterized by gradual and progressive loss of neural tissue and/or neural tissue function.
  • a neurodegenerative disease is a class of neurological disorder or disease, and where the neurological disease is characterized by a gradual and progressive loss of neural tissue, and/or altered neurological function, typically reduced neurological function as a result of a gradual and progressive loss of neural tissue.
  • neurodegenerative diseases include for example, but are not limited to, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease, Amyotrophic Lateral Sclerosis (ALS, also termed Lou Gehrig's disease) and Multiple Sclerosis (MS), polyglutamine expansion disorders (e.g., HD, dentatorubropallidoluysian atrophy, Kennedy's disease (also referred to as spinobulbar muscular atrophy), spinocerebellar ataxia (e.g., type 1, type 2, type 3 (also referred to as Machado- Joseph disease), type 6, type 7, and type 17)), other trinucleotide repeat expansion disorders (e.g., fragile X syndrome, fragile XE mental retardation, Friedreich's ataxia, myotonic dystrophy, spinocerebellar ataxia type 8, and spinocerebellar ataxia type 12), Alexander disease, Alper's disease, ataxia telangiectasia, Batten disease (also referred to as Spielqu
  • Alzheimer's Disease refers to all form of dementia, identified as a degenerative and terminal cognitive disorder.
  • the disease may be static, the result of a unique global brain injury, or progressive, resulting in long-term decline in cognitive function due to damage or disease in the body beyond what might be expected from normal aging.
  • the betaamyloid protein, or Ap involved in Alzheimer's has several different molecular forms that collect between neurons. It is formed from the breakdown of a larger protein, called amyloid precursor protein. One form, beta-amyloid 42, is thought to be especially toxic. An abnormal level of this protein is found in the Alzheimer’s brain, wherein the protein clump together to form plaques between neurons, leading to neuron function disruption.
  • the method disclosed herein can treat, decrease, mitigate, and/or prevent Alzheimer’s disease and/or a symptom thereof (e.g., A accumulation, an increase in levels of Tau protein, CNS inflammation, decline in cognitive function, and/or loss of memory). It should be understood and herein contemplated that the extent of effect of treating, decreasing, mitigating, and/or preventing Alzheimer’s disease and/or a symptom thereof is relative to a control (e.g., a subject not being administered with the composition).
  • a control e.g., a subject not being administered with the composition.
  • the timing of a neurodegenerative disease onset can often not be predicted.
  • the disclosed methods of treating, preventing, reducing, and/or inhibiting a neurodegenerative disease can be used prior to or following the onset of a neurodegenerative disease.
  • the disclosed methods can be employed 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 years, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 months, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18,
  • Alzheimer’s disease can be diagnosed by physical and neurological exam (e.g., assessment of overall neurological health by testing reflexes, muscle tone and strength, ability to get up from a chair and walk across the room, sense of sight and hearing, coordination, and/or balance), mental status and neuropsychological testing, and/or brain image (e.g., MRI or CT scan).
  • a method for treating a neurodegenerative disease comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of the composition disclosed herein.
  • Also disclosed herein is a method for treating a neurodegenerative disease, comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor.
  • CAV-1 caveolin-1
  • compositions described herein may be in any appropriate dosage form.
  • the dosage forms can be adapted for administration by any appropriate route.
  • Appropriate routes include, but are not limited to, oral (including buccal or sublingual), rectal, epidural, intracranial, intraocular, inhaled, intranasal, topical (including buccal, sublingual, or transdermal), vaginal, intraurethral, parenteral, intracranial, subcutaneous, intramuscular, intravenous, intraperitoneal, intradermal, intraosseous, intracardiac, intraarticular, intravenous, intrathecal, intravitreal, intracerebral, gingival, subgingival, intracerebroventricular, and intradermal.
  • Such formulations may be prepared by any method known in the art.
  • the composition is contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
  • disclosed herein is a method of treating a neurodegenerative disease, comprising administering to a subject a composition that is capable of crossing bloodbrain barrier, wherein the composition comprises the composition disclosed herein.
  • a method of treating a neurodegenerative disease comprising administering to a subject a composition that is capable of crossing bloodbrain barrier, wherein the composition comprises a caveolin-1 (CAV-1) inhibitor.
  • CAV-1 caveolin-1
  • the pharmaceutically acceptable carrier comprises a nanoparticle or docosahexaenoic acid (DHA).
  • DHA docosahexaenoic acid
  • the disclosed nanoparticles or liposomes may be engineered to a ligand binding to a receptor expressed ubiquitously expression on a cell of CNS or brain (e.g., a ligand binding to low density lipoprotein receptor-related protein (LPR)-1 and/or LPR-2).
  • LPR low density lipoprotein receptor-related protein
  • lymphangiogenesis involves the formation of new lymphatic vessels from pre-existing lymphatics; this occurs during embryonic development, wound healing and in various pathological contexts, including cancer.
  • Therapeutic lymphangiogenesis can be therapy strategy for the treatment of lymphedema.
  • Lymphedema refers to localized swelling of the body (such as at arms and legs) caused by an abnormal accumulation of lymph. Lymphedema includes primary and secondary lymphedema.
  • Primary lymphedema is an inherited condition caused by problems with the development of lymph vessels in the body. Specific causes of primary lymphedema include: Milroy's disease (congenital lymphedema).
  • Secondary lymphedema means a lymphedema caused by inflammatory or neoplastic obstruction of lymphatic vessels, and includes without limitation accumulation of ascites fluid due to peritoneal carcinomatosis or edema of the arm or other limbs following surgery or radiotherapy for breast cancer and other tumor types. Secondary lymphedema may also result from a trauma, a crush injury, hip or knee surgery, amputations, blood clots, vein grafts from cardiac surgery, chronic infections, or longstanding circulatory problems such as chronic venous insufficiency or diabetes.
  • a method of preventing, inhibiting, mitigating, and/or treating lymphedema in a subject comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
  • the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor.
  • the method disclosed herein can treat, decrease, mitigate, and/or prevent lymphedema and/or a symptom thereof (e.g., swelling of part or all of the arm or leg, including fingers or toes, restricted range of motion, and/or hardening and thickening of the skin (fibrosis)).
  • lymphedema and/or a symptom thereof e.g., swelling of part or all of the arm or leg, including fingers or toes, restricted range of motion, and/or hardening and thickening of the skin (fibrosis)
  • a control e.g., a subject not being administered with the composition.
  • the polynucleotide disclosed herein can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector).
  • the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
  • the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
  • the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR-Casl3.
  • the composition disclosed herein further comprises a stimulator for VEGFR3.
  • the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)).
  • the stimulator comprises Pioglitazone.
  • VEGFR3 stimulator for examples, VEGFC
  • VEGFC can improve cognition in aged subjects, it also elicits deleterious consequence such as vascular leakage and angiogenesis.
  • AIBP can protect against vascular leakage caused by VEGFR3 stimulatory.
  • compositions disclosed herein that comprises a stimulator for VEGFR3 and an APO Al binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide.
  • composition disclosed herein that comprises i) a stimulator for VEGFR3 and ii) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide in some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide.
  • the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide.
  • the compositions disclosed herein that comprises a stimulator for VEGFR3 and a CAV-1 inhibitor.
  • the CAV-1 inhibitor is a CSD peptide.
  • lymphedema onset can often not be predicted.
  • the disclosed methods of treating, preventing, reducing, and/or inhibiting lymphedema can be used prior to or following the onset of lymphedema.
  • the disclosed methods can be employed 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 years, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 months, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3 days, 60, 48, 36, 30, 24, 18, 15, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 hour prior to onset of lymphedema; or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 hours, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 45, 60, 90 days, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 or more years after onset of lymphedema.
  • Lymphedema can be diagnosed by MRI scan, CT scan that can reveal blockages in the lymphatic system, Doppler ultrasound (for checking blood flow and pressure by bouncing high-frequency sound waves (ultrasound) off red blood cells), and/or radionuclide imaging of the lymphatic system.
  • Also disclosed herein is a method for treating lymphedema, comprising diagnosing a subject as having lymphedema; and administering to the subject a therapeutically effective amount of the composition disclosed herein.
  • Also disclosed herein is a method for treating lymphedema, comprising diagnosing a subject as having lymphedema; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor.
  • CAV-1 caveolin-1
  • the composition is contained in or conjugated to a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier comprises a nanoparticle or a liposome.
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle.
  • the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
  • the disclosed nanoparticles or liposomes may be engineered to a ligand binding to receptor or markers expressed on lymphatic endothelial cells, such as CD34, Proxl, podoplanin, LYVE-1, and/or VEGFR-3.
  • TD is the first functional lymphatic vessel formed in the zebrafish trunk, situated between the dorsal aorta (DA) and PCV.
  • the PL string was normally formed in control embryos, but was completely abolished in -82% of apoalbp2 ⁇ / ⁇ animals and formed in a few segments (0-30%) in -8% of animals.
  • Similar developmental defects of TD and PLs were observed by morpholino antisense oligo (MO)- mediated apoalbp2 knockdown, which were restored to a large extent by co-injection of apocilbp2 mRNA (FIGS. 9A-9C).
  • Impaired TD formation can also be found in apoalbp2fi lyvel.Dsred zebrafish (FIGS. 9D-9E), which express DsRed in the lymphatic and venous ECs.
  • AIBP enhances LEC lineage specification in zebrafish and in the mouse embryonic stem cells (mESCs) to LEC differentiation model.
  • the defects in PL formation encouraged us to explore whether Aibp2 has any effect on lymphatic progenitor specification by immunostaining of Proxl at 36 hpf.
  • Proxl is the master transcription factor that dictates LEC fate specification, and is used as a readout of LEC identity.
  • - 3 of Proxl- positive LEC progenitors per 6 segments were found in the PCV.
  • Aibp2 knockout markedly reduced the number of lymphatic precursors to -1 (FIGS. 2A and 2B), indicating that Aibp2 regulates the development of lymphatic progenitors.
  • lymphatic vessel formation was also analyzed in embryos injected with apoalbp2 MO at 36 hpf. Reduced numbers of LEC progenitors found in Aibp2 morphants corroborates the Aibp2 effect on lymphatic progenitor specification (FIGS. 10A and 10B).
  • mEBs Mouse embryoid bodies prepared from mESCs can be differentiated into the derivatives of ectodermal, mesodermal, and endodermal tissues and recapitulate certain developmental processes; the LECs emerge from the mesoderm. mEBs were prepared and LEC generation was assessed in the differentiation medium containing BMP4 and bFGF followed by additional supplement on day 3 through day 7 with recombinant VEGFA and VEGFC in combination or with AIBP alone (FIG. 2C).
  • LECs were identified by the expression of PECAM, LYVE1 and PROXL Compared with control cells, VEGFA and VEGFC co-treatment increased both the mRNA and protein expression levels of these LEC-associated markers, LYVE1 and PROXI, at day 5 and day 7 of differentiation (FIGS. 2D-2G).
  • AIBP incubation strikingly increased LEC progenitor specification as evidenced by robust expression of the LEC markers (FIGS. 2D-2G), comparable to the effect of VEGFA/C co-administration.
  • a greater percentage of PECAM + /LYVE1 + LEC population was detected by FACS analysis at day 7 of LEC differentiation in the presence of AIBP or VEGFA/C (FIGS. 11A and 1 IB).
  • Aibp2-mediated cholesterol efflux is essential for proper lymphangiogenesis .
  • free cholesterol levels in apoalbp2 ⁇ / ⁇ embryos were first measured. Indeed, Aibp2 depletion significantly increases free cholesterol content in apoalbp2 ⁇ / ⁇ embryos (FIG. 3A).
  • apoalbp2 ⁇ / ⁇ animals were treated with atorvastatin, a cholesterol-lowering drug, and assessed the attendant effect on lymphangiogenesis. It was found that -93% of apoalbp2 ⁇ / ⁇ embryos displayed ⁇ 50% TD formation, and within this population, -80% of the embryos lacked the TD.
  • atorvastatin treatment substantially reduced the percentage of apoalbp2 ⁇ / ⁇ mutants with incomplete TD development, showing that atorvastatin-mediated reduction of cell cholesterol content rescues lymphatic vessel formation (FIGS. 3B and 3C).
  • APOA1 was overexpressed, the major protein constituent of HDL, in apoalbp2 knockout zebrafish.
  • CAV-1 a structural protein organizing the formation of caveolae, binds cholesterol directly, thereby forming cholesterol-rich microdomains, which provide a platform to facilitate membrane-anchored receptor signaling.
  • CAV-1 ablation in mice eliminates caveolae. Both the expression of CAV-1 and the formation of caveolae are dependent on cellular cholesterol levels. In many cases, CAV-1 deficiency alters plasma membrane receptor signaling competence through disruption of caveolae.
  • VEGFR3 signaling is required for the maintenance of the LEC progenitor fate, as well as LEC proliferation and migration.
  • CAV-1 in caveolae was reported to repress VEGFR3 activity in the ECs.
  • Next experiment was performed to assess that AIBP-mediated cholesterol efflux regulates lymphangiogenesis through CAV-1 -dependent VEGFC/VEGFR3 signaling. As shown in FIGS.
  • methyl beta cyclodextrin (MpCD)-mediated cholesterol depletion profoundly increased VEGFC-induced VEGFR3 activation, indicating that caveolae abundance and associated CAV-1 bioavailability inhibit VEGFR3 signaling.
  • MpCD methyl beta cyclodextrin
  • VEGFR3 was present in the caveolar fractions that were positive for CAV-1 (FIG. 4C).
  • AIBP and HDLs treatment in concert which disrupts caveolae, decreased CAV-1 localization in the caveolar fraction and induced VEGFR3 redistribution from the caveolar to the non-caveolar domains (FIG. 4C).
  • hLECs were treated with AIBP, HD L , or their combination, followed by stimulating cells with VEGFC.
  • AIBP or HDLa alone did not significantly affect VEGFC-induced activation of AKT and ERK, the two downstream effector kinases.
  • AIBP and HDLa in combination similarly to the treatment with MpCD (FIG.
  • VEGFC-induced VEGFR3 activation As shown by markedly increased phosphorylation of AKT and ERK (FIGS. 4D-4F). These results show that AIBP-mediated cholesterol efflux disrupts plasma membrane caveolae, thereby facilitating lymphangiogenesis by augmenting VEGFR3 activation.
  • Cav- 1 is a core component of caveolae, it is also a scaffolding protein that provides a spatially restricted platform for proper signaling of cell surface receptors.
  • Cav-1 contains a signaling motif that interacts with a variety of membrane receptors and modulates their activities.
  • VEGFR3 contains such a conserved motif (FIG. 5A), showing regulation of its signaling by CAV-1.
  • a VEGFR3 mutant VEGFR3 AAA that lacks the three conserved amino acids in the hypothetical CAV-1 binding domain was generated.
  • EGFP, VEGFR3, or VEGFR3 AAA was overexpressed in hLECs using lentivirus-mediated gene transfer and examined Cav-1/VEGFR3 interaction. As illustrated in FIG. 5B, wild-type VEGFR3 but not VEGFR3 AAA binds CAV-1 in hLECs.
  • VEGFR3 AAA mutant was ectopically expressed in hLECs using lentivirus-mediated transfection, followed by stimulation with VEGFC, and assessed VEGFR3 activation.
  • the results show that the triple Ala point mutations, which disrupt its interaction with CAV-1 (FIG. 5B), results in potent VEGFC-induced VEGFR3 phosphorylation as well as AKT and ERK activation compared to wild-type VEGFR3 (FIGS. 5C-5F).
  • Cciv-1 regulates LEC progenitor development.
  • cav-l ⁇ / ⁇ mutant zebrafish were generated (FIGS. 12A-12C), in which Western blot analysis revealed the absence of Cav-1 protein (FIG. 12D).
  • FIG. 12E Western blot analysis revealed the absence of Cav-1 protein
  • FIG. 12E Cav-1 knockout zebrafish showed greater numbers of Proxl labeled LEC progenitors at 36 hpf (FIGS. 6A and 6B). However, ID formation appeared normal in these animals (FIGS. 6C and 6D).
  • Cav- 1 depletion or overexpression resulted in profoundly increased or reduced expression of lymphatics-associated genes at 96 hpf, respectively (FIGS. 13A and 13B).
  • FIGS. 13A and 13B show that Cav-1 suppresses lymphatic progenitor development.
  • VEGFC-containing pellets were implanted, in the presence or absence of AIBP and the core protein component of HDL - APO Al or the CAV-1 blocking peptide (CAV-1 scaffolding domain, CSD) --- into the corneas of B6 mice.
  • APOA1 was used due to technical difficulties in the preparation of HDL pellets.
  • VEGFC pellet implantation elicited robust lymphangiogenesis (FIG. 7B).
  • co-administration with AIBP and cholesterol acceptor APOA1 further increased lymphangiogenesis (FIGS. 7B and 7C).
  • supplement of CSD augmented lymphatic vessel formation (FIGS. 7B and 7C).
  • AIBP expression was examined in human skin specimens derived from 7 subjects. Paired cutaneous normal and lymphedematous biopsy specimens were obtained from each of these individuals. Immunohistochemistry staining of AIBP showed that it is robustly expressed in the epidermis and the sweat glands (FIG. 8A). In human lymphedema, AIBP expression is mildly but significantly reduced in the epidermis but not in the dermis or sweat glands, when compared to the paired normal cutaneous specimens (FIGS. 8A-8D).
  • the comparable expression in the dermis and sweat gland excludes the possibility of tissue swelling elicited reduction of AIBP levels in the epidermis of lymphedema biopsy samples.
  • the observed abnormality in the lymphedematous epidermis indicates that reduced AIBP expression is associated with impairment of cutaneous lymphatic function.
  • lymphatic development in which AIBP-mediated cholesterol efflux disrupts caveolae and, consequently, reduces CAV-1 bioavailability, thereby mitigating the CAV-1 inhibition of VEGFR3 activation and augmenting lymphatic fate commitment and lymphangiogenesis.
  • AIBP-mediated cholesterol efflux disrupts lipid rafts/caveolae, which results in restricted angiogenesis by two mechanisms: 1) it impedes rafts/caveolae anchored proangiogenic VEGFR2 clustering, endocytosis and signaling, thereby restricting angiogenesis, and 2) it translocates y-secretase from lipid rafts/caveolae to the non-raft/caveolar domain, facilitating the cleavage of NOTCH receptor and augmenting anti -angiogenic NOTCH signaling.
  • AIBP instructs hematopoietic stem and progenitor cell specification in development through activation of SREBP2, which in turn transactivates NOTCH for hematopoiesis.
  • SREBP2 which in turn transactivates NOTCH for hematopoiesis.
  • AIBP increases lymphangiogenesis and instructs LEC progenitor specification.
  • AIBP-mediated cholesterol efflux attenuates the inhibitory effect of CAV-1 on VEGFR3 signaling.
  • disruption of CAV-1 and VEGFR3 binding augments VEGFR3 signaling and LEC progenitor development and lymphangiogenesis.
  • LECs are derived from blood ECs (BEC), their functions and molecular identifications differ. The fundamental differences between BECs and LECs contribute to the disparate effects of AIBP on angiogenesis and lymphangiogenesis.
  • RhoB ablation augments lymphangiogenesis but impairs angiogenesis in the cutaneous wound healing model.
  • Notch signaling limits angiogenesis, Notch has been reported to inhibit or augment lymphangiogenesis in a context-dependent fashion.
  • AIBP-mediated CAV-1 dependent regulation ofVEGFR3 signaling and lymphangiogenesis requires LEC fate commitment and subsequent sprouting.
  • the LEC fate acquisition, LEC sprouting from the embryonic CV, and dorsolateral migration under the control of VEGFC/VEGFR3/PROX1 signaling axis is highly conserved. Restricted expression of PROXI protein dictates LEC identity , which is critical for future LECs to migrate out of the PCV.
  • AIBP is evolutionarily conserved from zebrafish to human. Zebrafish have two apoalbp genes that encode Aibpl and Aibp2 proteins, and Aibp2 demonstrates functional properties resembling those of human AIBP.
  • a positive feedback loop between VEGFR3 and PROXI is necessary to maintain the identity of LEC progenitors in mice and zebrafish.
  • CAV-1 has been reported to inhibit VEGFR3 signaling in ECs.
  • the studies herein show that Cav-1 knockdown or overexpression in zebrafish increases or decreases expression of lymphatic genes, respectively.
  • loss of Cav-1 strongly enhances lymphatic progenitor specification and rescues impaired lymphangiogenesis.
  • the TD formation appears normal in Cav-1 null zebrafish (FIGS. 6C and 6D). Indeed, there are considerable variations in the number of LECs required to form a complete TD.
  • CAV-1 deficiency augments lymphangiogenesis in mouse development and in adult cornea with VEGFC stimulation (FIGS. 7A-7C and FIG. 14).
  • VEGFR3 is present in caveolae and contains a binding motif for CAV-1 (Fig. 5A).
  • the disruption of the CAV-1 and VEGFR3 interaction increases VEGFR3 activation in response to VEGFC treatment (FIGS. 5C-5F).
  • AIBP effect on ERK signaling ERK activation controls LEC fate specification and LEC sprouting.
  • Fig. 4D-F Fig. 4D-F
  • Aibp2 knockdown in zebrafish abrogates Erkl activation while robustly facilitating Akt activation.
  • VEGFR3 homodimer activates ERK while VEGFR2/VEGR3 heterodimer activates AKT.
  • lymphatic vessel formation is blunted at later developmental stages in zebrafish, and no significant difference was detected in tail lymphangiogenesis between AIBP knockout neonatal mice and control littermates, which can be due to the rescue effect of non-venous origin derived lymphangiogenesis.
  • the Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including tumor metastasis, cardiovascular disease, obesity, and diabetic mellitus.
  • Therapeutic augmentation of lymphangiogenesis has been documented to improve lymphatic structure and function.
  • the mechanistic study of AIBP-regulated lymphangiogenesis aids in the development of new therapies for the diseases associated with lymphatic dysfunction.
  • meningeal lymphatics are essential for transportation of brain interstitial fluid and cerebral fluid. Dysfunction of the meningeal lymphatics is associated with dementia, such as Alzheimer disease (AD) pathology. VEGFC-mediated enhancement of meningeal lymphatics function improves cognition in aged mice, and disruption of meningeal lymphatic vessels in transgenic AD mouse models aggravates P-amyloid
  • the CRISPR/Cas9-mediated gene knockout technique was performed as previously described. Briefly, the target sequence of caveolin 1 (Cav-1, NCBI Gene ID: 323695) was 5’- GACGTGATCGCCGAGCCTGC-3’ (SEQ ID NO: 8).
  • the guide-RNA (gRNA) sequence was subcloned into pT7-gRNA vector and gRNA was synthesized using HiScribeTM T7 Quick High Yield RNA Synthesis (New England Biolabs).
  • the pCS2-nCas9n was linearized using Notl and Cas9 mRNA was synthesized using mMESSAGE mMACHINE SP6 Kit (Invitrogen). Twenty- five ng/01 gRNA and 200 ng/pl Cas9 mRNA were mixed and injected into one-cell stage wild type embryos. The founder zebrafish were raised to adult. Following genotype verification using Sanger DNA sequencing, cavl +/ ⁇ animals were used for experiments.
  • VEGFC pellet preparation and implantation were carried out as previously described with minor modifications. Briefly, pellets containing recombinant VEGFC (5 mg/ml), AIBP (10 mg/ml) in combination with apoA-1 (15 mg/ml), CAV-1 scaffolding domain (CSD; 5 mg/ml), VEGFC with AIBP and APOA1 or with CSD were prepared by mixing with 10% sucralfate solution (w/v in PBS) and 12% poly-HEMA (w/v in ethanol).
  • the aforementioned components were mixed as following: 5 pl of poly-HEMA, 1 pl of sucralfate, 2 pl VEGFC, 2 pl AIBP, 2 pl APOA1, 2 pl CSD, and the final volume adjusted to 10 pl with PBS.
  • the mixture droplet was put on the PARAFILM, UV irradiated for 15 minutes, and air-dried in the cell culture hood. Dried pellets were used immediately or stored at 4 °C before usage.
  • the mouse was anesthetized with isoflurane, and one drop of 0.5% proparacaine HCI was applied to the cornea. Five minutes later, one mouse eye was properly orientated under a dissection microscope. A gentle cut was made with a von Graefe cataract knife from the center of the cornea, and a pocket with ⁇ 1.5-2 mm 2 size was generated by inserting the knife toward comeal limbus, with a few gentle waggles inside. The pellets were then inserted into the cornea pocket using forceps and von Graefe cataract knife, with the pellet flattened to secure the implantation. The ophthalmic antibiotic ointment was applied topically to the injured eye. Following 10 days of pellet implantation, the cornea was dissected and washed in PBS for later processing.
  • Tissue biopsy Cutaneous punch biopsy was performed as previously described. After informed consent was obtained, two contiguous 6 mm full thickness punch biopsy specimens were derived from the medial aspect of the forearm or calf of the affected extremity with an AcuPunch disposable device. Biopsy specimens were immediately place in formalin and processed for subsequent studies.
  • AIBP antiserum were generated in rabbits using tag-free recombinant human AIBP proteins.
  • AIBP antibody was subsequently purified from the AIBP antiserum using affinity purification, where AIBP antigen column was prepared by covalently conjugating recombinant AIBP to the aminereactive, beaded-agarose resin (Thermo Fisher, Pierce NHS-Activated Agarose, Cat # 26196).
  • the 5um human specimens were deparaffinized, and rehydrated serially through xylene, 100%, 80%, and 70% ethanol, and finally rinsed in milliQ water.
  • the resulting sections were subjected to antigen recovery by immersing in 100°C citrate buffer (pH6.0) for 10 minutes. An incubation with 3% hydrogen peroxide was applied to eliminate the endogenous peroxidase activity. Following rinsing in 1XTBST (diluted from 10X TBST wash buffer, Dako, Cat # S3006), the slides were further washed and blocked in protein block serum-free buffer (Dako, Cat # X0909). The slides were incubated with AIBP antibody for 1 hour at room temperature.
  • MO and mRNA injections Morpholino antisense oligos (MOs) were synthesized by Gene Tools. The following MOs were used in this study 1) control: 5’- CCTCTTACCTCAGTTACAATTTATA-3’ (SEQ ID NO: 14), and 2) apoalbp2 (NCBI Gene ID: 557840): 5’-GTGGTTCATCTTGATTTATTCGGC-3’ (SEQ ID NO: 15). The working concentrations of control and aopcilbp2 MOs were 0.2 mM and 0.1 mM, respectively.
  • Equal volume of aopcilbp2 MO (0.1 mM) and aopocilbp2 mRNA (200 ng/pl) were mixed to perform the rescue assays.
  • the working concentration of human APOA1 mRNA was 100 ng/pl.
  • One nl of MO, mRNA or the combination of MO with mRNA were injected into one-cell stage embryo using the microinjector FemtoJet® 4i (Eppendorf).
  • the hLECs were pretreated with 200 ng/ml AIBP protein, 100 pg/ml HDL3, or the combination of AIBP with HDL3 for 4 hours, washed with PBS, and followed by incubation with 100 ng/ml VEGFC (R&D, Cat # 9919) in pre-warmed serum-free ECM medium for 20 min.
  • Lenti-virus infection The hLECs were transduced with the third generation lentiviral system as previous described (81). After the cells were infected with human VEGFR3, VEGFR3AAA or CAV1 overexpression Lenti-virus for 96 hours, cells were subjected to 5 pg/ml puromycin (ThermoFisher, Cat #A1113803)) selection to generate pool cells stably expressing the transduced genes.
  • the antibodies used were: anti-Phospho-Akt (Ser473) (Cell Signaling, #4060S), anti-AKT (Cell Signaling, #4685 S), anti- Phospho-ERKl/2 (Cell Signaling, #4370S), anti-ERKl/2 (Cell Signaling, #9102S), rabbit anti- P-Tubulin (Cell Signaling, #2148S), anti-Phospho-tyrosine (clone 4G10, EMD Millipore, #05- 321), anti-Flt4 (Santa Cruz, C20, Cat No. sc321), anti-CAVl (Cell Signaling, Cat No.
  • the mESCs were cultured in feeder- and serum-free environment using ESGRO®-2i medium supplemented with leukemia inhibitory factor (LIF), GSK3P inhibitor and MEK1/2 inhibitor (EMD Millipore, Cat #. SF016- 100) according to the manufacturer’s instruction. Briefly, T25 flask was coated with 0.1% gelatin solution for 30 min at room temperature, and then 1 x 106 cells were plated onto gelatinized T25 flask in ESGRO®-2i medium and incubated at 37 °C with 5% CO2. The media were changed daily and mESCs were sub-cultured at a ratio of 1:5 when cells reached 60-90% confluence.
  • LIF leukemia inhibitory factor
  • GSK3P inhibitor GSK3P inhibitor
  • MEK1/2 inhibitor EMD Millipore, Cat #. SF016- 100
  • LEC differentiation medium ESGRO CompleteTM basal medium, Cat #. SCR002-500
  • ESGRO CompleteTM basal medium ESGRO CompleteTM basal medium, Cat #. SCR002-500
  • BMP4 10 ng/ml bFGF
  • 50 ng/ml VEGFA 50 ng/ml VEGFC
  • Differentiated cells were collected on days 3, 5 and 7 for qPCR, western blot, or FACS analysis.
  • FACS Fluorescence Activated Cell Sorting
  • VEGFR3 site-direct mutation VEGFR3 mutation was generated using a PCR- based direct site mutation strategy. Briefly, the plasmid containing wild type VEGFR3 was amplified using primer pair VEGFR3- AAA -F (5’- ACGCAGAGTGACGTGGCGTCCGCTGGGGTGCTTCTCGCGGAGATCTTCTCTCTGGGG GCC-3 ) (SEQ ID NO: 36) and VEGFR3- AAA -R (5’- AGAGAGAAGATCTCCGCGAGAAGCACCCCAGCGGACGCCACGTCACTCTGCGTGGT GTAC-3’) (SEQ ID NO: 37).
  • the PCR program was 95 °C for 2 min, 18 cycles of 95 °C for 30 s, 68 °C for 1 min per kilobase of plasmid length, then 68 °C for 7 min and hold at 4 °C.
  • One pl Dpnl was added to the PCR products to disrupt the plasmid template and incubated at 37 °C for
  • IF staining of mouse cornea and tail skin For IF staining, cornea and tail skin were fixed in 4% paraformaldehyde for 2 hr at room temperature. Subsequently, samples were washed 5 times with 0.05% tween/PBS (0.05% PBS-tw) and blocked with the blocking buffer (1% goat or donkey serum, 1% BSA, and 0.5% Triton X-100 in PBS) for 3-4 hr at room temperature. Samples were incubated with LYVE-1 antibody (1: 1000; Angiobio, Cat # 11-034) and CD31 antibody (1: 1000; BD Biosciences, Cat.
  • ROI region of interest
  • AD Alzheimer’s disease
  • meningeal lymphatics are essential for the transportation of brain interstitial fluid and cerebral fluid containing Ap aggregates. Indeed, VEGFC-mediated enhancement of meningeal lymphatics function improves cognition in aged mice. Conversely, disruption of meningeal lymphatic vessels in transgenic AD mouse models aggravates Ap accumulation. However, VEGFC can also elicit deleterious consequence such as vascular leakage.
  • AIBP APOA1 binding protein
  • AD Alzheimer’s disease
  • Meningeal lymphatics and AD The neuropathological hallmark of AD is the extracellular deposition of Ap aggregates and intracellular accumulation of dysfunctional tau, thereby precipitating neuronal dysfunction and behavioral changes.
  • the A protein was initially identified in the meningeal tissue of AD patients.
  • the brains are constantly perfused by cerebrospinal fluid (CSF) and brain interstitial fluid (ISF), which removes macromolecules, including Ap protein aggregates and extracellular dysfunctional tau.
  • CSF cerebrospinal fluid
  • ISF brain interstitial fluid
  • Recent studies show that the aging-associated Ap accumulation is attributed to the progressive attenuation of meningeal lymphatic function in the brain.
  • the meningeal lymphatics drains cerebral ISF/CSF from the central nerve system into the cervical lymph nodes in mice.
  • VEGFC vascular endothelial growth factor
  • AIBP improves VEGFC potency.
  • AIBP a secreted protein, by promoting cholesterol efflux from endothelial cell (EC), limits angiogenesis.
  • AIBP increases the binding sites of high-density lipoprotein (HDL) on ECs and promotes HDL-EC dissociation, a process that depletes the free cholesterol content of endothelial lipid rafts via the cholesterol transporter.
  • HDL high-density lipoprotein
  • AIBP acts on microglia and reduces neuroinflammation.
  • cholesterol deregulation precipitates neuronal Ap deposition
  • AIBP improves lipid metabolism and bestows multiple beneficial effects on AD.
  • the studies show that AIBP protects against vascular leakage, which makes AIBP a key modulator that increases the pro-lymphangiogenic potential of VEGFC but limits its effect on angiogenesis and vascular leakage.
  • AIBP also accelerates cellular cholesterol efflux, reduces neuroinflammation, and impairs y-secretase activity in lipid rafts, all of which further contribute to effective AD treatment.
  • AIBP-based therapy for AD introduces multi-pronged salutary effects to improve neurological function, thereby alleviating dementia.
  • AIBP-mediated cholesterol efflux promotes adult lymphangiogenesis in mice.
  • the biochemical analysis using human LECs shows that AIBP, by alleviating CAV-1 repression of VEGFR3 activation, facilitates lymphangiogenesis. It is further determined whether this mechanism controls lymphangiogenesis in mice.
  • the corneas of adult mice lack lymphatic vasculature, and is widely used as a model to study injury or growth factor-induced lymphangiogenesis ( Figure 17A, top left panel of next page).
  • VEGFC-containing pellets were implanted, in the presence or absence of AIBP or the CAV-1 modifying peptide (CAV-1 scaffolding domain, CSD) - into the corneas of B6 mice.
  • AIBP enhances differentiation of LECs from mouse embryonic stem cells (mESCs). Aibp dictates LEC fate and controls lymphangiogenesis in zebrafish. To determine the conserved role of AIBP in lymphatics, murine LEC specification was investigated.
  • the mouse embryoid bodies prepared from mESCs can be differentiated into the derivatives of ectodermal, mesodermal, and endodermal tissues and recapitulate certain developmental processes 19. The LECs emerge from the mesoderm.
  • Mouse embryoid bodies were prepared and LEC generation was assessed in the differentiation medium containing BMP4 & bFGF followed by additional supplement on day 3 through day 7 with recombinant VEGFA and VEGFC in combination or with AIBP alone (Figure 18A).
  • LECs were identified by the expression of PECAM, LYVE1 and PROXI.
  • VEGFA and VEGFC co-treatment increased both the mRNA and protein expression levels of these LEC associated markers LYVE1 and PROXI at day 5 and day 7 of differentiation.
  • AIBP incubation strikingly increased LEC specification as evidenced by robust expression of the LEC markers ( Figures 18B and 18C), comparable to the effect of VEGFA/C co-administration.
  • Nano-AIBP/VEGFC Preparation and characterization of Nano-AIBP/VEGFC. Generation of Nano-AIBP/VEGFC particles.
  • the nanoporous silicon particles (pSi) are fabricated with well- controlled shape, size, pore sizes and surface chemistries through a sol-gel method.
  • pSi poly(lactic- co -glycolic) acid (PLGA)
  • PLGA- pSi composite microspheres
  • 200 nm pSi with an average pore size of 2 nm is engineered, which can be loaded with endotoxin-low human VEGFC alone or AIBP+VEGFC.
  • PLGA (50:50) can be dissolved in dichloromethane (10 w/v %), and PLGA-pSi microspheres with an average diameter of 10 pm are prepared through a modified S/O/W emulsion method. AIBP-loaded particles are then lyophilized and encapsulated in Cy5 -labeled 5% PLGA (50:50) shell. The kinetics of AIBP/VEGFC release from the nanoparticles into cell culture medium and its stability are determined using Western blot.
  • Nano-AIBP/VEGFC shows a prolonged and sustainable release effect. The release kinetics can be adjusted to achieve an optimal effect of AIBP/VEGFC release by varying the polymeric compound of the nanoparticles. Nano-AIBP/VEGFC is more stable and biologically more potent than unpacked recombinant AIBP/VEGFC when placed at 31° C.
  • mice express humanized AD-prone APP in the relevant cells at physiological levels.
  • APP NL ' G ' F knock-in mice and their wild type littermates receive intracisternal injection of empty nanoparticle, Nano-VEGFC, Nano-AIBP/VEGFC, and recombinant AIBP/VEGFC. 12 mice are used for each treatment group for each sex. One month after administration, these mice are subjected to a battery of cognitive behavioral assays.
  • the cognitive tests include the novel object recognition (NOR), Y- maze, Morris water maze (MWM), radial arm water maze (RAWM), and fear conditioning. Following these assays, mice are euthanized and perfused with 10% Formalin. Brains are collected and the brain sections subjected to immunostaining for VEGFC, AIBP and Ap. The dura mater containing meningeal lymphatics is isolated and immunostained using LYVE1 and VEGFR3 antibodies. Images are taken using a confocal microscope and analyzed using Image). Recombinant AIBP (Sinobiological) and VEGFC (R&D) are used.
  • NOR novel object recognition
  • MLM Morris water maze
  • RAWM radial arm water maze
  • VEGFC overexpression can improve cognitive function in APP NL ' G ' F mice. More importantly, the combined AIBP and VEGFC can further improve cognitive function in APPNL-G-F mice Such findings prove the principle that AIBP can improve cognition. In addition, the improved cognition is associated with augmented lymphatic function (as assessed by LYVE1 and VEGFR3 immunostaining) and attenuated A accumulation. These results further show that AIBP-elicited benefits on cognition are mediated by improved lymphatic functions and therefore better drainage of brain Ap.
  • Cognitive function is examined one month following VEGFC and AIBP administration, as used in the prior studies. Chances are that the Nano-AIBP/VEGFC combination can confer a protective effect at a different time point. Thus, a longitudinal analysis of cognitive function is performed, which enables to detect the optimal time window for the treatment. In addition, for each mouse study, post hoc histology is performed to validate VEGFC and/or AIBP overexpression.
  • This study tests the translational potential of AIBP/VEGFC co-delivery for the treatment of AD.
  • the study shows: 1): AIBP/VEGFC improves dementia in a preclinical mouse model compared to VEGFC alone 2) the mechanisms of how AIBP/VEGFC functions to ameliorate AD.
  • the following components are used:
  • nanoparticle-mediated protein delivery is to use nanoparticle-mediated AIBP and VEGFC mRNA delivery.
  • AAV adeno-associated virus
  • VEGFC mRNA delivery.
  • the impact of adeno-associated virus (AAV)-mediated AIBP/VEGFC expression on AD is assessed, which is the current standard platform of gene therapy.
  • the drug targeting the AIBP downstream CAV1 - CSD is tested (See Figure 17) that can achieve similar effect on lymphangiogenesis.
  • AIBP-mediated cholesterol efflux promotes lymphangiogenesis is revealed. Increasing lymphatic function using AIBP can improve AD associated symptom such as dementia.
  • the studies on AD require brain injection of viruses/proteins and mouse neurobehavioral tests, including those for cognitive functions.
  • SEQ ID NO: 1 AIBP protein sequence, human
  • SEQ ID NO: 2 (AIBP polynucleotide sequence)
  • SEQ ID NO: 3 (Amino acid sequence for VEGFA)
  • SEQ ID NO: 4 polynucleotide sequence for VEGFA
  • SEQ ID NO: 5 amino acid sequence for VEGFC
  • SEQ ID NO: 6 polynucleotide sequence for VEGFC
  • SEQ ID NO: 7 polypeptide sequence of CAV-1, Q2TNI1
  • SEQ ID NO: 8 target sequence of CAV-1 by CRISPR-Cas9
  • SEQ ID NO: 9 (siRNA sequence for CAV-1)
  • SEQ ID NO: 10 (siRNA sequence for CAV-1)
  • SEQ ID NO: 11 siRNA sequence for CAV-1
  • SEQ ID NO: 40 (DNA, synthetic)
  • SEQ ID NO: 41 (DNA, synthetic)
  • SEQ ID NO: 42 (DNA, synthetic)
  • SEQ ID NO: 43 protein, Danio rerio
  • SEQ ID NO: 44 protein, synthetic

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Toxicology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Neurosurgery (AREA)
  • Biotechnology (AREA)
  • Neurology (AREA)
  • Public Health (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Environmental Sciences (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Animal Husbandry (AREA)
  • Cell Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed are compositions and uses thereof for treating neurodegenerative diseases and/or lymphedema.

Description

COMPOSITIONS AND METHODS FOR INCREASING
LYMPHANGIOGENESIS
I. CROSS-REFERENCE TO RELATED APPLICATIONS
1. This application claims the benefit of U.S. Provisional Application No. 63/091,048, filed October 13, 2020, which is expressly incorporated herein by reference in its entirety.
IL STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
2. This invention was made with government support under grant number HL132155 awarded by the National Institutes of Health. The government has certain rights in the invention.
III. BACKGROUND
3. Lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, dietary lipid transport and immune surveillance. The Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including lymphedema, Alzheimer’s disease, tumor metastasis, cardiovascular disease, obesity, and diabetic mellitus. What are needed are new compositions and methods for increasing lymphangiogenesis and treating the related diseases and disorders.
IV. SUMMARY
4. Disclosed herein are compositions and uses thereof for improving lymphangiogenesis. In some aspects, disclosed herein is a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor. The polynucleotide can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector). In some embodiments, the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide. In some embodiments, the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody. In some embodiments, the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR- Casl3, or CRISPR-Casl3d that ablate either DNA or mRNA. 5. In some embodiments, the composition disclosed herein further comprises a stimulator for VEGFR3. In one example, the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)). In one example, the stimulator comprises Pioglitazone.
Figure imgf000003_0001
Pioglitazone
6. The composition disclosed herein can be contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier. In some embodiments, the composition is contained in a nanoparticle. In some embodiments, the composition is conjugated to docosahexaenoic acid (DHA).
7. Also disclosed is a method for treating a neurodegenerative disease in a subject, comprising administering to the subject a therapeutically effective amount of the composition disclosed herein, wherein the composition comprises i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor. The composition can be administered to the subject intrathecally or intracranially. In some embodiments, the neurodegenerative disease comprises Alzheimer’s disease, Parkinson's disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, or Multiple Sclerosis. In some embodiments, the composition increases lymphangiogenesis in brain.
8. Also disclosed is a method for treating a neurodegenerative disease, comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor disclose herein.
9. Also disclosed is a method for treating a neurodegenerative disease, comprising administering to a subject a composition that is capable of crossing blood-brain barrier, wherein the composition comprises a caveolin-1 (CAV-1) inhibitor.
10. Also disclosed herein is a method for treating lymphedema in a subject, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
V. BRIEF DESCRIPTION OF THE DRAWINGS
11. The accompanying drawings, which are incorporated in and constitute a part of this specification, illustrate several embodiments and together with the description illustrate the disclosed compositions and methods. 12. Figures 1A-1D show effect of Aibp2 knockout on lymphatic vessel development. Figure 1A shows lymphatic developmental defects in apoalbp^ zebrafish. Maxiprojection confocal images of TD formation in the flilcc.egfp and apocilbp2~/~,' flilcc.egfp null zebrafish at 5 dpf. Arrows show the TD, and stars denote absent TD. Figure IB shows quantitative data of TD formation in Figure 1A. n=15. Figure 1C shows loss of Aibp2 disrupts PL formation in flilcc.egfp zebrafish. Confocal images of PL in the horizontal myoseptum at 48 hpf. Arrows indicate PLs, and stars show absent PLs. Figure ID shows quantitative data of the PL string in Figure 1C. n=20. Scale: 25 pm.
13. Figures 2A-2G show AIBP effect on LEC lineage commitment. Figure 2A shows impaired LEC specification in apocilbp2~'f flilcc. egfp zebrafish. Control and cipocilbp2 knockout zebrafish were fixed with 4% PFA at 36 hpf, whole mount immunostaining was performed using Proxl and EGFP antibodies, and images were captured using confocal microscopy. Figure 2B shows enumeration of LEC progenitors in Figure 2A. Arrows show specified LEC progenitors. Figure 2C shows scheme illustration of mESC to LEC differentiation. Figures 2D to 2G show that AIBP augments LEC differentiation from mESCs. The embryoid bodies (EBs) of mESCs were prepared and cultured in the EC differentiation medium containing 2 ng/ml BMP4 and 10 ng/ml bFGF for 3 days. Recombinant VEGFA (50 ng/ml) and VEGFC (50 ng/ml) in combination or AIBP (100 ng/ml) alone were supplemented at day 3 and kept in culture for additional 4 days. The resulting cells were harvested for qPCR analysis of endothelial cell (EC) marker Pecaml expression (Figure 2D) and LEC-associated Lyvel expression (Figure 2E) at the indicated time points. Western blot analyses of PECAM, LYVE1 and PROXI expression (Figure 2F) and the quantitative data (Figure G). C: control; V: VEGFA + VEGFC; A: AIBP. *, p<0.05; **, p<0.00I; ****, p<0.000I. Scale: 25 pm.
14. Figures 3A-3E. Effect of cholesterol reduction on lymphatic vessel development. Figure 3A shows free cholesterol (FC) content in control and Aibp2 null zebrafish. The 5 dpf zebrafish trunk of control or Aibp2 null zebrafish were dissected, total lipids extracted, and free cholesterol levels measured. The FC content were normalized to the protein levels. Fifty embryos were pooled for each measurement. Mean±SE; *, p<0.05. Figure 3B shows that inhibition of cholesterol synthesis rescues TD formation in Aibp2 null animals. TD formation in control zebrafish, Aibp2 null zebrafish treated with 1 pM atorvastatin or control vehicle ethanol at 5 dpf. Figure 3C shows quantitative data of TD formation in B. n=20. Figure 3D shows that APOA1 -mediated cholesterol efflux restores TD formation. Confocal images of TD formation in control zebrafish, Aibp2 null zebrafish, or Aibp2 knockout zebrafish with 100 ng human APOA1 mRNA overexpression. Figure 3E shows quantitative data of TD formation in Figure 3D. Arrows show TD, and stars denote absent TD. n=18. Scale: 25 pm. *, p<0.05.
15. Figures 4A-4F show effect of cholesterol efflux on VEGFR3 signaling. Figures 4A and 4B show cholesterol removal potentiates VEGFR3 signaling. Figure 4A shows that hLECs were serum-starved, and treated with 10 mM MpCD for 30 min, and cells were further stimulated with 100 ng/ml VEGFC. The resulting cells were lysed and blotted with Cav-1 or GAPDH antibodies. Figure 4B shows that hLECs were treated as in Figure 4A and cell lysates were immunoprecipitated using VEGFR3 antibody. Immunoblotting was performed using anti- phosphotyrosine (4G10) and VEGFR3 antibodies. Figure 4C shows that AIBP-mediated cholesterol efflux disrupts caveolae and reduces CAV-1 levels in the caveolar fractions. hLECs were treated with recombinant 200 ng/ml AIBP, 100 pg/ml HDL3, or both in serum-free EBM2 for 6 hours, and the cells were subjected to sucrose-mediated ultracentrifugation. The resulting fractions were collected for Western blot analysis as indicated. Figure 4D shows that AIBP- mediated cholesterol efflux increases VEGFR3 signaling. hLECs were serum-starved and treated as in Figure 4C, and further stimulated with 100 ng/ml VEGFC. The resulting cells were lyzed and immunoblotted as indicated. Figures 4E and 4F show quantitative data of AKT activation (Figure 4E) and ERK activation (Figure 4F). Mean±SD, n=3 independent repeats. *, p<0.05; **, p<0.0I; ns: not significant.
16. Figures 5A-5F show effect of CAV-1 on VEGFR3 signaling. Figure 5A shows conserved CAV-1 binding site on VEGFR3 in human (Hu), mouse (Ms), and zebrafish (Zf). Figure 5B shows that VEGFR3AAA loses its binding to CAV-1. hLECs were transfected with control EGFP (Ctrl), VEGFR3-EGFP (R3), or VEGFR3AAA-EGFP (RS^) using lentivirus- mediated gene transfer. After 72 hours, the resulting cells were lyzed and immunoprecipitated with EGFP antibody coupled to the magnetic Dynabeads and immunoblotted using VEGFR3 and CAV-1 antibodies. Immunoblotting of overexpressed VEGFR3-EGFP and VEGFR3AAA- EGFP (R3/R3AAA-EGFP) were detected using VEGFR3 antibody. The input lysates were shown on the right. Figure 5C shows that VEGFR3AAA increases VEGFR3 signaling. hLECs were transduced as in Figure 5B, and the resulting cells were serum starved and treated with 100 ng/ml VEGFC for 20 min, cells were then lysed and immunoblotted as indicated. Quantitative data of VEGFR3 activation (Figure 5D), AKT activation (Figure 5E), and ERK activation (Figure 5F) were shown. n=3 independent repeats. *, p<0.05; **, p<0.0I; ***, p<0.001. The sequences in Figure 5 include DVWSFGVLLWEIFSL (SEQ ID NO: 38) and DVWSYGVTVWELMTF (SEQ ID NO: 39). 17. Figures 6A-6D show effect of CAV-1 on Aibp2-regulated LEC specification. Figure 6A shows that Cav-1 deficiency rescues LEC specification in Aibp2 knockdown animals. Max projection confocal images of control, Aibp2-deficient, Cav-1 null, or Cav-1/Aibp2 double deficient zebrafish. The animals were PFA fixed at 36 hpf and immunostained using Proxl and EGFP antibodies. Arrows indicate LEC progenitors. Figure 6B shows quantitative data of LEC progenitors in (Figure 6A). Figure 6C shows that Cav-1 deficiency corrects lymphatic defects in Aibp2 knockdown animals. Confocal imaging of TD formation in the indicated genetically modified flilcc.egfp zebrafish at 5 dpf. Arrows show TD, and stars denote absent TD. Figure 6D shows quantitative data of TD formation in (Figure 6C). **, p<0.01; ****, p<0.0001; ns: not significant, n=20. Scale, Scale: 25 pm.
18. Figure 7A-7C show AIBP effect on comeal lymphangiogenesis and LEC specification. Figure 7A shows illustration of murine cornea lymphangiogenesis assay. Figure 7B shows representative images of murine comeal lymphangiogenesis by implantation of pellets containing the indicated recombinant proteins and immunostained using LYVE1 & CD31 antibodies. Enlarged images of the boxed regions (scale bar, 1000 pm) are shown in the lower panels (scale bar, 500 pm). Figure 7C shows quantification of LYVE1+ lymphatic vessel area per cornea. Ctrl: control; VC: VEGFC; AA: AIBP+APOA1; CSD: CAV-1 scaffolding domain peptide. **, p<0.01; ****, p<0.0001.
19. Figures 8A-8D show AIBP expression in cutaneous lymphedema. Figure 8A shows immunohistochemistry analysis of AIBP expression. Paired normal and lymphedema cutaneous biopsies (n=7 patients) were fixed with formalin and the 5 pm paraffin sections were prepared. The sections were deparaffinized and immunostained using our non-commercial AIBP antibody. Figures 8B to 8D show the AIBP signal in the epidermis (Figure 8B), sweat glands (Figure 8C), and dermis (Figure 8D) and were quantified using ImageJ. The enlarged areas of green and yellow boxes are shown on the right. The images were taken separately and stitched together using the EVOS microscope. NL: normal leg; LY: lymphedema. **p<0.0I.
20. Figures 9A-9E show AIBP regulates lymphatic vessel development in zebrafish. Figure 9A shows lymphatic developmental defects in Aibp2 knockdown zebrafish. Confocal images of PL at 48 hpf and TD formation at 5 dpf in the control and Aibp2 null zebrafish. Arrows show PL or TD, and stars denote absent PL or TD. Quantitative data of PL (Figure 9B) and TD formation (Figure 9C) in Figure 9A. n=25 in Figure 9A and =30 in Figure 9C. Scale: 25 pm. Figures 9D and 9E show TD development in apoalbp2-/- and control (CT) flilcc.egfp; lyvel.DsRed zebrafish (Figure 9D) and the quantification (Figure 9E). Scale bar: 100 pm. **, p<0.0I. 21. Figure 10A- 1 OB show that AIBP regulates lymphatic vessel development in zebrafish. Figure 10A shows that Aibp2 ablation disrupts LEC specification. Confocal images of LEC progenitors in the CV of control or Aibp2 knockdown zebrafish at 36 hpf. Figure 10B shows quantitative data of LEC progenitors in Figure 10A. Control or apoalbp2 morphants were fixed at 36 hpf, immunostained using Proxl and EGFP antibodies, and images were captured using confocal microscopy. Arrows indicate LEC progenitors. Scale: 25 pm. **p<0.01.
22. Figures 11A-1 IB show FACS analysis of murine ESC-derived CD31+LYVE1+ LECs. Figure 11A shows that AIBP induces LEC lineage commitment from mESCs. The murine ESCs were subjected to mesoderm and then endothelial differentiation as described in Figure 2C. At day 1 and day 7, the resulting cells were dissociated, immunostained with CD31 and LYVE1 antibodies, fixed with 4% PFA, and used for FACS analysis. The percentage of CD31+ and LYVE1+ cells were shown. Figure 1 IB shows quantitative data of FACS-sorted CD31+ and LYVEl+ cells in Figure 11A. **p<0.01.
23. Figures 12A-12E show generation of Cav-1 knockout zebrafish. Figure 12A depicts diagram showing position of the target site and its sequence (underline) in zebrafish apoalbp2 locus. PAM sequence (GGG) is shown in red. Figure 12B shows Sanger sequencing result of heterozygous mutants revealed an 8-bp genomic DNA fragment insertion from the target site. The PCR amplicons that span the mutated apoalbp2 region were ligated into a T- vector and subsequently transformed into competent cells. Single positive colonies were selected for sequencing. Figure 12C shows that the 8-bp insertion resulted in a frame shift that generates a mutated protein. Figure 12D shows Western analysis of pooled 26 hpf zebrafish (n= 15) show the absence of Cav-1 expression. Figure 12E shows no gross phenotypic defect observed in Cav- 1 knockout zebrafish. Zebrafish embryos at the indicated developmental stages were collected and images of live zebrafish embryos captured, hpf: hour(s) post fertilization, dpf: days postfertilization. WT: wild type. The sequences in Figure 12 include GACGTGATCGCCGAGCCTGCCGG (SEQ ID NO: 40), GATCGCCGAGCCTGCCGGCACCTACAGCTTCGACG (SEQ ID NO: 41), GATCGCCGAGCACCTACAGCTTCGACGGCGTGTGG (SEQ ID NO: 42), MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP KHLNDDVVKVDFEDVIAEPAGTYSFDGVWKASFTTFTVTKYWCYRLLTALVGIPLALV WGIFFAILSFIHIWAVVPCVKSYLIEIHCISRVYSICVHTFCDPLFEAMGKCFSNVRVTATK VV (SEQ ID NO: 43), MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP KHLNDDVVKVDFEDVIAEHLQLRRRVEGELHHLHSNQILVLQAADSAGGHPTRPGMG HLLRHPLLHPHLGRGALREELPNRDPLHQSSLLHLCAHLLRPTLGHGEMLRPGHCYGG (SEQ ID NO: 44).
24. Figures 13A-13B show quantitative PCR analysis of genes regulating lymphatic development in zebrafish. Figure 13A shows that loss of Cav-1 increases LEC gene expression. qPCR analysis of the indicated genes regulating lymphangiogenesis in control and Cav-1 null zebrafish at 96 hpf. Figure 13B shows that Cav-1 overexpression reduces LEC gene expression. The zebrafish embryo at one cell stage was injected with Cav-1 mRNA, and the resulting animals or control animals were harvested at 96 hpf, and total RNA extracted for reverse transcription. The indicated genes regulating lymphangiogenesis were analyzed using qPCR in control and Cav-1 overexpressing zebrafish. n=30 per sample. **, p<0.0I; ***, p<0.001; ****, pO.OOOl.
25. Figures 14A-14B show that CAV-1 knockout increases tail lymphangiogenesis in neonatal mice. Figure 14A shows that the CAV-1 knockout mice were purchased from JAX (stock No. 007083). The tail epidermis of CAV-1 knockout mice and control littermates were dissected from the similar anatomical locations and immunostained using LYVE-1 antibodies. Figure 14B shows that quantification of lymphatic vessel length in Figure 14A were performed using ImageJ. *, p<0.05.
26. Figures 15A-15D show that AIBP/APOA1 or CSD per se has no effect on adult lymphangiogenesis. Figures 15A and 15C show that PEG pellets containing recombinant AIBP and APOA1 were prepared and implanted into the corneas of B6 mice, control was implanted with control pellets. Figures 15B and 15D show quantification of lymphatic vessel area in Figure 15A and 15C.
27. Figures 16A-16B depict graphical Abstract. The lymphatics facilitate the drainage the cerebrospinal fluid (CSF) and interstitial fluid (ISF) from the brain. Figure 16B shows the underlying cause of AD is the formation of extracellular Ap aggregation and Tau neurofibrillary tangles. Impaired lymphatic drainage contributes to the A aggregation. Figure 16C shows that nanoparticle mediated delivery enables sustained release of AIBP/VEGFC, which can augment lymphatic vessel growth and lymphatic function, thereby improving AD. ISF: interstitial fluid.
28. Figures 17A-17C show that AIBP-CAV1 axis promotes comeal lymphangiogenesis. Figure 17A shows illustration of murine cornea lymphangiogenesis assay. Figure 17B shows representative whole mount images of murine comeal lymphangiogenesis by implantation of PEG pellets containing the indicated recombinant proteins and immunostained using LYVE1 & CD31 antibodies. Enlarged images of the boxed regions (scale bar, 1000 pm) are shown in the lower panels (scale bar, 500 pm). Figure 17C shows quantification of LYVE1+ lymphatic vessel area per cornea. VC: VEGFC; CSD: Cavl scaffolding domain peptide. ***p<0.001; *p<0.05.
29. Figures 18A-18C show that AIBP increases LEC lineage specification. Figure 18A shows scheme of mESC to LEC differentiation. Figures 18B-18C show that AIBP augments LEC differentiation from mESCs. The embryoid bodies of mESCs were prepared and cultured in the EC differentiation medium containing BMP4 and bFGF for 3 days. Recombinant VEGFA and VEGFC in combination or AIBP alone were supplemented at day 3 and kept in culture for additional 4 days. Western blot analyses of EC-associated PECAM, LEC marker LYVE1 and PROXI expression (Figure 18B) and the quantitative data (Figure 18C). C: control; V: VEGFA + VEGFC; A: AIBP.
30. Figures 19A-19C show that AIBP protects vascular integrity. Figure 19A shows that confluent HRMECs were stimulated for 1 hour with 100 ng/ml VEGF, in the presence or absence of 200 ng/ml recombinant AIBP, followed by immunostaining with anti-VE-Cad antibody. Images are representative of 5 different fields. Stars show paracellular gaps. Scale: 25 pm. Figure 19B shows scheme of permeability assay. Figure 19C shows measurements of leaked FITC-dextran (70 kDa) in the bottom chamber with the indicated treatments. *p<0.05.
31. Figure 20 illustrates the effect of three CAV 1 modifying peptides on comeal lymphangiogenesis. Figure 20 shows representative images of murine comeal lymphangiogenesis by implantation of pellets containing the indicated peptide and immunostained using LYVE1 & CD31 antibodies, and quantification of LYVE1+ lymphatic vessel area per cornea. VC: VEGFC; CSD: CAV-1 scaffolding domain peptide; CAV1-M: CAV1 modulator; CAVl-i: CAV1 inhibitor.
VI. DETAILED DESCRIPTION
32. Before the present compounds, compositions, articles, devices, and/or methods are disclosed and described, it is to be understood that they are not limited to specific synthetic methods or specific recombinant biotechnology methods unless otherwise specified, or to particular reagents unless otherwise specified, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
A. Definitions
33. As used in the specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a pharmaceutical carrier” includes mixtures of two or more such carriers, and the like.
34. Ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another embodiment includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another embodiment. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as “about” that particular value in addition to the value itself. For example, if the value “10” is disclosed, then “about 10” is also disclosed. It is also understood that when a value is disclosed that “less than or equal to” the value, “greater than or equal to the value” and possible ranges between values are also disclosed, as appropriately understood by the skilled artisan. For example, if the value “10” is disclosed the “less than or equal to 10”as well as “greater than or equal to 10” is also disclosed. It is also understood that the throughout the application, data is provided in a number of different formats, and that this data, represents endpoints and starting points, and ranges for any combination of the data points. For example, if a particular data point “10” and a particular data point 15 are disclosed, it is understood that greater than, greater than or equal to, less than, less than or equal to, and equal to 10 and 15 are considered disclosed as well as between 10 and 15. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
35. In this specification and in the claims that follow, reference will be made to a number of terms which shall be defined to have the following meanings:
36. "Activate", "activating", “stimulate” and "activation" mean to increase an activity, response, condition, or other biological parameter. This may also include, for example, a 10% increase in the activity, response, "or condition, as compared to the native or control level. Thus, the increase can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
37. “Activators” or “inhibitors” of expression or of activity are used to refer to inhibitory or activating molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., ligands, agonists, antagonists, and their homologs and mimetics. Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or protease activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein, e.g., antagonists. Activators are agents that, e.g., induce or activate the expression of a described target protein or bind to, stimulate, increase, open, activate, facilitate, enhance activation or protease inhibitor activity, sensitize or up regulate the activity of described target protein (or encoding polynucleotide), e.g., agonists. Samples or assays comprising described target protein that are treated with a potential activator or inhibitor are compared to control samples without the inhibitor or activator to examine the extent of effect. Control samples are assigned a relative activity value of 100% inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%. Activation of the described target protein is achieved when the activity value relative to the control is 110%, optionally 150%, optionally 200, 300%, 400%, 500%, or 1000-3000% or more.
38. “Administration” to a subject includes any route of introducing or delivering to a subject an agent. Administration can be carried out by any suitable route, including oral, topical, intravenous, subcutaneous, transcutaneous, transdermal, intramuscular, intra-joint, parenteral, intra-arteriole, intradermal, intraventricular, intracranial, intraperitoneal, intralesional, intranasal, rectal, vaginal, by inhalation, via an implanted reservoir, or via a transdermal patch, and the like. Administration includes self-administration and the administration by another. In some embodiments, the composition disclosed herein is administered intrathecally or intracranially. In some embodiments, the composition disclosed herein is administered topically (e.g., using a skin ointment).
39. The term “agonist” refers to a composition that binds to a receptor and activates the receptor to produce a biological response. The terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of agonists specifically mentioned herein, including, but not limited to, salts, esters, amides, reagents, active metabolites, isomers, fragments, analogs, and the like. When the term “agonist” is used, then, or when a particular agent is specifically identified, it is to be understood that the term includes the agent per se as well as pharmaceutically acceptable, pharmacologically active salts, esters, amides, reagents, conjugates, active metabolites, isomers, fragments, analogs, etc. Accordingly, the term “VEGFR3 agonist” can include any one or more agents which upon administration to a subject, can activate VEGFR3.
40. “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. 41. The term “biocompatible" generally refers to a material and any metabolites or degradation products thereof that are generally non-toxic to the recipient and do not cause significant adverse effects to the subject.
42. “Biological sample” refers to a sample of biological material obtained from a subject. Biological samples include all clinical samples useful for detection of disease or disorder in subjects. Appropriate samples include any conventional biological samples, including clinical samples obtained from a human or veterinary subject. Exemplary samples include, without limitation, cells, cell lysates, blood smears, cytocentrifiige preparations, cytology smears, bodily fluids (e.g., blood, plasma, serum, saliva, sputum, urine, bronchial alveolar lavage, semen, cerebrospinal fluid (CSF), etc.), tissue biopsies or autopsies, fine-needle aspirates, and/or tissue sections.
43. “Complementary” or “substantially complementary” refers to the hybridization or base pairing or the formation of a duplex between nucleotides or nucleic acids, such as, for instance, between the two strands of a double stranded DNA molecule or between an oligonucleotide primer and a primer binding site on a single stranded nucleic acid. Complementary nucleotides are, generally, A and T/U, or C and G. Two single-stranded RNA or DNA molecules are said to be substantially complementary when the nucleotides of one strand, optimally aligned and compared and with appropriate nucleotide insertions or deletions, pair with at least about 80% of the nucleotides of the other strand, usually at least about 90% to 95%, and more preferably from about 98 to 100%. Alternatively, substantial complementarity exists when an RNA or DNA strand will hybridize under selective hybridization conditions to its complement. Typically, selective hybridization will occur when there is at least about 65% complementary over a stretch of at least 14 to 25 nucleotides, at least about 75%, or at least about 90% complementary. See Kanehisa (1984) Nucl. Acids Res. 12:203.
44. As used herein, the term “comprising” is intended to mean that the compositions and methods include the recited elements, but not excluding others. “Consisting essentially of’ when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. “Consisting of’ shall mean excluding more than trace elements of other ingredients and substantial method steps for administering the compositions of this invention. Embodiments defined by each of these transition terms are within the scope of this invention. 45. “Composition” refers to any agent that has a beneficial biological effect.
Beneficial biological effects include both therapeutic effects, e.g., treatment of a disorder or other undesirable physiological condition, and prophylactic effects, e.g., prevention of a disorder or other undesirable physiological condition (e.g., a neurodegenerative disorder or lymphadema). The terms also encompass pharmaceutically acceptable, pharmacologically active derivatives of beneficial agents specifically mentioned herein, including, but not limited to, a vector, polynucleotide, cells, salts, esters, amides, reagents, active metabolites, isomers, fragments, analogs, and the like. When the term “composition” is used, then, or when a particular composition is specifically identified, it is to be understood that the term includes the composition per se as well as pharmaceutically acceptable, pharmacologically active vector, polynucleotide, salts, esters, amides, reagents, conjugates, active metabolites, isomers, fragments, analogs, etc.
46. A “control” is an alternative subject or sample used in an experiment for comparison purposes.
47. “Diagnosis” refers to the process of identifying a disease by its signs, symptoms and results of various tests. The conclusion reached through that process is also called "a diagnosis."
48. "Encoding" refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom, Thus, a gene encodes a protein if transcription and translation of mRNA.
49. "Expression vector", or “vector”, comprises a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed. An expression vector comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system. Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses (e.g., lentiviruses, retroviruses, adenoviruses, and adeno- associated viruses) that incorporate the recombinant polynucleotide.)
50. The “fragments,” whether attached to other sequences or not, can include insertions, deletions, substitutions, or other selected modifications of particular regions or specific amino acids residues, provided the activity of the fragment is not significantly altered or impaired compared to the nonmodified peptide or protein. These modifications can provide for some additional property, such as to remove or add amino acids capable of disulfide bonding, to increase its bio-longevity, to alter its secretory characteristics, etc. In any case, the fragment must possess a bioactive property, such as regulating the transcription of the target gene.
51. The term "gene" or "gene sequence" refers to the coding sequence or control sequence, or fragments thereof. A gene may include any combination of coding sequence and control sequence, or fragments thereof. Thus, a "gene" as referred to herein may be all or part of a native gene. A polynucleotide sequence as referred to herein may be used interchangeably with the term "gene”, or may include any coding sequence, non-coding sequence or control sequence, fragments thereof, and combinations thereof. The term "gene" or "gene sequence" includes, for example, control sequences upstream of the coding sequence (for example, the ribosome binding site).
52. "Inhibit", "inhibiting," and "inhibition" mean to decrease an activity, response, condition, disease, or other biological parameter. This can include but is not limited to the complete ablation of the activity, response, condition, or disease. This may also include, for example, a 10% reduction in the activity, response, condition, or disease as compared to the native or control level. Thus, the reduction can be a 10, 20, 30, 40, 50, 60, 70, 80, 90, 100%, or any amount of reduction in between as compared to native or control levels.
53. Nucleic acid: A deoxyribonucleotide or ribonucleotide polymer, which can include analogues of natural nucleotides that hybridize to nucleic acid molecules in a manner similar to naturally occurring nucleotides. In a particular example, a nucleic acid molecule is a single stranded (ss) DNA or RNA molecule, such as a probe or primer. In another particular example, a nucleic acid molecule is a double stranded (ds) nucleic acid, such as a target nucleic acid. Examples of modified nucleic acids are those with altered sugar moieties, such as a locked nucleic acid (LN A).
54. Nucleotide: The fundamental unit of nucleic acid molecules. A nucleotide includes a nitrogen-containing base attached to a pentose monosaccharide with one, two, or three phosphate groups attached by ester linkages to the saccharide moiety. The major nucleotides of DNA are deoxyadenosine 5 '-triphosphate (dATP or A), deoxyguanosine 5'- triphosphate (dGTP or G), deoxycytidine 5 '-triphosphate (dCTP or C) and deoxythymidine 5'- triphosphate (dTTP or T). The major nucleotides of RNA are adenosine 5 '-triphosphate (ATP or A), guanosine 5 '-triphosphate (GTP or G), cytidine 5 '-triphosphate (CTP or C) and uridine 5'- triphosphate (UTP or U).
55. "Pharmaceutically acceptable" component can refer to a component that is not biologically or otherwise undesirable, i.e., the component may be incorporated into a pharmaceutical formulation of the invention and administered to a subject as described herein without causing significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the formulation in which it is contained. When used in reference to administration to a human, the term generally implies the component has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug Administration.
56. "Pharmaceutically acceptable carrier" (sometimes referred to as a “carrier”) means a carrier or excipient that is useful in preparing a pharmaceutical or therapeutic composition that is generally safe and non-toxic, and includes a carrier that is acceptable for veterinary and/or human pharmaceutical or therapeutic use. The terms "carrier" or "pharmaceutically acceptable carrier" can include, but are not limited to, phosphate buffered saline solution, water, emulsions (such as an oil/water or water/oil emulsion) and/or various types of wetting agents.
57. As used herein, the term “carrier” encompasses any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, lipid, stabilizer, or other material well known in the art for use in pharmaceutical formulations. The choice of a carrier for use in a composition will depend upon the intended route of administration for the composition. The preparation of pharmaceutically acceptable carriers and formulations containing these materials is described in, e.g., Remington's Pharmaceutical Sciences, 21st Edition, ed. University of the Sciences in Philadelphia, Eippincott, Williams & Wilkins, Philadelphia, PA, 2005. Examples of physiologically acceptable carriers include saline, glycerol, DMSO, buffers such as phosphate buffers, citrate buffer, and buffers with other organic acids; antioxidants including ascorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or nonionic surfactants such as TWEEN™ (ICI, Inc.; Bridgewater, New Jersey), polyethylene glycol (PEG), and PLURONICS™ (BASF; Florham Park, NJ). To provide for the administration of such dosages for the desired therapeutic treatment, compositions disclosed herein can advantageously comprise between about 0.1% and 99% by weight of the total of one or more of the subject compounds based on the weight of the total composition including carrier or diluent.
58. The term "polynucleotide" refers to a single or double stranded polymer composed of nucleotide monomers (DNA or RNA). 59. The term "polypeptide" refers to a compound made up of a single chain of D- or L-amino acids or a mixture of D- and L-amino acids joined by peptide bonds.
60. The terms “peptide,” “protein,” and “polypeptide” are used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another.
61. The term "promoter" as used herein is defined as a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence.
62. As used herein, the term "promoter/regulatory sequence" means a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product. The promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
63. “Recombinant” used in reference to a gene refers herein to a sequence of nucleic acids that are not naturally occurring in the genome of the bacterium. The non-naturally occurring sequence may include a recombination, substitution, deletion, or addition of one or more bases with respect to the nucleic acid sequence originally present in the natural genome of the bacterium.
64. The term “increased” or “increase” as used herein generally means an increase by a statically significant amount; for the avoidance of any doubt, “increased” means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3 -fold, or at least about a 4-fold, or at least about a 5 -fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
65. The term “reduced”, “reduce”, “reduction”, or “decrease” as used herein generally means a decrease by a statistically significant amount. However, for avoidance of doubt, “reduced” means a decrease by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (i.e. absent level as compared to a reference sample), or any decrease between 10-100% as compared to a reference level.
66. Sequence identity: The similarity between two nucleic acid sequences is expressed in terms of the similarity between the sequences, otherwise referred to as sequence identity.
67. Sequence identity is frequently measured in terms of percentage identity, similarity, or homology; a higher percentage identity indicates a higher degree of sequence similarity. The NCBI Basic Local Alignment Search Tool (BLAST), Altschul et al, J. Mol. Biol. 215:403-10, 1990, is available from several sources, including the National Center for Biotechnology Information (NCBI, Bethesda, MD), for use in connection with the sequence analysis programs blastp, blastn, blastx, tblastn and tblastx. It can be accessed through the NCBI website. A description of how to determine sequence identity using this program is also available on the website.
68. When less than the entire sequence is being compared for sequence identity, homologs will typically possess at least 75% sequence identity over short windows of 10-20 amino acids, and can possess sequence identities of at least 85% or at least 90% or 95% depending on their similarity to the reference sequence. Methods for determining sequence identity over such short windows are described on the NCBI website.
69. These sequence identity ranges are provided for guidance only; it is entirely possible that strongly significant homologs could be obtained that fall outside of the ranges provided.
70. Subject: Any mammal, such as humans, non-human primates, pigs, sheep, horses, dogs, cats, cows, rodents and the like. In two non-limiting examples, a subject is a human subject or a murine subject. Thus, the term "subject" includes both human and veterinary subjects.
71. The terms “treat,” “treating,” “treatment,” and grammatical variations thereof as used herein, include partially or completely delaying, alleviating, mitigating or reducing the intensity of one or more attendant symptoms of a disorder or condition and/or alleviating, mitigating or impeding one or more causes of a disorder or condition. Treatments according to the invention may be applied preventively, prophy tactically, pallatively or remedially. Prophylactic treatments are administered to a subject prior to onset (e.g., before obvious signs of a neurodegenerative disorder or lymphedema), during early onset (e.g. , upon initial signs and symptoms of a neurodegenerative disorder or lymphedema), or after an established development of a neurodegenerative disorder or lymphedema. Prophylactic administration can occur for several days to years prior to the manifestation of symptoms of a disorder.
72. “Therapeutically effective amount” or “therapeutically effective dose” of a composition (e.g. a composition comprising an agent) refers to an amount that is effective to achieve a desired therapeutic result. In some embodiments, a desired therapeutic result is the control of a neurodegenerative disorder or lymphedema. In some embodiments, a desired therapeutic result is the control of a neurodegenerative disorder or lymphedema, or a symptom of a neurodegenerative disorder or lymphedema. Therapeutically effective amounts of a given therapeutic agent will typically vary with respect to factors such as the type and severity of the disorder or disease being treated and the age, gender, and weight of the subject. The term can also refer to an amount of a therapeutic agent, or a rate of delivery of a therapeutic agent (e.g., amount over time), effective to facilitate a desired therapeutic effect. The precise desired therapeutic effect will vary according to the condition to be treated, the tolerance of the subject, the agent and/or agent formulation to be administered (e.g., the potency of the therapeutic agent, the concentration of agent in the formulation, and the like), and a variety of other factors that are appreciated by those of ordinary skill in the art. In some instances, a desired biological or medical response is achieved following administration of multiple dosages of the composition to the subject over a period of days, weeks, or years.
73. Throughout this application, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.
B. Compositions
74. Lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, dietary lipid transport and immune surveillance. The Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including lymphedema, tumor metastasis, cardiovascular disease, obesity, Alzheimer’s disease, and diabetic mellitus. Therapeutic augmentation of lymphangiogenesis has been documented to improve lymphatic structure and function. Earlier studies show that the lymphatic system is derived from the embryonic cardinal vein (CV), where lymphatic endothelial cells (LEC) progenitors are specified and subsequently migrate and establish the lymphatic network. Recent studies indicate that a subset of lymphatics can be developed by non-canonical mechanisms. During murine development, the assembly of the lymphatic vascular network is initiated at approximately embryonic day 9.5 (E9.5) under the control of VEGFC signaling through its cognate receptor VEGFR3. LEC fate commitment occurs through SOX18-induced expression of PROX-1, which, in concert with the orphan nuclear factor NR2F2 (also known as COUP-TFII), dictates LEC differentiation from the CV. In addition, GATA2, HHEX, and PROXI itself have recently been shown to upregulate PROXI expression. The newly specified LECs subsequently bud from the CV and migrate (E10.0- E11.5) in a dorsolateral fashion to form lymph sacs and, thereby establish the entire lymphatic vessels. VEGFC-induced VEGFR3 signaling is the major driver of lymphangiogenesis in vertebrates. In addition to the regulation of LEC sprouting, VEGFR3 signaling also controls LEC progenitor homeostasis by maintaining PROXI expression levels in a positive feedback loop. In zebrafish, Vegfc/Vegfr3 signaling increases Proxl+ LEC progenitors in the axis vasculature. Mice deficient either in CCBE1, a critical matrix protein regulating VEGFC bioavailability, in VEGFC, or in VEGFR3 show fewer PROXI -positive LECs in the CV.
75. The developmental venous origin of the lymphatic endothelium and the requirement of VEGFC/VEGFR3 signaling for lymphangiogenesis are highly conserved from zebrafish to mice to humans. In zebrafish, from 30 to 32 hours post fertilization (hpf), bipotential precursor cells in the posterior cardinal veins (PCV) express Proxl and undergo a Vegfc/Vegfr3- dependent cell division to generate lymphatic and venous daughter cells. In response to the Vegfc cue, the daughter cells on the PCV progressively acquire the LEC fate at 36 hpf; among these, approximately half form parachordal LECs (PL) at the horizontal myoseptum by 48 hpf. These PLs subsequently migrate ventrally and dorsally along the arterial intersegmental vessels (ISVs) at ~60 hpf, to form the thoracic duct (TD), intersegmental lymphatic vessels and dorsal longitudinal lymphatic vessels (DLLV). Wnt5b has been reported to function upstream of Proxl to regulate proper lymphatic specification. Nevertheless, the dynamics of LEC fate determination and the molecular mechanism of this process remain to be further understood.
76. It was demonstrated that the secreted protein AIBP limits angiogenesis in a noncell autonomous fashion. Mechanistically, extracellular AIBP binds ECs, accelerates cholesterol efflux from ECs to high-density lipoprotein (HDL) and reduces lipid raft/caveola abundance, which in turn disrupts VEGFR2 signaling, thereby restricting angiogenesis. Lymphatic vessels are structurally and functionally related to blood vessels. Many genes that are required for angiogenesis, such as VEGFR3, FGF, D114-Notch, angiopoietin-Tie2, Ephrin B2, and TGFp family member ALK1, Epsin, and others, also function in lymphangiogenesis. Thus, the role AIBP-regulated cholesterol metabolism in lymphangiogenesis was studied. The studies herein reveal a previously unidentified role of AIBP in lymphangiogenesis, in which AIBP augments VEGFC-engaged VEFGR3 signaling in a CAV-1 -dependent fashion.
77. Further, it has been shown that APOA1 binding protein (AIBP) polypeptide and/or polynucleotide can used be treat neurodegenerative disorders. Dudau M et al. Arguments for Caveolin-1 Knockout Mice as an Alzheimer’s Disease Model, Austin Alzheimer s J Parkinsons Dis 3(1): idl028 (2016) indicates that caveolin-1 (CAV-1) knockout mice exhibit Alzheimer’s disease-like symptoms and can be used as Alzheimer’s disease model. But this is a genetic model and the mice lose CAV-1 from one cell stage in development, i.e., this cannot not considered as a normal mouse from the beginning of conception. In contrast, the present disclosure shows that administration of CAV-1 inhibitor and/or AIBP, in the presence of a VEGFR3 stimulator (e.g., VEGFC), surprisingly improves meningeal lymphatics functions and treats a neurodegenerative disease (e.g., Alzheimer’s disease).
78. Disclosed herein is a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor.
79. In some embodiments, the AIBP polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 1 or a fragment thereof. In a particular embodiment, the AIBP polypeptide comprises a sequence of SEQ ID NO: 1. The polynucleotide can be a DNA or an RNA. In some embodiments, the polynucleotide encoding the AIBP polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 2 or a fragment thereof. In a particular embodiment, the polynucleotide encoding the AIBP polypeptide comprises a sequence of SEQ ID NO: 2. In some embodiments, the polynucleotide is an RNA encoded by a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 2 or a fragment thereof.
80. Caveolin-1 (CAV-1) is an oncogenic membrane protein associated with endocytosis, extracellular matrix organization, cholesterol distribution, cell migration and signaling. The present disclosure shows that CAV-1 suppresses VEGFR3 activation in lymphatic endothelial cells. “CAV-1” refers herein to a polypeptide that, in humans, is encoded by the C4F7 gene. In some embodiments, the CAV-1 polypeptide is that identified in one or more publicly available databases as follows: HGNC: 1527, Entrez Gene: 857, Ensembl: ENSG00000105974, OMIM: 601047, UniProtKB: Q03135. In some embodiments, the CAV-1 polypeptide comprises the sequence of SEQ ID NO: 7, or a polypeptide sequence having at or greater than about 80%, about 85%, about 90%, about 95%, or about 98% homology with SEQ ID NO: 7, or a polypeptide comprising a portion of SEQ ID NO: 7. The CAV-1 polypeptide of SEQ ID NO: 7 may represent an immature or pre-processed form of mature CAV-1, and accordingly, included herein are mature or processed portions of the CAV-1 polypeptide in SEQ ID NO: 7.
81. As disclosed herein, CAV-1 suppresses VEGFR3 activation in lymphatic endothelial cells. Inhibition or deletion of CAV-1 increases lymphangiogenesis. Accordingly, in some embodiments, the composition disclose herein further comprises a CAV-1 inhibitor, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
82. “Inhibitors” of expression or of activity are used to refer to inhibitory molecules, respectively, identified using in vitro and in vivo assays for expression or activity of a described target protein, e.g., antagonists and their homologs and mimetics. Inhibitors are agents that, e.g., inhibit expression or bind to, partially or totally block stimulation or activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of the described target protein. Control samples (untreated with inhibitors) are assigned a relative activity value of 100%. Inhibition of a described target protein is achieved when the activity value relative to the control is about 80%, optionally 50% or 25, 10%, 5% or 1%.
83. It should be understood that the caveolin-1 (CAV-1) is a scaffolding protein that provides a spatially restricted platform for proper signaling of cell surface receptors. CAV-1 contains a signaling motif that interacts with a variety of membrane receptors and modulates their activities (e.g., the activities of VEGFR3). Accordingly, a CAV-1 inhibitor can be an inhibitor for one or more other factors (e.g., one or more genes, proteins, mRNA) involved in the CAV-1 -involving pathway or signaling platform. The CAV-1 inhibitor can include any one or more agents which upon administration to a subject, can inhibit CAV-1. The CAV-1 inhibitor can directly affect CAV-1, for example, by binding to the CAV-1 protein or preventing the transcription or translation of a CAV-1 gene. Alternatively, the CAV-1 inhibitor can inhibit one or more other factors (e.g., one or more genes, proteins, mRNA) involved in the CAV-1 pathway. In some examples, the CAV-1 inhibitor can be a peptide that competes with the CAV- 1 such that to inhibit the interaction of CAV-1 and a bind protein thereof (including, for example, VEGFR3).
84. In some embodiments, the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux. In some embodiments, the CAV-1 inhibitor comprises methyl beta cyclodextrin.
85. In some embodiments, the CAV-1 inhibitor is an antibody (e.g., an antagonizing/inhibitory antibody of CAV-1).
86. The terms “antibody” and “antibodies” are used herein in a broad sense and include polyclonal antibodies, monoclonal antibodies, and bi-specific antibodies. In addition to intact immunoglobulin molecules, also included in the term “antibodies” are fragments or polymers of those immunoglobulin molecules, and human or humanized versions of immunoglobulin molecules or fragments thereof. Antibodies are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end.
87. The antibodies can be tested for their desired activity using the in vitro assays described herein, or by analogous methods, after which, their in vivo therapeutic and/or prophylactic activities are tested according to known clinical testing methods. There are five major classes of human immunoglobulins: IgA, IgD, IgE, IgG and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG-1, IgG-2, IgG-3, and IgG-4; IgA-1 and IgA -2. One skilled in the art would recognize the comparable classes for mouse. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
88. The term “antibody” encompasses chimeric antibodies and hybrid antibodies, with dual or multiple antigen or epitope specificities, and fragments, such as F(ab’)2, Fab’, Fab, Fv, scFv, a single-chain variable fragment (scFv) antibody, or a VHH antibody and the like, including hybrid fragments. Thus, fragments of the antibodies that retain the ability to bind their specific antigens are provided.
89. Accordingly, in some embodiments, the composition disclosed herein comprises a CAV-1 inhibitor, wherein the CAV-1 inhibitor is an antibody, and wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, and/or a VHH antibody. 90. In some embodiments, the CAV-1 inhibitor comprises a CAV-1 gene editing tool, including, for example, a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, CRISPR-Casl3, or CRISPR-Casl3d. Methods of designing siRNA, shRNA, CRISPR-Cas9, CRISPR-Casl3, or CRISPR-Casl3d for CAV-1 are well-known in the art.
91. In one example, the CAV-1 inhibition method described herein comprises using CRISPR-Cas9 that targets a CAV-1 polynucleotide sequence. In some embodiments, CAV-1 polynucleotide sequence is at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 8 or a fragment thereof.
92. In one example, the CAV-1 inhibitor comprises a siRNA targeting CAV-1, wherein the siRNA comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 9, 10, or 11 or a fragment thereof.
93. In some embodiments, the CAV-1 inhibitor comprises a Caveolin-l(Cav-l) scaffolding domain (CSD) (CSD) peptide that can compete with the plasma membrane CAV-1, inhibit the interaction of the proteins and CAV-1, and re -store the functions of CAV-1 binding proteins. In some embodiments, the CSD peptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 12 or a fragment thereof.
94. The CAV-1 inhibitor can be a polynucleotide sequence encoding the CSD peptide disclosed herein. Accordingly, in one example, the composition disclosed herein comprises a first polynucleotide encoding an AIBP polypeptide and a second polynucleotide encoding a CSD polypeptide. In another example, the composition disclosed herein comprises a polynucleotide that encodes an AIBP polypeptide and a CSD polypeptide.
95. “VEGFR3” refers herein to a polypeptide that, in humans, is encoded by the FLT4 gene. In some embodiments, the VEGFR3 polypeptide is that identified in one or more publicly available databases as follows: HGNC: 3767; Entrez Gene: 2324; Ensembl: ENSG00000037280; OMIM: 136352; UniProtKB: P35916. The VEGFR3 tyrosine kinase is expressed mainly in lymphatic vessels. VEGFR3 transmits signals for lymphatic endothelial migration, survival, and proliferation and is involved in the biology and pathology of the lymphatic vasculature. As shown herein, augmenting VEGFR3 activation can increase lymphangiogenesis, thereby treating lymphedema and/or a neurodegenerative disease. Accordingly, in some embodiments, the composition disclosed herein further comprises a stimulator, activator, or agonist of VEGFR3. In some embodiments, the stimulator, activator, and/or agonist of VEGFR3 comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof. In some embodiments, the VEGFA polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 3 or a fragment thereof. In some embodiments, the VEGFC polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 5 or a fragment thereof. In some embodiments, the stimulator, activator, or agonist of VEGFR3 is VEGFC(C156S). In some embodiments, VEGFC(C156S) is encoded by a polynucleotide sequence at least about at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 13 or a fragment thereof. In some embodiments, the stimulator, activator, or agonist of VEGFR3 is a small molecule (e.g., Pioglitazone).
96. The composition disclosed herein can further comprises at least about one polynucleotide encoding a VEGFA polypeptide and/or a VEGFC polypeptide. Accordingly, in some embodiments, the composition disclosed herein comprises a first polynucleotide encoding an AIBP polypeptide, a second polynucleotide encoding a CSD polypeptide, a third polynucleotide encoding a VEGFA polypeptide, and/or a fourth polynucleotide encoding a VEGFC polypeptide, or any combination thereof. In another example, the composition disclosed herein comprises a polynucleotide that encodes an AIBP polypeptide, a CSD polypeptide, a VEGFA polypeptide, and/or a VEGFC polypeptide, or any combination thereof. In some embodiments, the polynucleotide encoding VEGFA polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 4 or a fragment thereof. In some embodiments, the polynucleotide encoding VEGFC polypeptide comprises a sequence at least about 60% (for example, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99%) identical to SEQ ID NO: 6 or a fragment thereof.
97. The polynucleotide disclosed herein can be contained in a vector that can be used to deliver the polynucleotide to cells, either in vitro or in vivo. The vectors and the delivery methods can largely be broken down into two classes: viral based delivery systems and non-viral based delivery systems. For example, the nucleic acids can be delivered through a number of direct delivery systems such as, electroporation, lipofection, calcium phosphate precipitation, plasmids, viral vectors, viral nucleic acids, phage nucleic acids, phages, cosmids, or via transfer of genetic material in cells or carriers such as cationic liposomes. Appropriate means for transfection, including viral vectors, chemical transfectants, or physico-mechanical methods such as electroporation and direct diffusion of DNA, are described by, for example, Wolff, J. A., et al., Science, 247, 1465-1468, (1990); and Wolff, J. A. Nature, 352, 815-818, (1991). Such methods are well known in the art and readily adaptable for use with the compositions and methods described herein. In certain cases, the methods will be modified to specifically function with large DNA molecules. Further, these methods can be used to target certain diseases and cell populations by using the targeting characteristics of the carrier.
98. Transfer vectors can be any nucleotide construction used to deliver genes into cells (e.g., a plasmid), or as part of a general strategy to deliver genes, e.g., as part of recombinant retrovirus or adenovirus (Ram et al. Cancer Res. 53:83-88, (1993)).
99. As used herein, plasmid or viral vectors are agents that transport the disclosed polynucleotides (e.g., a polynucleotide encoding an AIBP polypeptide, a CSD peptide, a VEGFA polypeptide, and/or a VEGFC polypeptide, or a combination thereof) into the cell without degradation and include a promoter yielding expression of the gene in the cells into which it is delivered. In some embodiments, the polypeptides are derived from either a virus or a retrovirus. Viral vectors can be, for example, Adenovirus, Adeno-associated virus, Herpes virus, Vaccinia virus, Polio virus, AIDS virus, neuronal trophic virus, Sindbis and other RNA viruses, including these viruses with the HIV backbone. Also preferred are any viral families which share the properties of these viruses which make them suitable for use as vectors. A preferred embodiment is a viral vector which has been engineered so as to suppress the immune response of the host organism, elicited by the viral antigens.
100. Viral vectors can have higher transaction (ability to introduce genes) abilities than chemical or physical methods to introduce genes into cells. Typically, viral vectors contain, nonstructural early genes, structural late genes, an RNA polymerase III transcript, inverted terminal repeats necessary for replication and encapsulation, and promoters to control the transcription and replication of the viral genome. When engineered as vectors, viruses typically have one or more of the early genes removed and a gene or gene/promotor cassette is inserted into the viral genome in place of the removed viral DNA.
101. In some embodiments, the polynucleotide disclosed herein is contained in an adeno-associated virus (AAV) vector. This defective parvovirus is a preferred vector because it can infect many cell types and is nonpathogenic to humans. AAV type vectors can transport about 4 to 5 kb and wild type AAV is known to stably insert into chromosome 19. Vectors which contain this site-specific integration property are preferred. The AAV vector can further comprise the herpes simplex virus thymidine kinase gene, HSV-tk, and/or a marker gene, such as the gene encoding the green fluorescent protein, GFP.
102. In another type of AAV virus, the AAV contains a pair of inverted terminal repeats (ITRs) which flank at least one cassette containing a promoter which directs cell-specific expression operably linked to a heterologous gene. Heterologous in this context refers to any nucleotide sequence or gene which is not native to the AAV or B19 parvovirus. Typically, the AAV and B 19 coding regions have been deleted, resulting in a safe, noncytotoxic vector. The AAV ITRs, or modifications thereof, confer infectivity and site-specific integration, but not cytotoxicity, and the promoter directs cell-specific expression. US Patent No. 6,261,834 is herein incorporated by reference for material related to the AAV vector. Accordingly, in some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide. In some embodiments, disclosed herein is an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide. 103. The disclosed vectors thus provide DNA molecules which are capable of integration into a mammalian chromosome without substantial toxicity. The inserted genes in viral and retroviral can contain promoters, and/or enhancers to help control the expression of the desired gene product.
104. The AAV used herein can be an AAV serotype AAV-5, AAV-6, AAV-8 or AAV-9; a rhesus-derived AAV, or the rhesus-derived AAV AAVrh.l0hCLN2; an organ-tropic AAV, or a neurotropic AAV; and/or an AAV capsid mutant or AAV hybrid serotype. In alternative embodiments, the AAV is engineered to increase efficiency in targeting a specific cell type that is non-permissive to a wild type (wt) AAV and/or to improve efficacy in infecting only a cell type of interest. It is well known in the art how to engineer an adeno-associated virus (AAV) capsid in order to increase efficiency in targeting specific cell types that are non- permissive to wild type (wt) viruses and to improve efficacy in infecting only the cell type of interest; see e.g., Wu et al., Mol. Ther. 2006 September; 14(3):316-27. Epub 2006 Jul. 7; Choi, et al., Curr. Gene Ther. 2005 June; 5(3):299-310.
105. For example, in alternative embodiments, serotypes AAV-8, AAV-9, AAV-DJ or AAV-DJ/8™ (Cell Biolabs, Inc., San Diego, Calif.), which have increased uptake in brain tissue in vivo, are used to deliver the polynucleotide disclosed herein for expression in the CNS.
106. In some embodiments, the composition disclosed herein is contained in or conjugated to a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutically acceptable carrier can be used to deliver the compositions to a CNS or a brain in vivo. For example, the carrier can be the ones described in U.S. Patent Publication No. 20060083737, incorporated by reference herein in its entirety. In some embodiments, the pharmaceutically acceptable carrier can be used to deliver the compositions to lymphatic endothelial cells.
107. In some embodiments, the composition disclosed herein is conjugated to docosahexaenoic acid (DHA).
108. Liposomes can be made using any method, e.g., as described in Park, et al., U.S. Application Publication No. 20070042031, including method of producing a liposome by encapsulating a composition disclosed herein, the method comprising providing an aqueous solution in a first reservoir; providing an organic lipid solution in a second reservoir, and then mixing the aqueous solution with the organic lipid solution in a first mixing region to produce a liposome solution, where the organic lipid solution mixes with the aqueous solution to substantially instantaneously produce a liposome encapsulating the active agent; and immediately then mixing the liposome solution with a buffer solution to produce a diluted liposome solution. 109. The nanoparticle used herein can be any nanoparticle useful for the delivery of nucleic acids and/or polypeptides. The term “nanoparticle” as used herein refers to a particle or structure which is biocompatible with and sufficiently resistant to chemical and/or physical destruction by the environment of such use so that a sufficient number of the nanoparticles remain substantially intact after delivery to the site of application or treatment and whose size is in the nanometer range. In some embodiments, the nanoparticle comprises a lipid-like nanoparticle. See, for example, WO/2016/187531A1, WO/2017/ 176974, WO/2019/027999, or Li, B et al. An Orthogonal array optimization of lipid-like nanoparticles for mRNA delivery in vivo. Nano Lett. 2015, 15, 8099-8107; which are incorporated herein by reference in their entireties. In some embodiments, the nanoparticle is a porous silica nanoparticle (pSi). In some embodiments, the nanoparticle comprises poly (lactide-co-glycolide) (PLGA). Porous silica nanoparticles are well known in the art. See, for example, US Patent No. 10,143,660; US Application Publication No. 2013/0216807; International Publication No. 2013/056132; which are incorporated herein by reference in their entireties.
110. Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers. For example, a contemplated nanoparticle may include about 10 to about 99 weight percent of a one or more block co-polymers that include a biodegradable polymer and polyethylene glycol, and about 0 to about 50 weight percent of a biodegradable homopolymer. Polymers can include, for example, both biostable and biodegradable polymers, such as microcrystalline cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, polyalkylene oxides such as polyethylene oxide (PEG), polyanhydrides, poly(ester anhydrides), polyhydroxy acids such as polylactide (PLA), polyglycolide (PGA), poly(lactide-co-glycolide) (PLGA), poly-3 -hydroxybutyrate (PHB) and copolymers thereof, poly-4-hydroxybutyrate (P4HB) and copolymers thereof, polycaprolactone and copolymers thereof, and combinations thereof.
111. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. 112. In some embodiments, the nanoparticle has a diameter from about 1 nm to about 1000 nm. In some embodiments, the nanoparticle has a diameter less than, for example, about 1000 nm, about 950 nm, about 900 nm, about 850 nm, about 800 nm, about 750 nm, about 700 nm, about 650 nm, about 600 nm, about 550 nm, about 500 nm, about 450 nm, about 400 nm, about 350 nm, about 300 nm, about 290 nm, about 280 nm, about 270 nm, about 260 nm , about 250 nm, about 240 nm, about 230 nm, about 220 nm, about 210 nm, about 200 nm, about 190 nm, about 180 nm, about 170 nm, about 160 nm, about 150 nm, about 140 nm, about 130 nm, about 120 nm, about 110 nm, about 100 nm, about 90 nm, about 80 nm, about 70 nm, about 60 nm, about 50 nm, about 40 nm, about 30 nm, about 20 nm, or about 10 nm. In some embodiments, the nanoparticle has a diameter, for example, from about 20 nm to about 1000 nm, from about 20 nm to about 800 nm, from about 20 nm to about 700 nm, from about 30 nm to about 600 nm, from about 30 nm to about 500 nm, from about 40 nm to about 400 nm, from about 40 nm to about 300 nm, from about 40 nm to about 250 nm, from about 50 nm to about 250 nm, from about 50 nm to about 200 nm, from about 50 nm to about 150 nm, from about 60 nm to about 150 nm, from about 70 nm to about 150 nm, from about 80 nm to about 150 nm, from about 90 nm to about 150 nm, from about 100 nm to about 150 nm, from about 110 nm to about 150 nm, from about 120 nm to about 150 nm, from about 90 nm to about 140 nm, from about 90 nm to about 130 nm, from about 90 nm to about 120 nm, from 100 nm to about 140 nm, from about 100 nm to about 130 nm, from about 100 nm to about 120 nm, from about 100 nm to about 110 nm, from about 110 nm to about 120 nm, from about 110 nm to about 130 nm, from about 110 nm to about 140 nm, from about 90 nm to about 200 nm, from about 100 nm to about 195 nm, from about 110 nm to about 190 nm, from about 120 nm to about 185 nm, from about 130 nm to about 180 nm, from about 140 nm to about 175 nm, from 150 nm to 175nm, or from about 150 nm to about 170 nm.
113. In some embodiments, the nanoparticle has a pore size from about 0. 1 nm to about 50 nm (including, for example, about 0. 1 nm, about 0.2 nm, about 0.3 nm, about 0.4 nm, about 0.5 nm, about 0.6 nm, about 0.7 nm, about 0.8 nm, about 0.9 nm, about 1.0 nm, about 1.2 nm, about 1.4 nm, about 1.6 nm, about 1.8 nm, about 2.0 nm, about 2.2 nm, about 2.4 nm, about 2.6 nm, about 2.8 nm, about 3.0 nm, about 3.5 nm, about 4.0 nm, about 4.5 nm, about 5 nm, about 5.5 nm, about 6 nm, about 6.5 nm, about 7 nm, about 7.5 nm, about 8 nm, about 8.5 nm, about 9 nm, about 9.5 nm, about 10 nm, about 15 nm, about 20 nm, about 25 nm, about 30 nm, about 35 nm, about 40 nm, about 45 nm, or about 50 nm).
114. Further, disclosed nanoparticles or liposomes may be able to efficiently bind to or otherwise associate with a biological entity, for example, a particular membrane component or cell surface receptor on a target cell (e.g., a receptor that facilitates delivery into the CNS or brain, or a receptor on lymphatic endothelial cells). For example, the disclosed nanoparticles or liposomes may be engineered to a ligand binding to a receptor ubiquitously expression on a cell of CNS or brain (e.g., a ligand binding to low density lipoprotein receptor-related protein (LPR)- 1 and/or LPR-2) or binding to receptor or markers expressed on lymphatic endothelial cells, such as CD34, Proxl, podoplanin, LYVE-1, and/or VEGFR-3.
C. Methods of treating neurodegenerative diseases
115. Disclosed herein is a method of increasing, enhancing, improving, and/or stimulating lymphangiogenesis in subject, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor. In some embodiments, the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor. The extent of effect of increasing, enhancing, improving, and/or stimulating lymphangiogenesis is relative to a control (e.g., a healthy subject or a subject not being administered with the composition).
116. It should be herein contemplated that dysfunction of the meningeal lymphatics is associated with dementia, such as Alzheimer disease (AD) pathology, and/or neurodegenerative disease. The methods that enhance meningeal lymphatic function can improve cognition in the diseased subjects. Accordingly, disclosed herein is also a method of preventing, inhibiting, mitigating, and/or treating a neurodegenerative disease in a subject, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor. In some embodiments, the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor. It should be understood and herein contemplated that the extent of effect of preventing, inhibiting, mitigating, and/or treating a neurodegenerative disease is relative to a control (e.g., a subject not being administered with the composition).
117. The polynucleotide disclosed herein can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector). In some embodiments, the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide. In some embodiments, the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody. In some embodiments, the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR-Casl3.
118. In some embodiments, the composition disclosed herein further comprises a stimulator for VEGFR3. In one example, the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)). In one example, the stimulator comprises Pioglitazone. Although VEGFR3 stimulator (for examples, VEGFC) can improve cognition in aged subjects, it also elicits deleterious consequence such as vascular leakage and angiogenesis. AIBP protect against vascular leakage caused by VEGFR3 activation. In some embodiments, the composition disclosed herein that comprises i) a stimulator for VEGFR3 and ii) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, in some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide.
119. Whether caveolin-1 promotes or inhibits angiogenesis remains under debate. Some studies shows that Cav-1 knockout mice exhibit hyperpermeable vasculature and increased angiogenesis; whereas, in some other studies, caveolin-1 inhibition mitigates angiogenesis in various vascular beds. The study herein shows that administration of a VEGFR3 stimulator, i.e., VEGFC, in combination with a caveolin-1 inhibitor (e.g., a CSD peptide) promotes lymphangiogenesis compared to VEGFC alone. AIBP or CSD can mitigate/prevent neovascularization, thereby reducing the associated vascular leakage. Accordingly, in some examples, the compositions disclosed herein that comprises a stimulator for VEGFR3 and a CAV-1 inhibitor. In some embodiments, the CAV-1 inhibitor is a CSD peptide.
120. The composition disclosed herein can be contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier. In some embodiments, the composition is contained in a nanoparticle. In some embodiments, the composition is conjugated to docosahexaenoic acid (DHA). In some embodiments, the nanoparticle is a porous silica nanoparticle (pSi). In some embodiments, the nanoparticle comprises poly(lactide-co-glycolide) (PLGA). In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
121. As used herein, the term “neurodegenerative disease” refers to a varied assortment of central nervous system disorders characterized by gradual and progressive loss of neural tissue and/or neural tissue function. A neurodegenerative disease is a class of neurological disorder or disease, and where the neurological disease is characterized by a gradual and progressive loss of neural tissue, and/or altered neurological function, typically reduced neurological function as a result of a gradual and progressive loss of neural tissue. Examples of neurodegenerative diseases include for example, but are not limited to, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's Disease, Amyotrophic Lateral Sclerosis (ALS, also termed Lou Gehrig's disease) and Multiple Sclerosis (MS), polyglutamine expansion disorders (e.g., HD, dentatorubropallidoluysian atrophy, Kennedy's disease (also referred to as spinobulbar muscular atrophy), spinocerebellar ataxia (e.g., type 1, type 2, type 3 (also referred to as Machado- Joseph disease), type 6, type 7, and type 17)), other trinucleotide repeat expansion disorders (e.g., fragile X syndrome, fragile XE mental retardation, Friedreich's ataxia, myotonic dystrophy, spinocerebellar ataxia type 8, and spinocerebellar ataxia type 12), Alexander disease, Alper's disease, ataxia telangiectasia, Batten disease (also referred to as Spielmeyer-Vogt-Sjogren-Batten disease), Canavan disease, Cockayne syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, ischemia stroke, Krabbe disease, Lewy body dementia, multiple system atrophy, Pelizaeus-Merzbacher disease, Pick's disease, primary lateral sclerosis, Refsum's disease, Sandhoff disease, Schilder's disease, spinal cord injury, spinal muscular atrophy (SMA), SteeleRichardson-Olszewski disease, Tabes dorsalis, and the like. In some embodiments, the neurodegenerative disease is Alzheimer's disease.
122. “Alzheimer's Disease” as used herein refers to all form of dementia, identified as a degenerative and terminal cognitive disorder. The disease may be static, the result of a unique global brain injury, or progressive, resulting in long-term decline in cognitive function due to damage or disease in the body beyond what might be expected from normal aging. The betaamyloid protein, or Ap, involved in Alzheimer's has several different molecular forms that collect between neurons. It is formed from the breakdown of a larger protein, called amyloid precursor protein. One form, beta-amyloid 42, is thought to be especially toxic. An abnormal level of this protein is found in the Alzheimer’s brain, wherein the protein clump together to form plaques between neurons, leading to neuron function disruption.
123. Accordingly, the method disclosed herein can treat, decrease, mitigate, and/or prevent Alzheimer’s disease and/or a symptom thereof (e.g., A accumulation, an increase in levels of Tau protein, CNS inflammation, decline in cognitive function, and/or loss of memory). It should be understood and herein contemplated that the extent of effect of treating, decreasing, mitigating, and/or preventing Alzheimer’s disease and/or a symptom thereof is relative to a control (e.g., a subject not being administered with the composition).
124. It is understood and herein contemplated that the timing of a neurodegenerative disease onset can often not be predicted. The disclosed methods of treating, preventing, reducing, and/or inhibiting a neurodegenerative disease can be used prior to or following the onset of a neurodegenerative disease. In one aspect, the disclosed methods can be employed 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 years, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 months, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18,
17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3 days, 60, 48, 36, 30, 24, 18, 15, 12, 10, 9, 8, 7, 6,
5, 4, 3, 2, or 1 hour prior to onset of a neurodegenerative disease; or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 hours, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19,
20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 45, 60, 90 days, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months, 2,
3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 or more years after onset of a neurodegenerative disease.
125. Alzheimer’s disease can be diagnosed by physical and neurological exam (e.g., assessment of overall neurological health by testing reflexes, muscle tone and strength, ability to get up from a chair and walk across the room, sense of sight and hearing, coordination, and/or balance), mental status and neuropsychological testing, and/or brain image (e.g., MRI or CT scan). 126. Also disclosed herein is a method for treating a neurodegenerative disease, comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of the composition disclosed herein.
127. Also disclosed herein is a method for treating a neurodegenerative disease, comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor.
128. The compositions described herein may be in any appropriate dosage form. The dosage forms can be adapted for administration by any appropriate route. Appropriate routes include, but are not limited to, oral (including buccal or sublingual), rectal, epidural, intracranial, intraocular, inhaled, intranasal, topical (including buccal, sublingual, or transdermal), vaginal, intraurethral, parenteral, intracranial, subcutaneous, intramuscular, intravenous, intraperitoneal, intradermal, intraosseous, intracardiac, intraarticular, intravenous, intrathecal, intravitreal, intracerebral, gingival, subgingival, intracerebroventricular, and intradermal. Such formulations may be prepared by any method known in the art.
129. In some embodiments, the composition is contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
130. In some aspects, disclosed herein is a method of treating a neurodegenerative disease, comprising administering to a subject a composition that is capable of crossing bloodbrain barrier, wherein the composition comprises the composition disclosed herein.
131. In some aspects, disclosed herein is a method of treating a neurodegenerative disease, comprising administering to a subject a composition that is capable of crossing bloodbrain barrier, wherein the composition comprises a caveolin-1 (CAV-1) inhibitor.
132. In some embodiments, the pharmaceutically acceptable carrier comprises a nanoparticle or docosahexaenoic acid (DHA).
133. The disclosed nanoparticles or liposomes may be engineered to a ligand binding to a receptor expressed ubiquitously expression on a cell of CNS or brain (e.g., a ligand binding to low density lipoprotein receptor-related protein (LPR)-1 and/or LPR-2).
D. Methods of treating lymphedema
134. As discussed above, the process of lymphangiogenesis involves the formation of new lymphatic vessels from pre-existing lymphatics; this occurs during embryonic development, wound healing and in various pathological contexts, including cancer. Therapeutic lymphangiogenesis can be therapy strategy for the treatment of lymphedema. Lymphedema refers to localized swelling of the body (such as at arms and legs) caused by an abnormal accumulation of lymph. Lymphedema includes primary and secondary lymphedema. Primary lymphedema is an inherited condition caused by problems with the development of lymph vessels in the body. Specific causes of primary lymphedema include: Milroy's disease (congenital lymphedema). "Secondary lymphedema" means a lymphedema caused by inflammatory or neoplastic obstruction of lymphatic vessels, and includes without limitation accumulation of ascites fluid due to peritoneal carcinomatosis or edema of the arm or other limbs following surgery or radiotherapy for breast cancer and other tumor types. Secondary lymphedema may also result from a trauma, a crush injury, hip or knee surgery, amputations, blood clots, vein grafts from cardiac surgery, chronic infections, or longstanding circulatory problems such as chronic venous insufficiency or diabetes. Accordingly, disclosed herein is also a method of preventing, inhibiting, mitigating, and/or treating lymphedema in a subject, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and/or ii) a caveolin-1 (CAV-1) inhibitor. In some embodiments, the composition comprising the APOA1 binding protein (AIBP) polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV-1 inhibitor.
135. Accordingly, the method disclosed herein can treat, decrease, mitigate, and/or prevent lymphedema and/or a symptom thereof (e.g., swelling of part or all of the arm or leg, including fingers or toes, restricted range of motion, and/or hardening and thickening of the skin (fibrosis)). It should be understood and herein contemplated that the extent of effect of treating, decreasing, mitigating, and/or preventing lymphedema and/or a symptom thereof is relative to a control (e.g., a subject not being administered with the composition).
136. The polynucleotide disclosed herein can be contained in a vector (including, for example, a viral vector such as an adeno-associated virus (AAV) vector). In some embodiments, the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide. In some embodiments, the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody. In some embodiments, the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR-Casl3.
137. In some embodiments, the composition disclosed herein further comprises a stimulator for VEGFR3. In one example, the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof (e.g., VEGFC(C156S)). In one example, the stimulator comprises Pioglitazone. Although VEGFR3 stimulator (for examples, VEGFC) can improve cognition in aged subjects, it also elicits deleterious consequence such as vascular leakage and angiogenesis. AIBP can protect against vascular leakage caused by VEGFR3 stimulatory. Accordingly, in some embodiments, the compositions disclosed herein that comprises a stimulator for VEGFR3 and an APO Al binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide. In some embodiments, the composition disclosed herein that comprises i) a stimulator for VEGFR3 and ii) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, in some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes an AIBP polypeptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFC polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide and ii) a polynucleotide that encodes a VEGFA polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, ii) a polynucleotide that encodes a VEGFC polypeptide, and iii) a polynucleotide that encodes an AIBP polypeptide. In some embodiments, the method disclosed herein comprises administering a subject in need an AAV vector comprising i) a polynucleotide that encodes a CSD peptide, iii) a polynucleotide that encodes a VEGFA polypeptide, and ii) a polynucleotide that encodes an AIBP polypeptide.
138. Animals with caveolin-1 deleted exhibit hyperpermeable vasculature and increased tumor angiogenesis. Conversely, the study herein shows that administration of a VEGFR3 stimulator and a caveolin-1 inhibitor (e.g., a CSD peptide) promotes lymphangiogenesis and mitigate s/prevents angiogenesis and vascular leakage. Accordingly, in some examples, the compositions disclosed herein that comprises a stimulator for VEGFR3 and a CAV-1 inhibitor. In some embodiments, the CAV-1 inhibitor is a CSD peptide.
139. It is understood and herein contemplated that the timing of lymphedema onset can often not be predicted. The disclosed methods of treating, preventing, reducing, and/or inhibiting lymphedema can be used prior to or following the onset of lymphedema. In one aspect, the disclosed methods can be employed 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 years, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 months, 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3 days, 60, 48, 36, 30, 24, 18, 15, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 hour prior to onset of lymphedema; or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 hours, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 45, 60, 90 days, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 15, 18, 24, 30, 36, 48, 60 or more years after onset of lymphedema.
140. Lymphedema can be diagnosed by MRI scan, CT scan that can reveal blockages in the lymphatic system, Doppler ultrasound (for checking blood flow and pressure by bouncing high-frequency sound waves (ultrasound) off red blood cells), and/or radionuclide imaging of the lymphatic system.
141. Also disclosed herein is a method for treating lymphedema, comprising diagnosing a subject as having lymphedema; and administering to the subject a therapeutically effective amount of the composition disclosed herein.
142. Also disclosed herein is a method for treating lymphedema, comprising diagnosing a subject as having lymphedema; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor.
143. In some embodiments, the composition is contained in or conjugated to a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutically acceptable carrier comprises a nanoparticle or a liposome. Accordingly, in some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, and ii) a polynucleotide that encodes a VEGFC peptide is contained or conjugated to a nanoparticle. In some embodiments, the composition comprising i) a polynucleotide that encodes an AIBP polypeptide, ii) a polynucleotide that encodes a VEGFC peptide, and iii) a polynucleotide that encodes a CSD peptide is contained or conjugated to a nanoparticle.
144. The disclosed nanoparticles or liposomes may be engineered to a ligand binding to receptor or markers expressed on lymphatic endothelial cells, such as CD34, Proxl, podoplanin, LYVE-1, and/or VEGFR-3.
VIE EXAMPLE
Example 1. AIBP-CAV1-VEGFR3 AXIS DICTATES LYMPHATIC CELL FATE AND CONTROLS LYMPHANGIOGENESIS
A. Results
145. Depletion of Aibp2 impairs lymphangiogenesis in zebrafish. Lymphatic vessel formation was investigated in the recently generated Aibp2 knockout zebrafish. The apoalbp2~/~ zebrafish were crossbred with flila:egfp zebrafish that express EGFP in the both the lymphatic and blood ECs, generating apoalbp2~/~,'flila:egfp. The TD is the first functional lymphatic vessel formed in the zebrafish trunk, situated between the dorsal aorta (DA) and PCV. At 5 days post fertilization (dpf), control siblings have a clearly demarcated TD running between the DA and PCV, whereas -90% of apoalbp2~/~ embryos displayed <30% length of normal TD. Notably, -80% of mutants showed a complete loss of this primary lymphatic vessel (FIGS. 1A and IB). Since the TD arises from the migration of PLs from the horizontal myoseptum, the development of PLs in the presence or absence of Aibp2 was assessed. As illustrated in FIGS. 1C and ID at 48 hpf, the PL string was normally formed in control embryos, but was completely abolished in -82% of apoalbp2~/~ animals and formed in a few segments (0-30%) in -8% of animals. Similar developmental defects of TD and PLs were observed by morpholino antisense oligo (MO)- mediated apoalbp2 knockdown, which were restored to a large extent by co-injection of apocilbp2 mRNA (FIGS. 9A-9C). Impaired TD formation can also be found in apoalbp2fi lyvel.Dsred zebrafish (FIGS. 9D-9E), which express DsRed in the lymphatic and venous ECs. Taken together, these data show that Aibp2 regulates lymphangiogenesis through controlling LEC fate specification.
146. AIBP enhances LEC lineage specification in zebrafish and in the mouse embryonic stem cells (mESCs) to LEC differentiation model. The defects in PL formation encouraged us to explore whether Aibp2 has any effect on lymphatic progenitor specification by immunostaining of Proxl at 36 hpf. Proxl is the master transcription factor that dictates LEC fate specification, and is used as a readout of LEC identity. In control siblings, - 3 of Proxl- positive LEC progenitors per 6 segments were found in the PCV. By contrast, Aibp2 knockout markedly reduced the number of lymphatic precursors to -1 (FIGS. 2A and 2B), indicating that Aibp2 regulates the development of lymphatic progenitors. The lymphatic vessel formation was also analyzed in embryos injected with apoalbp2 MO at 36 hpf. Reduced numbers of LEC progenitors found in Aibp2 morphants corroborates the Aibp2 effect on lymphatic progenitor specification (FIGS. 10A and 10B).
147. Next determined was whether the role of AIBP in lymphatic progenitor specification is evolutionarily conserved. Mouse embryoid bodies (mEBs) prepared from mESCs can be differentiated into the derivatives of ectodermal, mesodermal, and endodermal tissues and recapitulate certain developmental processes; the LECs emerge from the mesoderm. mEBs were prepared and LEC generation was assessed in the differentiation medium containing BMP4 and bFGF followed by additional supplement on day 3 through day 7 with recombinant VEGFA and VEGFC in combination or with AIBP alone (FIG. 2C). From day 0 onwards during differentiation, LECs were identified by the expression of PECAM, LYVE1 and PROXL Compared with control cells, VEGFA and VEGFC co-treatment increased both the mRNA and protein expression levels of these LEC-associated markers, LYVE1 and PROXI, at day 5 and day 7 of differentiation (FIGS. 2D-2G). AIBP incubation strikingly increased LEC progenitor specification as evidenced by robust expression of the LEC markers (FIGS. 2D-2G), comparable to the effect of VEGFA/C co-administration. A greater percentage of PECAM+/LYVE1+ LEC population was detected by FACS analysis at day 7 of LEC differentiation in the presence of AIBP or VEGFA/C (FIGS. 11A and 1 IB). These results demonstrate that AIBP regulation of LEC fate is conserved across evolution.
148. Aibp2-mediated cholesterol efflux is essential for proper lymphangiogenesis . Previous studies document that AIBP accelerates cholesterol efflux from ECs. Thus, free cholesterol levels in apoalbp2~/~ embryos were first measured. Indeed, Aibp2 depletion significantly increases free cholesterol content in apoalbp2~/~ embryos (FIG. 3A). Then apoalbp2~/~ animals were treated with atorvastatin, a cholesterol-lowering drug, and assessed the attendant effect on lymphangiogenesis. It was found that -93% of apoalbp2~/~ embryos displayed <50% TD formation, and within this population, -80% of the embryos lacked the TD. In contrast, atorvastatin treatment substantially reduced the percentage of apoalbp2~/~ mutants with incomplete TD development, showing that atorvastatin-mediated reduction of cell cholesterol content rescues lymphatic vessel formation (FIGS. 3B and 3C). In addition, APOA1 was overexpressed, the major protein constituent of HDL, in apoalbp2 knockout zebrafish. Similarly, enforced human APOA1 expression corrected TD formation in apoalbp2 knockout animals, as evidenced by -76% of apoalbp2 /_ zebrafish receiving APOA1 mRNA show rescued TD development (FIGS. 3D and 3E). These results indicate that Aibp2 regulates lymphatic vessel formation through cholesterol metabolism.
149. AIBP-mediated cholesterol efflux disrupts caveolae and enhances VEGFC/VEGFR3 signaling in human LECs. CAV-1, a structural protein organizing the formation of caveolae, binds cholesterol directly, thereby forming cholesterol-rich microdomains, which provide a platform to facilitate membrane-anchored receptor signaling. CAV-1 ablation in mice eliminates caveolae. Both the expression of CAV-1 and the formation of caveolae are dependent on cellular cholesterol levels. In many cases, CAV-1 deficiency alters plasma membrane receptor signaling competence through disruption of caveolae.
150. Following LEC specification from the venous ECs, VEGFR3 signaling is required for the maintenance of the LEC progenitor fate, as well as LEC proliferation and migration. CAV-1 in caveolae was reported to repress VEGFR3 activity in the ECs. Next experiment was performed to assess that AIBP-mediated cholesterol efflux regulates lymphangiogenesis through CAV-1 -dependent VEGFC/VEGFR3 signaling. As shown in FIGS. 4A and 4B, compared with VEGFC treatment in human LECs (hLECs), methyl beta cyclodextrin (MpCD)-mediated cholesterol depletion profoundly increased VEGFC-induced VEGFR3 activation, indicating that caveolae abundance and associated CAV-1 bioavailability inhibit VEGFR3 signaling. Given that AIBP-mediated cholesterol efflux disrupts caveolae, whether AIBP exerts its effect on CAV-1 -regulated VEGFC/VEGFR3 signaling via modulation of caveolae was tested. Detergent-free, discontinuous gradient ultracentrifugation analysis of hLECs was performed, which were pre-incubated with control media, AIBP, HDLs, or their combination. VEGFR3 was present in the caveolar fractions that were positive for CAV-1 (FIG. 4C). AIBP and HDLs treatment in concert, which disrupts caveolae, decreased CAV-1 localization in the caveolar fraction and induced VEGFR3 redistribution from the caveolar to the non-caveolar domains (FIG. 4C). To further investigate the effect of AIBP-mediated cholesterol efflux on VEGFR3 activation, hLECs were treated with AIBP, HD L , or their combination, followed by stimulating cells with VEGFC. AIBP or HDLa alone did not significantly affect VEGFC-induced activation of AKT and ERK, the two downstream effector kinases. However, AIBP and HDLa in combination, similarly to the treatment with MpCD (FIG. 4B), facilitated VEGFC-induced VEGFR3 activation as shown by markedly increased phosphorylation of AKT and ERK (FIGS. 4D-4F). These results show that AIBP-mediated cholesterol efflux disrupts plasma membrane caveolae, thereby facilitating lymphangiogenesis by augmenting VEGFR3 activation.
151. Disrupted CAV-1 and VEGFR3 interaction augments VEGFR3 signaling in hLECs. Whereas Cav- 1 is a core component of caveolae, it is also a scaffolding protein that provides a spatially restricted platform for proper signaling of cell surface receptors. Cav-1 contains a signaling motif that interacts with a variety of membrane receptors and modulates their activities. VEGFR3 contains such a conserved motif (FIG. 5A), showing regulation of its signaling by CAV-1. A VEGFR3 mutant VEGFR3AAA that lacks the three conserved amino acids in the hypothetical CAV-1 binding domain was generated. EGFP, VEGFR3, or VEGFR3AAA was overexpressed in hLECs using lentivirus-mediated gene transfer and examined Cav-1/VEGFR3 interaction. As illustrated in FIG. 5B, wild-type VEGFR3 but not VEGFR3AAA binds CAV-1 in hLECs.
152. To test the effect of the VEGFR3AAA mutant on VEGFR3 signaling, EGFP, VEGFR3 and VEGFR3AAA were ectopically expressed in hLECs using lentivirus-mediated transfection, followed by stimulation with VEGFC, and assessed VEGFR3 activation. The results show that the triple Ala point mutations, which disrupt its interaction with CAV-1 (FIG. 5B), results in potent VEGFC-induced VEGFR3 phosphorylation as well as AKT and ERK activation compared to wild-type VEGFR3 (FIGS. 5C-5F). Taken together, these data show that AIBP-mediated cholesterol efflux disrupts plasma membrane caveolae and mitigates CAV-1- mediated inhibition of VEGFR3 signaling in LEC.
153. Cciv-1 regulates LEC progenitor development. To explore the role of Cav-1 in lymphatic vessel formation, cav-l~/~ mutant zebrafish were generated (FIGS. 12A-12C), in which Western blot analysis revealed the absence of Cav-1 protein (FIG. 12D). Morphologically, Cav- 1 null zebrafish are comparable to control animals from 6 hpfto 5 dpf (FIG. 12E). Cav-1 knockout zebrafish showed greater numbers of Proxl labeled LEC progenitors at 36 hpf (FIGS. 6A and 6B). However, ID formation appeared normal in these animals (FIGS. 6C and 6D). Cav- 1 depletion or overexpression resulted in profoundly increased or reduced expression of lymphatics-associated genes at 96 hpf, respectively (FIGS. 13A and 13B). Thus, these data show that Cav-1 suppresses lymphatic progenitor development.
154. Loss of Cav-1 rescues defective lymphangiogenesis in Aihp2-deficient zebrafish. As shown above, Aibp2 knockout increases free cholesterol content (FIG. 3A), which increases caveola content, as cholesterol is enriched in the caveolae. Given that CAV-1 -organized caveolae negatively regulate VEGFR3 signaling, the effect of Cav- 1 deletion on lymphangiogenesis was examined in Aibp2-deficient animals. Absence of Cav-1 rescued impaired lymphatic progenitor development in Aibp2 knockdown animals as revealed by recovered Proxl staining (FIGS. 6A and 6B). Consistent with this rescue effect, defective TD formation was restored in the Cav-1- and Aibp2- double deficient animals (FIGS. 6C and 6D). The data show that Cav-1 functions downstream of Aibp2-mediated cholesterol efflux.
155. AIBP-mediated cholesterol efflux promotes adult lymphangiogenesis in mice. Since Cav-1 depletion rescues the TD development in Aibp2 morphants, the next experiment was performed to determine whether Aibp2, by alleviating Cav-1 repression of Vegfr3, facilitates lymphangiogenesis. This mechanism was further investigated in a murine model to determine whether this mechanism is evolutionarily conserved. Indeed, loss of CAV-1 significantly augmented tail lymphangiogenesis in P3 neonatal mice as revealed by LYVE-1 antibody staining of lymphatic vessels in the similar anatomic location (FIGS. 14A and 14B). The corneas of adult mice lack lymphatic vasculature, thereby providing a widely used model to study injury or growth factor-induced lymphangiogenesis (FIG. 7 A). To explore the role of cholesterol efflux in adult lymphangiogenesis, VEGFC-containing pellets were implanted, in the presence or absence of AIBP and the core protein component of HDL - APO Al or the CAV-1 blocking peptide (CAV-1 scaffolding domain, CSD) --- into the corneas of B6 mice. APOA1 was used due to technical difficulties in the preparation of HDL pellets. As expected, VEGFC pellet implantation elicited robust lymphangiogenesis (FIG. 7B). Remarkably, co-administration with AIBP and cholesterol acceptor APOA1 further increased lymphangiogenesis (FIGS. 7B and 7C). Similarly, supplement of CSD augmented lymphatic vessel formation (FIGS. 7B and 7C). However, no pro-lymphangiogenic effect was found with AIBP/APOA1 or CSD treatment alone (FIG. 15). Thus, consistent with the studies in zebrafish, AIBP-mediated cholesterol efflux increases lymphangiogenesis in neonatal and adult mice. Additional CSD peptides were also tested. Both peptides, Cav-i and Cav-M, showed the effects of improving lymphangiogenesis (FIG. 21).
156. Reduced AIBP expression in the affected epidermis of lymphedema patients. To probe the association of AIBP content with lymphatic vessel dysfunction, AIBP expression was examined in human skin specimens derived from 7 subjects. Paired cutaneous normal and lymphedematous biopsy specimens were obtained from each of these individuals. Immunohistochemistry staining of AIBP showed that it is robustly expressed in the epidermis and the sweat glands (FIG. 8A). In human lymphedema, AIBP expression is mildly but significantly reduced in the epidermis but not in the dermis or sweat glands, when compared to the paired normal cutaneous specimens (FIGS. 8A-8D). The comparable expression in the dermis and sweat gland excludes the possibility of tissue swelling elicited reduction of AIBP levels in the epidermis of lymphedema biopsy samples. The observed abnormality in the lymphedematous epidermis indicates that reduced AIBP expression is associated with impairment of cutaneous lymphatic function.
B. Discussion.
157. The studies shown herein reveal a molecular mechanism underlying lymphatic development, in which AIBP-mediated cholesterol efflux disrupts caveolae and, consequently, reduces CAV-1 bioavailability, thereby mitigating the CAV-1 inhibition of VEGFR3 activation and augmenting lymphatic fate commitment and lymphangiogenesis.
158. Opposing roles of AIBP in angiogenesis and lymphangiogenesis . Previous studies have shown that AIBP-mediated cholesterol efflux disrupts lipid rafts/caveolae, which results in restricted angiogenesis by two mechanisms: 1) it impedes rafts/caveolae anchored proangiogenic VEGFR2 clustering, endocytosis and signaling, thereby restricting angiogenesis, and 2) it translocates y-secretase from lipid rafts/caveolae to the non-raft/caveolar domain, facilitating the cleavage of NOTCH receptor and augmenting anti -angiogenic NOTCH signaling. Recently, it was demonstrated that AIBP instructs hematopoietic stem and progenitor cell specification in development through activation of SREBP2, which in turn transactivates NOTCH for hematopoiesis. Here, it is shown that AIBP increases lymphangiogenesis and instructs LEC progenitor specification. At the molecular level, AIBP-mediated cholesterol efflux attenuates the inhibitory effect of CAV-1 on VEGFR3 signaling. Thus, disruption of CAV-1 and VEGFR3 binding augments VEGFR3 signaling and LEC progenitor development and lymphangiogenesis. Although LECs are derived from blood ECs (BEC), their functions and molecular identifications differ. The fundamental differences between BECs and LECs contribute to the disparate effects of AIBP on angiogenesis and lymphangiogenesis.
159. It is not without precedent that one protein may have the opposing effects on angiogenesis and lymphangiogenesis. For instance, RhoB ablation augments lymphangiogenesis but impairs angiogenesis in the cutaneous wound healing model. Although Notch signaling limits angiogenesis, Notch has been reported to inhibit or augment lymphangiogenesis in a context-dependent fashion.
160. AIBP-mediated CAV-1 dependent regulation ofVEGFR3 signaling and lymphangiogenesis. Lymphatic vessel formation requires LEC fate commitment and subsequent sprouting. The LEC fate acquisition, LEC sprouting from the embryonic CV, and dorsolateral migration under the control of VEGFC/VEGFR3/PROX1 signaling axis is highly conserved. Restricted expression of PROXI protein dictates LEC identity , which is critical for future LECs to migrate out of the PCV. AIBP is evolutionarily conserved from zebrafish to human. Zebrafish have two apoalbp genes that encode Aibpl and Aibp2 proteins, and Aibp2 demonstrates functional properties resembling those of human AIBP. Although the degree of phenotypic severity varies, most of the Aibp2 mutants show a partial or complete loss of PLs in the horizontal myoseptum region and, thus, lacking large portion or all of the TD arising from PLs. Furthermore, these defects are attributed to a failure of LEC lineage commitment, as Aibp2 loss- of-fiinction impairs lymphatic precursor specification (FIGS. 2A and 2B).
161. A positive feedback loop between VEGFR3 and PROXI is necessary to maintain the identity of LEC progenitors in mice and zebrafish. CAV-1 has been reported to inhibit VEGFR3 signaling in ECs. The studies herein show that Cav-1 knockdown or overexpression in zebrafish increases or decreases expression of lymphatic genes, respectively. Moreover, loss of Cav-1 strongly enhances lymphatic progenitor specification and rescues impaired lymphangiogenesis. Despite reduced number of LEC progenitors, the TD formation appears normal in Cav-1 null zebrafish (FIGS. 6C and 6D). Indeed, there are considerable variations in the number of LECs required to form a complete TD. CAV-1 deficiency augments lymphangiogenesis in mouse development and in adult cornea with VEGFC stimulation (FIGS. 7A-7C and FIG. 14). In hLECs, VEGFR3 is present in caveolae and contains a binding motif for CAV-1 (Fig. 5A). The disruption of the CAV-1 and VEGFR3 interaction increases VEGFR3 activation in response to VEGFC treatment (FIGS. 5C-5F). These results show that AIBP- mediated cholesterol efflux facilitates lymphangiogenesis, which is achieved through abolishing the CAV-1 -mediated inhibition of VEGFR3 signaling.
162. AIBP effect on ERK signaling. ERK activation controls LEC fate specification and LEC sprouting. A modest but sustained ERK activation in mice, resulting from the expression of a RAF mutant, results in lymphatic hyperplasia. Expression of the Vegfr3 mutant that abolishes Erk activation in zebrafish impairs development of Proxl+ LECs. These data indicate that AIBP enhances VEGFC-mediated ERK activation in hLECs (Fig. 4D-F). Conversely, Aibp2 knockdown in zebrafish abrogates Erkl activation while robustly facilitating Akt activation. Notably, VEGFR3 homodimer activates ERK while VEGFR2/VEGR3 heterodimer activates AKT. These studies are consistent with the dominant role of ERK in lymphatic vessel development.
163. Conserved role of AIBP in the regulation of LEC fate and lymphangiogenesis . In the mESCs to the LEC differentiation model, it was found that recombinant AIBP incubation documents a remarkable effect on the LEC specification arising from mesodermal origin. Given that AIBP alone shows no observable effect on VEGFR3 phosphorylation, this AIBP effect on LEC fate commitment is due to a synergistic effect of AIBP and increased VEGFR signaling in the induced mesoderm. Although AIBP is unlikely to initiate a ligand-independent signaling, it fine-tunes VEGFC/VEGFR3/ERK signaling.
164. Earlier studies suggest that LECs emerge from the venous origin. However, other mechanisms of lymphatic vessel formation have been reported recently, such as in the zebrafish facial lymphatics, murine dermal and cardiac lymphatic vessel formation, and zebrafish brain. The Aibp2 effect on lymphatic vessel formation is blunted at later developmental stages in zebrafish, and no significant difference was detected in tail lymphangiogenesis between AIBP knockout neonatal mice and control littermates, which can be due to the rescue effect of non-venous origin derived lymphangiogenesis.
165. It often takes years to develop secondary lymphedema, an acquired damage to the lymphatic system occasioned by surgical disruption, radiotherapy, or infection. Genetic studies show that small changes in an array of lymphangiogenic genes, in the absence of a single dominant mutation, can precipitate secondary lymphedema. The AIBP staining of paired human normal/lymphedema cutaneous biopsy specimens disclosed that AIBP expression is significantly reduced in the epidermis in the settings of lymphedema, when compared to the paired, normal counterpart. Whereas it is a mild change, this can be indicative of a general attenuation of lymphangiogenic competence in the lymphedematous tissues.
166. The Lymphatic contribution is implicated in the pathogenesis of a variety of diseases, including tumor metastasis, cardiovascular disease, obesity, and diabetic mellitus. Therapeutic augmentation of lymphangiogenesis has been documented to improve lymphatic structure and function. The mechanistic study of AIBP-regulated lymphangiogenesis aids in the development of new therapies for the diseases associated with lymphatic dysfunction.
167. In addition, the meningeal lymphatics are essential for transportation of brain interstitial fluid and cerebral fluid. Dysfunction of the meningeal lymphatics is associated with dementia, such as Alzheimer disease (AD) pathology. VEGFC-mediated enhancement of meningeal lymphatics function improves cognition in aged mice, and disruption of meningeal lymphatic vessels in transgenic AD mouse models aggravates P-amyloid
(AP) accumulation. Whether AIBP or CSD-mediated enhancement of lymphatic function can rescue cognitive impairment in the aged mice and in the mouse AD model with Ap accumulation is assessed.
C. Materials and Methods
168. Generation of Cav-1 knockout zebrafish using CRISPR/Cas9. The CRISPR/Cas9-mediated gene knockout technique was performed as previously described. Briefly, the target sequence of caveolin 1 (Cav-1, NCBI Gene ID: 323695) was 5’- GACGTGATCGCCGAGCCTGC-3’ (SEQ ID NO: 8). The guide-RNA (gRNA) sequence was subcloned into pT7-gRNA vector and gRNA was synthesized using HiScribeTM T7 Quick High Yield RNA Synthesis (New England Biolabs). The pCS2-nCas9n was linearized using Notl and Cas9 mRNA was synthesized using mMESSAGE mMACHINE SP6 Kit (Invitrogen). Twenty- five ng/01 gRNA and 200 ng/pl Cas9 mRNA were mixed and injected into one-cell stage wild type embryos. The founder zebrafish were raised to adult. Following genotype verification using Sanger DNA sequencing, cavl+/~ animals were used for experiments.
169. Mouse cornea lymphangiogenesis. VEGFC pellet preparation and implantation were carried out as previously described with minor modifications. Briefly, pellets containing recombinant VEGFC (5 mg/ml), AIBP (10 mg/ml) in combination with apoA-1 (15 mg/ml), CAV-1 scaffolding domain (CSD; 5 mg/ml), VEGFC with AIBP and APOA1 or with CSD were prepared by mixing with 10% sucralfate solution (w/v in PBS) and 12% poly-HEMA (w/v in ethanol). The aforementioned components were mixed as following: 5 pl of poly-HEMA, 1 pl of sucralfate, 2 pl VEGFC, 2 pl AIBP, 2 pl APOA1, 2 pl CSD, and the final volume adjusted to 10 pl with PBS. The mixture droplet was put on the PARAFILM, UV irradiated for 15 minutes, and air-dried in the cell culture hood. Dried pellets were used immediately or stored at 4 °C before usage.
170. The mouse was anesthetized with isoflurane, and one drop of 0.5% proparacaine HCI was applied to the cornea. Five minutes later, one mouse eye was properly orientated under a dissection microscope. A gentle cut was made with a von Graefe cataract knife from the center of the cornea, and a pocket with ~ 1.5-2 mm2 size was generated by inserting the knife toward comeal limbus, with a few gentle waggles inside. The pellets were then inserted into the cornea pocket using forceps and von Graefe cataract knife, with the pellet flattened to secure the implantation. The ophthalmic antibiotic ointment was applied topically to the injured eye. Following 10 days of pellet implantation, the cornea was dissected and washed in PBS for later processing.
171. Tissue biopsy. Cutaneous punch biopsy was performed as previously described. After informed consent was obtained, two contiguous 6 mm full thickness punch biopsy specimens were derived from the medial aspect of the forearm or calf of the affected extremity with an AcuPunch disposable device. Biopsy specimens were immediately place in formalin and processed for subsequent studies.
172. Immunohistochemistry (IHC) staining of human lymphedema tissues. AIBP antiserum were generated in rabbits using tag-free recombinant human AIBP proteins. AIBP antibody was subsequently purified from the AIBP antiserum using affinity purification, where AIBP antigen column was prepared by covalently conjugating recombinant AIBP to the aminereactive, beaded-agarose resin (Thermo Fisher, Pierce NHS-Activated Agarose, Cat # 26196).
173. The 5um human specimens were deparaffinized, and rehydrated serially through xylene, 100%, 80%, and 70% ethanol, and finally rinsed in milliQ water. The resulting sections were subjected to antigen recovery by immersing in 100°C citrate buffer (pH6.0) for 10 minutes. An incubation with 3% hydrogen peroxide was applied to eliminate the endogenous peroxidase activity. Following rinsing in 1XTBST (diluted from 10X TBST wash buffer, Dako, Cat # S3006), the slides were further washed and blocked in protein block serum-free buffer (Dako, Cat # X0909). The slides were incubated with AIBP antibody for 1 hour at room temperature. Following proper TBST wash, the slides were sequentially incubated with biotinylated antirabbit (1:200, Vector, Cat # BA-9500) and Avidin conjugated alkaline phosphatase (Vector, Cat # AK-5000) for 30 minutes at room temperature. AP blue substrate mixture (Vector, Cat # SK- 5300) was administered for final color development. The slides were mounted in the aqueous mounting medium. 174. Zebrafish husbandry. All the wild-type AB and transgenic zebrafish lines were maintained as previously described and in accordance with Houston Methodist Research Institute Animal Care and Use Committee (IACUC) regulations and under the appropriate project protocols.
175. MO and mRNA injections. Morpholino antisense oligos (MOs) were synthesized by Gene Tools. The following MOs were used in this study 1) control: 5’- CCTCTTACCTCAGTTACAATTTATA-3’ (SEQ ID NO: 14), and 2) apoalbp2 (NCBI Gene ID: 557840): 5’-GTGGTTCATCTTGATTTATTCGGC-3’ (SEQ ID NO: 15). The working concentrations of control and aopcilbp2 MOs were 0.2 mM and 0.1 mM, respectively. Equal volume of aopcilbp2 MO (0.1 mM) and aopocilbp2 mRNA (200 ng/pl) were mixed to perform the rescue assays. The working concentration of human APOA1 mRNA was 100 ng/pl. One nl of MO, mRNA or the combination of MO with mRNA were injected into one-cell stage embryo using the microinjector FemtoJet® 4i (Eppendorf).
176. Chemical treatments. For 4OHT treatment, embryos were treated with 5 pM 4OHT (Sigma, Cat # H7904,) from 6 to 36 hours post fertilization (hip). For atorvastatin (Sigma, Cat # PZ0001) treatment, embryos were treated with 1 pM atorvastatin from 6 to 120 hpf. All control embryos were incubated with equal amount of ethanol at the indicated time points.
177. Cell culture. Human dermal lymphatic microvascular endothelial cells (hLECs) (Angio-Proteomie, Cat # cAP-0003) were cultured in Endothelial Cell Medium (ScienCell, Cat # 1001). Methyl-P-cyclodextrin (Sigma, Cat # C4555) was prepared in serum-free ECM medium with a working concentration of 10 mM. HDL3 isolation was performed as previous described. The hLECs were pretreated with 200 ng/ml AIBP protein, 100 pg/ml HDL3, or the combination of AIBP with HDL3 for 4 hours, washed with PBS, and followed by incubation with 100 ng/ml VEGFC (R&D, Cat # 9919) in pre-warmed serum-free ECM medium for 20 min.
178. Lenti-virus infection. The hLECs were transduced with the third generation lentiviral system as previous described (81). After the cells were infected with human VEGFR3, VEGFR3AAA or CAV1 overexpression Lenti-virus for 96 hours, cells were subjected to 5 pg/ml puromycin (ThermoFisher, Cat #A1113803)) selection to generate pool cells stably expressing the transduced genes.
179. Quantitative PCR. The qPCR was performed as previously described. The gene information and the primers used in this study were: murine reference gene Efla (GeneBank Accession # NM 010106.2), Efla-F (5’-GATCGATCGTCGTTCTGGTAAG-3’) (SEQ ID NO: 16) and Efla-R (5’-AGTGGAGGGTAGTCAGAGAAG-3’) (SEQ ID NO: 17); CD31 (GeneBank Accession # NM 008816.3), CD31-F (5’- CAACAGAGCCAGCAGTATGA) (SEQ ID NO: 18) and CD31-R (5’- TGACAACCACCGCAATGA-3’) (SEQ ID NO: 19); Lyvel (GeneBank Accession # NM 053247.4), Lyvel-F (5’- CCTTGTTGGCTGAGACTGTAA) (SEQ ID NO: 20) and Lyvel-R (5’- CTAGAGAACACCAGCAACAGTAA-3’) (SEQ ID NO: 21); zebrafish reference gene efla (GeneBank Accession # NM_131263.1), efla-F (5’- ATGCCCTTGATGCCATTCT-3’) (SEQ ID NO: 22) and efla-R (5’- CCCACAGGTACAGTTCCAATAC-3’) (SEQ ID NO: 23); lyvel (GeneBank Accession # XM 687593.6), lyvel-F (5’-GACAGCTCCCAAACAACAATAAA-3’) (SEQ ID NO: 24) and lyvel-R (5’-CTGAGAGGTTGAATGAGAGGAAG-3’) (SEQ ID NO: 25); vegfc (Genebank Accession # NM 205734.1), vegfc-F (5’- CGTCTCTTGATGTCTCGGAATG-3’) (SEQ ID NO: 26) and vegfc-R (5’- GCTGTTACTTTGGATCCCTCTC-3’) (SEQ ID NO: 27); vegfr3 (Genebank Accession # NM_130945.1), vegfr3-F (5’-CAGTGTGGTCACCTGGAATAA-3’) (SEQ ID NO: 28)and vegfr3-R (5’-TGGAGCAGTAGAAGCCAATAAA-3’) (SEQ ID NO: 29); proxl (Genebank Accession # NM_131405.2), proxl-F (5’- CGTGATGGATCAAGAGGAAAGA-3’) (SEQ ID NO: 30) and proxl-R (5’- CTACCTGGGACATTGCTGTATT-3’) (SEQ ID NO: 31); soxl8 (Genebank Accession # XM 001337666.3), soxl8-F (5’-GGAAGATGTGGGTCTGTCTTC-3’) (SEQ ID NO: 32) and soxl8-R (5’-TGGGAATGCTGGAGGTTATG-3’) (SEQ ID NO: 33)); coupTFII (Genebank Accession # NM 131183.1), coupTFII-F (5 CACAGGTCGCTAACCTATTT-3 ’) (SEQ ID NO: 34) and coupTFII -R (5 ’-ACAAGGGCTAGTGTACTGAATG-3’) (SEQ ID NO: 35).
180. Western blot and Immunoprecipitation. Zebrafish or hLECs were lysed on ice with the RIPA buffer (Pierce) and western blotting was performed as previously described. Immunoprecipitation was performed using the Pierce Classic Magnetic IP/Co-IP Kit (Thermo Scientific, Cat No. 88804) following the manufacturer’s instruction. The antibodies used were: anti-Phospho-Akt (Ser473) (Cell Signaling, #4060S), anti-AKT (Cell Signaling, #4685 S), anti- Phospho-ERKl/2 (Cell Signaling, #4370S), anti-ERKl/2 (Cell Signaling, #9102S), rabbit anti- P-Tubulin (Cell Signaling, #2148S), anti-Phospho-tyrosine (clone 4G10, EMD Millipore, #05- 321), anti-Flt4 (Santa Cruz, C20, Cat No. sc321), anti-CAVl (Cell Signaling, Cat No. 3238S), anti-GAPDH (Cell Signaling, #2118S), anti-mLYVEl (Angiobio, Cat No. 11034), anti-CD31 (Abeam, Cat No. ab28364), mouse anti-LAMIN A/C (Cell Signaling, Cat No. 4777S), goat antiGuinea Pig HRP -conjugated antibody (Jackson Labs, Cat No. 106-035-003), goat anti-mouse HRP -conjugated antibody (Jackson Labs, Cat No. 115-035- 003), and goat anti-rabbit HRP- conjugated antibody (Jackson Labs, Cat # 111-035-144). 181. Mouse embryonic stem cell (mESC) culture. The mESCs were cultured in feeder- and serum-free environment using ESGRO®-2i medium supplemented with leukemia inhibitory factor (LIF), GSK3P inhibitor and MEK1/2 inhibitor (EMD Millipore, Cat #. SF016- 100) according to the manufacturer’s instruction. Briefly, T25 flask was coated with 0.1% gelatin solution for 30 min at room temperature, and then 1 x 106 cells were plated onto gelatinized T25 flask in ESGRO®-2i medium and incubated at 37 °C with 5% CO2. The media were changed daily and mESCs were sub-cultured at a ratio of 1:5 when cells reached 60-90% confluence.
182. Lymphatic endothelial cell (LEC) differentiation. The mESCs differentiation to the endothelial lineage was performed as previously described with the following modifications. Briefly, mESCs were seeded onto collagen IV-coated plates and maintained in LEC differentiation medium (ESGRO Complete™ basal medium, Cat #. SCR002-500) supplemented with 2 ng/ml BMP4, 10 ng/ml bFGF, 50 ng/ml VEGFA and 50 ng/ml VEGFC for 7 days. Differentiated cells were collected on days 3, 5 and 7 for qPCR, western blot, or FACS analysis.
183. Fluorescence Activated Cell Sorting (FACS) analysis. Cells were dissociated using enzyme-free Hanks’ Balanced Salt Solution (Gibco, Cat No. 13150016), resuspended in
1 x DPBS supplemented with 2% heat-inactivated FBS. Cells were then stained with APC anti- CD31 antibody (Bio-legend, Cat No. 102409), Alexa Fluor 488 conjugated LYVE1 Monoclonal Antibody (ALY7), eBioscience™ (Cat No. 53-0443-82) and DAPI, and analyzed by BD LSR II flow cytometer. Data were analyzed using the FlowJo software.
184. VEGFR3 site-direct mutation. VEGFR3 mutation was generated using a PCR- based direct site mutation strategy. Briefly, the plasmid containing wild type VEGFR3 was amplified using primer pair VEGFR3-AAA-F (5’- ACGCAGAGTGACGTGGCGTCCGCTGGGGTGCTTCTCGCGGAGATCTTCTCTCTGGGG GCC-3 ) (SEQ ID NO: 36) and VEGFR3-AAA-R (5’- AGAGAGAAGATCTCCGCGAGAAGCACCCCAGCGGACGCCACGTCACTCTGCGTGGT GTAC-3’) (SEQ ID NO: 37). The PCR program was 95 °C for 2 min, 18 cycles of 95 °C for 30 s, 68 °C for 1 min per kilobase of plasmid length, then 68 °C for 7 min and hold at 4 °C. One pl Dpnl was added to the PCR products to disrupt the plasmid template and incubated at 37 °C for
2 hours, and then 5 pl PCR products were transformed into stbl3 competent cells. The resulting clones were selected, plasmid extracted, and DNA sequenced. The one with expected mutation was used for subsequent experiments.
185. Proxl staining. Zebrafish larvae at 36 hpf were fixed in 4% paraformaldehyde for 2 hours at room temperature. Following fixation, larvae were washed with PBS, dehydrated through 25%, 50%, 70% and 100% methanol/PBS series and stored at - 20 °C in 100% methanol for later usage. Before usage, larvae were rehydrated through 75%, 50% and 25% methanol/PBS series, washed with PBS/0.1% Tween (PBST), and permeabilized in pre-chilled acetone for 20 min. The resulting animals were then washed with PBST and blocked in 10% BSA/PBST overnight at 4 °C. Following incubation with Proxl antibody (R&D, Cat No. AF2727) and GFP antibody (ThermoFisher, Cat # A 10262) for 3 days at 4 °C, and after proper wash with PBST, the larvae were incubated overnight at 4 °C in dark with secondary antibody diluted in 1% BSA/PBST. Following extensive PBST wash, the immunostained animals were imaged using Olympus FV 3000 confocal microscope.
186. Dissection of the tail skin of neonatal mice. After euthanization, the tails of Cavl /_or littermate control at P3 were dissected, with the tail tips cut off. The tail bones and extra connective tissues were then manually removed. Tails were incubated in 20 mM EDTA/PBS overnight at 4°C to facilitate dermis isolation. The dissected dermis was washed with PBS for subsequent immunostaining as described below.
187. Immuno-fluorescence (IF) staining of mouse cornea and tail skin. For IF staining, cornea and tail skin were fixed in 4% paraformaldehyde for 2 hr at room temperature. Subsequently, samples were washed 5 times with 0.05% tween/PBS (0.05% PBS-tw) and blocked with the blocking buffer (1% goat or donkey serum, 1% BSA, and 0.5% Triton X-100 in PBS) for 3-4 hr at room temperature. Samples were incubated with LYVE-1 antibody (1: 1000; Angiobio, Cat # 11-034) and CD31 antibody (1: 1000; BD Biosciences, Cat. 553369) diluted in the blocking solution for 2 days and with secondary antibodies (1 : 1000; Jackson ImmunoResearch Inc., AB_2338052 and AB_2338372) overnight at 4 °C following proper wash with 0.5% Triton X-100/PBS (0.5% PBS-tx). The immunostained tissues were flat-mounted in the mounting medium (VECTASHIELD, H-1000) and stored at 4 °C before imaging.
188. Confocal imaging and image quantification. Flatmounted tissues were imaged using Nikon Al confocal microscope. Auto-stitching function was applied to generate the whole cornea image. Z-stacked images were maxi -projected and processed using Image J. Color balance and threshold were adjusted using the same threshold for image quantification.
189. For quantification of tail lymphatic vessels, images were converted to binary image by applying “Erode” or “Dilate” options as exampled (imagej.nih.gov/ij/docs/menus/process.html), and images were skeletonized. Skeleton length was quantified, and pixel value was converted to micrometer (pm).
190. For quantification of cornea lymphatic vessel (LV) and blood vessel (BV), region of interest (ROI) on each image were chosen based on the location of the VEGFC containing pellets. ROI images were processed using Image J to quantify percentage of LV or BV coverage.
191. Data analysis. Statistical comparisons between two groups were analyzed using Student t test. One-way ANOVA was used to compare the means of multiple groups. Data were expressed as the mean ± SD if n<5 or as the mean ± SEM if n > 5. p < 0.05 was considered statistically significant. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.
EXAMPLE 2. AIBP-CAV1-VEGFR3 AXIS DICTATES LYMPHATIC CELL FATE AND MODULATES ALZHEIMER’S DISEASE
192. Aging is the major risk factor for cognitive impairment, including Alzheimer’s disease (AD). Thus far, the cause of aging -associated dementia is not fully understood, and disease-modifying therapies are limited (only one was recently approved by the FDA). The two major hallmarks of AD are the accumulation of extracellular senile plaques, comprising of amyloid-beta (Ap) peptide aggregates, and the formation of intracellular aggregates of hyperphosphorylated tau. Attenuated extracellular transport of A and dysfunctional tau underlie AD and AD-related disease (ADRD). How to target the transport process, thereby eliminating AD- and ADRD-promoting components, is underexplored.
193. The meningeal lymphatics are essential for the transportation of brain interstitial fluid and cerebral fluid containing Ap aggregates. Indeed, VEGFC-mediated enhancement of meningeal lymphatics function improves cognition in aged mice. Conversely, disruption of meningeal lymphatic vessels in transgenic AD mouse models aggravates Ap accumulation. However, VEGFC can also elicit deleterious consequence such as vascular leakage.
194. It is shown herein that APOA1 binding protein (AIBP) enhances VEGFC- mediated lymphatic growth and that AIBP limits vascular leakage. In this study, slow-release nanoparticles loaded with recombinant VEGFC and AIBP are engineered, which can improve the brain lymphatic function and facilitate the removal of the AD-associated Ap aggregates and/or dysfunction Tau. Furthermore, AIBP alleviates other AD components by promoting cell cholesterol efflux, reducing neuroinflammation, and repressing y-secretase activity that generates the AD-prone Ap aggregates. Collectively, these studies guide the development of a potential paradigm shifting therapy for AD.
195. Lack of effective treatment for Alzheimer’s disease. AD is the most common cause of severe memory loss in the elderly. One out of nine people aged 65 years and about one third of people aged 85 years and older suffer from AD. Due to the aging of populations worldwide, AD is reaching epidemic proportions, posing a large socioeconomic burden. Despite the tremendous efforts in the AD research field, there is no cure for the disease. It is estimated that any new medicine that could delay the onset of AD by just 5 years could decrease the number of AD patients by 43%. Therefore, there is an urgent need for novel therapies for AD.
196. Meningeal lymphatics and AD. The neuropathological hallmark of AD is the extracellular deposition of Ap aggregates and intracellular accumulation of dysfunctional tau, thereby precipitating neuronal dysfunction and behavioral changes. As a matter of fact, the A protein was initially identified in the meningeal tissue of AD patients. The brains are constantly perfused by cerebrospinal fluid (CSF) and brain interstitial fluid (ISF), which removes macromolecules, including Ap protein aggregates and extracellular dysfunctional tau. Recent studies show that the aging-associated Ap accumulation is attributed to the progressive attenuation of meningeal lymphatic function in the brain. The meningeal lymphatics drains cerebral ISF/CSF from the central nerve system into the cervical lymph nodes in mice. Its dysfunction impairs the drainage of macromolecules in the brain fluid, resulting in neurological dysfunction and cognitive defects. Improving meningeal lymphatic function with pro- lymphangiogenic VEGFC augments the removal of macromolecular components and, consequently, rescues brain perfusion and cognition function. However, high levels of VEGFC delivery, while better promoting lymphangiogenesis, elicits neovascularization and vascular leakage.
197. AIBP improves VEGFC potency. AIBP, a secreted protein, by promoting cholesterol efflux from endothelial cell (EC), limits angiogenesis. Mechanistically, AIBP increases the binding sites of high-density lipoprotein (HDL) on ECs and promotes HDL-EC dissociation, a process that depletes the free cholesterol content of endothelial lipid rafts via the cholesterol transporter. Through a similar mechanism, AIBP acts on microglia and reduces neuroinflammation. As cholesterol deregulation precipitates neuronal Ap deposition, AIBP improves lipid metabolism and bestows multiple beneficial effects on AD. The studies show that AIBP protects against vascular leakage, which makes AIBP a key modulator that increases the pro-lymphangiogenic potential of VEGFC but limits its effect on angiogenesis and vascular leakage.
198. The data strongly show that, for the first time, AIBP connects cholesterol efflux with lymphangiogenesis. Increasing brain lymphangiogenesis is regarded as one of the potential therapeutic strategies to improve AD. Thus, these studies of the role of AIBP in meningeal lymphatic regeneration can have the major importance in developing new therapies that can ameliorate dementia and provide benefits to patients with AD and ADRD.
199. In addition to the effect of AIBP on augmenting brain lymphatic function, as focused in this proposal, AIBP also accelerates cellular cholesterol efflux, reduces neuroinflammation, and impairs y-secretase activity in lipid rafts, all of which further contribute to effective AD treatment. Conceivably, the application of AIBP-based therapy for AD introduces multi-pronged salutary effects to improve neurological function, thereby alleviating dementia.
200. AIBP-mediated cholesterol efflux promotes adult lymphangiogenesis in mice. The biochemical analysis using human LECs shows that AIBP, by alleviating CAV-1 repression of VEGFR3 activation, facilitates lymphangiogenesis. It is further determined whether this mechanism controls lymphangiogenesis in mice. The corneas of adult mice lack lymphatic vasculature, and is widely used as a model to study injury or growth factor-induced lymphangiogenesis (Figure 17A, top left panel of next page). To explore the role of cholesterol efflux in adult lymphangiogenesis, VEGFC-containing pellets were implanted, in the presence or absence of AIBP or the CAV-1 modifying peptide (CAV-1 scaffolding domain, CSD) - into the corneas of B6 mice. Five days later, the corneas were dissected and lymphatic vessel growth was assessed using anti-LYVEl antibody, the golden standard in the field. VEGFC pellet implantation elicited robust lymphangiogenesis (Figure 17B). Co-administration with AIBP or with CSD further increased lymphangiogenesis (Figures 17B-17C). Thus, AIBP-mediated cholesterol efflux increases lymphangiogenesis in adult mice. The role of AIBP in promoting lymphangiogenesis was also validated using zebrafish model.
201. AIBP enhances differentiation of LECs from mouse embryonic stem cells (mESCs). Aibp dictates LEC fate and controls lymphangiogenesis in zebrafish. To determine the conserved role of AIBP in lymphatics, murine LEC specification was investigated. The mouse embryoid bodies prepared from mESCs can be differentiated into the derivatives of ectodermal, mesodermal, and endodermal tissues and recapitulate certain developmental processes 19. The LECs emerge from the mesoderm. Mouse embryoid bodies were prepared and LEC generation was assessed in the differentiation medium containing BMP4 & bFGF followed by additional supplement on day 3 through day 7 with recombinant VEGFA and VEGFC in combination or with AIBP alone (Figure 18A). From day 0 onwards during differentiation, LECs were identified by the expression of PECAM, LYVE1 and PROXI. Compared to control cells, VEGFA and VEGFC co-treatment increased both the mRNA and protein expression levels of these LEC associated markers LYVE1 and PROXI at day 5 and day 7 of differentiation. AIBP incubation strikingly increased LEC specification as evidenced by robust expression of the LEC markers (Figures 18B and 18C), comparable to the effect of VEGFA/C co-administration. These results show that AIBP regulation of LEC fate is conserved across evolution. 202. AIBP protects VEGF elicited disruption of vascular junctions. AIBP represses angiogenesis. To determine the role of AIBP in vascular integrity, primary human retinal microvascular ECs (HMREC), after reaching confluence for 3 days, were pre-incubated with recombinant AIBP, and then stimulated with VEGF. Immunostaining of VE-Cadherin (VE- Cad) showed that VEGF treatment disrupted adherens junction structure, as evidenced by the presence of gaps between ECs. Preincubation with AIBP preserved the structural integrity (Figure 19A). Consistent with this, using the transwell permeability assay, recombinant AIBP pre-treatment reduced VEGF-induced leakage of FITC-dextran (70 kDa) across the endothelial monolayer formed on the transwell (Figures 19B and 19C). Thus, these data show that AIBP plays a previously unrecognized role in protecting EC junctions.
203. Preparation and characterization of Nano-AIBP/VEGFC. Generation of Nano-AIBP/VEGFC particles. The nanoporous silicon particles (pSi) are fabricated with well- controlled shape, size, pore sizes and surface chemistries through a sol-gel method. By coating pSi with a polymer (poly(lactic- co -glycolic) acid (PLGA), composite microspheres (PLGA- pSi) that allows the controlled release of the payload for over a month were obtained, while preserving its stability and activity. Briefly, 200 nm pSi with an average pore size of 2 nm is engineered, which can be loaded with endotoxin-low human VEGFC alone or AIBP+VEGFC. PLGA (50:50) can be dissolved in dichloromethane (10 w/v %), and PLGA-pSi microspheres with an average diameter of 10 pm are prepared through a modified S/O/W emulsion method. AIBP-loaded particles are then lyophilized and encapsulated in Cy5 -labeled 5% PLGA (50:50) shell. The kinetics of AIBP/VEGFC release from the nanoparticles into cell culture medium and its stability are determined using Western blot.
204. Nano-AIBP/VEGFC shows a prolonged and sustainable release effect. The release kinetics can be adjusted to achieve an optimal effect of AIBP/VEGFC release by varying the polymeric compound of the nanoparticles. Nano-AIBP/VEGFC is more stable and biologically more potent than unpacked recombinant AIBP/VEGFC when placed at 31° C.
205. To test the hypothesis that AIBP treatment improves cognitive function in PPNL G-F miCe. The APPNL'G'F knock-in mice express humanized AD-prone APP in the relevant cells at physiological levels. At 6 months of age, APPNL'G'F knock-in mice and their wild type littermates receive intracisternal injection of empty nanoparticle, Nano-VEGFC, Nano-AIBP/VEGFC, and recombinant AIBP/VEGFC. 12 mice are used for each treatment group for each sex. One month after administration, these mice are subjected to a battery of cognitive behavioral assays. The cognitive tests include the novel object recognition (NOR), Y- maze, Morris water maze (MWM), radial arm water maze (RAWM), and fear conditioning. Following these assays, mice are euthanized and perfused with 10% Formalin. Brains are collected and the brain sections subjected to immunostaining for VEGFC, AIBP and Ap. The dura mater containing meningeal lymphatics is isolated and immunostained using LYVE1 and VEGFR3 antibodies. Images are taken using a confocal microscope and analyzed using Image). Recombinant AIBP (Sinobiological) and VEGFC (R&D) are used.
206. VEGFC overexpression can improve cognitive function in APPNL'G'F mice. More importantly, the combined AIBP and VEGFC can further improve cognitive function in APPNL-G-F mice Such findings prove the principle that AIBP can improve cognition. In addition, the improved cognition is associated with augmented lymphatic function (as assessed by LYVE1 and VEGFR3 immunostaining) and attenuated A accumulation. These results further show that AIBP-elicited benefits on cognition are mediated by improved lymphatic functions and therefore better drainage of brain Ap.
207. Cognitive function is examined one month following VEGFC and AIBP administration, as used in the prior studies. Chances are that the Nano-AIBP/VEGFC combination can confer a protective effect at a different time point. Thus, a longitudinal analysis of cognitive function is performed, which enables to detect the optimal time window for the treatment. In addition, for each mouse study, post hoc histology is performed to validate VEGFC and/or AIBP overexpression.
208. Multiple memory tests were chosen in these studies. The NOR, MWM, and fear conditioning tests were commonly used to characterize the recognition and spatial memories in AD mouse models. Y-maze test and RAWM (Figure 19B) have been used to detect cognitive deficits in APPNL'G'Fmice. Thus, these five cognitive tests are included in the current studies to characterize the various types of memory. For each mouse, the least stressful NOR and Y-maze test are performed, followed by the MWM and RAWM, and the most stressful fear conditioning is performed at last. In addition, each test is separated by at least one week to avoid the confounding issue.
209. This study tests the translational potential of AIBP/VEGFC co-delivery for the treatment of AD. The study shows: 1): AIBP/VEGFC improves dementia in a preclinical mouse model compared to VEGFC alone 2) the mechanisms of how AIBP/VEGFC functions to ameliorate AD. The following components are used:
• Purchase recombinant AIBP (Sinobiological) and VEGFC (R&D)
• Manufacture VEGFC and AIBP/VEGFC slow release nanoparticles, 5 months
• Validation of the slow release nanoparticles carrying VEGFC or AIBP/VEGFC (shortened as Nano-VEGFC or Nano-AIBP/VEGFC) in test tubes and on cell culture models, • Prepare 4 groups of APPNL'G'F knock-in mice (12/group)
• Administer Nanoparticle alone, Nano-VEGFC, and Nano-AIBP/VEGFC into the mouse brain
• Memory tests
• Brain lymphatic tissue collection, immuno staining and imaging
210. An alternative approach to nanoparticle-mediated protein delivery is to use nanoparticle-mediated AIBP and VEGFC mRNA delivery. The impact of adeno-associated virus (AAV)-mediated AIBP/VEGFC expression on AD is assessed, which is the current standard platform of gene therapy. The drug targeting the AIBP downstream CAV1 - CSD is tested (See Figure 17) that can achieve similar effect on lymphangiogenesis.
211. AIBP-mediated cholesterol efflux promotes lymphangiogenesis is revealed. Increasing lymphatic function using AIBP can improve AD associated symptom such as dementia. The studies on AD require brain injection of viruses/proteins and mouse neurobehavioral tests, including those for cognitive functions.
212. These studies (a) delineate the mechanisms by which AIBP regulates pro- lymphangiogenic VEGFR3 signaling (e.g., AIBP receptor in lymphatics and the associated signaling cascade) and determine its regulation of meningeal lymphatics and cognition, (b) identify the critical cell type that synthesizes and produces AIBP and its acting sites in the brain (e.g. hippocampus, amygdala), (c) examine the therapeutic potential of combined AIBP and A neutralization or immunotherapy in pre-clinical AD models, and (d) perform single-cell RNA- seq analysis and metabolic studies in VEGFC and AIBP/VEGFC treated murine brains to systemically explore the full beneficial spectrum.
VIII. SEQUENCES
SEQ ID NO: 1 (AIBP protein sequence, human)
MRRGRAGPGRAGGARSASWMSRLRALLGLGLLVAGSRLPRIKSQTIACRSGPTWWGPQ
RLNSGGRWDSEVMASTVVKYLSQEEAQAVDQELFNEYQFSVDQLMELAGLSCATAIA
KAYPPTSMSRSPPTVLVICGPGNNGGDGLVCARHLKLFGYEPTIYYPKRPNKPLFTALVT
QCQKMDIPFLGEMPAEPMTIDELYELVVDAIFGFSFKGDVREPFHSILSVLKGLTVPIASI
DIPSGWDVEKGNAGGIQPDLLISLTAPKKSATQFTGRYHYLGGRFVPPALEKKYQLNLP
PYPDTECVYRLQ
SEQ ID NO: 2 (AIBP polynucleotide sequence)
ATGTCCAGGCTGCGGGCGCTGCTGGGCCTCGGGCTGCTGGTTGCGGGCTCGCGCGT
GCCGCGGATCAAAAGCCAGACCATCGCCTGTCGCTCGGGACCCACCTGGTGGGGAC
CGCAGCGGCTGAACTCGGGTGGCCGCTGGGACTCAGAGGTCATGGCGAGCACGGTG
GTGAAGTACCTGAGCCAGGAGGAGGCCCAGGCCGTGGACCAGGAGCTATTTAACGA
ATACCAGTTCAGCGTGGACCAACTTATGGAACTGGCCGGGCTGAGCTGTGCTACAG
CCATCGCCAAGGCATATCCCCCCACGTCCATGTCCAGGAGCCCCCCTACTGTCCTGG
TCATCTGTGGCCCGGGGAATAATGGAGGAGATGGTCTGGTCTGTGCTCGACACCTC
AAACTCTTTGGCTACGAGCCAACCATCTATTACCCCAAAAGGCCTAACAAGCCCCTC
TTCACTGCATTGGTGACCCAGTGTCAGAAAATGGACATCCCTTTCCTTGGGGAAATG
CCCGCAGAGCCCATGACGATTGATGAACTGTATGAGCTGGTGGTGGATGCCATCTIT
GGCTTCAGCTTCAAGGGCGATGTTCGGGAACCGTTCCACAGCATCCTGAGTGTCCTG
AAGGGACTCACTGTGCCCATTGCCAGCATCGACATTCCCTCAGGTGCTGGGATCCAG
AAGGTGGGGTGGGGGAGATTGGGGCCCTACCCTCCTGACTCTTGCCCACACCAGGT
CTAA
SEQ ID NO: 3 (Amino acid sequence for VEGFA)
MNFLLSWVHWSLALLLYLHHAKWSQAAPMAEGGGQNHHEVVKFMDVYQRSYCHPIE
TLVDIFQEYPDEIEYIFKPSCVPLMRCGGCCNDEGLECVPTEESNITMQIMRIKPHQGQHI
GEMSFLQHNKCECRPKKDRARQENPCGPCSERRKHLFVQDPQTCKCSCKNTDSRCKAR
QLELNERTCRCDKPRR
SEQ ID NO: 4 (polynucleotide sequence for VEGFA) ATGAACTTTCTGCTGTCTTGGGTGCATTGGAGCCTTGCCTTGCTGCTCTACCTCCACC ATGCCAAGTGGTCCCAGGCTGCACCCATGGCAGAAGGAGGAGGGCAGAATCATCAC GAAGTGGTGAAGTTCATGGATGTCTATCAGCGCAGCTACTGCCATCCAATCGAGAC CCTGGTGGACATCTTCCAGGAGTACCCTGATGAGATCGAGTACATCTTCAAGCCATC CTGTGTGCCCCTGATGCGATGCGGGGGCTGCTGCAATGACGAGGGCCTGGAGTGTG TGCCCACTGAGGAGTCCAACATCACCATGCAGATTATGCGGATCAAACCTCACCAA
GGCCAGCACATAGGAGAGATGAGCTTCCTACAGCACAACAAATGTGAATGCAGACC AAAGAAAGATAGAGCAAGACAAGAAAAAAAATCAGTTCGAGGAAAGGGAAAGGG GCAAAAACGAAAGCGCAAGAAATCCCGGTATAAGTCCTGGAGCGTGTACGTTGGTG CCCGCTGCTGTCTAATGCCCTGGAGCCTCCCTGGCCCCCATCCCTGTGGGCCTTGCT CAGAGCGGAGAAAGCATTTGTTTGTACAAGATCCGCAGACGTGTAAATGTTCCTGC AAAAACACAGACTCGCGTTGCAAGGCGAGGCAGCTTGAGTTAAACGAACGTACTTG
CAGATGTGACAAGCCGAGGCGGTGA
SEQ ID NO: 5 (amino acid sequence for VEGFC)
MHLLGFFSVACSLLAAALLPGPREAPAAAAAFESGLDLSDAEPDAGEATAYASKDLEE QLRSVSSVDELMTVLYPEYWKMYKCQLRKGGWQHNREQANLNSRTEETIKFAAAHYN TEILKSIDNEWRKTQCMPREVCIDVGKEFGVATNTFFKPPCVSVYRCGGCCNSEGLQCM NTSTSYLSKTLFEITVPLSQGPKPVTISFANHTSCRCMSKLDVYRQVHSIIRRSLPATLPQC QAANKTCPTNYMWNNHICRCLAQEDFMFSSDAGDDSTDGFHDICGPNKELDEETCQCV CRAGLRPASCGPHKELDRNSCQCVCKNKLFPSQCGANREFDENTCQCVCKRTCPRNQP
LNPGKCACECTESPQKCLLKGKKFHHQTCSCYRRPCTNRQKACEPGFSYSEEVCRCVPS YWKRPQMS
SEQ ID NO: 6 (polynucleotide sequence for VEGFC)
ATGCACTTGCTGGGCTTCTTCTCTGTGGCGTGTTCTCTGCTCGCCGCTGCGCTGCTCC CGGGTCCTCGCGAGGCGCCCGCCGCCGCCGCCGCCTTCGAGTCCGGACTCGACCTCT CGGACGCGGAGCCCGACGCGGGCGAGGCCACGGCTTATGCAAGCAAAGATCTGGA GGAGCAGTTACGGTCTGTGTCCAGTGTAGATGAACTCATGACTGTACTCTACCCAGA ATATTGGAAAATGTACAAGTGTCAGCTAAGGAAAGGAGGCTGGCAACATAACAGA GAACAGGCCAACCTCAACTCAAGGACAGAAGAGACTATAAAATTTGCTGCAGCACA
TTATAATACAGAGATCTTGAAAAGTATTGATAATGAGTGGAGAAAGACTCAATGCA TGCCACGGGAGGTGTGTATAGATGTGGGGAAGGAGTTTGGAGTCGCGACAAACACC TTCTTTAAACCTCCATGTGTGTCCGTCTACAGATGTGGGGGTTGCTGCAATAGTGAG GGGCTGCAGTGCATGAACACCAGCACGAGCTACCTCAGCAAGACGTTATTTGAAAT
TACAGTGCCTCTCTCTCAAGGCCCCAAACCAGTAACAATCAGTTTTGCCAATCACAC
TTCCTGCCGATGCATGTCTAAACTGGATGTTTACAGACAAGTTCATTCCATTATTAG
ACGTTCCCTGCCAGCAACACTACCACAGTGTCAGGCAGCGAACAAGACCTGCCCCA
CCAATTACATGTGGAATAATCACATCTGCAGATGCCTGGCTCAGGAAGATTTTATGT
TTTCCTCGGATGCTGGAGATGACTCAACAGATGGATTCCATGACATCTGTGGACCAA
ACAAGGAGCTGGATGAAGAGACCTGTCAGTGTGTCTGCAGAGCGGGGCTTCGGCCT
GCCAGCTGTGGACCCCACAAAGAACTAGACAGAAACTCATGCCAGTGTGTCTGTAA
AAACAAACTCTTCCCCAGCCAATGTGGGGCCAACCGAGAATTTGATGAAAACACAT
GCCAGTGTGTATGTAAAAGAACCTGCCCCAGAAATCAACCCCTAAATCCTGGAAAA
TGTGCCTGTGAATGTACAGAAAGTCCACAGAAATGCTTGTTAAAAGGAAAGAAGTT
CCACCACCAAACATGCAGCTGTTACAGACGGCCATGTACGAACCGCCAGAAGGCTT
GTGAGCCAGGATTTTCATATAGTGAAGAAGTGTGTCGTTGTGTCCCTTCATATTGGA
AAAGACCACAAATGAGCTAA
SEQ ID NO: 7 (polypeptide sequence of CAV-1, Q2TNI1)
MSGGKYVDSEGHLYTVPIREQGNIYKPNNKAMADELSEKQVYDAHTKEID
LVNRDPKHLNDDVVKIDFEDVIAEPEGTHSFDGIWKASFTTFTVTKYWFY
RLLSALFGIPMALIWGIYFAILSFLHIWAVVPCIKSFLIEIQCISRVYSI
YVHTVCDPLFEAVGKIFSNVRINLQKEI
SEQ ID NO: 8 (target sequence of CAV-1 by CRISPR-Cas9)
GACGTGATCGCCGAGCCTGC
SEQ ID NO: 9 (siRNA sequence for CAV-1)
CCAUCAAUUUGGAGACUAU
SEQ ID NO: 10 (siRNA sequence for CAV-1)
CCACUCAGCAACUGAAUGA
SEQ ID NO: 11 (siRNA sequence for CAV-1)
GUACCUGAGUCUCCAGAAA SEQ ID NO: 12 (CSD sequence)
RQIKIWFQNRRMKWKKDGIWKASFTTFTVTKYWFYR
SEQ ID NO: 13 (VEGFC C156S)
ATGCACTTGCTGGGCTTCTTCTCTGTGGCGTGTTCTCTGCTCGCCGCTGCGCTGCTCC
CGGGTCCTCGCGAGGCGCCCGCCGCCGCCGCCGCCTTCGAGTCCGGACTCGACCTCT
CGGACGCGGAGCCCGACGCGGGCGAGGCCACGGCTTATGCAAGCAAAGATCTGGA
GGAGCAGTTACGGTCTGTGTCCAGTGTAGATGAACTCATGACTGTACTCTACCCAGA
ATATTGGAAAATGTACAAGTGTCAGCTAAGGAAAGGAGGCTGGCAACATAACAGA
GAACAGGCCAACCTCAACTCAAGGACAGAAGAGACTATAAAATTTGCTGCAGCACA
TTATAATACAGAGATCTTGAAAAGTATTGATAATGAGTGGAGAAAGACTCAATGCA
TGCCACGGGAGGTGTGTATAGATGTGGGGAAGGAGTTTGGAGTCGCGACAAACACC
TTCTTTAAACCTCCAAGTGTGTCCGTCTACAGATGTGGGGGTTGCTGCAATAGTGAG
GGGCTGCAGTGCATGAACACCAGCACGAGCTACCTCAGCAAGACGTTATTTGAAAT
TACAGTGCCTCTCTCTCAAGGCCCCAAACCAGTAACAATCAGTTTTGCCAATCACAC
TTCCTGCCGATGCATGTCTAAACTGGATGTTTACAGACAAGTTCATTCCATTATTAG
ACGTTCCCTGCCAGCAACACTACCACAGTGTCAGGCAGCGAACAAGACCTGCCCCA
CCAATTACATGTGGAATAATCACATCTGCAGATGCCTGGCTCAGGAAGATTTTATGT
TTTCCTCGGATGCTGGAGATGACTCAACAGATGGATTCCATGACATCTGTGGACCAA
ACAAGGAGCTGGATGAAGAGACCTGTCAGTGTGTCTGCAGAGCGGGGCTTCGGCCT
GCCAGCTGTGGACCCCACAAAGAACTAGACAGAAACTCATGCCAGTGTGTCTGTAA
AAACAAACTCTTCCCCAGCCAATGTGGGGCCAACCGAGAATTTGATGAAAACACAT
GCCAGTGTGTATGTAAAAGAACCTGCCCCAGAAATCAACCCCTAAATCCTGGAAAA
TGTGCCTGTGAATGTACAGAAAGTCCACAGAAATGCTTGTTAAAAGGAAAGAAGTT
CCACCACCAAACATGCAGCTGTTACAGACGGCCATGTACGAACCGCCAGAAGGCTT
GTGAGCCAGGATTTTCATATAGTGAAGAAGTGTGTCGTTGTGTCCCTTCATATTGGA
AAAGACCACAAATGAGCTAA
SEQ ID NO: 14
CCTCTTACCTCAGTTACAATTTATA
SEQ ID NO: 15
GTGGTTCATCTTGATTTATTCGGC SEQ ID NO: 16
GATCGATCGTCGTTCTGGTAAG
SEQ ID NO: 17
AGTGGAGGGTAGTCAGAGAAG
SEQ ID NO: 18
CAACAGAGCCAGCAGTATGA
SEQ ID NO: 19
TGACAACCACCGCAATGA
SEQ ID NO: 20
CCTTGTTGGCTGAGACTGTAA
SEQ ID NO: 21
CTAGAGAACACCAGCAACAGTAA
SEQ ID NO: 22
ATGCCCTTGATGCCATTCT
SEQ ID NO: 23
CCCACAGGTACAGTTCCAATAC
SEQ ID NO: 24
GACAGCTCCCAAACAACAATAAA
SEQ ID NO: 25
CTGAGAGGTTGAATGAGAGGAAG
SEQ ID NO: 26
CGTCTCTTGATGTCTCGGAATG SEQ ID NO: 27
GCTGTTACTTTGGATCCCTCTC
SEQ ID NO: 28
CAGTGTGGTCACCTGGAATAA
SEQ ID NO: 29
TGGAGCAGTAGAAGCCAATAAA
SEQ ID NO: 30
CGTGATGGATCAAGAGGAAAGA
SEQ ID NO: 31
CTACCTGGGACATTGCTGTATT
SEQ ID NO: 32
GGAAGATGTGGGTCTGTCTTC
SEQ ID NO: 33
TGGGAATGCTGGAGGTTATG
SEQ ID NO: 34
CACAGGTCGCTAACCTATTT
SEQ ID NO: 35
ACAAGGGCTAGTGTACTGAATG
SEQ ID NO: 36
ACGCAGAGTGACGTGGCGTCCGCTGGGGTGCTTCTCGCGGAGATCTTCTCTCTGGGG
GCC
SEQ ID NO: 37
AGAGAGAAGATCTCCGCGAGAAGCACCCCAGCGGACGCCACGTCACTCTGCGTGGT
GTAC SEQ ID NO: 38 (Protein, synthetic)
DVWSFGVLLWEIFSL
SEQ ID NO: 39 (Protein, synthetic)
DVWSYGVTVWELMTF
SEQ ID NO: 40 (DNA, synthetic)
GACGTGATCGCCGAGCCTGCCGG
SEQ ID NO: 41 (DNA, synthetic)
GATCGCCGAGCCTGCCGGCACCTACAGCTTCGACG
SEQ ID NO: 42 (DNA, synthetic)
GATCGCCGAGCACCTACAGCTTCGACGGCGTGTGG
SEQ ID NO: 43 (protein, Danio rerio)
MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP
KHLNDDVVKVDFEDVIAEPAGTYSFDGVWKASFTTFTVTKYWCYRLLTALVGIPLALV
WGIFFAILSFIHIWAVVPCVKSYLIEIHCISRVYSICVHTFCDPLFEAMGKCFSNVRVTATK vv
SEQ ID NO: 44 (protein, synthetic)
MTSGYKDGTPEEEYAHSPFIRKQGNIYKPNNKEMDNDSINEKTLQDVHTKEIDLVNRDP
KHLNDDVVKVDFEDVIAEHLQLRRRVEGELHHLHSNQILVLQAADSAGGHPTRPGMG
HLLRHPLLHPHLGRGALREELPNRDPLHQSSLLHLCAHLLRPTLGHGEMLRPGHCYGG

Claims

IX. CLAIMS What is claimed is:
1. A composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, or ii) a caveolin-1 (CAV-1) inhibitor.
2. The composition of claim 1, wherein the composition comprises i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and ii) a caveolin-1 (CAV-1) inhibitor.
3. The composition of claim 1, wherein the AIBP polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 1.
4. The composition of claim 1, wherein the AIBP polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 1.
5. The composition of claim 1, wherein the polynucleotide comprises a nucleic acid sequence 90% identical to SEQ ID NO: 2.
6. The composition of claim 1, wherein the polynucleotide comprises a nucleic acid sequence 95% identical to SEQ ID NO: 2.
7. The composition of any one of claims 1-6, wherein the polynucleotide is contained in a vector.
8. The composition of claim 7, wherein the vector is a viral vector.
9. The composition of claim 8, wherein the viral vector is an adeno-associated virus (AAV) vector.
10. The composition of any one of claims 1-9, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
— 63 —
11. The composition of claim 10, wherein the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux.
12. The composition of claim 10 or 11, wherein the CAV-1 inhibitor comprises methyl beta cyclodextrin.
13. The composition of claim 10, wherein the CSD peptide comprises a sequence 90% identical to SEQ ID NO: 12.
14. The composition of claim 10, wherein the CSD peptide comprises a sequence 95% identical to SEQ ID NO: 12.
15. The composition of claim 10, wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
16. The composition of claim 10, wherein the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9, or CRISPR- Casl3.
17. The composition of any one of claims 1-16, further comprising a stimulator for VEGFR3.
18. The composition of claim 17, wherein the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
19. The composition of claim 17 or 18, wherein the stimulator is VEGFC(C156S).
20. The composition of claim 17, wherein the stimulator comprises Pioglitazone.
21. The composition of claim 17, wherein the VEGFA polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 3.
22. The composition of claim 17, wherein the VEGFA polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 3.
— 64 —
23. The composition of claim 17, wherein the VEGFC polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 5.
24. The composition of claim 17, wherein the VEGFC polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 5.
25. The composition of any one of claims 1-24, wherein the composition is contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
26. The composition of claim 25, wherein the pharmaceutically acceptable carrier is a nanoparticle or docosahexaenoic acid (DHA).
27. The composition of claim 26, wherein the nanoparticle is a porous silica nanoparticle (pSi).
28. The composition of claim 26 or 27, wherein the nanoparticle comprises poly(lactide-co-glycolide) (PLGA) .
29. A method for treating a neurodegenerative disease in a subject, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, or ii) a caveolin-1 (CAV-1) inhibitor.
30. The method of claim 29, comprising administering to subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and ii) a caveolin-1 (CAV-1) inhibitor.
31. The method of claim 29 or 30, wherein the AIBP polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 1.
32. The method of claim 29 or 30, wherein the AIBP polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 1.
— 65 —
33. The method of claim 29 or 30, wherein the polynucleotide comprises a nucleic acid sequence 90% identical to SEQ ID NO: 2.
34. The method of claim 29 or 30, wherein the polynucleotide comprises a nucleic acid sequence 95% identical to SEQ ID NO: 2.
35. The method of any one of claims 29-34, wherein the polynucleotide is contained in a vector.
36. The method of claim 35, wherein the vector is a viral vector.
37. The method of claim 36, wherein the viral vector is an adeno-associated virus (AAV) vector.
38. The method of any one of claims 29-37, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
39. The method of claim 38, wherein the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux.
40. The method of claim 38 or 39, wherein the CAV-1 inhibitor comprises methyl beta cyclodextrin.
41. The method of claim 40, wherein the CSD peptide comprises a sequence 90 % identical to SEQ ID NO: 12.
42. The method of claim 40, wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
43. The method of claim 40, wherein the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9 or CRISPR-Casl3.
44. The method of any one of claims 29-43, wherein the composition further comprises a stimulator of VEGFR3.
— 66 —
45. The method of claim 44, wherein the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
46. The method of claim 44 or 45, wherein the stimulator comprises VEGFC(C156S).
47. The method of claim 44, wherein the stimulator comprises Pioglitazone.
48. The method of claim 45, wherein the VEGFA polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 3.
49. The method of claim 45, wherein the VEGFA polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 3.
50. The method of claim 45, wherein the VEGFC polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 5.
51. The method of claim 45, wherein the VEGFC polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 5.
52. The method of any one of claims 29-51, wherein the composition is contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
53. The method of claim 52, wherein the pharmaceutically acceptable carrier comprises a nanoparticle or docosahexaenoic acid (DHA).
54. The method of claim 53, wherein the nanoparticle is a porous silica nanoparticle (pSi).
55. The method of claim 53 or 54, wherein the nanoparticle comprises poly (lactide- co-glycolide) (PLGA).
— 67 —
56. The method of any one of claims 29-55, wherein the composition is administered intrathecally or intracranially.
57. The method of any one of claims 29-56, wherein the neurodegenerative disease comprises Alzheimer’s disease, Parkinson's disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, or Multiple Sclerosis.
58. The method of any one of claims 29-57, wherein the composition increases lymphangiogenesis in brain.
59. A method for treating a neurodegenerative disease, comprising diagnosing a subject as having a neurodegenerative disease; and administering to the subject a therapeutically effective amount of a caveolin-1 (CAV-1) inhibitor.
60. The method of claim 59, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
61. The method of claim 60, wherein the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux.
62. The method of claim 60 or 61, wherein the CAV-1 inhibitor comprises methyl beta cyclodextrin.
63. The method of claim 59, wherein the CSD peptide comprises a sequence 90% identical to SEQ ID NO: 12.
64. The method of claim 59, wherein the CSD peptide comprises a sequence 95% identical to SEQ ID NO: 12.
65. The method of claim 59, wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
66. The method of claim 59, wherein the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), a CRISPR-Cas9, or CRISPR-Casl3.
67. The method of any one of claims 59-66, further comprising administering a therapeutically effective amount of an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide.
68. The method of claim 67, wherein the AIBP polypeptide or the polynucleotide that encodes the AIBP polypeptide is administered simultaneously or subsequentially with the CAV- 1 inhibitor of any one of claims 59-66.
69. The method of claim 67 or 68, wherein the AIBP polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 1.
70. The method of claim 67 or 68, wherein the AIBP polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 1
71. The method of claim 67 or 68, wherein the polynucleotide comprises a nucleic acid sequence 90% identical to SEQ ID NO: 2.
72. The method of claim 67 or 68, wherein the polynucleotide comprises a nucleic acid sequence 95% identical to SEQ ID NO: 2.
73. The method of any one of claims 59-72, wherein the polynucleotide is contained in a vector.
74. The method of claim 73, wherein the vector is a viral vector.
75. The method of claim 74, wherein the viral vector is an adeno-associated virus (AAV) vector.
76. The method of any one of claims 59-75, further comprising administering to the subject a therapeutically effective amount of a stimulator for VEGFR3.
77. The method of claim 76, wherein the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
78. The method of claim 76 or 77, wherein the stimulator comprises VEGFC(C156S).
79. The method of claim 76, wherein the stimulator comprises Pioglitazone.
80. The method of claim 77, wherein the VEGFA polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 3.
81. The method of claim 77, wherein the VEGFA polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 3.
82. The method of claim 77, wherein the VEGFC polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 5.
83. The method of claim 77, wherein the VEGFC polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 5.
84. The method of any one of claims 59-83, wherein the composition is contained in or conjugated to a pharmaceutically acceptable carrier.
85. The method of any one of claims 84, wherein the pharmaceutically acceptable carrier comprises a nanoparticle or docosahexaenoic acid (DHA).
86. The method of claim 85, wherein the nanoparticle is a porous silica nanoparticle (pSi).
87. The method of claim 85 or 86, wherein the nanoparticle comprises poly(lactide- co-glycolide) (PLGA).
88. The method of any one of claims 59-87, wherein the composition is administered intrathecally or intracranially.
89. The method of any one of claims 59-89, wherein the neurodegenerative disease comprises Alzheimer’s disease, Parkinson's disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, or Multiple Sclerosis.
90. The method of any one of claims 59-89, wherein the composition increases lymphangiogenesis in brain.
91. A method of treating a neurodegenerative disease, comprising administering to a subject a composition that is capable of crossing blood-brain barrier, wherein the composition comprises a caveolin-1 (CAV-1) inhibitor.
92. The method of claim 91, wherein the composition further comprises an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide.
93. The method of claim 91 or 92, wherein the composition further comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
94. The method of any one of claims 91-93, wherein the composition is contained in or conjugated to a pharmaceutically acceptable carrier that is capable of crossing blood-brain barrier.
95. The method of claim 94, wherein the pharmaceutically acceptable carrier comprises a nanoparticle or docosahexaenoic acid (DHA).
96. The method of claim 95, wherein the nanoparticle is a porous silica nanoparticle (pSi).
97. The method of claim 95 or 96, wherein the nanoparticle comprises poly(lactide- co-glycolide) (PLGA).
98. The method of any one of claims 91-97, further comprising administering to the subject an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide.
— 71 —
99. The method of any one of claims 91-98, further comprising administering to the subject a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
100. The method of any one of claims 91-99, wherein the neurodegenerative disease comprises Alzheimer’s disease, Parkinson's disease, Huntington's Disease, Amyotrophic Lateral Sclerosis, or Multiple Sclerosis.
101. A method for treating lymphedema in a subj ect, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, or ii) a caveolin-1 (CAV-1) inhibitor.
102. The method of claim 101, comprising administering to the subject a therapeutically effective amount of a composition comprising i) an APOA1 binding protein (AIBP) polypeptide or a polynucleotide that encodes the AIBP polypeptide, and ii) a caveolin-1 (CAV-1) inhibitor.
103. The method of claim 101 or 102, wherein the AIBP polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 1.
104. The method of claim 101 or 102, wherein the AIBP polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 1.
105. The method of claim 101 or 102, wherein the polynucleotide comprises a nucleic acid sequence 90% identical to SEQ ID NO: 2.
106. The method of claim 101 or 102, wherein the polynucleotide comprises a nucleic acid sequence 95% identical to SEQ ID NO: 2.
107. The method of any one of claims 101-106, wherein the polynucleotide is contained in a vector.
108. The method of claim 107, wherein the vector is a viral vector.
— 72 —
109. The method of claim 108, wherein the viral vector is an adeno-associated virus (AAV) vector.
110. The method of any one of claims 101-109, wherein the CAV-1 inhibitor comprises a small molecule, a CAV-1 gene editing tool, an antibody, or a CAV-1 scaffolding domain (CSD) peptide.
111. The method of claim 110, wherein the CAV-1 inhibitor comprises a form of cyclodextrin that is capable of promoting cholesterol efflux.
112. The method of claim 110 or 111, wherein the CAV-1 inhibitor comprises methyl beta cyclodextrin.
113. The method of claim 110, wherein the CSD peptide comprises a sequence 90 % identical to SEQ ID NO: 12.
114. The method of claim 110, wherein the antibody comprises a conventional antibody, a Fab antibody, a single-chain variable fragment (scFv) antibody, or a VHH antibody.
115. The method of claim 110, wherein the CAV-1 gene editing tool comprises a small interfering RNA (siRNA), a short hairpin RNA (shRNA), CRISPR-Cas9 or CRISPR- Casl3.
116. The method of any one of claims 101-115, wherein the composition further comprises a stimulator of VEGFR3.
117. The method of claim 116, wherein the stimulator comprises a VEGFA polypeptide, a VEGFC polypeptide, or a variant thereof.
118. The method of claim 116 or 117, wherein the stimulator comprises VEGFC(C156S).
119. The method of claim 116, wherein the stimulator comprises Pioglitazone.
— 73 —
120. The method of claim 117, wherein the VEGFA polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 3.
121. The method of claim 117, wherein the VEGFA polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 3.
122. The method of claim 117, wherein the VEGFC polypeptide comprises an amino acid sequence 90% identical to SEQ ID NO: 5.
123. The method of claim 117, wherein the VEGFC polypeptide comprises an amino acid sequence 95% identical to SEQ ID NO: 5.
124. The method of any one of claims 101-123, wherein the composition is contained in or conjugated to a pharmaceutically acceptable carrier.
125. The method of claim 124, wherein the pharmaceutically acceptable carrier comprises a nanoparticle.
126. The method of any one of claims 101-125, wherein the composition increases lymphangiogene sis .
— 74 —
PCT/US2021/045918 2020-10-13 2021-08-13 Compositions and methods for increasing lymphangiogenesis WO2022081245A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063091048P 2020-10-13 2020-10-13
US63/091,048 2020-10-13

Publications (1)

Publication Number Publication Date
WO2022081245A1 true WO2022081245A1 (en) 2022-04-21

Family

ID=81208537

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/045918 WO2022081245A1 (en) 2020-10-13 2021-08-13 Compositions and methods for increasing lymphangiogenesis

Country Status (1)

Country Link
WO (1) WO2022081245A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180360993A1 (en) * 2015-12-10 2018-12-20 The Regents Of The University Of California Compositions and methods for treating or ameliorating neuroinflammation, neurodegeneration, neuropathic pain, and migraine
US10731219B1 (en) * 2019-09-19 2020-08-04 Dasman Diabetes Institute Method for preventing progression to metabolic syndrome

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180360993A1 (en) * 2015-12-10 2018-12-20 The Regents Of The University Of California Compositions and methods for treating or ameliorating neuroinflammation, neurodegeneration, neuropathic pain, and migraine
US10731219B1 (en) * 2019-09-19 2020-08-04 Dasman Diabetes Institute Method for preventing progression to metabolic syndrome

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE NUCLEOTIDE [online] 23 November 2018 (2018-11-23), "Homo sapiens NAD(P)HX epimerase (NAXE), mRNA", XP055934568, retrieved from ncbi Database accession no. NM_144772.3 *
DATABASE UniProtKB [online] 23 September 2008 (2008-09-23), "Full=NAD(P)H-hydrate epimerase {ECO:0000256|HAMAPRule:MF_03159};", XP055934566, retrieved from UniProt Database accession no. B4DP80 *

Similar Documents

Publication Publication Date Title
AU2019201337B2 (en) Methods and compositions for treating aging-associated impairments
Müller et al. Astrocyte-derived CNTF switches mature RGCs to a regenerative state following inflammatory stimulation
Hao et al. Long-term protective effects of AAV9-mesencephalic astrocyte-derived neurotrophic factor gene transfer in parkinsonian rats
US8007790B2 (en) Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
RU2699706C2 (en) Substances and methods of modulating tendon healing
JP2021525707A (en) Methods for treating spinal cord injury
JP2009256374A (en) Nitric oxide donor for treatment of disease and injury
Gao et al. Induced neural stem cells modulate microglia activation states via CXCL12/CXCR4 signaling
Muñoz-Manchado et al. Neuroprotective and reparative effects of carotid body grafts in a chronic MPTP model of Parkinson's disease
US20230301967A1 (en) Methods and compositions for the treatment of retinopathy and other ocular diseases
US9028810B2 (en) Composition for inducing migration of neural stem cells containing periostin as effective ingredient
Guo et al. NF-KappaB pathway is involved in bone marrow stromal cell-produced pain relief
JP2011513290A (en) New use of VEGFxxxb
EP2904009B1 (en) Compounds for treating the remyelination blockade in diseases associated with the expression of herv-w envelope protein
JPWO2005094846A1 (en) Prion disease therapeutic agent and method for producing the same
WO2022081245A1 (en) Compositions and methods for increasing lymphangiogenesis
CN113302302A (en) Double-stranded RNA and use thereof
JP2021506794A (en) Peptides and other drugs to treat pain and increase pain sensitivity
Jain et al. Neuroprotection in Alzheimer disease
EP4137148A1 (en) Small extracellular vesicles expressing a dominant negative ampk alpha 1 mutant for use in the treatment of obesity
Bao miR101 aggravates cell apoptosis in cerebral ischemic area by downregulating JAK2
Schepers PDE4 gene inhibition: a novel approach to treat demyelinating disorders
KR101358601B1 (en) The method for preparing dopamine neurons
KR20240042501A (en) Small extracellular vesicles expressing dominant-negative AMPK alpha 1 variants for use in the treatment of obesity
US10487148B2 (en) Methods and compositions for treating aging-associated impairments

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: 21880732

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21880732

Country of ref document: EP

Kind code of ref document: A1