US20220403341A1 - Exosomes from clonal progenitor cells - Google Patents

Exosomes from clonal progenitor cells Download PDF

Info

Publication number
US20220403341A1
US20220403341A1 US17/592,200 US202217592200A US2022403341A1 US 20220403341 A1 US20220403341 A1 US 20220403341A1 US 202217592200 A US202217592200 A US 202217592200A US 2022403341 A1 US2022403341 A1 US 2022403341A1
Authority
US
United States
Prior art keywords
cell
exosomes
progenitor cell
exosome
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/592,200
Inventor
Dana Larocca
Mohammad Hassanipour
Paola A. Bignone
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Recyte Therapeutics Inc
Original Assignee
Recyte Therapeutics Inc
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 Recyte Therapeutics Inc filed Critical Recyte Therapeutics Inc
Priority to US17/592,200 priority Critical patent/US20220403341A1/en
Assigned to ReCyte Therapeutics, Inc. reassignment ReCyte Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Larocca, Dana
Assigned to ReCyte Therapeutics, Inc. reassignment ReCyte Therapeutics, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HASSANIPOUR, MOHAMMAD, Bignone, Paola A.
Publication of US20220403341A1 publication Critical patent/US20220403341A1/en
Assigned to JUVENESCENCE LIMITED reassignment JUVENESCENCE LIMITED SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AGEX THERAPEUTICS, INC., ReCyte Therapeutics, Inc., REVERSE BIOENGINEERING, INC.
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/069Vascular Endothelial cells
    • C12N5/0692Stem cells; Progenitor cells; Precursor cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/44Vessels; Vascular smooth muscle cells; Endothelial cells; Endothelial progenitor cells
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1353Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from bone marrow mesenchymal stem cells (BM-MSC)

Definitions

  • the field of the invention relates to exosomes isolated from progenitor cells.
  • Exosomes are believed to contain important signaling molecules that may provide the source of trophic factors responsible for some regenerative benefits seen in cell replacement therapy. As such they would provide an alternative to some cell based therapies that would be easier to manufacture on a large scale and potentially safer to administer to a subject in need of cell therapy. In particular, the risk associated with transmission of infectious agents such as viruses may be lower compared to transplanting whole cells. Moreover, the risk of immune rejection of the exosomes relative to transplanted cells may also be lower. Accordingly, exosomes may provide an attractive alternative or adjunct to cell based therapies and cell based regenerative medicine.
  • Exosomes are 30 to 120 nm vesicles secreted by a wide range of mammalian cell types.
  • the vesicles are enclosed by a lipid bilayer and are larger than LDL which has a size of 22 nm, but smaller than a red blood cell, which is 6000 to 8000 nm in diameter and has a thickness of 2000 nm.
  • Exosomes are found both in cells growing in vitro as well as in vivo. They can be isolated from tissue culture media as well as bodily fluids such as plasma, urine, milk and cerebrospinal fluid. George et al. (1982) Blood 60:834; Martinez et al. (2005) Am J Physiol Health Cir Physiol 288:H1004. Exosomes originate from the endosomal membrane compartment. They are stored in intraluminal vesicles within multivesicular bodies of the late endosome. Multivesicular bodies are derived from the early endosome compartment and contain within them smaller vesicular bodies that include exosomes. Exosomes are released from the cell when multivesicular bodies fuse with the plasma membrane. Methods of isolating exosomes from cells has been described, see e.g. US Patent Application Publication No. 20120093885
  • Exosomes contain a variety of molecules including proteins, lipids and nucleic acids such as DNA, mRNA and miRNA. Their contents are believed to play a part in cell to cell communication involving the release of the exosome from one cell and the binding/fusion of the exosome with a second cell, wherein the contents of the exosomal compartment are released within the second cell.
  • exosomes derived from endothelial progenitor cells may act as vehicle for mRNA transport among cells. These exosomes were shown to incorporate into normal endothelial cells by interacting with the ⁇ 4 ⁇ 1 integrin. Once incorporated into the endothelial cells, the exosomes stimulated an angiogenic program. Deregibus et al. (2007) Blood 110:2440. Similar results were obtained in vivo using severe combined immunodeficient mice. Exosome stimulated endothelial cells implanted subcutaneously in Matrigel (a murine sarcoma extract) organized into a patent vessel network connected with the murine vasculature. Deregibus, supra. Bruno et al. (2009) J Am Soc Nephrol 20:1053; Herrera et al. (2010) J Cell Mol Med 14:1605.
  • MiRNAs are small, non-coding regulatory RNAs that can have a wide range of effects on multiple RNA targets, thus having the potential to have greater phenotypic influence than coding RNAs.
  • MiRNA profiles of exosomes often differ from those of the parent cells. Profiling studies have demonstrated that miRNAs are not randomly incorporated into exosomes but rather a subset of miRNAs is preferentially packaged into exosomes, suggesting an active sorting mechanism of exosomal miRNAs. Guduric-Fuchs et al. (2014) Nucleic Acid Res. 42:9195; Ohshima et al. (2010) PloS One 5(10):e13247.
  • exosomes contain a variety of molecules, many believed to play an important role in cell signaling, exosomes would prove useful in research and industry and would have applications as therapeutics, diagnostics and in screening assays. Frequently, however, the availability of reproducible, essentially identical populations of exosomes is limited by the fact that most sources of exosomes are cells that senesce and thus have limited replicative capacity. Accordingly, there is a need for exosomes that are derived from a clonal source that has an extended replicative capacity that is greater than most adult or fetal derived cells. The invention described infra meets this need and as well as other needs in the field.
  • compositions comprising exosomes obtained from progenitor cell lines, as well as methods of making and using exosomes obtained from progenitor cell lines.
  • Embryonic progenitors are cell lines derived under a variety of culture conditions from pluripotent stem cells, such as human embryonic stem (hES) cells or induced pluripotent stem (iPS) cells.
  • the progenitor cell lines are clonal and while they do, in most instances, senesce, they also possess longer telomeres compared to adult or fetal derived tissue or cells (such as adult stem cells) and accordingly have enhanced replicative capacity relative to those cell types. Because of their clonality and their enhanced replicative capacity they provide a suitable source of exosomes that will offer the benefit of uniformity with regard to the exosome composition and abundance relative to exosomes derived from their typical sources such as adult cells or adult stem cells.
  • the invention provides an exosome isolated from a progenitor cell line, such as clonal progenitor cell line.
  • the invention provides an exosome isolated from a human progenitor cell line, such as a clonal human progenitor cell line.
  • the invention provides an exosome isolated from endothelial progenitor cell.
  • the invention provides an exosome isolated from a clonal human endothelial progenitor cell.
  • the invention provides an exosome isolated from the 30-MV2-6 human clonal progenitor cell line.
  • the invention provides an exosome isolated from a human clonal progenitor cell that expresses CD31 and CD34.
  • the invention provides an exosome isolated from a human progenitor cell line, wherein the human progenitor cell is not an adult stem cell.
  • the invention provides an exosome isolated from a human progenitor cell line, wherein the human progenitor cell is not a mesenchymal stem cell (MSC).
  • MSC mesenchymal stem cell
  • the invention provides an exosome isolated from a cell that has not been transfected with an exogenous gene.
  • the invention provides an exosome isolated from a cell that has been transfected with an exogenous gene, wherein the gene is not c-myc.
  • the invention provides an exosome isolated from a cell that does not overexpress c-myc.
  • the invention provides an exosome isolated from the 30-MV2-6 clonal human progenitor cell line.
  • the invention provides an exosome isolated from a cell expressing one or more genes chosen from the genes listed in Table 1.
  • the invention provides an exosome isolated from a cell expressing a plurality of the genes chosen from the genes listed in Table 1.
  • the invention provides an exosome isolated from a cell expressing the genes listed in Table 1.
  • the invention provides an exosome containing CD63.
  • the invention provides an exosome containing one or more miRNAs listed in Table 2 or Table 4.
  • the invention provides an exosome containing one or more angiogenic miRNAs.
  • the invention provides an exosome containing miR-126.
  • the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains one or more miRNAs listed in Table 2 or Table 4.
  • the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains one or more angiogenic miRNAs.
  • the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains miR-126.
  • the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains CD63.
  • the invention provides an exosome that induces a cell to form vascular tube like structures.
  • the invention provides an exosome that induces a cell to form branching vascular tube like structures.
  • the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome.
  • the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell has the ability to form vascular tube like structures.
  • the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell is an endothelial cell.
  • the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell is a human umbilical vein endothelial cell (HUVEC).
  • a human umbilical vein endothelial cell HUVEC
  • the progenitor cell may be a human progenitor cell, such as a human embryonic progenitor cell.
  • a human embryonic progenitor cell is the 30-MV2-6 cell line.
  • the progenitor cell may be, for example, a clonal progenitor cell line, an oligoprogenitor cell line.
  • the progenitor cell may express one or more genes listed in Table 1.
  • the progenitor cell may express a plurality of the genes listed in Table 1.
  • the progenitor cell line may express the genes listed in Table 1.
  • the progenitor cell line may express CD31 and CD34.
  • the invention provides a method of isolating an exosome from a progenitor cell, such as a clonal progenitor cell comprising 1) culturing the progenitor cell in a suitable media or buffer for a time sufficient to allow the cells to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • a progenitor cell such as a clonal progenitor cell comprising 1) culturing the progenitor cell in a suitable media or buffer for a time sufficient to allow the cells to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a clonal progenitor cell
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell comprising 1) culturing the human clonal progenitor cell in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • the invention provides a method of isolating an exosome from a 30-MV2-6 human clonal progenitor cell line comprising 1) culturing the 30-MV2-6 human clonal progenitor cell line in a suitable media or buffer for a time sufficient to allow the 30-MV2-6 human clonal progenitor cell line to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a 30-MV2-6 human clonal progenitor cell line.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing CD31 and CD34 comprising 1) culturing the human clonal progenitor cell line expressing CD31 and CD34 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing CD31 and CD34 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing CD31 and CD34.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing one or more of the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing one or more of the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing one or more of the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing one or more of the genes listed in Table 1.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing a plurality of the genes listed in Table 1.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing the genes listed in Table 1.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell wherein the human clonal progenitor cell line has not been transfected with an exogenous gene comprising 1) culturing the human clonal progenitor cell line that has not been transfected with an exogenous gene in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has not been transfected with an exogenous gene to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that has not been transfected with an exogenous gene.
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell, wherein the human clonal progenitor cell line has been transfected with an exogenous gene, wherein the gene is not c-myc, comprising 1) culturing the human clonal progenitor cell line that has been transfected with an exogenous gene, wherein the exogenous gene is not c-myc, in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has been transfected with an exogenous gene, wherein the exogenous gene is not c-myc, to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that has been transfected with an exogenous gene,
  • the invention provides a method of isolating an exosome from a human clonal progenitor cell wherein the human clonal progenitor cell line does not overexpress c-myc comprising 1) culturing the human clonal progenitor cell line that does not overexpress c-myc in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has not been transfected with an exogenous gene to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that does not overexpress c-myc.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting an endothelial cell with an exosome isolated from a progenitor cell thereby inducing or enhancing an endothelial cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a clonal progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human clonal progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human endothelial progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human clonal endothelial progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing one or more genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing a plurality of genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing the genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing the markers CD31 and CD34 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a 30-MV2-6 cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • the invention provides a method of regeneration a tissue or an organ comprising contacting one or more cells capable of regenerating a tissue or an organ with an exosome isolated from a progenitor cell.
  • the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a progenitor cell.
  • the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a human clonal endothelial progenitor cell.
  • the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a 30-MV2-6 cell.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a progenitor cell.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from an endothelial progenitor cell.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a 30-MV2-6 human endothelial progenitor cell.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing CD31 and CD34.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing one or more genes listed in Table 1.
  • the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing a plurality of genes listed in Table 1.
  • the invention provides a kit comprising an exosome isolated from a progenitor cell and at least one container.
  • FIG. 1 shows a graph of the size and concentration of exosomes isolated from
  • FIG. 2 A shows three photomicrographs, the first showing the effects on vascular tube like formation in HUVECs grown in the presence of basal media supplemented with exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (top); the second showing the effects on vascular tube formation in HUVECs grown in base media supplemented with PBS, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV-2-6 (middle); and the third showing the effects on vascular tube like formation in HUVECs grown in complete medium, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (bottom).
  • FIG. 2 B is a graph quantifying four parameters: cell covered area; total tube length; total number of branching points and total number of loops in HUVECs grown in the presence of exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“MV2-6 EXO”); HUVECs grown in basal media+PBS, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“Basal PBS”); and HUVECs grown in complete media, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“Complete”).
  • MV2-6 EXO HUVECs grown in basal media+PBS, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6
  • Base PBS HUVECs grown in complete media, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“Complete”).
  • FIG. 3 A is photomicrograph showing that hES derived perivascular cells form aggregates when cultured in the presence of complete EGM-MV2 media with serum and growth factors.
  • FIG. 3 B is a photomicrograph showing that hES derived perivascular cells form incomplete tubes when cultured in EGM-MV2 basal media.
  • FIGS. 3 C-E are photomicrographs showing that increasing doses of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6 resulted in increasing tube formation in hES derived perivascular cells.
  • FIG. 3 F is a graph showing the cell covered area of hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5 ⁇ 10 7 , 5.0 ⁇ 10 7 , 10.0 ⁇ 10 7 ) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3 G is a graph showing the total number of branching points in hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5 ⁇ 10 7 , 5.0 ⁇ 10 7 , 10.0 ⁇ 10 7 ) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3 H is a graph showing the total tube length of vascular tube like structures formed by hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5 ⁇ 10 7 , 5.0 ⁇ 10 7 , 10.0 ⁇ 10 7 ) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3 I is a graph showing the total number of loops formed by hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplement with increasing doses (2.5 ⁇ 10 7 , 5.0 ⁇ 10 7 , 10.0 ⁇ 10 7 ) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 4 shows comparison of in vitro angiogenic activity of exosomes isolated from the human clonal endothelial cell line 30-MV2-6 versus exosomes isolated from bone marrow mesenchymal stem cells (BM-MSC).
  • BM-MSC bone marrow mesenchymal stem cells
  • 30-MV2-6 exosomes derived from 30-MV2-6 cell line
  • MSC exosomes derived from BM-MSC
  • BM basal medium, negative control
  • CM complete growth medium. Results were normalized to tube length obtained using complete growth medium (CM).
  • FIG. 5 shows that the angiogenic activity of exosomes isolated from the 30-MV2-6 cell line is dose dependent (panel A) and at least six times more potent than the angiogenic activity of exosomes isolated from BM-MSCs (panel B).
  • FIG. 6 A depicts analysis of miRNA content in 30-MV2-6 derived exosomes versus BM-MSC derived exosomes.
  • 30-MV2-6 exosome RNA was compared to BM-MSC RNA for miRNA content using an 84 miRNA PCR array.
  • the scatter plot indicates miRNAs with greater than 6-fold differences.
  • the upper circled dot represents miRNA miR-126-3p, which is expressed at 77.6-fold higher in 30-MV2-6 exosomes than in BM-MSC exosomes.
  • the lower circled dot represents miRNA miR-376c-3p which is expressed at 752-fold lower in 30-MV2-6 exosomes than in MB-MSC exosomes.
  • FIG. 6 B is a bar graph illustrating the differences (measured as fold-change) in miRNA expression profile between 30-MV2-6-derived exosomes versus BM-MSC-derived exosomes.
  • FIG. 7 consists of 5 photomicrographs showing in vivo angiogenic activity of human clonal endothelial cell line 30-MV2-6 in the Matrigel plug assay in immunocompromised mice. Blood vessel-like structures are seen in the exosome treated plugs (panels A and C) but not in the control plug (panel E). Endothelial cell content is confirmed by staining with anti-Von Willebrand factor antibody (panels B and D).
  • FIG. 8 shows that the in vitro angiogenic activity of exosomes derived from 30-MV2-6 cells grown in T-flasks is comparable to the in vitro angiogenic activity of exosomes derived from 30-MV2-6 cells grown in the Quantum cell expansion bioreactor.
  • Panel A depicts tube network formation and panel B depicts quantitative analysis of tube length using Image J angiogenesis analyzer software.
  • FIG. 9 is a bar graph illustrating the effect of oxygen level and conditioning medium used on the angiogenic activity of exosomes derived from 30-MV2-6 cells.
  • Use of PBS versus basal medium as the conditioning medium had no significant effect on the angiogenic activity of the 30-MV2-6 exosomes.
  • no significant effect on the angiogenic activity was observed in conditioning the cells in 1% versus 5% oxygen.
  • FIG. 10 shows ELISA detection of CD63 on intact exosomes to determine exosome concentration.
  • the standard curve was prepared using exosomes (from HT1080 cells) of known concentration as determined by nanoparticle analysis (NTA). Quantitation of exosomes in samples of unknown concentration was calculated from OD value at 450 nm. The assay assumes CD63 content of exosomes derived from various different cells remains relatively constant.
  • FIG. 11 depicts the high proliferative capacity and stable angiogenic exosome production of the clonal human embryonic progenitor cell line 30-MV2-6.
  • the 30-MV2-6 cells continue to proliferate in cell culture past 50 population doublings (panel A).
  • exosomes that retain their angiogenic activity as measured by the in vitro tube formation assay may be prepared from 30-MV2-6 cells that have been cultured for at least 50 population doublings (panel B).
  • FIG. 12 is a heat map of Illumina gene expression array (Illumina, Hayward, Calif.) data showing that the clonal embryonic epithelial progenitor cell lines derived from the embryonic stem cell line ESI-017 have similar endothelial specific gene expression pattern as various adult endothelial cells from different sources.
  • Illumina gene expression array Illumina, Hayward, Calif.
  • the invention provides exosomes isolated from clonal progenitor cells, such as human clonal progenitor cells derived from a human pluripotent stem cell. Because the cells are clonal and have enhanced replicative capacity in vitro, the invention provides a means of producing the same exosomes over and over again. This provides for a consistent product allowing either the researcher or clinician to alleviate any concerns regarding both the quality and the consistency of the exosomes in any application. Accordingly, the invention also provides methods of making the progenitor cells from which the exosomes are derived and methods of isolating the exosomes from these cells. The invention also contemplates uses, cell cultures and kits comprising the exosomes all of which are described infra.
  • the term “about” means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45% to 55%.
  • CD31 also known as PECAM-1 (platelet endothelial cell adhesion molecule) is a protein in the immunoglobulin superfamily found on the surface of platelets, monocytes, neutrophils, and some types of T-cells. CD31 makes up a large portion of endothelial cell intercellular junctions. CD31 is encoded in humans by the PECAM-1 gene and is commonly used as a marker for endothelial cells.
  • CD34 is a cell surface glycoprotein that functions as a cell-cell adhesion factor.
  • the CD34 protein is a member of a family of single-pass transmembrane sialomucin proteins that are expressed on early hematopoietic and vascular-associated tissue.
  • CD34 is encoded in humans by the CD34 gene. It is commonly used as a marker for hematopoietic and/or vascular endothelial cells.
  • clonal refers to a population of cells obtained by the expansion of a single cell into a population of cells all derived from that original single cell and not containing other cells.
  • the terms “clonal progenitor cell”, “embryonic clonal progenitor cell”, “clonal progenitor cell line” and “embryonic clonal progenitor cell line” each refer to progenitor cell lines that are derived clonally, i.e., derived by the expansion of a single cell into a population of cells all derived from that original single cell and not containing other cells.
  • embryonic stem cell refers to a pluripotent cell that is derived from a blastocysts, such as an in vitro fertilized blastocyst.
  • Embryonic stem cells include human embryonic stem cells, which are available as established cell lines. The established cell lines are available commercially from numerous public cell banks, e.g. WiCell and private corporations, e.g. ESI BIO.
  • human pluripotent cell or “human pluripotent stem cell” as used herein refers to a human cell which is capable of differentiating into at least one cell type found in or derived from each of the three primary germ layers. Some human pluripotent stem cells have the ability to differentiate into all cells found in or derived from each of the three primary germ layers. Examples of human pluripotent stem cells include human embryonic stem cells (Thomson (1998) Science 282:1145), human embryonic germ cells (Shamblott et al. (2001) PNAS 98:113 and induced pluripotent cells (Takahashi et al. (2007) Cell 131:861.
  • induced pluripotent stem cell refers to a pluripotent cell that has been genetically reprogrammed using any technique known in the art from an adult somatic cell back to the developmentally less mature pluripotent state.
  • miRNA refers to microRNA which includes RNA species that are 21-25 nt long and may be single- or double-stranded.
  • MicroRNAs are short, non-coding RNA molecules that have been found in animals, including humans, and in plants.
  • the term encompasses small interfering RNA (siRNA) and small temporal RNA (stRNA), as well as miRNA proper.
  • miRNAs are transcribed as parts of longer RNA molecules and processed in the nucleus by the dsRNA ribonuclease Drosha to hairpin structures 70-100 nucleotides long. These are transported to the cytoplasm where they are digested to 21-23-mers by the dsRNA ribonuclease Dicer. Single-stranded miRNAs bind to complementary sequences in mRNA thereby inhibiting translation.
  • miR-126 is a human microRNA that is specifically expressed in endothelial cells, throughout capillaries and in larger blood vessels. miR-126 plays a role in angiogenesis by regulating the expression levels of various genes by pre- and post-transcription mechanisms. As used herein, the term “miR-126” refers to all of the following: the stem-loop miR-126, miR-126-3p (3′ arm of the hairpin precursor) and miR-126-5p (5′ arm of the hairpin precursor). miRNA naming conventions are described in Kozomara and Griffiths-Jones, (2014) Nucleic Acids Res. 42(Database issue):D68. The terms “miR-126-3p” and “hsa-miR-126-3p” are also used interchangeably throughout this application.
  • nucleic acid means at least two nucleotides covalently linked together.
  • an oligonucleotide is an oligomer of 6, 8, 10, 12, 20, 30 or up to 100 nucleotides.
  • an oligonucleotide is an oligomer of at least 6, 8, 10, 12, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 400, or 500 nucleotides.
  • a “polynucleotide” or “oligonucleotide” may comprise DNA, RNA, cDNA, PNA or a polymer of nucleotides linked by phosphodiester and/or any alternate bonds.
  • peptide refers to two or more amino acids joined by a peptide bond.
  • a peptide can, in some instances, be a portion of a full length protein.
  • protein refers to a full length protein, i.e. one having all of the amino acids coded for by the mRNA that encodes the particular protein. Also included in the definition are modified proteins where one or more amino acids have been cleaved (e.g. a signal sequence) as a result of the protein being secreted from a cell.
  • progenitor cell line refers to a line of cells that is more differentiated (developed) compared to a pluripotent cell, such as iPS cell or an hES cell, but is not terminally differentiated. Progenitor cells will have enhanced replicative capacity compared to a terminally differentiated cell which typically has senesced. Progenitor cells may also have longer telomere lengths compared to a cell that has terminally differentiated. Progenitor cell lines, when cultured, may be able double in population size at least 5, at least 10, at least 20, at least 30, at least 40, at least 50 times.
  • progenitor cell lines may be able to double in population size 5-400 times, 10-300 times, 20-200 times, 30-80 times, 40-60 times.
  • a progenitor cell line is an embryonic progenitor cell.
  • Embryonic progenitor cell is obtained from a pluripotent cell such as an iPS cell or a hES as previously described. See West et al. (2008) Regen Med 3:287; US Patent Application Publication Nos. 20080070303 20100184033.
  • subject includes, but is not limited to, humans, non-human primates and non-human vertebrates such as wild, domestic and farm animals including any mammal, such as cats, dogs, cows, sheep, pigs, horses, rabbits, rodents such as mice and rats.
  • the term “subject,” refers to a male. In some embodiments, the term “subject,” refers to a female.
  • suitable media refers to a solution that can be used to grow cells in culture.
  • a suitable media may include a formulation of salts and/or buffering reagents.
  • a suitable media may include any or all of the following: salts, sugars, amino acids, proteins, growth factors, cytokines, and hormones, additives such as serum, albumin, antibiotics, insulin, selenium and transferrin.
  • Suitable culture media includes for example commercially available culture media such as DMEM, MEM Stem Pro and the like.
  • treat can refer to both therapeutic treatment or prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological condition, symptom, disorder or disease, or to obtain beneficial or desired clinical results.
  • the term may refer to both treating and preventing.
  • beneficial or desired clinical results may include, but are not limited to one or more of the following: alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease.
  • Treatment includes eliciting a clinically significant response. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.
  • the exosomes are derived from a clonal progenitor cell
  • the exosomes will be of uniform quality and composition.
  • the exosomes isolated from a clonal progenitor cell will not vary as a result of genetic variation of the source cell.
  • the molecular composition of the contents and the bio-physical characteristics of the vesicles will be consistent and reproducible.
  • the invention provides an overabundance of the exosomes of the invention. This is in direct contrast with exosomes obtained from other sources known in the art where the paucity of the cell type or the problem of senescence limits the availability of a reproducible exosome.
  • the cells giving rise to the exosomes of the invention are neither transformed nor malignant, thus avoiding any possible concern regarding carcinogenesis of the exosomes.
  • the exosomes of the invention may have diameter ranging from about 20 nm-130 nm; from about 30 nm-120 nm; about 40 nm-110 nm; about 50 nm-100 nm; about 85 nm-95 nm. In some embodiments the exosomes of the invention have a diameter of about 90 nm. In some embodiments the exosomes of the invention have a diameter of about 88 nm.
  • the exosomes of the invention may serve as depots for the delivery of therapeutic molecules of any kind.
  • the exosomes of the invention can be engineered to contain therapeutic molecules such as nucleic acids, proteins, peptides, small molecules such as drugs and the like. Any technique known in the art can be used to load the exosomes of the invention with a desired therapeutic molecule.
  • cationic lipids could be used to transfect the exosomes with a desired nucleic acid such as DNA, RNA, include mRNA and miRNA.
  • HIV tat protein could be used to transport protein or peptide therapeutics into the exosomes of the invention.
  • the therapeutic molecules can be chosen, engineered or designed to have any desired therapeutic effect. For example molecules associated with enhanced angiogenesis could be loaded into the exosomes of the invention, e.g. VEGF.
  • Exosomes may be isolated from any suitable cell that contains exosomes. Described infra are several exemplary cell and cell types that may be used to implement this method. The method may involve seeding the cell at an appropriate density in a tissue culture vessel and then incubating the cells in a suitable media or buffer for a suitable period of time. In some embodiments the cells may be permitted to attach to the culture vessel before the exosomes are isolated. In other embodiments the cells may be kept in suspension while the exosomes are isolated. The cells may be permitted to replicate in culture before the exosomes are isolated. Alternatively, the exosomes may be isolated from the cells that have not replicated, or replicated minimally (e.g. less than 1 doubling).
  • the cell density may range from about 1 k/cm 2 -100 k/cm 2 , 10 k/cm 2 -90 k/cm 2 , 20 k/cm 2 -80 k/cm 2 , 30 k/cm 2 -70 k/cm 2 , 40 k/cm 2 -60 k/cm 2 .
  • the cells are seeded at a density (cells per unit area) of 40 k/cm 2 .
  • the method may be practiced by incubating the cells in the media or buffer for about 1-72 hours, 1-48 hours, 2-24 hours, 3-18 hours, 4-16 hours, 5-10 hours. In some embodiments the cells are incubated for about 16 hours.
  • the isotonic solution may be harvested.
  • the isotonic solution may be pipetted or decanted into another vessel such as a centrifuge tube.
  • a precipitating agent may be added to the isotonic solution at this time to facilitate the precipitation of the exosomes in the solution.
  • precipitating agents include a solution that is about 15% polyethylene glycol.
  • a commercially prepared precipitating agent may be used, e.g., Total Exosome Isolation Reagent (Life Technologies, Carlsbad, Calif.).
  • the harvested cell conditioned isotonic solution may be centrifuged at about a temperature of 2° C., 4° C., 6° C., 8° C., 10° C., 12° C., 14° C., 16° C., 18° C., 20° C., 22° C., 24° C., 26° C. In one embodiment the harvested cell conditioned isotonic solution are centrifuged at about a temperature of 4° C.
  • the volume of buffer may be about 0.01 volumes-about 0.09 volumes, about 0.02 volumes to about 0.08 volumes; about 0.03 volumes to about 0.07 volumes of the precipitating solution.
  • the harvested cell conditioned isotonic solution is resuspended in PBS at a volume equivalent to about 0.01 volumes of the precipitating solution.
  • the harvested exosomes may be used immediately or frozen and stored, e.g., at ⁇ 20° C., for later use.
  • progenitor cells serve as the source of the exosomes described infra.
  • the progenitor cell may be from any animal or plant.
  • the exosome may be from a mammal, such as a human, a non-human primate, a horse, a cow, a sheep, a goat, a pig, a cat, a dog, a rabbit, a guinea pig, a rodent such as a mouse or a rat.
  • a progenitor cell will not have an essentially unlimited replicative capacity as typically found in embryonic stem cells, but will nonetheless have, a result of their longer telomeres, a greater replicative capacity compared to adult primary cells or tissues (e.g. primary cells) or adult stem cells.
  • the progenitor cell may be derived from a pluripotent stem cell, such as an embryonic stem cell or an induced pluripotent stem cell.
  • the progenitor cell may be a clonal cell or an oligoclonal cell.
  • An oligoclonal cell would include a population of cells similar cells, e.g. phenotypically or genetically.
  • the progenitor cell may be a clonal human embryonic progenitor cell.
  • the progenitor cell may be a clonal human embryonic endothelial progenitor cell.
  • the progenitor cell may be a clonal embryonic progenitor cell that expresses CD31 and CD36.
  • the progenitor cell may be a clonal embryonic progenitor cell expressing one or more genes listed in Table 1.
  • the progenitor cell may be a clonal embryonic progenitor cell expressing a plurality of the genes listed in Table 1.
  • the progenitor cell may be a clonal embryonic progenitor cell expressing the genes listed in Table 1.
  • progenitor cells are clonal cells obtained from pluripotent stem cells they will provide an almost unlimited source of the same exosomes. This is due to two factors: the genetic identity of the original cellular source material and the enhanced telomere lengths found in early progenitors which provide for enhanced replicative capacity relative to adult tissue or cells or adult stem cells. Moreover, unlike adult stem cells which are typically available in very small numbers and are difficult to expand in culture, the clonal embryonic progenitors described infra are available in large numbers and are relatively easy to expand in culture.
  • the progenitor cell is not an adult stem cell. In some embodiments of the invention the progenitor cell is not an MSC. In some embodiments the clonal progenitor cell is not transfected or engineered to express an exogenous gene. In some embodiments the clonal progenitor cell is not transfected to express an oncogene. In some embodiments the clonal progenitor does not express c-myc. In other embodiments the clonal progenitor cell is transfected or engineered to express an exogenous gene. Examples of suitable exogenous genes include the catalytic component of human telomerase, e.g. hTERT.
  • exosomes described herein may be used in therapeutic, research and diagnostic applications.
  • the exosomes described infra may be added to a cell culture to enhance one or more phenotypic traits of the cells.
  • the exosomes of the invention may be added to a cell culture to inhibit one or more phenotypic traits of the cells.
  • the exosomes of the invention may be added to a cell culture to provide a new phenotypic trait of the cells.
  • the exosomes of the invention may be added to a culture of endothelial cells to enhance the ability of the cells to form vascular tube like structures.
  • the exosomes of the invention may be added to any cell having the ability to form vascular tube like structures to enhance the cells ability to form tube like structures.
  • the exosomes of the invention are contacted with a cell thereby providing at least one new phenotypic trait to the cell.
  • the exosomes of the invention may confer the ability to form vascular tube like structures to cell lacking the ability to form vascular tube like structures before it was contacted with the exosomes of the invention.
  • the exosomes of the invention may be added to a culture of perivascular cells to enhance the ability of the perivascular cells to form vascular tube like structures.
  • the invention provides a method of increasing the length of a vascular tube like structure formed by a cell such as an endothelial relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the invention provides a method of increasing the length of a vascular tube like structure formed by a cell such as a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the invention provides a method of increasing the branching of a vascular tube like structure formed by an endothelial cell relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the invention provides a method of increasing the branching of a vascular tube like structure formed by a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the invention provides a method of increasing the number of loops in the vascular tube like structures formed by an endothelial cell relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the invention provides a method of increasing the number of loops in the vascular tube like structures formed by a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • the exosomes of the invention may be administered therapeutically to a subject in need of treatment.
  • the exosomes of the invention may be administered to a subject in need of treatment for any disease requiring the enhanced ability to form vascular tube like structures.
  • the exosomes of the invention may be used to treat a subject suffering from cardiovascular disease, heart failure, infarction, chronic wounds, ulcer, clogged vessels or arteries, damaged vessels, stenotic vessels, arteriosclerosis, angina, peripheral vascular disease, Alzheimer's disease, ischemia, diabetes, cancer, cell replacement transplant or therapy, tissue and cell regenerative therapy and Parkinson's disease.
  • the exosomes may be used as depot to deliver therapeutic molecules such as small molecules, nucleic acids, proteins and peptides.
  • the exosomes of the invention may be directly administered to a subject in need of treatment or an in vitro cell culture.
  • the exosomes can be provided enclosed within a matrix or scaffold.
  • Suitable matrices or scaffolds may include a matrix or scaffold comprised of one or more extracellular matrix proteins, e.g. laminin, fibronectin and the like.
  • Other suitable matrices or scaffolds include Matrigel® which is a murine sarcoma extract.
  • the matrix or scaffold may be a hydrogel.
  • the hydrogel may be comprised of hylauronate and gelatin (see U.S. Pat. Nos. 8,324,184; 7,928,069).
  • the exosomes of the invention may be delivered in HyStem (Biotime, Inc., Alameda Calif.).
  • the exosomes of the invention may be used to isolate one or more nucleic acids, proteins or peptides expressed by a progenitor cell serving as the source of the exosome.
  • the proteins or peptides isolated from the exosomes of the invention can be used to make antibodies to the isolated proteins or peptides (See Harlow et al. Antibodies: A Lab Manual 2 nd Edition; Cold Spring Harbor Press 2013).
  • the exosomes of the invention may be used in drug screening assays.
  • the exosomes described infra enhance vascular tube formation in vitro
  • the exosomes can be used to screen for drugs that enhance or inhibit this capability.
  • a cell culture comprising cells having the ability to form vascular tube like structures may be contacted with the exosomes of the invention and a drug candidate may be applied to the same cell culture either before, after or simultaneously with the exosomes to determine the effect of the drug the ability of the exosomes to enhance vascular tube formation in the cell culture.
  • the effects can be compared to untreated cells and cells treated only with the exosomes of the invention.
  • Modes of administration for a therapeutic can be, but are not limited to, sublingual, injectable (including short-acting, depot, implant and pellet forms injected subcutaneously or intramuscularly), or by use of vaginal creams, suppositories, vaginal rings, rectal suppositories, intrauterine devices, and transdermal forms such as patches and creams.
  • Specific modes of administration will depend on the indication.
  • the selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response.
  • the amount of therapeutic to be administered is that amount which is therapeutically effective.
  • the dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular animal treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician).
  • compositions containing the therapeutic of the present disclosure and a suitable carrier can be solid dosage forms which include, but are not limited to, tablets, capsules, cachets, pellets, pills, powders and granules; topical dosage forms which include, but are not limited to, solutions, powders, fluid emulsions, fluid suspensions, semi-solids, ointments, pastes, creams, gels and jellies, and foams; and parenteral dosage forms which include, but are not limited to, solutions, suspensions, emulsions, and dry powder; comprising an effective amount of a polymer or copolymer of the present disclosure.
  • the active ingredients can be contained in such formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • pharmaceutically acceptable diluents fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like.
  • the means and methods for administration are known in the art and an artisan can refer to various pharmacologic references for guidance. For example, Modern Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Gilman's The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980) can be consulted
  • compositions of the present disclosure can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • the compositions can be administered by continuous infusion subcutaneously over a period of about 15 minutes to about 24 hours.
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions can be formulated readily by combining the therapeutic with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the therapeutic of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinyl pyrrolidone (PVP).
  • disintegrating agents can be added, such as, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores can be provided with suitable coatings.
  • suitable coatings can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active therapeutic doses.
  • compositions which can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers.
  • the active therapeutic can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the pharmaceutical compositions can take the form of, e.g., tablets or lozenges formulated in a conventional manner.
  • the therapeutic for use according to the present disclosure is conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or
  • compositions of the present disclosure can also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the therapeutic of the present disclosure can also be formulated as a depot preparation.
  • Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • compositions can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions of the present disclosure can be applied to a plaster, or can be applied by transdermal, therapeutic systems that are consequently supplied to the organism.
  • compositions can include suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as, e.g., polyethylene glycols.
  • compositions of the present disclosure can also be administered in combination with other active ingredients, such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • active ingredients such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • the disintegrant component comprises one or more of croscarmellose sodium, carmellose calcium, crospovidone, alginic acid, sodium alginate, potassium alginate, calcium alginate, an ion exchange resin, an effervescent system based on food acids and an alkaline carbonate component, clay, talc, starch, pregelatinized starch, sodium starch glycolate, cellulose floc, carboxymethylcellulose, hydroxypropylcellulose, calcium silicate, a metal carbonate, sodium bicarbonate, calcium citrate, or calcium phosphate.
  • the diluent component may include one or more of mannitol, lactose, sucrose, maltodextrin, sorbitol, xylitol, powdered cellulose, microcrystalline cellulose, carboxymethylcellulose, carboxyethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, methylhydroxyethylcellulose, starch, sodium starch glycolate, pregelatinized starch, a calcium phosphate, a metal carbonate, a metal oxide, or a metal aluminosilicate.
  • the optional lubricant component when present, comprises one or more of stearic acid, metallic stearate, sodium stearylfumarate, fatty acid, fatty alcohol, fatty acid ester, glycerylbehenate, mineral oil, vegetable oil, paraffin, leucine, silica, silicic acid, talc, propylene glycol fatty acid ester, polyethoxylated castor oil, polyethylene glycol, polypropylene glycol, polyalkylene glycol, polyoxyethylene-glycerol fatty ester, polyoxyethylene fatty alcohol ether, polyethoxylated sterol, polyethoxylated castor oil, polyethoxylated vegetable oil, or sodium chloride.
  • the invention provides a kit comprising exosomes isolated from a progenitor cell, such as a human clonal progenitor cell.
  • the progenitor cell may be an endothelial progenitor cell, such as human clonal embryonic progenitor cell, e.g. 30MV2-6.
  • the exosomes may be provided in one or more containers.
  • the exosomes may be provided in a suitable buffer, e.g. PBS or a suitable media, such as a commercially available cell culture media, e.g. DMEM.
  • the kit may further contain a cell having the ability to form vascular tube like structures.
  • the cell may be an endothelial cell, e.g.
  • the cells may be provided in a suitable media, e.g. DMEM or the like or alternatively the cells may be provided in a buffer such as PBS. In some embodiments the cells may be provided frozen in a suitable freezing media such as a commercially available media supplemented with DMSO.
  • the kit may optionally include instructions as to how to reconstitute the exosomes, culture the cells and/or contact the cells with exosomes so as to enhance vascular tube like formation.
  • the invention provides a kit comprising a human clonal embryonic progenitor cell, such as 30-MV2-6.
  • the cell may be provided in at least one container in suitable media or buffer.
  • the kit may include buffers and/or media for isolating exosomes from the cells.
  • the kit may contain one or more vessels, e.g. a multi-well plate for culturing the cells.
  • the kit may further contain a cell line capable of forming vascular tube like structures such as endothelial cells. Suitable cells include endothelial cells such as HUVEC and/or a a perivascular cell. Any or all of the cells may be provided frozen in a suitable media, e.g.
  • the kit may optionally include instructions as to how to culture the cells and/or contact the endothelial cells with exosomes isolated from the progenitor cells so as to enhance or induce vascular tube like formation.
  • exosome of 1 wherein the exosome enhances the formation of vascular tube like formations when contacted with an endothelial cell.
  • exosome of 1 further comprising a pharmaceutical carrier.
  • a method of isolating an exosome from a clonal progenitor cell comprising 1) culturing the clonal progenitor cell in a suitable media or buffer for a time sufficient to allow the clonal progenitor cell to exocytose exosomes into the culture media or buffer; 2) harvesting the media or buffer from the cell culture of step 1; and 3) isolating the exosomes from the media or buffer of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • step 3 comprises centrifuging the harvested media of buffer.
  • step 2 The method of 14, wherein after step 2, the method further comprises a step of incubating harvested media or buffer.
  • a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome.
  • a method of inducing or enhancing a cell's ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a progenitor cell.
  • progenitor cell line is a human progenitor cell line.
  • progenitor cell line is a clonal progenitor cell line.
  • progenitor cell line is an endothelial progenitor cell line.
  • the method of 39 further comprising a pharmaceutical carrier.
  • Example 1 Preparation of Exosomes Derived from a Human Embryonic Progenitor Cell Line
  • Exosomes were prepared from a human embryonic progenitor cell line (PureStem® cell line, ESI Bio, Alameda, Calif.). PureStem® cell lines are scalable clonally pure embryonic progenitor cell lines derived from human embryonic stem (hES) cells (West et al. (2008) Regen Med. 3(3):287).
  • the 30-MV2-6 PureStem® cell line is a CD31 positive, CD34 positive, endothelial progenitor line derived from the ESI-017 embryonic stem cell line.
  • a gene expression profile of the 30-MV2-6 cells as analyzed by microarray is provided herein in Table 1, and includes genes yielding relative fluorescence units >1000 rfu.
  • the 30-MV2-6 cells were maintained in EGM-MV2 media (PromoCell) plus TGF ⁇ inhibitor (SB43154) (Cayman Chemical Co., Ann Arbor, Mich.) in a 5% CO 2 , 5% O 2 humidified cell culture incubator.
  • the 30-MV2-6 cells were seeded at a density of 40 k/cm 2 .
  • the culture media was removed and after two washes with phosphate buffered saline (PBS), (PBS) was added at 0.1 ml/cm 2 to produce conditioned medium from which exosomes were isolated.
  • PBS phosphate buffered saline
  • basal EGM-MV2 medium Basal EGM-MV2 medium (PromoCell, Heidelberg, Germany) without fetal calf serum or growth factor additives was substituted for PBS.
  • the media was conditioned by the cells in a humidified tissue culture incubator for 16 hours at 37° C. at 5% CO 2 and 1% O 2 .
  • the conditioned medium was collected and 0.5 volumes of Total Exosome Isolation Reagent (Life Technologies) was added and mixed well by vortexing until there was a homogenous solution.
  • a solution of 15% polyethylene glycol (Hampton Research, Aliso Viejo, Calif.), 1.5 M NaCl (Sigma, St Louis, Mo.) was substituted for the Total Exosome Isolation Reagent.
  • the sample was incubated at 4° C. for at least 16 hours to precipitate the exosomes, followed by centrifugation at 10,000 ⁇ g for 1 hour at 4° C. The supernatant was removed and the pellet is resuspended in 0.01 volume of PBS.
  • Samples were adsorbed to the ELISA plate by incubation overnight in PBS.
  • the PBS was removed and wells were washed 3 times in ELISA wash buffer (Thermo Scientific, Waltham, Mass.) followed by incubation with primary anti-CD63 antibody (BD Pharmingen, Franklin Lakes, N.J.) for 1 hour at room temperature.
  • the primary antibody was removed followed by washing 3 times in wash buffer and incubation with secondary antibody (HRP conjugated anti-mouse) (Invitrogen, Grand Island, N.Y.)) at 1:3000 dilution for 1 hour at room temperature.
  • Angiogenic activity of exosomes was assayed using an in-vitro endothelial tube forming assay.
  • the assay was performed in triplicate in a ⁇ well slide (Ibidi, Verona, Wis.) or in single wells of a 96-well plate.
  • the wells were coated with reduced growth factor Matrigel (BD, Franklin Lakes, N.J.).
  • Human umbilical cord vascular endothelial cells that were grown to 70-80% confluence were plated at 5000-7000 cells per well in a ⁇ well slide in 50 ul of EGM-MV2 basal medium (Promocell, Heidelberg, Germany) (no supplements) containing up to 10 ⁇ l of exosomes in PBS or equivalent volume of PBS without exosomes as a negative control or in 50 ⁇ l of complete EGM-MV2 medium with growth factor supplements as a positive control.
  • the assay was performed in a 96-well plate using 60,000 to 90,000 cells per well in 280 ⁇ l of medium and 20 ⁇ l of exosomes or PBS. The cells are incubated at 37° C.
  • FIG. 2 A shows an increase in HUVEC endothelial tube formation when grown in the presence of 30-MV2-6 derived exosomes (in PBS) compared to basal medium with an equivalent amount of PBS (with no exosomes) added (negative control).
  • the quantified results indicate that total tube length, cell covered area, branch points and the number of loops were all increased by the addition of exosomes compared to basal medium indicating that the 30-MV2-6 exosomes are angiogenic.
  • Angiogenic activity of exosomes was also assessed by their ability to stimulate in vitro tube formation using human embryonic stem (hES) cell derived perivascular embryonic progenitor cells (PEPCs) (also called 017-PC-A) cells bearing pericyte and stemness markers (CD146, CD133, Podoplanin)(U.S. patent application Ser. No. 14/625,621, filed on Feb. 18, 2015).
  • the assay was performed as described for HUVECs except that hES cell derived PEPCs were used instead of HUVECs.
  • the assay was performed in triplicate using ⁇ well slides (Ibidi, Verona, Wis.).
  • the hES pericytes that were grown in defined medium differ from HUVECs in their response to complete medium.
  • HUVECs respond to complete EGM-MV2 medium in the tube forming assay with robust tube formation ( FIG. 2 A ).
  • hES PEPCs migrate to form foci consisting of cellular aggregates ( FIG. 3 A ) when grown on Matrigel in complete EGM-MV2 medium and thus exhibited reduced tube formation.
  • the hES PEPCs grown in defined medium form incomplete tubes ( FIG. 3 B ) similar to HUVEC in basal medium ( FIG. 2 A ).
  • the hES PEPCs grown in the presence of 30-MV2-6 exosomes displayed an increase in tube formation as shown in FIG. 3 C- 3 E .
  • the tube formation was dose responsive and quantitative analysis indicated an increase in all 4 tube formation parameters ( FIG. 3 F- 3 I ).
  • hES derived PEPCs respond to exosomes with increased tube formation but respond to complete medium with reduced tube formation compared to basal medium.
  • 30-MV2-6 exosomes induce angiogenesis in a non-angiogenic cell type that does not respond to the angiogenic factors present in EGM-MV2 complete medium.
  • Example 3 Comparison of Angiogenic Activity of Exosomes Derived from a Human Embryonic Progenitor Cell Line and Exosomes Derived from Adult Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs)
  • Exosomes were prepared from an embryonic stem cell derived PureStem® cell line, 30-MV2-6, and from adult bone marrow-derived mesenchymal stem cells (BM-MSCs) from two different commercial sources (Lonza and Promocell), according to methods described in Example 1.
  • the angiogenic activity was assessed using the in vitro endothelial tube formation assay described in Example 2. Briefly, the exosomes (2 ⁇ 10 8 particles/50 ⁇ l) were incubated with human umbilical cord vascular endothelial (HUVEC) cells for 12-16 hours on low growth factor Matrigel using a ⁇ -well slide (Ibidi, Verona, Wis.).
  • HUVEC human umbilical cord vascular endothelial
  • Tube length was assessed by image capture and analyzed using Angiogenesis Analyzer in ImageJ (http://rsb.info.nih.gov/ij/) image processing program. Total tube length formed per image/ ⁇ -well was calculated relative to total tube length formed for HUVEC in complete EGM-MV2 medium (CM) containing angiogenic growth factors (VEGF and FGF2) and fetal bovine serum (FBS).
  • CM EGM-MV2 medium
  • FGF angiogenic growth factors
  • FBS fetal bovine serum
  • Exosomes derived from the 30-MV2-6 cell line were compared to early passage BM-MSCs from the two different sources, Promocell (Heidelberg, Germany) ( FIG. 4 , panel A) and from Lonza (Basel, Switzerland) ( FIG. 4 , panel B). In both cases the angiogenic activity of 30-MV2-6 derived exosomes was greater than the angiogenic activity of BM-MSC derived exosomes.
  • the total tube length of HUVECs incubated in basal medium with 30-MV2-6 derived exosomes (in PBS) was similar to HUVECs incubated in complete EGM-MV2 medium and significantly greater than BM-MSC derived exosomes (from either source) or HUVEC incubated in basal EGM-MV2 medium and PBS alone.
  • the total tube length resulting from BM-MSC derived exosomes was on average slightly higher than PBS in basal medium (BM) but the difference was not statistically significant (p ⁇ 0.05).
  • BM-MSC and 30-MV2-6 derived exosomes were further compared in a dose response experiment to determine differences in their potency.
  • Exosomes were prepared and their concentration determined using nanoparticle tracking analysis (NTA; Nanosight, Malvern Instruments Ltd, Malvern, Worcestershire, UK).
  • ELISA was used to confirm the presence of transpanins CD63, CD81 and CD9 that are typically expressed on exosomes.
  • the 30-MV2-6 exosomes were tested at doses ranging from 50 million to 400 million exosomes per well.
  • the BM-MSC exosomes were tested at doses ranging from 400 to 1200 million exosomes per well, because no significant activity was observed at 200 million exosomes per well. The results, shown in FIG.
  • FIG. 5 indicate that the angiogenic response of HUVEC cells to 30-MV2-6 exosomes is dose responsive starting at 50 million per well and saturating at doses of ⁇ 200 million exosomes per well ( FIG. 5 , panel A).
  • the BM-MSC exosomes showed a dose response of increasing angiogenic activity at doses from 400 million to 1200 million exosomes per well ( FIG. 5 , panel B).
  • the potency of 30-MV2-6 derived exosomes was at least 6-fold greater than that of BM-MSC derived exosomes, having the equivalent activity at 200 million exosomes per well as BM-MSC derived exosomes at 1200 million exosomes per well ( FIG. 5 , panel B).
  • Example 4 Comparison of miRNA Content in Exosomes Derived from a Human Embryonic Progenitor Cell Line Versus Exosomes Derived from Adult BM-MSCs
  • the exosome cDNA was amplified by polymerase chain reaction (PCR) using the miScript pre-AMP PCR kit (Qiagen) and miScript pre-AMP pathway primer mix (human miFinder MBHS-001Z; Qiagen) according to the manufacturer's recommended protocol. Relative miRNA levels were assessed for 84 human miRNAs by quantitative PCR using the human miFinder miScript miRNA PCR array (#331221; Qiagen) according to the manufacturer's recommended protocol. The results were analyzed using the ⁇ CT method of relative quantitation available at (http://perdataanalysis.sabiosciences.com/mirna).
  • MiR-126 is a known angiogenic miRNA (“angiomiR”) that is endothelial cell-specific and has been shown to regulate both vascular integrity and developmental angiogenesis.
  • angiomiR angiogenic miRNA
  • RNA from 30-MV2-6 exosomes was also used to compare the miRNA content of angiogenic versus non-angiogenic exosomes.
  • Exosomes derived from HT1080 cells are not angiogenic in the HUVEC in vitro angiogenesis assay at 2.0 ⁇ 10 8 exosomes, a dose at which 30-MV2-6 exosomes show maximum angiogenic activity.
  • HT1080 exosome RNA was analyzed on the miFinder miScript PCR array of 84 human miRNAs as described above and compared to 30-MV2-6 and BM-MSC exosome RNA (Table 4).
  • HT1080 exosomes contain the lowest amount of miRNA for all miRNAs tested, except for miR-96-5p and miR-142-3p, which are lowest in 30-MV2-6 exosomes.
  • the miRNA miR-142-3p is expressed at highest levels in HT1080 exosomes and is known to repress several inhibitors of oncogenic transformation. It is mimicked by Kaposi sarcoma viral miRNA, miR-K10a. Forte et al. (2015) J Virol 89(4): 2333.
  • the miR-96-5p miRNA is present in highest levels in BM-MSC exosomes and is higher in HT1080 exosomes than 30-MV2-6 exosomes.
  • This miRNA is thought to be involved in osteogenic and adipogenic differentiation in BM-MSCs (Laine et al. (2012) J Cell Biochem. 113(8):2687) but is also involved in tumor cell proliferation (Lin et al. (2010) Plos One 5(12):e15797; Haflidadottir et al. (2013) Plos One 8(8):e72400). Strikingly the miRNA with the highest levels in 30-MV2-6 exosomes relative to HT1080 exosomes is miR-126-3p.
  • This known angiogenic miRNA is present at a 9618-fold higher level in 30-MV2-6 exosomes than HT1080 exosomes.
  • MiR-155 is 4-fold higher in 30-MV2-6 exosomes than BM-MSC or HT1080 exosomes and is anti-angiogenic but pro-arteriogenic.
  • 6 are known to be involved in angiogenesis.
  • the miRNAs miR-18a-5p, miR-20a-5p, miR-424-5p, miR-17-5p, and miR-7-5p miRNAs have anti-angiogenic activity.
  • anti-angiogenic miRNAs are more than 2.5-fold enriched in 30-MV2-6 exosomes compared to BM-MSC exosomes.
  • the pro-angiogenic miR-106b is 9-fold enriched in 30-MV2-6 but only 4-fold enriched in BM-MSC exosomes compared to HT1080 exosomes. It is needed for neovascularization after hind limb ischemia. Semo et al. (2014) Eur Heart J. 35(45):3212.
  • miRNAs including miR-143-3p (Climent et al. (2015) Circ Res. 116(11):1753), miR-223-3p (Dai et al. (2014) Plos One 9(10):e108468), miR-222-3p (Suarez and Sessa (2009) Circ Res. 104(4):442), miR-15a, miR-15b and miR-16 (Spinetti et al. (2013) Circ Res. 112(2):335; Liu et al. (2012) Cell Physiol Biochem. 29(5-6):851)) were enriched for and/or present at highest level in the BM-MSC-derived exosomes.
  • Example 5 Comparison of Angiogenic Activity of Exosomes Derived from Various Clonal Embryonic Stem Cell Lines and Exosomes Derived from the Parental Pluriopotent Stem Cell Lines
  • hPS human pluripotent stem
  • the resulting cell lines are not immortalized but have higher replicative potential than primary cell lines because of their long telomere length that is near that of the parental hPS cell line from which they are derived.
  • a wide diversity of cell types was produced by exposing hPS cells to an array of cell culture medium, cell matrix, and growth conditions followed by selective pressure for clonal growth and scalability. Over 140 such cell types have been determined to be distinct by analysis of total transcribed RNA using standard Illumina microarrays.
  • the in vitro angiogenesis assay (described in detail in Example 2) was used to screen clonal embryonic progenitor cells for production of angiogenic exosomes. As shown in Table 5, most embryonic endothelial progenitor cell-derived exosomes have angiogenic activity in the range of 30-MV2-6 derived exosomes (+; relative tube length (RTL)>0.75 and ⁇ 1.25). The 30-MV2-9 exosomes scored highest (++; RTL>1.25). Two endothelial progenitor lines scored negative ( ⁇ ; RTL ⁇ 0.75).
  • Exosomes from an osteochondral line, primary fibroblasts (BJ), BM-MSCs, and a human sarcoma cell line (HT1080) were also negative.
  • the two clonal smooth muscle cell progenitor cell lines and one clonal pericyte line tested were positive in the in vitro vascular tube formation assay.
  • Exosomes prepared from conditioned medium of the parental human embryonic stem cell lines H9 (WA09) and ESI-017 were also positive in the in vitro vascular tube formation assay.
  • mice Male Nu/J mice aged 6-8 weeks; 2 plugs/mouse; 2 mice/group) were injected subcutaneously with approximately 300 ⁇ l of Matrigel containing PBS, 4 ⁇ 10 8 /ml exosomes, or 150 ng/ml bFGF plus 60 ng/ml VEGF (positive control). The plugs were removed at day 14 after implant followed by fixation and paraffin embedding. The sections were stained with hematoxylin and Eosin (H&E) for histological examination and stained with von Willabrand factor antibody for detection of endothelial cells.
  • H&E hematoxylin and Eosin
  • the exosome containing plugs show regions of infiltration of cells into the plug with vessel formation ( FIG. 7 , panels A and C).
  • the positive control plugs containing growth factors have regions of vessel formation (not shown)
  • Immunostaining with antibody against von Willabrand factor FIG. 7 , panels B and D
  • the PBS control plugs show less cell infiltration and no vessel formation ( FIG. 7 , panel E).
  • Clonal embryonic progenitor cell lines described here are advantageous over other sources of biologically active exosomes because of their scalability.
  • the parental pluripotent stem cell line to 30-MV2-6 which also produces angiogenic exosomes is costly to scale up because of the requirements for specialized medium and cell matrix (e.g. Matrigel).
  • Primary endothelial stem cells or mesenchymal stromal cells rapidly lose differentiation and proliferative capacity upon culture in vitro.
  • MSCs begin to senesce in culture after 7-12 passages (approximately 10 population doublings) and show multiple changes including altered surface marker expression and increased autofluorescence.
  • human embryonic clonal progenitor lines such as the cell lines of the instant invention are grown under standard tissue culture conditions and medium and are highly scalable with typical replicative lifespans of 60 to 100 population doublings.
  • the Terumo Quantum Cell Expansion system (the bioreactor used in the instant example) is an automated hollow fiber cell culture platform designed for GMP compatible production of cells for use in cell therapy.
  • the bioreactor was seeded at a density of approximately 900 cells/cm 2 with approximately 4.0 ⁇ 10 7 30-MV2-6 cells (passage 9) and the cells were cultured for 13 days under their standard growth conditions of EGM-MV2 medium and 5% oxygen.
  • the exosomes were collected by exchanging the complete medium for conditioning medium (basal EGM-MV2 medium without serum added; alternatively PBS may be also used).
  • the conditioning medium was left in the bioreactor for 16 hours and collected for exosome purification.
  • the cells were harvested by exchanging medium with a 0.25% trypsin solution to remove cells for the reactor, tested for viability and counted. Cells were scaled over 10-fold from the initial 40 million to approximately 440 million.
  • the purified exosomes were quantified using CD63 detection ELISA (alternatively, nanoparticle tracking analysis as described in Example 1 may be used to quantify the exosomes).
  • the yield of exosomes from one bioreactor run is at least 2.3 ⁇ 10 10 , which is equivalent to the approximate exosome yield from 72 T-225 flasks of 30-MV2-6 cells.
  • the purified exosomes were tested for angiogenic activity at a dose of 2.0 ⁇ 10 6 exosomes per well in the in vitro tube formation assay (described in detail in Example 2). As shown in FIG. 8 , the angiogenic activity of exosomes prepared from media conditioned by 30-MV2-6 cells grown in T-flasks was equivalent to the angiogenic activity of exosomes prepared from medium conditioned by 30-MV2-6 cells grown in the Quantum Cell Expansion system.
  • hypoxia has been reported to increase exosome production from mammalian cells (Tadokoro et al. (2013) J Biol Chem. 288(48):34343; King et al. (2012) BMC Cancer 12:241). Furthermore, clonal embryonic progenitor cell lines are derived and maintained under low oxygen (5%). West et al. (2008) Regen Med. 3(3): 287. Therefore, 1% oxygen was tested for exosome production to determine if increasing hypoxia will increase exosome production or angiogenic activity.
  • Serum starvation is used to induce exosome production.
  • Nutrient deprivation was tested by using PBS as the conditioning medium. The use of PBS versus basal EGM-MV2 medium for conditioning the cells was also tested.
  • a standard curve is prepared from exosome samples of known concentration (ranging from 5 ⁇ 10 8 to 8 ⁇ 10 7 exosomes/mL).
  • the unknown samples are prepared in PBS or a buffer exchanged into PBS.
  • Samples of intact exosomes are bound to 96 well ELISA plate wells in PBS at 50 ⁇ l/well for at least 16 hours at 37° C.
  • the wells are washed 3 ⁇ 5 minutes in wash buffer (e.g. TBS-Tween).
  • the wells are incubated with a mouse monoclonal antibody prepared in a suitable blocking buffer (e.g. PBS containing exosome depleted FBS and 0.05% Tween 20) that recognizes the extracellular domain of CD63 on intact exosomes for 1 hour at room temperature.
  • a suitable blocking buffer e.g. PBS containing exosome depleted FBS and 0.05% Tween 20
  • the wells are washed again 3 ⁇ 5 minutes at room temperature.
  • the wells are incubated with a suitable secondary antibody in a blocking buffer for detection of mouse anti-CD63 antibody bound to exosomes on the plate surface (e.g. HRP conjugated goat anti-mouse IgG) for 1 hour at room temperature.
  • the wells are washed again 3 ⁇ 5 minutes at room temperature and the wells incubated with 50 ⁇ l of HRP substrate (e.g. Supersensitive TMB ELISA substrate) for 30 minutes at room temperature.
  • the wells are washed 3 ⁇ 5 minutes at room temperature and 50 ⁇ l of stop buffer (0.16M sulfuric acid) is added to provide a fixed endpoint.
  • the concentration of exosomes is quantitated by measuring the absorbance of each well at 450 nm.
  • FIG. 10 An example of a standard curve and quantitation of samples is shown in FIG. 10 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Vascular Medicine (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The invention provides methods, compositions, uses and kits relating to exosomes isolated from progenitor cells.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 16/270,295, filed Feb. 7, 2019, which is a divisional application that claims priority to U.S. application Ser. No. 14/748,215, filed Jun. 23, 2015, which claims priority to U.S. Provisional Application No. 62/020,869, filed on Jul. 3, 2014. The entire contents of the foregoing applications are hereby incorporated by reference.
  • FIELD OF THE INVENTION
  • The field of the invention relates to exosomes isolated from progenitor cells.
  • BACKGROUND
  • Exosomes are believed to contain important signaling molecules that may provide the source of trophic factors responsible for some regenerative benefits seen in cell replacement therapy. As such they would provide an alternative to some cell based therapies that would be easier to manufacture on a large scale and potentially safer to administer to a subject in need of cell therapy. In particular, the risk associated with transmission of infectious agents such as viruses may be lower compared to transplanting whole cells. Moreover, the risk of immune rejection of the exosomes relative to transplanted cells may also be lower. Accordingly, exosomes may provide an attractive alternative or adjunct to cell based therapies and cell based regenerative medicine.
  • Exosomes are 30 to 120 nm vesicles secreted by a wide range of mammalian cell types. Keller et al. (2006) Immunol Lett. 107(2):102; Camussi et al. (2010) Kidney International 78:838. The vesicles are enclosed by a lipid bilayer and are larger than LDL which has a size of 22 nm, but smaller than a red blood cell, which is 6000 to 8000 nm in diameter and has a thickness of 2000 nm. Keller et al. (2006) Immunol Lett. 107(2):102.
  • Exosomes are found both in cells growing in vitro as well as in vivo. They can be isolated from tissue culture media as well as bodily fluids such as plasma, urine, milk and cerebrospinal fluid. George et al. (1982) Blood 60:834; Martinez et al. (2005) Am J Physiol Health Cir Physiol 288:H1004. Exosomes originate from the endosomal membrane compartment. They are stored in intraluminal vesicles within multivesicular bodies of the late endosome. Multivesicular bodies are derived from the early endosome compartment and contain within them smaller vesicular bodies that include exosomes. Exosomes are released from the cell when multivesicular bodies fuse with the plasma membrane. Methods of isolating exosomes from cells has been described, see e.g. US Patent Application Publication No. 20120093885
  • Exosomes contain a variety of molecules including proteins, lipids and nucleic acids such as DNA, mRNA and miRNA. Their contents are believed to play a part in cell to cell communication involving the release of the exosome from one cell and the binding/fusion of the exosome with a second cell, wherein the contents of the exosomal compartment are released within the second cell.
  • It has been reported that exosomes derived from endothelial progenitor cells may act as vehicle for mRNA transport among cells. These exosomes were shown to incorporate into normal endothelial cells by interacting with the α4β1 integrin. Once incorporated into the endothelial cells, the exosomes stimulated an angiogenic program. Deregibus et al. (2007) Blood 110:2440. Similar results were obtained in vivo using severe combined immunodeficient mice. Exosome stimulated endothelial cells implanted subcutaneously in Matrigel (a murine sarcoma extract) organized into a patent vessel network connected with the murine vasculature. Deregibus, supra. Bruno et al. (2009) J Am Soc Nephrol 20:1053; Herrera et al. (2010) J Cell Mol Med 14:1605.
  • Of the various molecular cargo of exosomes, miRNAs have recently attracted a lot of attention due to their regulatory roles in gene expression. MiRNAs are small, non-coding regulatory RNAs that can have a wide range of effects on multiple RNA targets, thus having the potential to have greater phenotypic influence than coding RNAs. MiRNA profiles of exosomes often differ from those of the parent cells. Profiling studies have demonstrated that miRNAs are not randomly incorporated into exosomes but rather a subset of miRNAs is preferentially packaged into exosomes, suggesting an active sorting mechanism of exosomal miRNAs. Guduric-Fuchs et al. (2014) Nucleic Acid Res. 42:9195; Ohshima et al. (2010) PloS One 5(10):e13247.
  • Because exosomes contain a variety of molecules, many believed to play an important role in cell signaling, exosomes would prove useful in research and industry and would have applications as therapeutics, diagnostics and in screening assays. Frequently, however, the availability of reproducible, essentially identical populations of exosomes is limited by the fact that most sources of exosomes are cells that senesce and thus have limited replicative capacity. Accordingly, there is a need for exosomes that are derived from a clonal source that has an extended replicative capacity that is greater than most adult or fetal derived cells. The invention described infra meets this need and as well as other needs in the field.
  • SUMMARY OF THE INVENTION
  • In various embodiments described herein the invention provides compositions comprising exosomes obtained from progenitor cell lines, as well as methods of making and using exosomes obtained from progenitor cell lines.
  • The isolation of embryonic progenitor cells has been described. See West et al. (2008) Regen Med 3:287; US Patent Application Publication Nos. 20080070303 20100184033. Embryonic progenitors are cell lines derived under a variety of culture conditions from pluripotent stem cells, such as human embryonic stem (hES) cells or induced pluripotent stem (iPS) cells. The progenitor cell lines are clonal and while they do, in most instances, senesce, they also possess longer telomeres compared to adult or fetal derived tissue or cells (such as adult stem cells) and accordingly have enhanced replicative capacity relative to those cell types. Because of their clonality and their enhanced replicative capacity they provide a suitable source of exosomes that will offer the benefit of uniformity with regard to the exosome composition and abundance relative to exosomes derived from their typical sources such as adult cells or adult stem cells.
  • In certain embodiments the invention provides an exosome isolated from a progenitor cell line, such as clonal progenitor cell line.
  • In certain embodiments the invention provides an exosome isolated from a human progenitor cell line, such as a clonal human progenitor cell line.
  • In some embodiments the invention provides an exosome isolated from endothelial progenitor cell.
  • In some embodiments the invention provides an exosome isolated from a clonal human endothelial progenitor cell.
  • In other embodiments the invention provides an exosome isolated from the 30-MV2-6 human clonal progenitor cell line.
  • In further embodiments the invention provides an exosome isolated from a human clonal progenitor cell that expresses CD31 and CD34.
  • In certain embodiments the invention provides an exosome isolated from a human progenitor cell line, wherein the human progenitor cell is not an adult stem cell.
  • In further embodiments the invention provides an exosome isolated from a human progenitor cell line, wherein the human progenitor cell is not a mesenchymal stem cell (MSC).
  • In certain embodiments the invention provides an exosome isolated from a cell that has not been transfected with an exogenous gene.
  • In certain other embodiments the invention provides an exosome isolated from a cell that has been transfected with an exogenous gene, wherein the gene is not c-myc.
  • In yet other embodiments the invention provides an exosome isolated from a cell that does not overexpress c-myc.
  • In other embodiments the invention provides an exosome isolated from the 30-MV2-6 clonal human progenitor cell line.
  • In still other embodiments the invention provides an exosome isolated from a cell expressing one or more genes chosen from the genes listed in Table 1.
  • In further embodiments the invention provides an exosome isolated from a cell expressing a plurality of the genes chosen from the genes listed in Table 1.
  • In yet other embodiments the invention provides an exosome isolated from a cell expressing the genes listed in Table 1.
  • In some embodiments, the invention provides an exosome containing CD63.
  • In other embodiments, the invention provides an exosome containing one or more miRNAs listed in Table 2 or Table 4.
  • In further embodiments, the invention provides an exosome containing one or more angiogenic miRNAs.
  • In yet further embodiments, the invention provides an exosome containing miR-126.
  • In some embodiments the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains one or more miRNAs listed in Table 2 or Table 4.
  • In other embodiments the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains one or more angiogenic miRNAs.
  • In yet other embodiments the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains miR-126.
  • In further embodiments the invention provides an exosome isolated from a human clonal progenitor cell, wherein the exosome contains CD63.
  • In some embodiments the invention provides an exosome that induces a cell to form vascular tube like structures.
  • In other embodiments the invention provides an exosome that induces a cell to form branching vascular tube like structures.
  • In yet other embodiments the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome.
  • In certain embodiments the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell has the ability to form vascular tube like structures.
  • In further embodiments the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell is an endothelial cell.
  • In still further embodiments the invention provides a cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome, wherein the cell is a human umbilical vein endothelial cell (HUVEC).
  • In the cell culture embodiments described above the progenitor cell may be a human progenitor cell, such as a human embryonic progenitor cell. One example of a human embryonic progenitor cell is the 30-MV2-6 cell line.
  • In the cell culture embodiments described above the progenitor cell may be, for example, a clonal progenitor cell line, an oligoprogenitor cell line. The progenitor cell may express one or more genes listed in Table 1. The progenitor cell may express a plurality of the genes listed in Table 1. The progenitor cell line may express the genes listed in Table 1. The progenitor cell line may express CD31 and CD34.
  • In some embodiments the invention provides a method of isolating an exosome from a progenitor cell, such as a clonal progenitor cell comprising 1) culturing the progenitor cell in a suitable media or buffer for a time sufficient to allow the cells to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • In some embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell comprising 1) culturing the human clonal progenitor cell in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • In other embodiments the invention provides a method of isolating an exosome from a 30-MV2-6 human clonal progenitor cell line comprising 1) culturing the 30-MV2-6 human clonal progenitor cell line in a suitable media or buffer for a time sufficient to allow the 30-MV2-6 human clonal progenitor cell line to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a 30-MV2-6 human clonal progenitor cell line.
  • In still other embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing CD31 and CD34 comprising 1) culturing the human clonal progenitor cell line expressing CD31 and CD34 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing CD31 and CD34 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing CD31 and CD34.
  • In some embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing one or more of the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing one or more of the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing one or more of the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing one or more of the genes listed in Table 1.
  • In still other embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing a plurality of the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing a plurality of the genes listed in Table 1.
  • In further embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell line expressing the genes listed in Table 1 comprising 1) culturing the human clonal progenitor cell line expressing the genes listed in Table 1 in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line expressing the genes listed in Table 1 to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line expressing the genes listed in Table 1.
  • In some embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell wherein the human clonal progenitor cell line has not been transfected with an exogenous gene comprising 1) culturing the human clonal progenitor cell line that has not been transfected with an exogenous gene in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has not been transfected with an exogenous gene to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that has not been transfected with an exogenous gene.
  • In other embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell, wherein the human clonal progenitor cell line has been transfected with an exogenous gene, wherein the gene is not c-myc, comprising 1) culturing the human clonal progenitor cell line that has been transfected with an exogenous gene, wherein the exogenous gene is not c-myc, in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has been transfected with an exogenous gene, wherein the exogenous gene is not c-myc, to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that has been transfected with an exogenous gene, wherein the exogenous gene is not c-myc.
  • In still other embodiments the invention provides a method of isolating an exosome from a human clonal progenitor cell wherein the human clonal progenitor cell line does not overexpress c-myc comprising 1) culturing the human clonal progenitor cell line that does not overexpress c-myc in a suitable media or buffer for a time sufficient to allow the human clonal progenitor cell line that has not been transfected with an exogenous gene to exocytose exosomes into the culture media; 2) harvesting the media from the cell culture of step 1; and 3) isolating the exosomes from the media of step 2, thereby isolating an exosome from a human clonal progenitor cell line that does not overexpress c-myc.
  • In further embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In still further embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting an endothelial cell with an exosome isolated from a progenitor cell thereby inducing or enhancing an endothelial cells ability to form vascular tube like structures.
  • In other embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a clonal progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In further embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human clonal progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In certain embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human endothelial progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In yet other embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a human clonal endothelial progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In further embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing one or more genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In other embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing a plurality of genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In yet other embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing the genes listed in Table 1 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In still other embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a cell expressing the markers CD31 and CD34 thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In further embodiments the invention provides a method of inducing or enhancing a cells ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a 30-MV2-6 cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • In still other embodiments the invention provides a method of regeneration a tissue or an organ comprising contacting one or more cells capable of regenerating a tissue or an organ with an exosome isolated from a progenitor cell.
  • In yet other embodiments the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a progenitor cell.
  • In some embodiments the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a human clonal endothelial progenitor cell.
  • In further embodiments the invention provides a method of regenerating a vascular tissue or organ comprising contacting a cell capable of regenerating a vascular tissue or organ with an exosome isolated from a 30-MV2-6 cell.
  • In certain embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a progenitor cell.
  • In some embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell.
  • In further embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from an endothelial progenitor cell.
  • In certain embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from human clonal endothelial progenitor cell.
  • In yet other embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a 30-MV2-6 human endothelial progenitor cell.
  • In still other embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing CD31 and CD34.
  • In further embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing one or more genes listed in Table 1.
  • In some embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing a plurality of genes listed in Table 1.
  • In yet other embodiments the invention provides a method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a human clonal progenitor cell expressing the markers listed in Table 1.
  • In further embodiments the invention provides a kit comprising an exosome isolated from a progenitor cell and at least one container.
  • BRIEF DESCRIPTION OF DRAWINGS
  • For a fuller understanding of the nature and advantages of the present invention, reference should be had to the following detailed description taken in connection with the accompanying drawings, in which:
  • FIG. 1 shows a graph of the size and concentration of exosomes isolated from
      • a) human embryonic progenitor cell line 30-MV2-6; and b) the HT1080 cell line.
  • FIG. 2A shows three photomicrographs, the first showing the effects on vascular tube like formation in HUVECs grown in the presence of basal media supplemented with exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (top); the second showing the effects on vascular tube formation in HUVECs grown in base media supplemented with PBS, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV-2-6 (middle); and the third showing the effects on vascular tube like formation in HUVECs grown in complete medium, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (bottom).
  • FIG. 2B is a graph quantifying four parameters: cell covered area; total tube length; total number of branching points and total number of loops in HUVECs grown in the presence of exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“MV2-6 EXO”); HUVECs grown in basal media+PBS, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“Basal PBS”); and HUVECs grown in complete media, but without exposure to exosomes isolated from human embryonic progenitor cell line 30-MV2-6 (“Complete”).
  • FIG. 3A is photomicrograph showing that hES derived perivascular cells form aggregates when cultured in the presence of complete EGM-MV2 media with serum and growth factors.
  • FIG. 3B is a photomicrograph showing that hES derived perivascular cells form incomplete tubes when cultured in EGM-MV2 basal media.
  • FIGS. 3C-E are photomicrographs showing that increasing doses of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6 resulted in increasing tube formation in hES derived perivascular cells.
  • FIG. 3F is a graph showing the cell covered area of hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5×107, 5.0×107, 10.0×107) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3G is a graph showing the total number of branching points in hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5×107, 5.0×107, 10.0×107) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3H is a graph showing the total tube length of vascular tube like structures formed by hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplemented with increasing doses (2.5×107, 5.0×107, 10.0×107) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 3I is a graph showing the total number of loops formed by hES derived perivascular cells grown either in complete media (complete); basal media (base) or basal media supplement with increasing doses (2.5×107, 5.0×107, 10.0×107) of exosomes isolated from the human clonal endothelial progenitor cell line 30-MV2-6.
  • FIG. 4 shows comparison of in vitro angiogenic activity of exosomes isolated from the human clonal endothelial cell line 30-MV2-6 versus exosomes isolated from bone marrow mesenchymal stem cells (BM-MSC). Two different commercially available sources of BM-MSCs were used, Promocell (panel A) and Lonza (panel B). 30-MV2-6: exosomes derived from 30-MV2-6 cell line; MSC: exosomes derived from BM-MSC; BM: basal medium, negative control; CM: complete growth medium. Results were normalized to tube length obtained using complete growth medium (CM).
  • FIG. 5 shows that the angiogenic activity of exosomes isolated from the 30-MV2-6 cell line is dose dependent (panel A) and at least six times more potent than the angiogenic activity of exosomes isolated from BM-MSCs (panel B).
  • FIG. 6A depicts analysis of miRNA content in 30-MV2-6 derived exosomes versus BM-MSC derived exosomes. 30-MV2-6 exosome RNA was compared to BM-MSC RNA for miRNA content using an 84 miRNA PCR array. The scatter plot indicates miRNAs with greater than 6-fold differences. The upper circled dot represents miRNA miR-126-3p, which is expressed at 77.6-fold higher in 30-MV2-6 exosomes than in BM-MSC exosomes. The lower circled dot represents miRNA miR-376c-3p which is expressed at 752-fold lower in 30-MV2-6 exosomes than in MB-MSC exosomes.
  • FIG. 6B is a bar graph illustrating the differences (measured as fold-change) in miRNA expression profile between 30-MV2-6-derived exosomes versus BM-MSC-derived exosomes.
  • FIG. 7 consists of 5 photomicrographs showing in vivo angiogenic activity of human clonal endothelial cell line 30-MV2-6 in the Matrigel plug assay in immunocompromised mice. Blood vessel-like structures are seen in the exosome treated plugs (panels A and C) but not in the control plug (panel E). Endothelial cell content is confirmed by staining with anti-Von Willebrand factor antibody (panels B and D).
  • FIG. 8 shows that the in vitro angiogenic activity of exosomes derived from 30-MV2-6 cells grown in T-flasks is comparable to the in vitro angiogenic activity of exosomes derived from 30-MV2-6 cells grown in the Quantum cell expansion bioreactor. Panel A depicts tube network formation and panel B depicts quantitative analysis of tube length using Image J angiogenesis analyzer software. BM: basal medium, negative control; CM: complete growth medium.
  • FIG. 9 is a bar graph illustrating the effect of oxygen level and conditioning medium used on the angiogenic activity of exosomes derived from 30-MV2-6 cells. Use of PBS versus basal medium as the conditioning medium had no significant effect on the angiogenic activity of the 30-MV2-6 exosomes. Similarly, no significant effect on the angiogenic activity was observed in conditioning the cells in 1% versus 5% oxygen.
  • FIG. 10 shows ELISA detection of CD63 on intact exosomes to determine exosome concentration. The standard curve was prepared using exosomes (from HT1080 cells) of known concentration as determined by nanoparticle analysis (NTA). Quantitation of exosomes in samples of unknown concentration was calculated from OD value at 450 nm. The assay assumes CD63 content of exosomes derived from various different cells remains relatively constant.
  • FIG. 11 depicts the high proliferative capacity and stable angiogenic exosome production of the clonal human embryonic progenitor cell line 30-MV2-6. The 30-MV2-6 cells continue to proliferate in cell culture past 50 population doublings (panel A). Similarly, exosomes that retain their angiogenic activity as measured by the in vitro tube formation assay may be prepared from 30-MV2-6 cells that have been cultured for at least 50 population doublings (panel B).
  • FIG. 12 is a heat map of Illumina gene expression array (Illumina, Hayward, Calif.) data showing that the clonal embryonic epithelial progenitor cell lines derived from the embryonic stem cell line ESI-017 have similar endothelial specific gene expression pattern as various adult endothelial cells from different sources.
  • DETAILED DESCRIPTION
  • Before the present compositions and methods are described, it is to be understood that this invention is not limited to the particular processes, compositions, or methodologies described, as these may vary. It is also to be understood that the terminology used in the description is for the purpose of describing the particular versions or embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. Any methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the present disclosure.
  • The invention provides exosomes isolated from clonal progenitor cells, such as human clonal progenitor cells derived from a human pluripotent stem cell. Because the cells are clonal and have enhanced replicative capacity in vitro, the invention provides a means of producing the same exosomes over and over again. This provides for a consistent product allowing either the researcher or clinician to alleviate any concerns regarding both the quality and the consistency of the exosomes in any application. Accordingly, the invention also provides methods of making the progenitor cells from which the exosomes are derived and methods of isolating the exosomes from these cells. The invention also contemplates uses, cell cultures and kits comprising the exosomes all of which are described infra.
  • Definitions
  • As used herein, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to a “therapeutic” is a reference to one or more therapeutics and equivalents thereof known to those skilled in the art, and so forth.
  • As used herein, the term “about” means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of 45% to 55%.
  • “CD31”, also known as PECAM-1 (platelet endothelial cell adhesion molecule) is a protein in the immunoglobulin superfamily found on the surface of platelets, monocytes, neutrophils, and some types of T-cells. CD31 makes up a large portion of endothelial cell intercellular junctions. CD31 is encoded in humans by the PECAM-1 gene and is commonly used as a marker for endothelial cells.
  • “CD34” is a cell surface glycoprotein that functions as a cell-cell adhesion factor. The CD34 protein is a member of a family of single-pass transmembrane sialomucin proteins that are expressed on early hematopoietic and vascular-associated tissue. CD34 is encoded in humans by the CD34 gene. It is commonly used as a marker for hematopoietic and/or vascular endothelial cells.
  • As used herein, the term “clonal” refers to a population of cells obtained by the expansion of a single cell into a population of cells all derived from that original single cell and not containing other cells. The terms “clonal progenitor cell”, “embryonic clonal progenitor cell”, “clonal progenitor cell line” and “embryonic clonal progenitor cell line” each refer to progenitor cell lines that are derived clonally, i.e., derived by the expansion of a single cell into a population of cells all derived from that original single cell and not containing other cells.
  • The term “embryonic stem cell” as used herein refers to a pluripotent cell that is derived from a blastocysts, such as an in vitro fertilized blastocyst. Embryonic stem cells include human embryonic stem cells, which are available as established cell lines. The established cell lines are available commercially from numerous public cell banks, e.g. WiCell and private corporations, e.g. ESI BIO.
  • The term “human pluripotent cell” or “human pluripotent stem cell” as used herein refers to a human cell which is capable of differentiating into at least one cell type found in or derived from each of the three primary germ layers. Some human pluripotent stem cells have the ability to differentiate into all cells found in or derived from each of the three primary germ layers. Examples of human pluripotent stem cells include human embryonic stem cells (Thomson (1998) Science 282:1145), human embryonic germ cells (Shamblott et al. (2001) PNAS 98:113 and induced pluripotent cells (Takahashi et al. (2007) Cell 131:861.
  • The term “induced pluripotent stem cell” as used herein, refers to a pluripotent cell that has been genetically reprogrammed using any technique known in the art from an adult somatic cell back to the developmentally less mature pluripotent state.
  • The term “miRNA,” as used herein, refers to microRNA which includes RNA species that are 21-25 nt long and may be single- or double-stranded. MicroRNAs are short, non-coding RNA molecules that have been found in animals, including humans, and in plants. The term encompasses small interfering RNA (siRNA) and small temporal RNA (stRNA), as well as miRNA proper. miRNAs are transcribed as parts of longer RNA molecules and processed in the nucleus by the dsRNA ribonuclease Drosha to hairpin structures 70-100 nucleotides long. These are transported to the cytoplasm where they are digested to 21-23-mers by the dsRNA ribonuclease Dicer. Single-stranded miRNAs bind to complementary sequences in mRNA thereby inhibiting translation.
  • “miR-126” is a human microRNA that is specifically expressed in endothelial cells, throughout capillaries and in larger blood vessels. miR-126 plays a role in angiogenesis by regulating the expression levels of various genes by pre- and post-transcription mechanisms. As used herein, the term “miR-126” refers to all of the following: the stem-loop miR-126, miR-126-3p (3′ arm of the hairpin precursor) and miR-126-5p (5′ arm of the hairpin precursor). miRNA naming conventions are described in Kozomara and Griffiths-Jones, (2014) Nucleic Acids Res. 42(Database issue):D68. The terms “miR-126-3p” and “hsa-miR-126-3p” are also used interchangeably throughout this application.
  • The use of “nucleic acid,” “polynucleotide” or “oligonucleotide” or equivalents herein means at least two nucleotides covalently linked together. In some embodiments, an oligonucleotide is an oligomer of 6, 8, 10, 12, 20, 30 or up to 100 nucleotides. In some embodiments, an oligonucleotide is an oligomer of at least 6, 8, 10, 12, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 400, or 500 nucleotides. A “polynucleotide” or “oligonucleotide” may comprise DNA, RNA, cDNA, PNA or a polymer of nucleotides linked by phosphodiester and/or any alternate bonds.
  • The term “peptide,” as used herein, refers to two or more amino acids joined by a peptide bond. A peptide can, in some instances, be a portion of a full length protein.
  • The term “protein” as used herein, refers to a full length protein, i.e. one having all of the amino acids coded for by the mRNA that encodes the particular protein. Also included in the definition are modified proteins where one or more amino acids have been cleaved (e.g. a signal sequence) as a result of the protein being secreted from a cell.
  • By “pharmaceutically acceptable”, it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • The term “pluripotent cell” or “pluripotent stem cell” as used herein, refers to a cell which is capable of differentiating into at least one cell type found in or derived from each of the three primary germ layers. Some pluripotent stem cells have the ability to differentiate into all cells found in or derived from each of the three primary germ layers.
  • The term “progenitor cell line” as used herein refers to a line of cells that is more differentiated (developed) compared to a pluripotent cell, such as iPS cell or an hES cell, but is not terminally differentiated. Progenitor cells will have enhanced replicative capacity compared to a terminally differentiated cell which typically has senesced. Progenitor cells may also have longer telomere lengths compared to a cell that has terminally differentiated. Progenitor cell lines, when cultured, may be able double in population size at least 5, at least 10, at least 20, at least 30, at least 40, at least 50 times. In some instances progenitor cell lines may be able to double in population size 5-400 times, 10-300 times, 20-200 times, 30-80 times, 40-60 times. One example of a progenitor cell line is an embryonic progenitor cell. Embryonic progenitor cell is obtained from a pluripotent cell such as an iPS cell or a hES as previously described. See West et al. (2008) Regen Med 3:287; US Patent Application Publication Nos. 20080070303 20100184033.
  • The term “subject,” as used herein includes, but is not limited to, humans, non-human primates and non-human vertebrates such as wild, domestic and farm animals including any mammal, such as cats, dogs, cows, sheep, pigs, horses, rabbits, rodents such as mice and rats. In some embodiments, the term “subject,” refers to a male. In some embodiments, the term “subject,” refers to a female.
  • The term “suitable media,” as used herein, refers to a solution that can be used to grow cells in culture. A suitable media may include a formulation of salts and/or buffering reagents. A suitable media may include any or all of the following: salts, sugars, amino acids, proteins, growth factors, cytokines, and hormones, additives such as serum, albumin, antibiotics, insulin, selenium and transferrin. Suitable culture media includes for example commercially available culture media such as DMEM, MEM Stem Pro and the like.
  • A “therapeutically effective amount” of a composition such as a therapeutic agent described infra, e.g. an exosome, is a predetermined amount calculated to achieve the desired effect. In some embodiments, the effective amount is a prophylactic amount. In some embodiments, the effective amount is an amount used to medically treat the disease or condition. The specific dose of a composition administered according to this invention to obtain therapeutic and/or prophylactic effects will, of course, be determined by the particular circumstances surrounding the case, including, for example, the composition administered, the route of administration, and the condition being treated. It will be understood that the effective amount administered will be determined by the physician in the light of the relevant circumstances including the condition to be treated, the choice of composition to be administered, and the chosen route of administration. A therapeutically effective amount of composition of this invention is typically an amount such that when it is administered in a physiologically tolerable excipient composition, it is sufficient to achieve an effective systemic concentration or local concentration in the targeted tissue.
  • The terms “treat,” “treated,” or “treating,” as used herein, can refer to both therapeutic treatment or prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological condition, symptom, disorder or disease, or to obtain beneficial or desired clinical results. In some embodiments, the term may refer to both treating and preventing. For the purposes of this disclosure, beneficial or desired clinical results may include, but are not limited to one or more of the following: alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease. Treatment includes eliciting a clinically significant response. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.
  • Exosomes
  • Exosomes of the invention are double membrane bound vesicles secreted from cells of plants and animals, such as mammals including humans, non-human primates, dogs, cats, sheep, cows, pigs, horses, rabbits, mice, rats and guinea pigs to name but a few. Thus exosomes may be isolated from any cell type from any source. In some embodiments of the invention the exosomes of the invention may be secreted from a human cell, such as a human clonal progenitor cell. In some embodiments the exosomes may be secreted from an endothelial human clonal progenitor cell.
  • The exosomes may contain one or more markers expressed by their cell of origin. In some embodiments the exosomes contain CD63.
  • The exosomes may contain one or more miRNAs. In some embodiments, the exosomes of the invention contain one or more miRNAs chosen from Table 2 or 4. In some embodiments, the exosomes of the invention contain one or more angiogenic miRNAs. In some embodiments, the exosomes of the invention contain miR-126.
  • Where the exosomes are derived from a clonal progenitor cell, the exosomes will be of uniform quality and composition. Thus the exosomes isolated from a clonal progenitor cell will not vary as a result of genetic variation of the source cell. The molecular composition of the contents and the bio-physical characteristics of the vesicles will be consistent and reproducible. Moreover, because of the replicative capacity of the human embryonic progenitor cells, the invention provides an overabundance of the exosomes of the invention. This is in direct contrast with exosomes obtained from other sources known in the art where the paucity of the cell type or the problem of senescence limits the availability of a reproducible exosome. Moreover, in certain embodiments the cells giving rise to the exosomes of the invention, are neither transformed nor malignant, thus avoiding any possible concern regarding carcinogenesis of the exosomes.
  • The exosomes of the invention may have diameter ranging from about 20 nm-130 nm; from about 30 nm-120 nm; about 40 nm-110 nm; about 50 nm-100 nm; about 85 nm-95 nm. In some embodiments the exosomes of the invention have a diameter of about 90 nm. In some embodiments the exosomes of the invention have a diameter of about 88 nm.
  • The exosomes may be comprised of a lipid bilayer containing transmembrane proteins and may contain hydrophilic components within the vesicle of the exosome. The contents of the vesicle may be derived from the cytoplasm of the cell or from other vesicle structures within the cell, e.g., endosomes. The vesicle may contain nucleic acids, such as DNA, RNA including mRNA, miRNA as well as proteins and peptides.
  • The exosomes of the invention may serve as depots for the delivery of therapeutic molecules of any kind. The exosomes of the invention can be engineered to contain therapeutic molecules such as nucleic acids, proteins, peptides, small molecules such as drugs and the like. Any technique known in the art can be used to load the exosomes of the invention with a desired therapeutic molecule. For example cationic lipids could be used to transfect the exosomes with a desired nucleic acid such as DNA, RNA, include mRNA and miRNA. HIV tat protein could be used to transport protein or peptide therapeutics into the exosomes of the invention. The therapeutic molecules can be chosen, engineered or designed to have any desired therapeutic effect. For example molecules associated with enhanced angiogenesis could be loaded into the exosomes of the invention, e.g. VEGF.
  • The secreted exosomes of the invention can be contacted with a target cell (e.g. a cell that is not the same as the cell of origin for the exosome) such that the exosome is taken up by the target cell, e.g. endocytosed. Once inside the cell, the contents of the vesicle may be released into the cytoplasm where the molecules contained within the vesicle may act as signaling molecules in one or more signaling pathways thereby inhibiting or enhancing gene expression. The signaling molecules may act at the level of transcription or translation for example. In some instances, where the vesicles contain RNA, the RNA can be transcribed by the target cell. In some instances where the RNA is a miRNA the miRNA can inhibit gene expression.
  • Methods of Isolating Exosomes
  • Exosomes may be isolated from any suitable cell that contains exosomes. Described infra are several exemplary cell and cell types that may be used to implement this method. The method may involve seeding the cell at an appropriate density in a tissue culture vessel and then incubating the cells in a suitable media or buffer for a suitable period of time. In some embodiments the cells may be permitted to attach to the culture vessel before the exosomes are isolated. In other embodiments the cells may be kept in suspension while the exosomes are isolated. The cells may be permitted to replicate in culture before the exosomes are isolated. Alternatively, the exosomes may be isolated from the cells that have not replicated, or replicated minimally (e.g. less than 1 doubling).
  • To initiate the method the cells are seeded in a tissue culture method at a suitable cell density. The cell density (cells per unit area) may range from about 5 k/cm2, about 10 k/cm2, about 15 k/cm2, about 20 k/cm2, about 25 k/cm2, about 30 k/cm2, about 35 k/cm2, about 40 k/cm2, about 45 k/cm2, about 50 k/cm2, about 55 k/cm2, about 60 k/cm2, about 70 k/cm2, about 75 k/cm2. In some embodiments the cell density (cells per unit area) may range from about 1 k/cm2-100 k/cm2, 10 k/cm2-90 k/cm2, 20 k/cm2-80 k/cm2, 30 k/cm2-70 k/cm2, 40 k/cm2-60 k/cm2. In one embodiment the cells are seeded at a density (cells per unit area) of 40 k/cm2.
  • The cells may be seeded in any isotonic solution. In one embodiment a suitable solution may include a suitable buffer. Examples of suitable buffers may include phosphate buffered saline (PBS), HEPES and the like. In other embodiments the cells may be seeded in any suitable cell culture media, many of which are commercially available. Exemplary media include DMEM, RPMI, MEM, Media 199, HAMS and the like. In one embodiment the media is EGM-MV2. The media may be supplemented with one or more of the following: growth factors, cytokines, hormones, serum, such as fetal calf serum, serum substitutes such as knock out replacement serum or B27, antibiotics, vitamins and/or small molecule drugs. In one embodiment the media is supplemented with a TGFβ inhibitor, e.g. SB43154).
  • The method may be practiced by placing the cells in a suitable environment, such as a cell incubator heated to about 37° C. In some embodiments the cells may be incubated at room temperature. The incubator may be humidified and have an atmosphere that is about 5% CO2 and about 1% 02. In some embodiments the CO2 concentration may range from about 1-20%, 2-10%, 3-5%. In some embodiments the O2 concentration may range from about 1-20%, 2-10%, 3-5%.
  • The method may be practiced by incubating the cells in the media or buffer for about 1-72 hours, 1-48 hours, 2-24 hours, 3-18 hours, 4-16 hours, 5-10 hours. In some embodiments the cells are incubated for about 16 hours.
  • Incubation of the cells as described above allows for the exocytosis of the exosomes by the cells into the isotonic solution. After incubation of the cells in the isotonic solution as described above, the isotonic solution may be harvested. For example the isotonic solution may be pipetted or decanted into another vessel such as a centrifuge tube. A precipitating agent may be added to the isotonic solution at this time to facilitate the precipitation of the exosomes in the solution. Examples of precipitating agents include a solution that is about 15% polyethylene glycol. Alternatively, a commercially prepared precipitating agent may be used, e.g., Total Exosome Isolation Reagent (Life Technologies, Carlsbad, Calif.). The cells may then be incubated for a suitable time period e.g., 1-48 hours, 2-24 hours, 3-18 hours, 4-16 hours, 5-10 hours. In some embodiments the cells are incubated for about 16 hours. The cells may be incubated at a temperature of about 1° C., 5° C., 10° C., 15° C., 20° C., 25° C., 30° C. In one embodiment the cells are incubate at about 4° C.
  • After incubating the harvested cell conditioned isotonic solution with the precipitating reagent described above the harvested cell conditioned isotonic solution may be centrifuged at about 1,000×g, 2,000×g, 4,000×g, 6,000×g, 8,000×g; 10,0000×g; 12,000×g, 14,000×g, 16,000×g; 18,000×g. In one embodiment the harvested cell conditioned isotonic solution is centrifuged at about 10,000×g. The harvested cell conditioned isotonic solution may be centrifuged at about a temperature of 2° C., 4° C., 6° C., 8° C., 10° C., 12° C., 14° C., 16° C., 18° C., 20° C., 22° C., 24° C., 26° C. In one embodiment the harvested cell conditioned isotonic solution are centrifuged at about a temperature of 4° C.
  • After centrifugation the isotonic solution is removed and the exosomes are resuspended in a suitable buffer such as PBS. The volume of buffer may be about 0.01 volumes-about 0.09 volumes, about 0.02 volumes to about 0.08 volumes; about 0.03 volumes to about 0.07 volumes of the precipitating solution. In one embodiment the harvested cell conditioned isotonic solution is resuspended in PBS at a volume equivalent to about 0.01 volumes of the precipitating solution. The harvested exosomes may be used immediately or frozen and stored, e.g., at −20° C., for later use.
  • Progenitor Cells
  • In certain embodiments of the invention progenitor cells serve as the source of the exosomes described infra. The progenitor cell may be from any animal or plant. For example the exosome may be from a mammal, such as a human, a non-human primate, a horse, a cow, a sheep, a goat, a pig, a cat, a dog, a rabbit, a guinea pig, a rodent such as a mouse or a rat. Typically a progenitor cell will not have an essentially unlimited replicative capacity as typically found in embryonic stem cells, but will nonetheless have, a result of their longer telomeres, a greater replicative capacity compared to adult primary cells or tissues (e.g. primary cells) or adult stem cells.
  • The progenitor cell may be derived from a pluripotent stem cell, such as an embryonic stem cell or an induced pluripotent stem cell. The progenitor cell may be a clonal cell or an oligoclonal cell. An oligoclonal cell would include a population of cells similar cells, e.g. phenotypically or genetically. The progenitor cell may be a clonal human embryonic progenitor cell. The progenitor cell may be a clonal human embryonic endothelial progenitor cell. The progenitor cell may be a clonal embryonic progenitor cell that expresses CD31 and CD36. The progenitor cell may be a clonal embryonic progenitor cell expressing one or more genes listed in Table 1. The progenitor cell may be a clonal embryonic progenitor cell expressing a plurality of the genes listed in Table 1. The progenitor cell may be a clonal embryonic progenitor cell expressing the genes listed in Table 1.
  • Where the progenitor cells are clonal cells obtained from pluripotent stem cells they will provide an almost unlimited source of the same exosomes. This is due to two factors: the genetic identity of the original cellular source material and the enhanced telomere lengths found in early progenitors which provide for enhanced replicative capacity relative to adult tissue or cells or adult stem cells. Moreover, unlike adult stem cells which are typically available in very small numbers and are difficult to expand in culture, the clonal embryonic progenitors described infra are available in large numbers and are relatively easy to expand in culture.
  • In some embodiments the progenitor cell is not an adult stem cell. In some embodiments of the invention the progenitor cell is not an MSC. In some embodiments the clonal progenitor cell is not transfected or engineered to express an exogenous gene. In some embodiments the clonal progenitor cell is not transfected to express an oncogene. In some embodiments the clonal progenitor does not express c-myc. In other embodiments the clonal progenitor cell is transfected or engineered to express an exogenous gene. Examples of suitable exogenous genes include the catalytic component of human telomerase, e.g. hTERT.
  • Uses of Exosomes
  • The exosomes described herein may be used in therapeutic, research and diagnostic applications. For example the exosomes described infra may be added to a cell culture to enhance one or more phenotypic traits of the cells. The exosomes of the invention may be added to a cell culture to inhibit one or more phenotypic traits of the cells. The exosomes of the invention may be added to a cell culture to provide a new phenotypic trait of the cells.
  • The exosomes of the invention may be added to a culture of endothelial cells to enhance the ability of the cells to form vascular tube like structures. The exosomes of the invention may be added to any cell having the ability to form vascular tube like structures to enhance the cells ability to form tube like structures.
  • In some embodiments the exosomes of the invention are contacted with a cell thereby providing at least one new phenotypic trait to the cell. For example, the exosomes of the invention may confer the ability to form vascular tube like structures to cell lacking the ability to form vascular tube like structures before it was contacted with the exosomes of the invention.
  • In certain embodiments the exosomes of the invention may be added to a culture of perivascular cells to enhance the ability of the perivascular cells to form vascular tube like structures.
  • In some embodiments the invention provides a method of increasing the length of a vascular tube like structure formed by a cell such as an endothelial relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells. In some embodiments the invention provides a method of increasing the length of a vascular tube like structure formed by a cell such as a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells. In some embodiments the invention provides a method of increasing the branching of a vascular tube like structure formed by an endothelial cell relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells. In some embodiments the invention provides a method of increasing the branching of a vascular tube like structure formed by a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells. In still other embodiments the invention provides a method of increasing the number of loops in the vascular tube like structures formed by an endothelial cell relative to an endothelial cell that has not been treated with the exosomes of the invention comprising contacting the endothelial cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells. In yet other embodiments the invention provides a method of increasing the number of loops in the vascular tube like structures formed by a perivascular cell relative to a perivascular cell that has not been treated with the exosomes of the invention comprising contacting the perivascular cell with an exosome isolated from a progenitor cell such as a human clonal progenitor cell, e.g., 30-MV2-6 cells.
  • The exosomes of the invention may be administered therapeutically to a subject in need of treatment. For example the exosomes of the invention may be administered to a subject in need of treatment for any disease requiring the enhanced ability to form vascular tube like structures. The exosomes of the invention may be used to treat a subject suffering from cardiovascular disease, heart failure, infarction, chronic wounds, ulcer, clogged vessels or arteries, damaged vessels, stenotic vessels, arteriosclerosis, angina, peripheral vascular disease, Alzheimer's disease, ischemia, diabetes, cancer, cell replacement transplant or therapy, tissue and cell regenerative therapy and Parkinson's disease. The exosomes may be used as depot to deliver therapeutic molecules such as small molecules, nucleic acids, proteins and peptides.
  • The exosomes of the invention may be directly administered to a subject in need of treatment or an in vitro cell culture. Alternatively the exosomes can be provided enclosed within a matrix or scaffold. Suitable matrices or scaffolds may include a matrix or scaffold comprised of one or more extracellular matrix proteins, e.g. laminin, fibronectin and the like. Other suitable matrices or scaffolds include Matrigel® which is a murine sarcoma extract. The matrix or scaffold may be a hydrogel. The hydrogel may be comprised of hylauronate and gelatin (see U.S. Pat. Nos. 8,324,184; 7,928,069). In one embodiment the exosomes of the invention may be delivered in HyStem (Biotime, Inc., Alameda Calif.).
  • Using the methods described infra along with routine chromatographic techniques known in the art the exosomes of the invention may be used to isolate one or more nucleic acids, proteins or peptides expressed by a progenitor cell serving as the source of the exosome. Once isolated, the proteins or peptides isolated from the exosomes of the invention can be used to make antibodies to the isolated proteins or peptides (See Harlow et al. Antibodies: A Lab Manual 2nd Edition; Cold Spring Harbor Press 2013).
  • The exosomes of the invention may be used in drug screening assays. For example where the exosomes described infra enhance vascular tube formation in vitro, the exosomes can be used to screen for drugs that enhance or inhibit this capability. A cell culture comprising cells having the ability to form vascular tube like structures may be contacted with the exosomes of the invention and a drug candidate may be applied to the same cell culture either before, after or simultaneously with the exosomes to determine the effect of the drug the ability of the exosomes to enhance vascular tube formation in the cell culture. The effects can be compared to untreated cells and cells treated only with the exosomes of the invention.
  • Pharmaceutical Compositions
  • Modes of administration for a therapeutic (either alone or in combination with other pharmaceuticals) can be, but are not limited to, sublingual, injectable (including short-acting, depot, implant and pellet forms injected subcutaneously or intramuscularly), or by use of vaginal creams, suppositories, vaginal rings, rectal suppositories, intrauterine devices, and transdermal forms such as patches and creams.
  • Specific modes of administration will depend on the indication. The selection of the specific route of administration and the dose regimen is to be adjusted or titrated by the clinician according to methods known to the clinician in order to obtain the optimal clinical response. The amount of therapeutic to be administered is that amount which is therapeutically effective. The dosage to be administered will depend on the characteristics of the subject being treated, e.g., the particular animal treated, age, weight, health, types of concurrent treatment, if any, and frequency of treatments, and can be easily determined by one of skill in the art (e.g., by the clinician).
  • Pharmaceutical formulations containing the therapeutic of the present disclosure and a suitable carrier can be solid dosage forms which include, but are not limited to, tablets, capsules, cachets, pellets, pills, powders and granules; topical dosage forms which include, but are not limited to, solutions, powders, fluid emulsions, fluid suspensions, semi-solids, ointments, pastes, creams, gels and jellies, and foams; and parenteral dosage forms which include, but are not limited to, solutions, suspensions, emulsions, and dry powder; comprising an effective amount of a polymer or copolymer of the present disclosure. It is also known in the art that the active ingredients can be contained in such formulations with pharmaceutically acceptable diluents, fillers, disintegrants, binders, lubricants, surfactants, hydrophobic vehicles, water soluble vehicles, emulsifiers, buffers, humectants, moisturizers, solubilizers, preservatives and the like. The means and methods for administration are known in the art and an artisan can refer to various pharmacologic references for guidance. For example, Modern Pharmaceutics, Banker & Rhodes, Marcel Dekker, Inc. (1979); and Goodman & Gilman's The Pharmaceutical Basis of Therapeutics, 6th Edition, MacMillan Publishing Co., New York (1980) can be consulted.
  • The compositions of the present disclosure can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. The compositions can be administered by continuous infusion subcutaneously over a period of about 15 minutes to about 24 hours. Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • For oral administration, the compositions can be formulated readily by combining the therapeutic with pharmaceutically acceptable carriers well known in the art. Such carriers enable the therapeutic of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, but are not limited to, fillers such as sugars, including, but not limited to, lactose, sucrose, mannitol, and sorbitol; cellulose preparations such as, but not limited to, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and polyvinyl pyrrolidone (PVP). If desired, disintegrating agents can be added, such as, but not limited to, the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores can be provided with suitable coatings. For this purpose, concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active therapeutic doses.
  • Pharmaceutical preparations which can be used orally include, but are not limited to, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as, e.g., lactose, binders such as, e.g., starches, and/or lubricants such as, e.g., talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active therapeutic can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • For buccal administration, the pharmaceutical compositions can take the form of, e.g., tablets or lozenges formulated in a conventional manner.
  • For administration by inhalation, the therapeutic for use according to the present disclosure is conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit can be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the therapeutic and a suitable powder base such as lactose or starch.
  • The compositions of the present disclosure can also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • In addition to the formulations described previously, the therapeutic of the present disclosure can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • Depot injections can be administered at about 1 to about 6 months or longer intervals. Thus, for example, the compositions can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • In transdermal administration, the compositions of the present disclosure, for example, can be applied to a plaster, or can be applied by transdermal, therapeutic systems that are consequently supplied to the organism.
  • Pharmaceutical compositions can include suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as, e.g., polyethylene glycols.
  • The compositions of the present disclosure can also be administered in combination with other active ingredients, such as, for example, adjuvants, protease inhibitors, or other compatible drugs or compounds where such combination is seen to be desirable or advantageous in achieving the desired effects of the methods described herein.
  • In some embodiments, the disintegrant component comprises one or more of croscarmellose sodium, carmellose calcium, crospovidone, alginic acid, sodium alginate, potassium alginate, calcium alginate, an ion exchange resin, an effervescent system based on food acids and an alkaline carbonate component, clay, talc, starch, pregelatinized starch, sodium starch glycolate, cellulose floc, carboxymethylcellulose, hydroxypropylcellulose, calcium silicate, a metal carbonate, sodium bicarbonate, calcium citrate, or calcium phosphate.
  • In some embodiments, the diluent component may include one or more of mannitol, lactose, sucrose, maltodextrin, sorbitol, xylitol, powdered cellulose, microcrystalline cellulose, carboxymethylcellulose, carboxyethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, methylhydroxyethylcellulose, starch, sodium starch glycolate, pregelatinized starch, a calcium phosphate, a metal carbonate, a metal oxide, or a metal aluminosilicate.
  • In some embodiments, the optional lubricant component, when present, comprises one or more of stearic acid, metallic stearate, sodium stearylfumarate, fatty acid, fatty alcohol, fatty acid ester, glycerylbehenate, mineral oil, vegetable oil, paraffin, leucine, silica, silicic acid, talc, propylene glycol fatty acid ester, polyethoxylated castor oil, polyethylene glycol, polypropylene glycol, polyalkylene glycol, polyoxyethylene-glycerol fatty ester, polyoxyethylene fatty alcohol ether, polyethoxylated sterol, polyethoxylated castor oil, polyethoxylated vegetable oil, or sodium chloride.
  • Kits
  • In some embodiments the invention provides a kit comprising exosomes isolated from a progenitor cell, such as a human clonal progenitor cell. The progenitor cell may be an endothelial progenitor cell, such as human clonal embryonic progenitor cell, e.g. 30MV2-6. The exosomes may be provided in one or more containers. The exosomes may be provided in a suitable buffer, e.g. PBS or a suitable media, such as a commercially available cell culture media, e.g. DMEM. The kit may further contain a cell having the ability to form vascular tube like structures. The cell may be an endothelial cell, e.g. HUVEC and/or a perivascular cell. The cells may be provided in a suitable media, e.g. DMEM or the like or alternatively the cells may be provided in a buffer such as PBS. In some embodiments the cells may be provided frozen in a suitable freezing media such as a commercially available media supplemented with DMSO. The kit may optionally include instructions as to how to reconstitute the exosomes, culture the cells and/or contact the cells with exosomes so as to enhance vascular tube like formation.
  • In other embodiments the invention provides a kit comprising a human clonal embryonic progenitor cell, such as 30-MV2-6. The cell may be provided in at least one container in suitable media or buffer. The kit may include buffers and/or media for isolating exosomes from the cells. The kit may contain one or more vessels, e.g. a multi-well plate for culturing the cells. The kit may further contain a cell line capable of forming vascular tube like structures such as endothelial cells. Suitable cells include endothelial cells such as HUVEC and/or a a perivascular cell. Any or all of the cells may be provided frozen in a suitable media, e.g. freezing media such as a commercially available media supplemented with DMSO. The kit may optionally include instructions as to how to culture the cells and/or contact the endothelial cells with exosomes isolated from the progenitor cells so as to enhance or induce vascular tube like formation.
  • Additional Embodiments of the Invention
  • 1. An exosome isolated from a progenitor cell line.
  • 2. The exosome of 1, wherein the progenitor cell line is a human progenitor cell line.
  • 3. The exosome of 1, wherein the progenitor cell line is a clonal progenitor cell line.
  • 4. The exosome of 1, wherein the progenitor cell line is an endothelial progenitor cell line.
  • 5. The exosome of 1, wherein the exosome contains CD63.
  • 6. The exosome of 1, wherein the exosome contains one or more miRNAs listed in Table 2 or Table 4.
  • 7. The exosome of 1, wherein the exosome contains one or more angiogenic miRNAs.
  • 8. The exosome of 1, wherein the exosome contains miR-126.
  • 9. The exosome of 1, wherein the progenitor cell line expresses CD31 and
  • CD34.
  • 10. The exosome of 1, wherein the progenitor cell line expresses one or more genes listed in Table 1.
  • 11. The exosome of 1, wherein the progenitor cell line is 30-MV2-6.
  • 12. The exosome of 1, wherein the exosome enhances the formation of vascular tube like formations when contacted with an endothelial cell.
  • 13. The exosome of 1 further comprising a pharmaceutical carrier.
  • 14. A method of isolating an exosome from a clonal progenitor cell comprising 1) culturing the clonal progenitor cell in a suitable media or buffer for a time sufficient to allow the clonal progenitor cell to exocytose exosomes into the culture media or buffer; 2) harvesting the media or buffer from the cell culture of step 1; and 3) isolating the exosomes from the media or buffer of step 2, thereby isolating an exosome from a clonal progenitor cell.
  • 15. The method of 14, wherein the suitable media or buffer is PBS.
  • 16. The method of 14, wherein the suitable media or buffer is EGM-MV2.
  • 17. The method of 14, wherein after step 2 a precipitating agent is added to the media or buffer.
  • 18. The method of 14, wherein the precipitating agent comprises polyethylene glycol.
  • 19. The method of 14, wherein step 3 comprises centrifuging the harvested media of buffer.
  • 20. The method of 14, wherein the suitable time of step 1 is about 16 hours.
  • 21. The method of 14, wherein after step 2, the method further comprises a step of incubating harvested media or buffer.
  • 22. The method of 21, wherein the incubation step is performed for about 16 hours.
  • 23. The method of 21, wherein the incubation step is performed at about 4° C.
  • 24. A cell culture comprising an exosome isolated from a progenitor cell and a cell which was not the source of the isolated exosome.
  • 25. The exosome of 24, wherein the progenitor cell line is a human progenitor cell line.
  • 26. The exosome of 24, wherein the progenitor cell line is a clonal progenitor cell line.
  • 27. The exosome of 24, wherein the progenitor cell line is an endothelial progenitor cell line.
  • 28. The exosome of 24, wherein the exosome contains CD63.
  • 29. The exosome of 24, wherein the exosome contains one or more miRNAs listed in Table 2 or Table 4.
  • 30. The exosome of 24, wherein the exosome contains one or more angiogenic miRNAs.
  • 31. The exosome of 24, wherein the exosome contains miR-126.
  • 32. The exosome of 24, wherein the progenitor cell line expresses CD31 and CD34.
  • 33. The exosome of 24, wherein the progenitor cell line expresses one or more genes listed in Table 1.
  • 34. The exosome of 24, wherein the progenitor cell line is 30-MV2-6.
  • 35. The exosome of 24, wherein the exosome enhances the formation of vascular tube like formations when contacted with an endothelial cell.
  • 36. A method of inducing or enhancing a cell's ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a progenitor cell thereby inducing or enhancing a cells ability to form vascular tube like structures.
  • 37. The method of 36, wherein the cell capable of making vascular tube like structures is an endothelial cell.
  • 38. The method of 37, wherein the endothelial cell is a HUVEC.
  • 39. A method of treating a subject in need of vascular therapy comprising administering an exosome isolated from a progenitor cell.
  • 40. The method of 39, wherein the subject is human.
  • 41. The method of 39, wherein the subject exosome is administered to the subject to treat a condition chosen from cardiovascular disease, heart failure, infarction, chronic wounds, ulcer, clogged vessels or arteries, damaged vessels, stenotic vessels, arteriosclerosis, angina, peripheral vascular disease, Alzheimer's disease, ischemia, diabetes, cancer, cell replacement transplant or therapy, tissue and cell regenerative therapy and Parkinson's disease.
  • 42. The method of 39, wherein the progenitor cell line is a human progenitor cell line.
  • 43. The method of 39, wherein the progenitor cell line is a clonal progenitor cell line.
  • 44. The method of 39, wherein the progenitor cell line is an endothelial progenitor cell line.
  • 45. The method of 39, wherein the exosome contains CD63.
  • 46. The method of 39, wherein the exosome contains one or more miRNAs listed in Table 2 or Table 4.
  • 47. The method of 39, wherein the exosome contains one or more angiogenic miRNAs.
  • 48. The method of 39, wherein the exosome contains miR-126.
  • 49. The method of 39, wherein the progenitor cell line expresses CD31 and CD34.
  • 50. The method of 39, wherein the progenitor cell line expresses one or more genes listed in Table 1.
  • 51. The method of 39, wherein the progenitor cell line is 30-MV2-6.
  • 52. The method of 39, wherein the exosome enhances the formation of vascular tube like formations when contacted with an endothelial cell.
  • 53. The method of 39 further comprising a pharmaceutical carrier.
  • EXAMPLES Example 1: Preparation of Exosomes Derived from a Human Embryonic Progenitor Cell Line
  • Exosomes were prepared from a human embryonic progenitor cell line (PureStem® cell line, ESI Bio, Alameda, Calif.). PureStem® cell lines are scalable clonally pure embryonic progenitor cell lines derived from human embryonic stem (hES) cells (West et al. (2008) Regen Med. 3(3):287). The 30-MV2-6 PureStem® cell line is a CD31 positive, CD34 positive, endothelial progenitor line derived from the ESI-017 embryonic stem cell line. A gene expression profile of the 30-MV2-6 cells as analyzed by microarray is provided herein in Table 1, and includes genes yielding relative fluorescence units >1000 rfu.
  • TABLE 1
    30-MV2-6 P6,
    Gene symbol MBA_3877
    EEF1A1 29470.94
    EEF1A1 28581.27
    EEF1A1 28032.06
    TMSB4X 27627.77
    GNB2L1 27295.94
    TPT1 26985.77
    LOC100129758 26697.4
    LAIR1 26461.51
    F2R 26252
    LOC285176 26055.83
    RPL41 25872.51
    NAG18 25689.47
    LOC649150 25513.84
    FTL 25344.95
    LOC91561 25177.92
    LOC100132593 25023.94
    RPLP2 24889.1
    LOC388474 24755.51
    MGC16703 24611.59
    UBC 24480.02
    LOC389342 24350.84
    CLUAP1 24238.08
    RPS27 24117.06
    ZNF674 23992.58
    IMAA 23881.17
    RRP7B 23769.37
    C19orf31 23655.06
    ANXA2P2 23547.03
    LOC401206 23440.4
    LOC100133876 23333.52
    GGA1 23233.85
    MSH3 23140.74
    LOC729439 23047.74
    LOC440589 22950.21
    UBC 22847.36
    FTL 22745.35
    LOC100130553 22658.54
    LOC728658 22562.38
    ACTG1 22481.09
    LOC644604 22386.22
    RPL38 22292.94
    LOC642210 22201
    LOC148430 22112.95
    LOC100133465 22026.54
    LOC400963 21937.41
    TMSB10 21854.66
    LOC284393 21771.97
    RN7SL1 21682.27
    RPS29 21599.25
    LOC100133931 21519.28
    RPS12 21431.97
    ITIH5 21348.51
    LOC341457 21263.76
    LOC642250 21185.09
    KIAA0101 21102.53
    LOC389435 21019.57
    LOC100132488 20946.02
    ANXA2 20864.81
    LOC441034 20786.82
    RPS19 20710.05
    ACTB 20640.19
    LOC440589 20572.73
    LOC388720 20496.3
    LOC728658 20419.73
    FAM177A1 20346.14
    LOC387930 20271.08
    LOC440595 20196.18
    LOC653232 20127
    ORC6L 20058.03
    RPS25 19986.5
    ACTB 19922.52
    LOC100130980 19848.39
    PDCD7 19772.26
    RPL18A 19703.81
    LOC642892 19628.75
    LOC727808 19558.66
    LOC389223 19478.59
    ROCK2 19416.46
    CCR6 19350.04
    RPS15A 19290.37
    RPS11 19219.96
    RPL18 19149.27
    RPL6 19086.13
    RPS29 19022.98
    RPL38 18959.29
    RPL27A 18894.7
    RPS27 18834.63
    LOC387841 18770.86
    TUBA1A 18706.36
    LOC647361 18639.92
    RPL11 18576.44
    LOC100130446 18517.91
    LOC728553 18455.8
    UBA52 18391.57
    LOC728576 18328.88
    LOC100129553 18269.35
    MYL6 18203.91
    ACTG1 18140.81
    FTHL16 18076.95
    LOC441876 18016.04
    LOC343184 17954.72
    LOC391777 17893.68
    RPL9 17830.68
    RPS27A 17768.43
    LOC643863 17711.83
    RPS6 17652.27
    PSMD12 17594.21
    MYL6 17535.65
    LOC646195 17476.44
    RPL32 17418.94
    RPLP0 17356.64
    LOC729402 17294.36
    RPS17 17236.98
    RPL35A 17181.16
    RPL11 17118.82
    RPL18A 17060.66
    LOC100129158 17003.18
    LOC401019 16944.5
    LOC100133607 16889.64
    LOC729324 16834.91
    LOC645895 16775.01
    LOC649076 16721.19
    RPS16 16662.93
    PPIAL4A 16607.66
    RPL17 16550.82
    RPS10 16491.53
    LOC440733 16435.98
    RPL18A 16384.7
    LOC645899 16331.75
    LOC644029 16276.86
    VIM 16220.75
    LOC100129902 16168.98
    LOC440027 16117.11
    LOC728517 16063.24
    C10orf58 16009.07
    LOC728368 15951.03
    LOC439953 15892.32
    LOC651894 15837.98
    LOC644745 15784.83
    FARSLB 15733.5
    LOC388524 15675.03
    FTHL7 15621.63
    RPL27 15573.05
    RPL12 15524.71
    RPL37A 15473.36
    RPLP1 15423.13
    RPS20 15371.34
    RPS14 15315.83
    XPNPEP3 15265.39
    LOC389101 15209.69
    LOC402057 15164.27
    LOC643509 15114.24
    LOC284230 15063.66
    LOC642357 15015.66
    LOC652071 14963.6
    RPL39 14914.66
    LOC728576 14864.7
    LOC644464 14816.74
    ACTB 14769.08
    RPL30 14718.38
    RPS3 14668.78
    IFITM2 14620.77
    LOC644039 14568.97
    LOC645683 14518.14
    FAM115A 14471.98
    RPL19 14423.17
    LOC653162 14375.34
    LOC441775 14323.48
    LGALS1 14273.15
    RPS24 14227.73
    RPL3 14179.84
    RPS15 14130.03
    GNG11 14079.57
    CAV1 14033.15
    LOC729090 13979.81
    TM4SF1 13927.96
    NACA 13881.36
    ATP5EP2 13834.41
    LOC653881 13785.78
    RPL17 13738.14
    OAZ1 13689.32
    LOC645296 13641.2
    S100A10 13595.26
    RPL31 13545.91
    RPL24 13494.67
    LOC100128505 13442.05
    LOC645174 13394.88
    LOC648729 13345.11
    RPL8 13298.89
    LOC731096 13253.78
    LOC642741 13211.35
    LOC647276 13169.46
    LOC729903 13121.3
    LOC401019 13076.43
    LOC728128 13029.34
    CD81 12985.19
    LOC731542 12939.34
    LOC651436 12894.54
    RPL10A 12850.49
    LOC441013 12803.59
    RPS4X 12755.92
    GJC1 12710.73
    LDB2 12664.08
    RPS8 12614.56
    BTF3 12567.71
    WBP5 12522.85
    LOC650276 12477.88
    PFN1 12433.05
    RPL6 12389.93
    LOC100129141 12346.77
    ATP5B 12304.98
    CD93 12259.89
    VIM 12217.76
    LOC730754 12172.71
    HSPB1 12127.76
    LOC728453 12085.1
    EIF4A1 12041.45
    LOC389404 11999.19
    CD151 11957.83
    LOC647276 11912.95
    LOC729789 11868.46
    LOC728937 11828.98
    IFITM3 11788.35
    LILRB3 11744.43
    LOC646294 11705.26
    RPS2 11661.18
    FSCN1 11621.16
    LOC441246 11578.21
    LOC645387 11538.01
    LOC647099 11499.43
    PRCP 11460.34
    SOX18 11422.95
    LOC440575 11382.54
    LOC641814 11344.01
    KCNH6 11306.56
    LOC653314 11266.4
    LOC100133649 11225.89
    LOC729603 11184.33
    TUBA1C 11147.33
    LOC286444 11110.15
    LOC643531 11068.19
    LOC643284 11030.72
    AP2S1 10993.52
    PTRF 10952.09
    H3F3A 10913.04
    LOC100132742 10876.23
    LOC648210 10835.95
    EIF3E 10798.77
    RPL3 10762.44
    TXN 10725.63
    RPS29 10688.26
    MYL12A 10651.34
    GABPB2 10615.8
    RPS9 10581.37
    ATP5EP2 10543.17
    LOC647000 10509.38
    UBB 10473.09
    LOC388556 10436.98
    LOC728693 10403.82
    NGFRAP1 10370.85
    COX7C 10337.94
    GAPDH 10305.26
    GNAS 10271.86
    LOC400721 10233.35
    ICAM2 10200.27
    RPS3A 10164.91
    LOC100131713 10128.06
    B2M 10097.39
    UBB 10066.7
    CLEC2D 10033.68
    MGC26356 10004.77
    LOC100133273 9971.113
    TPT1 9939.753
    RPSA 9911.836
    RPS13 9882.49
    ENO1 9852.533
    LOC100132742 9822.571
    LOC729617 9790.951
    S100A10 9759.989
    LOC730187 9729.674
    LOC648000 9697.648
    LOC644464 9669.355
    RPS5 9640.032
    RPL14L 9608.673
    RPL36AL 9578.568
    NEDD8 9548.513
    RPS6 9520.098
    TGFBR2 9492.364
    RPLP1 9463.913
    LOC440926 9432.986
    TUBA1B 9407.654
    LOC284821 9377.909
    BTF3 9350.254
    LOC730246 9325.183
    LOC731365 9295.564
    LOC729466 9265.775
    LOC646200 9235.456
    ADAM15 9210.81
    RPS27L 9182.401
    AKR1D1 9154.34
    CYB5R3 9128.27
    RPS3A 9101.282
    RPS4X 9076.407
    CREB1 9049.403
    PDE4C 9022.809
    TFPI 8997.954
    LOC728782 8971.264
    LOC645387 8944.967
    SEPT2 8917.304
    GLTSCR2 8894.782
    SLC25A5 8867.191
    LOC646294 8843.528
    PECAM1 8815.434
    H3F3A 8791.886
    LOC649548 8767.153
    POTEF 8740.947
    TGFBR2 8714.794
    VWF 8689.385
    ITGB1 8666.613
    LOC729301 8641.795
    LOC100133812 8618.085
    EIF3L 8594.98
    LOC642947 8573.153
    DNCL1 8550.102
    TFPI 8526.81
    CDKN2AIPNL 8504.479
    VAMP5 8479.908
    CDH5 8455.61
    LRAP 8433.076
    RHOC 8409.996
    CDKN1A 8386.336
    S100A6 8366.138
    LOC645385 8343.072
    SNRPD2 8321.468
    ATP5A1 8298.988
    LDHA 8275.572
    EEF2 8253.723
    LOC389141 8231.352
    COX4I1 8212.02
    RPS9 8190.523
    LOC650152 8168.617
    CDC37 8145.008
    LOC643358 8121.837
    LOC100133233 8102.312
    YWHAQ 8082.7
    SNX3 8061.547
    AV762104 8040.466
    H3F3A 8017.827
    PFN1 7996.922
    GAPDH 7973.344
    YWHAZ 7953.511
    RPS14 7934.452
    RPL15 7915.846
    FAU 7885.26
    GPX1 7885.26
    LOC728620 7854.277
    RPL12 7833.73
    LOC648294 7816.09
    SLC16A12 7795.425
    LOC645715 7775.663
    RPS3A 7756.622
    ALDOA 7737.847
    CDK2AP1 7718.065
    TCEAL4 7699.396
    EEF1G 7679.361
    LOC646766 7659.651
    LOC100190938 7640.343
    C21orf55 7621.212
    EIF4G2 7602.998
    LOC100128731 7583.863
    LOC100133177 7566.641
    ZNF430 7547.497
    CCNI 7529.735
    RPL36 7510.359
    SERPINB6 7494.061
    LOC285053 7475.076
    EEF1B2 7455.902
    C11orf10 7436.825
    CALM3 7420.417
    LOC441087 7403.128
    TUBA1A 7386.972
    ZMAT3 7370.062
    KLF6 7351.817
    LOC643031 7336.027
    PABPC1 7319.402
    FKTN 7301.646
    CFL1 7282.935
    LOC644863 7265.614
    RPS27A 7248.343
    RPS17 7229.865
    COX7A2 7213.416
    RPS15A 7198.388
    LOC100134134 7180.453
    ATP5G2 7162.671
    IL18 7144.369
    LOC283412 7127.359
    NUCB1 7110.135
    LOC729798 7092.872
    LOC387867 7076.424
    PCBP1 7060.549
    MRLC2 7043.793
    LOC389517 7027.69
    LOC399900 7013.314
    SERF2 6997.182
    EEF1B2 6981.873
    MARCKS 6967.221
    NACA 6951.604
    LOC100129424 6936.077
    NPC2 6920.058
    RPL35 6904.67
    GPX4 6889.884
    C10orf58 6874.525
    TOMM7 6860.304
    SLC25A3 6845.603
    MDH2 6832.719
    RPL26 6817.137
    ATP5I 6800.876
    CTNNA1 6785.87
    RALA 6771.026
    FAM69B 6754.661
    CALM1 6738.351
    SHCBP1 6723.184
    SRP14 6708.369
    LOC646531 6693.502
    TACC1 6679.065
    DPYSL2 6663.472
    LOC100127993 6648.476
    LOC100130168 6634.481
    LOC285900 6620.755
    RPL7L1 6604.924
    PCBP2 6590.635
    PNPT1 6575.888
    HCG2P7 6562.18
    GPR116 6548.164
    H2AFZ 6534.282
    COX6C 6521.256
    ANXA5 6508.408
    NQO1 6495.337
    DAD1 6480.547
    COL4A1 6468.127
    ATP5A1 6454.348
    ZNF549 6440.91
    MYH9 6426.936
    SEC61G 6412.935
    FKBP1A 6399.774
    ARPC2 6387.262
    EIF4A2 6373.784
    EMP1 6360.16
    LOC729102 6346.829
    DDX5 6333.568
    HINT1 6318.728
    LOC645436 6305.671
    NOP10 6292.858
    PMP22 6279.687
    PSMB1 6265.957
    SQSTM1 6253.051
    LOC653737 6240.21
    HSPA8 6226.94
    TUBB 6214.39
    SHANK3 6201.642
    UQCRH 6188.65
    LOC730313 6176.263
    ATP5L 6163.733
    LOC100133372 6150.941
    LOC550643 6139.155
    TXN 6126.585
    DBI 6113.103
    TMBIM6 6100.574
    SEPT2 6088.34
    LOC642489 6075.764
    CDAN1 6062.012
    PLSCR3 6050.002
    LOC649049 6038.353
    JUND 6026.033
    YWHAH 6013.875
    GSTP1 6002.531
    HSP90AA1 5991.17
    SLC44A4 5979.221
    PSAP 5967.178
    CLDN5 5954.744
    LOC100130445 5943.334
    HNRNPD 5931.25
    SOD1 5919.922
    EEF1AL7 5909.133
    LOC647856 5897.997
    TM4SF18 5886.761
    PTBP1 5875.687
    RAN 5864.145
    RPL4 5852.968
    RAC1 5840.897
    CSTB 5829.299
    C14orf156 5817.476
    NME1-NME2 5807.022
    ITM2B 5796.124
    BGN 5785.701
    SCD 5776.214
    LOC645317 5764.427
    CMTM7 5753.074
    TOMM7 5741.794
    SEC61G 5730.923
    PPM1F 5719.127
    SLC25A3 5709.5
    ACVRL1 5699.607
    COMMD6 5689.162
    CLIC1 5677.854
    C17orf45 5667.08
    PRDX1 5656.452
    SAT1 5645.519
    SEPT9 5634.972
    ATP6AP2 5624.128
    CSDA 5613.424
    PRDX1 5602.954
    BTG1 5592.841
    MTCH1 5582.283
    LOC134997 5573.499
    LOC286444 5563.255
    RPS18 5552.792
    HLA-E 5543.25
    EDF1 5532.461
    ITGB1 5522.71
    MGST2 5511.287
    EIF3L 5500.726
    TM4SF18 5490.636
    NONO 5480.652
    ECSCR 5470.367
    PSAP 5460.726
    PSMA6 5451.676
    MARCKSL1 5442.01
    LOC729742 5432.988
    LOC100131387 5423.154
    NGFRAP1 5413.279
    MAP4K2 5402.065
    BEXL1 5392.217
    TBCA 5382.14
    EIF1 5371.946
    MCART1 5360.794
    MCM8 5351.053
    PSMB6 5340.972
    DAZAP2 5331.065
    QARS 5320.504
    LOC440055 5310.703
    APLNR 5301.342
    RPL13A 5290.971
    C14orf85 5281.173
    CNN3 5271.19
    LOC100132795 5260.873
    LAMA5 5251.119
    SLC44A1 5241.342
    LOC100131609 5232.272
    ARL16 5222.225
    LOC100129362 5212.558
    WSB1 5203.553
    TSPO 5193.677
    LOC645173 5184.411
    PRCP 5175.037
    ESD 5165.838
    HNRNPD 5156.636
    LOC648771 5148.032
    CST3 5139.557
    PRKAR1A 5130.244
    EPAS1 5121.617
    HSPA8 5112.767
    TPI1 5103.14
    NFIB 5094.79
    LOC646942 5086.126
    NCOA4 5077.186
    FLOT2 5069.23
    LOC729978 5060.045
    IGFBP4 5051.913
    LOC650646 5043.719
    ANP32B 5034.827
    CCND1 5026.154
    ATP5J 5017.307
    SHFM1 5008.422
    ATP5O 5000.003
    LOC440927 4992.161
    RHOA 4983.717
    H2AFY 4975.798
    CTGF 4966.956
    LOC728809 4957.689
    RALB 4949.349
    FABP5L2 4940.152
    NDUFB2 4932.081
    LOC646483 4923.669
    GNAI2 4916.183
    MRPL33 4908.738
    DDB1 4900.69
    LOC441073 4892.921
    LOC649447 4885.438
    WDR1 4877.46
    LOC400948 4868.795
    TIMP1 4860.48
    LOC402251 4852.983
    GNB1 4845.2
    RPL36AL 4837.16
    NOP10 4828.969
    CHCHD2 4821.331
    HIST1H4C 4813.682
    FLJ46309 4805.906
    ANGPT2 4798.277
    C20orf52 4790.903
    HOXB5 4782.823
    NDUFS5 4774.766
    UQCRQ 4766.396
    LOC402694 4758.111
    LOC644914 4750.406
    CXCR4 4742.489
    WBP2 4733.885
    COX5B 4725.663
    LOC646630 4717.094
    PRDX5 4704.898
    RPS26P11 4704.898
    NFKBIA 4694.165
    LOC728481 4686.756
    LOC100128084 4679.009
    SRGN 4670.712
    LOC645452 4663.261
    ARHGDIB 4655.951
    SUMO2 4648.817
    TRAM1 4641.106
    LDHA 4633.259
    MYH9 4625.853
    CTGLF7 4618.362
    LOC388654 4611.233
    CALM2 4604.143
    POFUT1 4596.614
    HDAC1 4588.697
    ROMO1 4581.606
    SHANK3 4574.51
    RPL5 4566.714
    NDUFAF3 4559.808
    GSTO1 4551.681
    SRP9 4544.382
    CCDC72 4537.007
    EIF3F 4529.952
    SRGN 4518.68
    RPS6P1 4518.68
    PFDN5 4508.432
    SCARB2 4500.946
    ESAM 4494.812
    HNRNPAB 4487.656
    EGLN2 4480.514
    LOC401537 4473.142
    EMP3 4466.904
    COX5A 4459.584
    SHC1 4452.944
    LOC648249 4445.896
    ANXA1 4438.55
    LOC728428 4431.064
    COMMD7 4424.43
    LOC392437 4417.459
    CSNK1E 4410.571
    GNS 4403.982
    LAMP1 4397.37
    DNAJA1 4390.667
    SPARC 4384.334
    SNRPG 4377.093
    RNF7 4367.271
    LOC729679 4367.271
    CAP1 4356.992
    LOC613037 4350.417
    FAM129B 4343.163
    PRDX5 4336.744
    SNHG5 4330.19
    LILRB1 4323.835
    ATP5J 4317.569
    CCT7 4311.306
    VDAC3 4304.92
    GIMAP8 4298.598
    PTTG1IP 4291.833
    PEA15 4285.228
    MDK 4279.023
    LOC728672 4272.423
    LTA4H 4265.782
    ARPC3 4259.694
    SFRS5 4253.734
    FABP5 4247.866
    B2M 4241.581
    JAM3 4235.323
    ATP5H 4229.135
    ZFAND5 4223.899
    UBE2E1 4217.975
    LOC100128266 4211.829
    NDUFA1 4206.047
    FKSG30 4199.931
    TUBB 4194.265
    LOC389168 4188.835
    C21orf24 4182.646
    PAM 4176.248
    LOC647340 4170.147
    ZNF14 4164.111
    MIF 4158.17
    COX6B1 4151.975
    NDUFS8 4145.751
    SF3B14 4139.69
    LOC389168 4132.783
    PSMD10 4126.234
    ATP5H 4119.575
    LOC644315 4113.929
    LOC643357 4107.532
    COX8A 4101.995
    HSP90B1 4095.739
    SAE1 4090.083
    YWHAB 4084.048
    LOC390345 4077.921
    RPS26L 4072.481
    SFRS6 4066.232
    CMTM3 4060.137
    NDUFB8 4054.134
    RPL7A 4048.417
    LASP1 4042.834
    LOC730029 4037.351
    GSTO1 4031.487
    HMGN1 4026.092
    HBXIP 4020.532
    LOC390557 4014.99
    KLF6 4009.341
    RTN4 4003.429
    AP2S1 3997.423
    TMEM17 3991.647
    LOC100132795 3986.28
    DYNLL1 3980.465
    UBE2D3 3973.772
    LOC92755 3968.05
    GPX4 3962.772
    APLN 3956.977
    LYVE1 3951.747
    AHNAK 3946.472
    LOC652624 3940.562
    ATP6V0E1 3934.799
    EIF4G2 3929.242
    FAM43A 3923.254
    LOC728873 3917.709
    PFDN5 3912.337
    LOC440737 3906.923
    HNRPM 3898.701
    CYB5B 3898.701
    LOC728126 3890.699
    RALGDS 3885.374
    GIMAP4 3880.145
    PPP2CA 3874.804
    CIRBP 3869.011
    C9orf80 3863.877
    CD34 3858.429
    ATP6AP1 3852.803
    MRFAP1 3847.485
    LOC649821 3842.339
    EZR 3837.019
    C20orf24 3831.573
    CD99L2 3825.978
    AIRE 3820.74
    PSMC1 3815.655
    C10orf10 3810.431
    LOC23117 3805.025
    ATP1A1 3799.669
    TKT 3794.411
    PSMB7 3789.396
    JUP 3784.532
    LOC643433 3779.673
    TMEM66 3774.548
    PSMC1 3769.582
    NDUFA3 3764.494
    MAGED1 3759.579
    C20orf24 3754.453
    LOC646785 3746.863
    LOC653226 3746.863
    SET 3739.565
    CRIP2 3734.717
    GLRX5 3730.108
    LOC100131196 3725.29
    PGD 3720.447
    TCEB2 3715.75
    BX537698 3711.047
    TMEM59 3706.181
    C8orf37 3701.639
    ZNF428 3696.942
    PHLDA1 3692.216
    TUBA1C 3687.356
    ERP29 3682.382
    RPL21 3677.297
    ESM1 3672.187
    LOC728139 3666.905
    FAM50A 3661.865
    LAMC1 3657.38
    UBE2I 3652.873
    ACTR2 3648.318
    RPS15A 3643.939
    C8orf45 3639.256
    B4GALT5 3635.08
    ADD1 3628.227
    FAM119A 3628.227
    LOC646819 3621.287
    EIF3B 3617.007
    C6orf48 3611.933
    HLA-A 3607.342
    ALKBH5 3602.68
    KHDRBS1 3598.262
    LOC100134648 3593.131
    SNRK 3588.844
    MAPRE1 3584.246
    APP 3579.695
    ATP5F1 3574.877
    DYNLRB1 3570.567
    RASIP1 3565.849
    LOC729926 3561.556
    CS 3556.809
    NUCKS1 3552.277
    C20orf100 3547.784
    SFRS5 3543.466
    FCGRT 3539.09
    ALDH9A1 3534.536
    JTB 3530.451
    DCTN2 3525.898
    FAM127A 3521.184
    EPN1 3516.76
    LOC402112 3512.346
    RRAGA 3507.74
    ARHGEF2 3503.294
    ITGB1 3498.718
    FKBP1A 3493.963
    NDUFA12 3489.713
    VEGFB 3485.396
    FEZ2 3480.915
    FKBP1A 3476.397
    TRMT112 3471.984
    PRKCH 3467.297
    LOC391370 3462.874
    RAB11A 3458.473
    S100A16 3454.14
    ROBLD3 3449.96
    TALDO1 3445.991
    RPL22 3441.584
    LOC644511 3437.516
    LOC127295 3431.188
    ANXA2 3431.188
    ARF4 3424.504
    AHR 3420.322
    TXNDC5 3415.883
    LOC646688 3411.785
    NDUFB3 3407.559
    AP1S2 3403.573
    DNAJC8 3399.217
    SFRS2 3394.83
    MATR3 3390.652
    ATP6V0C 3386.519
    C3orf34 3382.759
    NDUFB3 3378.886
    LOC728553 3374.77
    HNRPA2B1 3370.461
    OCIAD1 3366.672
    TMEM14C 3362.56
    IGFBP2 3358.728
    VAMP8 3354.294
    UQCRFS1 3350.384
    EIF3D 3346.006
    CTNNA1 3341.683
    SQLE 3337.535
    TSPAN18 3333.603
    RNASE1 3329.575
    CD99 3325.701
    ATP6V1E1 3321.748
    OSTC 3318.133
    PRR13 3313.96
    HNRPA1P4 3310.016
    LOC440353 3306.287
    ERGIC3 3301.974
    C2orf69 3297.976
    LOC100133477 3293.898
    LOC728698 3289.942
    LOC648390 3286.072
    HK1 3282.036
    PDHB 3278.294
    FLJ44124 3274.603
    TUG1 3270.77
    MORF4L2 3266.977
    AL15748 3263.189
    MYADM 3259.222
    DEGS1 3255.744
    LOC727865 3251.966
    LOC729236 3248.111
    LOC158345 3244.281
    PARK7 3240.659
    CS 3236.723
    BMS1P5 3233.004
    LOC390354 3229.485
    SNRPB2 3226.027
    PCBP2 3222.358
    LOC440043 3218.805
    LOC402175 3215.17
    TMBIM4 3211.304
    LOC730004 3207.415
    LOC374395 3203.745
    ZDHHC8 3200.006
    MFNG 3196.474
    AMY1C 3192.522
    VCL 3188.645
    GABARAPL2 3185.102
    TUBB2B 3181.562
    RCN1 3176.229
    PTK2 3176.229
    C14orf173 3170.849
    LOC399804 3166.942
    VKORC1 3163.391
    CCNY 3159.724
    PRNP 3156.361
    PTP4A2 3152.617
    NDUFB5 3148.956
    LOC100131801 3145.122
    NDUFA4 3141.587
    GAPDH 3137.906
    MRPS21 3134.596
    HSPD1 3131.109
    DARS 3127.618
    PLOD1 3124.315
    LOC347544 3120.537
    DUXAP3 3116.969
    POMP 3113.4
    GPIHBP1 3109.723
    PLS3 3106.332
    PGK1 3101.494
    ITPR3 3101.494
    HIATL2 3096.506
    ZNF486 3092.927
    MFSD10 3089.188
    PON2 3085.436
    NME1 3082.096
    LOC731985 3078.831
    ILF2 3075.404
    DSTN 3071.802
    SFRS9 3068.507
    DUSP19 3064.885
    GHITM 3061.454
    FAM124B 3057.985
    ATP6V1E1 3054.703
    PTTG1IP 3051.21
    HPCAL1 3047.698
    ZNF394 3042.981
    RAB7A 3042.981
    CAV1 3037.999
    DYNC1LI2 3033.362
    FASN 3033.362
    PTBP1 3028.638
    CCDC130 3025.44
    PSMA4 3022.079
    HMGN1 3018.673
    TMED3 3015.24
    CCT8 3011.764
    IL10 3008.282
    LOC645058 3005.028
    MORF4L1 3001.861
    SLC44A2 2998.583
    TMEM123 2995.415
    MAT2A 2992.155
    ADM 2988.951
    PRND 2985.625
    HNRNPK 2982.658
    NOL7 2979.434
    YBX1 2976.279
    LOC391656 2973.021
    CAMLG 2969.814
    FLNB 2966.365
    ARL6IP1 2963.044
    LOC399988 2959.773
    LOC100130562 2956.718
    RWDD1 2953.291
    LOC650518 2949.922
    TCEAL3 2947.075
    S100A4 2944.066
    EIF2S3 2940.487
    PRDX6 2937.178
    TSPAN9 2934.115
    GLO1 2931.327
    PSMD6 2928.123
    ILK 2924.745
    ACADVL 2921.625
    RHOC 2918.659
    PSME1 2915.555
    LOC387820 2912.308
    LDLR 2909.147
    TPM2 2904.888
    LOC728888 2904.888
    SEC11A 2900.347
    TEAD2 2897.152
    SLC25A6 2894.058
    BTBD2 2890.722
    NCL 2887.878
    LOC100132391 2884.602
    RPN1 2881.471
    TRIM8 2878.486
    HEXB 2875.395
    ZMAT3 2872.252
    MGST3 2869.117
    APP 2866.02
    LOC728244 2863.085
    ARGLU1 2860.002
    LEPROT 2857.123
    DDX51 2854.073
    CXXC5 2850.969
    AP1S2 2847.996
    LOC653314 2845.126
    SRP14P1 2842.19
    ACP1 2838.992
    C14orf153 2836.086
    C20orf30 2832.782
    UBA1 2829.927
    SNRPB 2826.874
    TXNIP 2823.728
    NUDT14 2820.921
    LOC642817 2818.102
    ATP1B1 2815.268
    CSNK2B 2812.341
    SNRPF 2809.605
    UXT 2806.481
    EIF3M 2803.284
    ALDOA 2800.582
    EFEMP1 2797.68
    STAU1 2794.503
    ANAPC13 2791.876
    DMC1 2788.981
    HNRNPH1 2785.906
    LPP 2783.35
    KRTCAP2 2780.536
    RPL14L 2777.793
    RPRC1 2774.822
    DKK3 2771.896
    BUB3 2769.109
    CAPZA2 2766.271
    MGC16121 2763.458
    EIF4B 2760.512
    MYH10 2757.669
    LOC100134159 2754.867
    ARL2 2751.979
    COLEC12 2749.382
    RHOJ 2746.622
    LOC401115 2743.952
    TIMM23 2741.358
    CARM1 2738.743
    PJA2 2736.055
    CMIP 2733.33
    TINP1 2730.681
    COPA 2728.078
    SSR4 2725.157
    LOC645688 2722.435
    PALM 2719.394
    UBE2D3 2716.675
    TMSL3 2714.115
    EID2B 2711.423
    TGM2 2708.749
    P4HB 2706.261
    NAT5 2703.492
    LOC653079 2700.971
    STX16 2698.241
    PUF60 2695.483
    SEC61B 2692.776
    KLF2 2690.108
    LOC441506 2687.565
    PSMA1 2684.755
    DAPP1 2682.076
    RAB10 2679.581
    TIMP2 2677.038
    NDUFA8 2674.402
    PRDX5 2671.801
    PSMA5 2668.989
    PIGY 2666.487
    PRSS23 2663.739
    ATP6V1F 2661.253
    C2orf28 2658.504
    PLS3 2655.947
    STARD7 2653.165
    FDFT1 2649.481
    LOC100130003 2649.481
    NUP62 2645.797
    PSMB3 2643.409
    FAM39E 2640.664
    LOC653505 2637.95
    TOMM6 2635.176
    AK095855 2631.407
    CAPNS1 2631.407
    LOC649553 2627.467
    RPL17 2625.058
    RBX1 2622.313
    CYBA 2619.898
    ARPC1A 2617.495
    VAMP3 2614.954
    LOC100133772 2612.487
    SUMO3 2609.9
    CD34 2607.309
    PRMT1 2605.064
    CD63 2602.476
    TPI1 2599.884
    BRI3 2597.245
    LMNA 2594.722
    SNRNP70 2592.098
    ID3 2589.513
    LOC442454 2587.179
    CAV2 2584.557
    POLR2G 2582.077
    LOC388707 2579.665
    ATP6V0E1 2577.161
    LOC654194 2574.714
    PHPT1 2572.017
    POLR2F 2569.517
    APEX1 2567.108
    EIF3K 2564.684
    LOC653226 2562.343
    C15orf24 2559.838
    IMPDH2 2557.397
    DUSP3 2553.815
    KPNB1 2553.815
    NDUFA11 2549.928
    CTGF 2547.565
    NDUFS4 2544.957
    BLZF1 2542.604
    RHEB 2540.184
    PRICKLE4 2537.998
    PTPLAD1 2535.534
    HSP90AA1 2533.024
    BANF1 2530.587
    COL4A5 2528.138
    SDCBP 2525.598
    LRRC37B2 2523.156
    LRRC32 2520.757
    GSTM1 2518.524
    TTC3 2516.152
    DYSF 2513.921
    ETS1 2511.662
    PON2 2509.463
    PDCD6 2507.209
    TOMM20 2504.969
    REPIN1 2502.723
    BOLA2 2499.166
    LOC391126 2499.166
    SIVA1 2495.534
    HPRT1 2493.245
    PRDX3 2491.116
    CDC16 2488.933
    ATOX1 2486.616
    RBM22 2484.209
    NUAK1 2481.962
    VPS29 2479.728
    VDAC1 2477.395
    EVL 2475.158
    TAGLN2 2473.013
    LY6E 2470.802
    GPR56 2468.456
    REEP5 2466.211
    ZNF69 2464.034
    LOC728590 2461.808
    EEF1D 2459.394
    POLR2H 2457.249
    PPP2R1A 2454.819
    PSMB5 2452.568
    C20orf43 2450.369
    SPCS1 2448.261
    ATF4 2444.895
    EIF4A1 2444.895
    OSTC 2441.645
    RASGRP3 2438.988
    LOC100128353 2436.823
    LOC648024 2434.574
    EHD4 2432.192
    VPS26A 2429.896
    ARAP3 2427.613
    SDHB 2425.444
    RPS6KA2 2423.269
    MRPS6 2420.95
    LOC648210 2418.767
    EIF3G 2416.711
    CNBP 2414.27
    GPR56 2412.085
    SH2B3 2409.804
    REXO2 2407.484
    RNF181 2405.349
    TSPAN3 2403.161
    ADCY4 2401.073
    HNRPUL1 2398.99
    LOC388339 2397.042
    DDX3X 2394.857
    GALK1 2392.643
    PPP1CC 2390.58
    HSP90AB1 2388.513
    FAM107B 2386.169
    CREB1 2384.065
    VIL2 2381.946
    SNRPF 2379.903
    TST 2377.882
    LOC730534 2375.733
    MKNK2 2373.554
    STC1 2371.352
    EIF3I 2369.413
    PPP1R11 2367.293
    MYLIP 2365.426
    SNRPB 2363.423
    SDCBP 2361.481
    PSMB4 2359.383
    YY1 2357.516
    NDUFS3 2355.493
    H1F0 2353.539
    THBS1 2351.476
    SMS 2349.239
    LOC391075 2347
    ARF1 2345.002
    ZMIZ1 2343.074
    RHOG 2341.097
    EIF4H 2339.187
    RAC2 2336.985
    PPA2 2334.924
    MSN 2332.827
    RPL23 2330.874
    ITGB4BP 2328.691
    MYL6B 2326.759
    MFGE8 2324.612
    ADSL 2322.313
    RPL10A 2320.322
    SHISA5 2318.275
    SGSM2 2316.145
    ARL5A 2314.145
    LOC644063 2312.122
    DHX15 2310.085
    LOC642956 2307.807
    DDOST 2305.894
    SDHALP1 2303.914
    EIF4H 2302.04
    MRPL22 2300.216
    LOC100132528 2298.284
    LOC653658 2296.237
    DYNC1I2 2294.276
    C20orf30 2292.387
    HNRNPR 2290.423
    G3BP2 2288.484
    ZNF682 2286.593
    PGRMC1 2284.582
    LOC728492 2282.552
    BAX 2280.554
    MGC4677 2278.547
    NNAT 2276.699
    SRRM2 2274.895
    GUK1 2272.81
    S100A4 2270.843
    TMEM14C 2268.872
    TIE1 2267.041
    IL32 2265.217
    RPS27 2263.252
    GSTM2 2261.182
    SUMF2 2259.346
    DDT 2257.489
    C20orf199 2255.455
    ARCN1 2253.645
    CSNK1G2 2251.65
    UCHL1 2249.632
    MDH1 2247.564
    ARL2BP 2245.594
    TEK 2243.715
    TCEB1 2241.851
    M6PRBP1 2239.955
    CAV2 2238.078
    HYAL2 2236.148
    PRKCDBP 2234.444
    NUDC 2232.664
    NBPF10 2230.705
    NDUFA2 2228.844
    LXN 2227.013
    LOC647000 2225.325
    C20orf160 2223.387
    PPM1G 2220.516
    UBL5 2220.516
    URM1 2217.626
    VASH1 2215.82
    XRCC6 2213.876
    PSMB2 2212.134
    TCEAL8 2210.404
    ZNHIT1 2208.435
    SETD3 2206.564
    NHP2 2204.765
    LOC100128288 2202.999
    SNRK 2200.977
    SDPR 2199.231
    ESYT1 2197.376
    GDI2 2195.532
    LOC100130561 2193.647
    CUTA 2191.759
    GJA4 2189.946
    SFRS4 2187.892
    TMED9 2186.194
    CATSPER2 2184.456
    RAI14 2182.639
    PSMD10 2180.94
    AB074172 2179.276
    UQCRHL 2177.506
    ARMET 2175.753
    MAP1LC3A 2173.799
    ZNF652 2172.067
    TPM1 2170.117
    LMO2 2168.384
    WDR18 2166.626
    PODXL 2164.78
    PCMT1 2163.02
    FERMT2 2161.297
    SNHG6 2158.576
    ACLY 2158.576
    TMEM98 2155.966
    GYPC 2154.276
    HNRNPM 2152.428
    FAM171A1 2150.789
    PXDN 2148.125
    NGRN 2148.125
    LSM2 2145.52
    CIB1 2143.783
    NDUFB7 2142.01
    ANKS1A 2140.256
    NPTN 2138.669
    EFNB2 2137.024
    C12orf57 2135.199
    PRDX4 2133.33
    BEX4 2131.534
    RDX 2129.679
    TPM1 2128.18
    LOC391811 2126.614
    HNRNPA0 2124.898
    RAB5B 2123.318
    AARS 2121.396
    RBM10 2119.742
    CKLF 2117.921
    C15orf63 2116.23
    ARPC5 2114.352
    DAB2 2112.622
    HLX 2110.92
    CD46 2109.263
    ARHGAP23 2107.661
    ERH 2105.924
    GLTP 2104.153
    OXA1L 2102.456
    ADAMTS9 2100.784
    TUBB6 2098.927
    PRPSAP1 2097.122
    LOC728533 2095.487
    CETN2 2092.925
    COMT 2092.925
    MIR1978 2090.444
    ATP1B3 2088.676
    TCF25 2086.963
    GSPT1 2085.414
    LOC728031 2083.839
    HOXB7 2082.164
    LOC644907 2080.516
    XPNPEP1 2078.884
    ZNF22 2077.23
    DPY30 2075.5
    LOC653773 2073.876
    LOC100128410 2072.218
    CCL14 2070.749
    LOC648622 2069.157
    CRCP 2067.613
    COMMD3 2066.041
    LOC728661 2064.404
    FEZ2 2062.769
    QRFPR 2061.197
    CD2BP2 2059.596
    LOC645138 2057.918
    ANXA2P1 2056.255
    ACTN1 2054.614
    SASH1 2052.96
    TPM2 2051.325
    RBM5 2049.709
    CTSL1 2048.076
    HSPD1 2046.443
    LOC728554 2044.892
    TEK 2043.162
    LOC148430 2041.445
    CLTA 2039.745
    DDX1 2038.117
    MGC87895 2036.414
    ENY2 2034.704
    C21orf58 2033.058
    EIF3H 2031.374
    PTOV1 2029.659
    C19orf10 2027.986
    NSA2 2026.356
    SLC2A3 2024.764
    PRDX3 2023.325
    GTF2A2 2021.712
    BCKDK 2019.916
    SPTLC1 2018.472
    SPCS2 2016.07
    C17orf61 2016.07
    C13orf15 2013.725
    GRAP 2012.201
    TXNDC17 2010.558
    GLG1 2009.077
    RING1 2007.525
    CDC16 2005.976
    FTHL12 2004.417
    JMJD8 2002.931
    DYNLRB1 2001.493
    LOC730740 2000
    GTPBP6 1998.578
    ADAM19 1997.024
    OCIAD1 1995.423
    ALPP 1993.895
    LOC645452 1992.252
    LOC730455 1990.712
    CIP29 1989.102
    HDHD1A 1987.7
    PRDX2 1986.069
    SSU72 1984.546
    TBCB 1983.009
    UBXN4 1981.49
    LAMP2 1979.835
    SOX7 1978.374
    TSPAN4 1977.045
    SH3BGRL 1975.596
    JAG2 1974.152
    LDHB 1972.673
    ANKRD30B 1971.064
    STOML2 1969.434
    MT2A 1967.846
    CKAP4 1966.368
    PABPC4 1965.115
    COX7A2L 1963.582
    BCAP31 1962.06
    VPS35 1960.581
    LOC730316 1959.032
    HSPE1 1957.463
    DECR1 1956.08
    NBPF20 1954.56
    HDAC2 1953.03
    DYNLT1 1951.625
    MTPN 1950.182
    ATP5E 1948.728
    CLCN7 1947.253
    KDELR1 1944.949
    GARS 1944.949
    FNTA 1941.981
    NME4 1941.981
    ADAR 1939.685
    SS18L2 1938.165
    SNHG7 1936.806
    CLIC4 1935.304
    MRPL22 1933.809
    PLIN2 1932.481
    NDEL1 1930.949
    LOC100131531 1929.469
    BC036485 1928.086
    UNC84B 1926.666
    SEC61A1 1925.247
    PPP2R2A 1923.752
    CCT2 1922.433
    HGS 1921.051
    RNASEK 1919.6
    EIF4A3 1918.241
    TUBB2C 1916.747
    APEX1 1915.376
    CCND3 1913.884
    RPAIN 1912.433
    LOC729841 1911.051
    UNC50 1909.61
    RPP21 1908.262
    LZTR1 1907.012
    ABCA1 1905.602
    NDUFV2 1904.214
    TAF15 1902.111
    F2R 1902.111
    GSTK1 1900.015
    LSM1 1898.52
    GRK5 1897.066
    RPS23 1895.565
    RPLP0 1894.206
    SRPX 1892.791
    SRP14 1891.423
    LOC642590 1890.132
    MRPL51 1888.738
    MTSS1 1887.426
    SPG7 1886.074
    ETFA 1884.622
    NCSTN 1883.33
    ARS2 1882.034
    LOC90586 1880.692
    TPD52L2 1879.422
    RHBDF1 1878.106
    MAPK3 1876.805
    RSL24D1 1875.555
    PIN1 1874.261
    CTSB 1872.918
    CCDC90B 1871.493
    NAP1L4 1870.159
    ATP9A 1868.852
    EVI1 1866.765
    POLR1D 1866.765
    SSTR1 1864.751
    PCID2 1863.457
    HIGD1A 1861.98
    MGC10997 1860.583
    STRAP 1859.285
    MRPL36 1857.963
    ARHGEF7 1856.522
    ATP5D 1855.212
    NR2F2 1853.965
    FNBP1 1852.702
    LMBRD1 1851.216
    LOC392437 1849.863
    DNAJB11 1847.944
    ELTD1 1847.944
    ZNF358 1846.034
    PPP6C 1844.683
    LOC646567 1843.41
    GOLGA7 1842.093
    ID1 1840.693
    UBXN1 1839.336
    IPO11 1838.036
    TNFRSF10B 1836.729
    C7orf59 1835.465
    SYT11 1834.199
    OSTF1 1832.859
    ZNHIT3 1831.481
    GOT2 1828.835
    LOC399748 1827.599
    HSBP1 1826.317
    EFNA1 1825.115
    IDH1 1823.88
    HNRPK 1822.669
    ANGPTL2 1821.346
    HOXB8 1820.111
    TMEM85 1818.718
    SIAH1 1817.475
    LOC285741 1816.02
    CFDP1 1814.757
    LOC652489 1813.434
    BANP 1812.163
    C2orf25 1810.833
    ARHGAP17 1809.513
    SMS 1808.21
    ATP6V1G1 1806.998
    TMED2 1805.879
    CTDSP2 1804.656
    PPP2CB 1803.45
    PSMD4 1802.198
    FKBP14 1800.996
    LUZP1 1799.739
    CTSL1 1798.517
    GLCE 1797.266
    DCTPP1 1795.899
    PCNP 1794.674
    MRPL37 1793.311
    SSBP1 1792.031
    BZW2 1790.698
    GLB1 1789.417
    STOM 1788.173
    ZYX 1786.884
    EIF5A 1785.669
    NUMB 1784.498
    PSMD7 1783.367
    FXR1 1782.152
    PARP4 1780.857
    RARS 1779.561
    RBMS1 1778.275
    FAM175A 1776.427
    SF3B1 1776.427
    LOC100128936 1774.579
    LOC644191 1773.38
    CHRNA5 1772.196
    EIF2AK1 1771.002
    MGC71993 1769.83
    LOC255167 1768.607
    CAB39 1767.443
    FGD5 1766.278
    HNRPR 1765.174
    RPS26 1763.91
    TAX1BP3 1762.65
    PSMC5 1761.404
    LOC642755 1760.256
    LOC202781 1758.944
    DKK3 1757.831
    ZMYND11 1756.637
    C19orf70 1755.48
    SVIL 1754.196
    SELS 1753.02
    NDUFB11 1751.891
    CPNE3 1750.703
    MRI1 1749.53
    LOC401397 1748.318
    TPRG1L 1747.141
    VAT1 1746.025
    TNFRSF1B 1744.824
    C5orf28 1743.673
    NOSIP 1742.474
    FER1L3 1741.29
    RPS28 1740.056
    TCEAL4 1738.878
    FAHD1 1737.738
    PQLC1 1736.625
    ATP6V1A 1734.904
    TAX1BP1 1734.904
    WDR6 1733.152
    GPS1 1731.944
    DAB2 1730.771
    LOC338870 1729.516
    HNRNPM 1728.303
    PUM1 1727.184
    SLC9A1 1725.915
    TRMT5 1724.831
    ATP1B1 1723.685
    EMD 1722.585
    PSMG2 1720.345
    CCDC23 1720.345
    C20orf24 1720.345
    UROD 1718.176
    PPP2R2B 1717.048
    COPB1 1715.868
    PUM1 1714.76
    CASP2 1713.646
    TFG 1712.523
    LOC728820 1711.473
    CHCHD9 1710.374
    CSE1L 1709.265
    LOC100131785 1708.063
    FAM120A 1706.955
    TMEM111 1705.855
    HMGN2 1704.702
    TNK2 1703.603
    PTPRF 1702.506
    C19orf56 1701.464
    C1orf85 1700.401
    CLDND1 1699.33
    SF3B5 1698.141
    CCDC56 1696.976
    HIGD2A 1695.904
    SRP54 1694.673
    MRPS18C 1693.505
    SLC38A2 1692.344
    SYPL1 1691.19
    PWP1 1690.03
    CYC1 1688.812
    VPS28 1687.69
    BSG 1686.67
    TRIOBP 1685.549
    IDH3B 1684.483
    FAM65A 1683.363
    IMP3 1682.299
    SCRN1 1681.224
    PLOD3 1680.136
    TSC22D1 1679.005
    UCKL1 1677.916
    C3orf54 1676.807
    TCEAL3 1675.69
    ZRANB2 1674.65
    SFRS18 1673.475
    PSMC2 1672.436
    TMEM147 1671.356
    CNDP2 1670.377
    BAT1 1669.32
    PSMD4 1667.617
    SYPL1 1667.617
    FAM96A 1666.04
    PFDN1 1664.983
    MRPS12 1663.866
    MXD4 1662.821
    PSMA6 1661.627
    LOC729279 1660.481
    LRP10 1659.336
    MRPL17 1658.299
    WSB1 1657.178
    ARL6IP5 1656.15
    HSD17B7 1655.049
    CYP1A1 1654.03
    RGL1 1653.014
    ARHGDIA 1651.931
    LSM3 1650.846
    LOC440359 1649.8
    LOC730744 1648.793
    SFRS14 1647.759
    LOC653566 1646.763
    LOC651894 1645.664
    CLDND1 1644.57
    PTDSS1 1643.545
    SSB 1642.473
    GRIPAP1 1641.337
    RRAS 1640.329
    PRPF8 1639.153
    CRTAP 1638.192
    AV737317 1637.094
    PDIA5 1635.993
    GYPC 1634.996
    LOC653086 1633.937
    SULT1A1 1632.887
    EXOSC10 1631.823
    SEC14L1 1630.73
    CMTM7 1629.708
    CDK4 1628.643
    SLC12A2 1627.639
    LOC100128062 1626.109
    DUSP22 1626.109
    LOC100129379 1624.099
    TMEM158 1624.099
    SH3GLB1 1622.482
    TGFBR3 1621.398
    SFRS1 1620.42
    C1QBP 1619.375
    MMS19L 1618.318
    FABP4 1617.363
    SYF2 1615.805
    LOC647285 1615.805
    POM121C 1614.33
    CTXN1 1613.323
    PMP22 1612.342
    DCTN3 1611.336
    EI24 1610.259
    SNURF 1609.211
    PFKP 1608.135
    AK3 1607.133
    NAP1L1 1606.141
    PSMB10 1605.125
    FIS1 1604.15
    RCN2 1603.109
    COPS5 1602.015
    UBE1 1600.951
    METAP2 1599.995
    DEGS1 1598.954
    TPP1 1597.891
    TCF4 1596.551
    PLD3 1596.551
    SLC20A1 1595.085
    BRD2 1593.958
    GTF2E2 1592.892
    CARHSP1 1591.833
    KIAA1949 1590.829
    PSMF1 1589.926
    DCTN1 1588.867
    LOC643668 1587.862
    C11orf59 1586.41
    CDC42EP4 1586.41
    LOC729317 1584.87
    ATXN2 1584.024
    ZDHHC16 1582.96
    KLHL3 1581.971
    FBXO11 1581.021
    HSD17B12 1579.986
    WDR54 1578.996
    THOC7 1578.022
    LOC286157 1577.048
    PGAM1 1576.049
    RRBP1 1575.086
    COPS3 1574.042
    TGOLN2 1572.574
    ATP5J2 1572.574
    PAICS 1571.114
    MYCT1 1570.123
    CCNG1 1569.125
    EIF1B 1568.22
    PTPLAD1 1567.188
    GLUD1 1566.225
    SRRM1 1565.277
    BCL2L1 1564.333
    SDHAF2 1563.342
    TMEM126B 1562.454
    COX6A1 1561.487
    LOC651198 1560.508
    TSC22D3 1559.555
    ACAT1 1558.552
    LOC389787 1557.626
    RALY 1556.622
    SSR2 1555.669
    MTUS1 1554.711
    RNF144 1553.795
    LOC100132727 1552.848
    JUN 1551.928
    NDUFAB1 1550.977
    MCM3 1550.039
    MRFAP1L1 1549.157
    PRNP 1548.212
    COL18A1 1547.275
    ECH1 1546.324
    LOC650369 1545.353
    CDC42EP5 1543.892
    ZNF738 1543.892
    HSPA9 1542.471
    KCTD12 1541.497
    SUZ12 1540.117
    RABGAP1 1540.117
    GTF2H5 1538.645
    DDA1 1537.81
    BIN1 1536.906
    DUSP1 1535.978
    STK24 1535.023
    ITGA5 1534.085
    DDX47 1533.17
    SEC31A 1532.248
    PNPT1 1531.378
    SPTBN1 1530.478
    GPSM1 1529.602
    EXOC7 1528.681
    PSMC6 1527.768
    GTPBP4 1526.921
    DBN1 1525.956
    GLT25D1 1525.037
    EIF2A 1523.676
    LIMCH1 1523.676
    CCNDBP1 1522.32
    TROVE2 1521.457
    RPS26L 1520.508
    AURKAIP1 1519.568
    WBP5 1518.619
    RPAIN 1517.721
    TPM3 1516.876
    KIAA1671 1516.034
    LOC653994 1515.152
    DBNL 1514.297
    NTAN1 1513.386
    BOLA2 1512.461
    TMEM44 1511.551
    EDN1 1510.177
    ITGB1BP1 1510.177
    CCDC109B 1508.421
    ZNF22 1508.421
    C4orf18 1507.078
    APOA1BP 1506.23
    SH3BGRL3 1505.32
    UBE2M 1504.415
    CAPNS1 1503.518
    SPOP 1502.701
    GNG10 1501.888
    PLSCR4 1500.944
    TMEM181 1500.131
    LOC388275 1499.324
    FEZ1 1498.408
    COX17 1497.494
    LMNA 1496.663
    C21orf33 1495.672
    ITM2C 1494.223
    DNMT1 1494.223
    ITM2C 1492.824
    PRR14 1492.013
    CYR61 1491.16
    BNIP3 1490.33
    IGF2R 1489.489
    SON 1488.169
    GFOD1 1488.169
    LOC641700 1486.891
    MRI1 1486.039
    PROCR 1485.086
    LOC732007 1484.229
    CTTN 1483.402
    LOC644799 1482.539
    RUSC1 1481.565
    GRN 1480.712
    ITGB5 1479.823
    GYG1 1478.946
    MRPL23 1478.119
    ASAP1 1477.166
    KPNA4 1476.338
    PSME2 1475.46
    PRAGMIN 1474.691
    RCC2 1473.898
    INPP1 1472.989
    LOC100132291 1472.102
    NHP2L1 1471.226
    ANAPC11 1470.023
    CGNL1 1470.023
    NFATC2IP 1468.795
    IARS2 1467.883
    TJP1 1467.001
    LOC729768 1466.205
    ATP6V1B2 1465.338
    LOC644774 1464.489
    CSF2RA 1463.664
    ANGPT2 1462.786
    ATP5J2 1462.035
    ANAPC5 1461.197
    BRMS1 1460.376
    ADRM1 1459.571
    C2orf28 1458.684
    DGUOK 1457.91
    SLC25A39 1456.678
    CHCHD10 1456.678
    DGUOK 1455.463
    PEBP1 1454.59
    PPP1R14B 1453.763
    TMEM205 1453.072
    GNL2 1452.202
    LOC646723 1451.384
    BCLAF1 1450.526
    PAPSS2 1449.722
    ROD1 1448.848
    C8orf59 1448.044
    CLNS1A 1447.207
    TAX1BP1 1446.445
    BNIP3L 1445.532
    NFKB1 1444.768
    LOC644934 1443.99
    ADAM15 1443.15
    PPP1CA 1442.281
    C17orf49 1441.421
    CGGBP1 1440.555
    AP3B1 1439.754
    ARPC4 1439.019
    TMEM87A 1438.202
    LOC440093 1437.389
    PRDX2 1436.503
    CENPB 1435.284
    RIOK3 1435.284
    PEPD 1434.051
    C7orf30 1433.277
    SF3B4 1432.503
    C7orf50 1431.741
    PAICS 1430.9
    DHRS7 1430.109
    SCHIP1 1429.311
    SMARCA4 1428.443
    LOC391833 1427.659
    CD151 1426.918
    C14orf112 1426.134
    CCT3 1425.327
    TSPAN17 1424.433
    HEBP1 1423.707
    PALMD 1422.87
    PSMA3 1422.059
    HCFC1R1 1420.912
    FAM96B 1420.912
    LOC728532 1419.695
    TRAPPC2L 1418.904
    BASP1 1418.075
    ZFYVE21 1416.821
    EDF1 1416.821
    TMEM43 1415.619
    KIAA1191 1414.827
    COMMD1 1414.003
    VEZF1 1413.213
    TMCO1 1412.417
    PSMD6 1411.674
    PAFAH1B3 1410.928
    C19orf43 1410.128
    CWC15 1409.341
    PHB2 1408.51
    FAM45A 1407.765
    SPTLC1 1407.045
    C2orf29 1406.196
    PGLS 1405.486
    GNPDA1 1404.659
    AIDA 1403.945
    FNBP1L 1403.256
    TCEAL8 1402.467
    WFS1 1401.669
    CYTSA 1400.85
    IFNGR2 1400.118
    MRPS24 1399.314
    SASH1 1398.612
    LOC728590 1397.854
    LSM5 1397.054
    NDUFB10 1396.327
    PTPRM 1395.602
    BIN1 1394.867
    MLLT11 1394.156
    KLHL5 1393.377
    CAMK2N1 1392.62
    IFI16 1391.929
    RAB2B 1391.157
    TSG101 1390.367
    ARHGAP21 1389.62
    TXNL2 1388.918
    EIF2B4 1388.222
    AKR7A2 1387.016
    PPP4C 1387.016
    MARCH7 1385.806
    EWSR1 1385.086
    MGEA5 1383.891
    JMJD8 1383.891
    TSPAN3 1382.76
    FAM62B 1381.996
    PLVAP 1381.302
    ATP1B1 1380.575
    LOC220433 1379.767
    KIAA1751 1378.533
    NOX4 1378.533
    BCAT1 1377.397
    TRAPPC5 1376.299
    DIABLO 1376.299
    ATG4A 1375.226
    EWSR1 1374.548
    LOC100130919 1373.854
    EIF6 1373.121
    SERF1B 1372.435
    C2orf25 1371.71
    ISCU 1370.6
    EMCN 1370.6
    NAE1 1369.468
    CBX2 1368.713
    GTF3A 1367.917
    FXYD5 1367.183
    RNU6-1 1366.377
    CIAO1 1365.645
    SF3A2 1364.954
    LOC729217 1364.264
    SMARCD1 1363.514
    YPEL5 1362.416
    MAT2B 1362.416
    CD9 1361.367
    CLK1 1360.635
    SHE 1359.905
    ARPC4 1359.19
    ENG 1358.525
    DPM1 1357.848
    RPS26L 1356.814
    SEC24C 1356.814
    WDFY1 1355.688
    ABCF1 1354.999
    SEPN1 1354.331
    CTPS2 1353.527
    JAK1 1352.825
    IGF2BP2 1352.016
    CALU 1351.325
    NSUN2 1350.603
    ETS2 1349.889
    PSMA4 1349.159
    NOTCH4 1348.37
    PPIL3 1347.714
    EBPL 1346.925
    UBE2A 1346.266
    TMEM14D 1345.427
    TSPO 1344.7
    UBE2E3 1344.022
    SNX17 1343.293
    AHCY 1342.54
    APH1A 1341.847
    CTTN 1341.154
    SH3GLB2 1340.142
    LOC646347 1340.142
    TUBB3 1339.084
    LSM4 1338.417
    MCM7 1337.661
    UBE2G2 1336.937
    CCL15 1336.199
    VPS37C 1335.507
    MRPL43 1334.784
    AKT1 1334.044
    TSC22D1 1333.376
    GLRX3 1332.713
    C3orf21 1332.065
    UFC1 1331.382
    DDEF2 1330.627
    HNRPDL 1329.926
    OAT 1329.173
    SAMM50 1328.509
    DDX42 1327.8
    TMEM189-UBE2V1 1327.136
    BRD9 1326.459
    TRABD 1325.811
    LYN 1325.106
    LOC100130516 1324.409
    UBAP2L 1323.751
    LYL1 1323.043
    NAT5 1322.032
    C19orf43 1322.032
    C22orf13 1320.994
    PAFAH1B1 1320.273
    HECW2 1319.607
    DDX39 1318.904
    NSMCE4A 1318.229
    NRP1 1317.531
    NLRP8 1316.868
    IFFO1 1316.188
    SERPINH1 1315.531
    TOMM20 1314.873
    SLC35B1 1314.205
    BMP6 1313.502
    ACTR10 1312.85
    AIMP2 1312.181
    PLRG1 1311.423
    TUBB6 1310.078
    SWAP70 1310.078
    LOC345041 1310.078
    PPA1 1308.715
    CUTA 1308.027
    MAGT1 1307.382
    LOC388621 1306.728
    TSPO 1306.076
    CKLF 1305.451
    ACTR1A 1304.807
    HSPA1B 1304.1
    DYNLRB1 1303.429
    LOC651149 1302.787
    TINF2 1302.134
    ACTL6A 1301.494
    CNIH4 1300.809
    NECAP2 1300.112
    AKT1 1299.402
    FLOT1 1298.767
    C6orf153 1298.085
    CUEDC2 1297.413
    AK90694 1296.433
    LOC728903 1296.433
    TXNRD1 1295.501
    AFAP1L1 1294.547
    CAPN11 1294.547
    UBE2L6 1293.604
    KIAA0355 1292.622
    LAPTM4B 1292.622
    RNU6-15 1291.31
    TGFB1I1 1291.31
    TNFRSF21 1290.303
    HNRNPAB 1289.261
    C14orf166 1289.261
    C3orf10 1288.225
    MAPK3 1287.591
    BUD31 1286.944
    CCDC50 1286.227
    DPM1 1285.552
    TSEN34 1284.894
    FAM32A 1284.298
    PTGR1 1283.698
    BTBD6 1283.024
    COQ5 1282.389
    DNAJA2 1281.734
    YTHDC1 1281.127
    CXCR4 1280.516
    SNCA 1279.908
    C19orf53 1279.337
    TMED10P 1278.664
    PIP5K2B 1278.034
    CTDSPL 1277.381
    CSE1L 1276.724
    LOC728973 1276.059
    ITM2A 1275.44
    SEPT15 1274.779
    DERA 1274.102
    THOC4 1273.114
    SNRPN 1273.114
    ATG12 1272.123
    SUPT16H 1271.505
    NINJ1 1270.853
    TRAF3IP2 1270.277
    LOC100132247 1269.647
    MMP1 1269.005
    GPN1 1268.349
    C16orf61 1267.703
    ZFP91 1267.065
    CLTA 1266.103
    RBM3 1266.103
    STK25 1265.098
    CD99L2 1264.439
    SEMA3E 1263.804
    MMRN1 1263.233
    FAM38A 1262.61
    CXXC5 1261.953
    FAM125A 1261.062
    COPE 1261.062
    CNRIP1 1260.09
    NDUFB6 1259.195
    AKR1A1 1259.195
    ATP2B4 1257.931
    SF3A3 1257.931
    ACSS2 1256.642
    SGSH 1256.642
    MRPL32 1255.638
    FBLN1 1254.976
    CKAP5 1254.373
    PPP1R15A 1253.721
    PCDHB2 1253.122
    FBXO21 1252.554
    TMEM183B 1251.959
    TYK2 1251.349
    EIF2B4 1250.723
    KIAA1310 1250.124
    UBE3C 1249.545
    ZNF207 1248.609
    TOMM22 1248.609
    AP2M1 1247.656
    RBM9 1247.018
    NAGK 1246.158
    SIVA 1246.158
    PGAM4 1245.267
    BRPF1 1244.707
    LOC653232 1244.138
    MRPL24 1243.487
    ITPRIPL2 1242.837
    RANBP1 1242.19
    PIR 1241.579
    NDUFS7 1241.024
    MRPL33 1240.402
    LOC731777 1239.449
    ACSL3 1239.449
    SYNCRIP 1238.21
    SCAMP1 1238.21
    HSPH1 1237.276
    SNORD13 1236.367
    ATP5C1 1236.367
    RNPS1 1235.512
    YRDC 1234.929
    FNBP4 1234.016
    SLC27A3 1234.016
    SNTB2 1233.115
    AK2 1232.525
    C9orf78 1231.887
    SHROOM4 1231.273
    CHMP5 1230.644
    KLHDC3 1229.734
    COL5A2 1229.734
    MKRN1 1228.856
    CLPTM1L 1228.232
    FZD4 1227.591
    AHCYL1 1226.997
    C11orf2 1226.102
    TCEA2 1226.102
    SERTAD2 1225.225
    ZNF581 1224.638
    TXNRD1 1224.021
    MRPS22 1223.439
    COPB2 1222.825
    EIF2B2 1222.166
    MPDZ 1221.614
    RABAC1 1220.997
    LRRFIP1 1220.45
    CCT7 1219.851
    LOC643336 1219.27
    EIF4E2 1218.658
    SNRNP70 1218.084
    UNC45A 1217.535
    EPRS 1216.981
    LOC653147 1216.359
    EIF4G2 1215.815
    CHIC2 1215.238
    RALY 1214.672
    COMMD4 1214.132
    HAGH 1213.555
    ATIC 1212.981
    SEMA6A 1212.362
    SDAD1 1211.776
    TMEM173 1211.176
    WDR61 1210.555
    UQCRC1 1209.94
    ERGIC3 1209.292
    C16orf58 1208.466
    FTHL12 1208.466
    TIGA1 1207.301
    ITGB5 1207.301
    ATP2B4 1206.435
    HSPC268 1205.868
    ACP5 1205.3
    CHST7 1204.734
    LOC728643 1204.165
    TMEM93 1203.577
    RNF5P1 1203.002
    IMMT 1202.47
    NOP56 1201.878
    STX5 1201.322
    TXNDC5 1200.716
    LOC100131905 1200.21
    PLEKHM2 1199.673
    LSM7 1199.112
    SPRY1 1198.584
    C19orf60 1198.021
    LSM14A 1197.449
    SRP54 1196.849
    AMZ2 1196.268
    FKBP9L 1195.389
    RAB8A 1195.389
    SPSB3 1194.239
    GIPC1 1194.239
    SLC29A1 1193.425
    MRPL3 1192.832
    CNIH 1192.266
    FAM127B 1191.741
    ATP6V0B 1191.148
    ATP1B3 1190.628
    IDH3B 1190.099
    TFDP1 1189.245
    TECR 1189.245
    GAR1 1188.402
    CDR2L 1187.799
    KIAA1147 1187.189
    IGFBP7 1186.642
    HRASLS3 1186.117
    PFN2 1185.528
    RPL7L1 1184.957
    TDP1 1184.364
    RASA1 1183.793
    BMS1 1183.25
    DRAP1 1182.717
    POLE3 1182.185
    NARF 1181.617
    EBNA1BP2 1181.081
    LOC644563 1180.512
    HECTD1 1179.933
    ATG4A 1179.092
    IRAK1 1179.092
    CCDC92 1178.273
    SNRPA1 1177.46
    CAPZB 1177.46
    SCAMP3 1176.606
    LOC642975 1176.084
    CNIH 1175.454
    TRAPPC4 1174.829
    NISCH 1174.275
    ADARB1 1173.705
    ECHS1 1173.163
    GSN 1172.576
    GOLGA3 1171.965
    TMEM183A 1171.472
    PREI3 1170.618
    COPZ1 1170.618
    RNF149 1169.794
    PRKRIR 1169.219
    KLHL9 1168.706
    RPL9 1168.134
    ANKRD9 1167.583
    MRPL14 1167.011
    CCBE1 1166.449
    VBP1 1165.62
    LAMB1 1165.62
    C12orf10 1164.491
    MRPS10 1164.491
    TWSG1 1163.691
    LOC100132585 1163.139
    MED6 1162.643
    GAK 1162.143
    HPS6 1161.618
    SOX4 1161.072
    CLSTN1 1160.531
    TAF1C 1159.974
    LIMS1 1159.434
    TRIM44 1158.63
    TNPO2 1158.63
    CHMP1B 1157.806
    ATP5G1 1157.298
    TUBG1 1156.749
    NDUFV1 1156.212
    MAP2K1 1155.693
    NOTCH1 1155.126
    UBE2F 1154.596
    POLR2I 1154.025
    RSL1D1 1153.5
    MRPL21 1152.919
    LOC648695 1152.101
    POLR2J3 1152.101
    DNAJB6 1151.286
    TMEM131 1150.796
    CHMP5 1150.222
    UNC84A 1149.634
    FAM84B 1149.095
    SOX7 1148.576
    NEDD8 1148.045
    CRELD2 1147.203
    EEF2K 1147.203
    SH2D3C 1146.37
    ACSS2 1145.794
    CNIH 1145.304
    RPL13 1144.794
    ARF4 1144.238
    ASAP2 1143.712
    HCFC1 1143.163
    SLC41A3 1142.617
    ARID1A 1142.087
    MRPL54 1141.562
    SNRPB2 1140.987
    PAIP2 1140.448
    ULK1 1139.961
    CALD1 1139.447
    MAPBPIP 1138.939
    HARS 1138.45
    HCLS1 1137.98
    SFRS17A 1137.448
    ASH2L 1136.926
    LOC441131 1136.194
    AIF1L 1136.194
    METAP1 1135.416
    TTC37 1134.883
    RNASET2 1134.356
    CARD10 1133.855
    ATP5SL 1133.309
    CTSC 1132.787
    GDPD5 1132.295
    C5orf15 1131.76
    C1orf123 1131.216
    MED28 1130.738
    ADD3 1130.209
    HES4 1129.346
    VPS28 1129.346
    SIRPA 1128.332
    PPP1R16B 1128.332
    ATP1A1 1127.569
    TOP2B 1127.102
    CLDN14 1126.581
    BRIX1 1126.127
    GLRX 1125.649
    PHRF1 1125.135
    ANXA7 1124.618
    PEX11B 1124.07
    LOC390466 1123.317
    FTHL8 1123.317
    RBM4 1122.305
    ACO1 1122.305
    FAF2 1121.286
    ZSCAN18 1121.286
    USO1 1120.278
    PDCD4 1120.278
    BOLA3 1119.499
    GIMAP6 1118.759
    EXOSC1 1118.759
    TNPO1 1117.953
    MRPL21 1117.431
    ETF1 1116.964
    TMEM109 1116.22
    PICALM 1116.22
    PPP2R5E 1115.468
    DHX15 1115.038
    RANGAP1 1114.519
    NUAK1 1114.015
    RAPGEF1 1113.275
    C1orf43 1113.275
    FAM38B 1112.526
    ATP1B3 1111.986
    AW276479 1111.51
    CNPY2 1111.025
    CORO1B 1110.552
    AV737943 1110.036
    ARL6IP6 1109.565
    SRF 1109.065
    GALNT11 1108.587
    DIMT1L 1108.078
    SEC14L1 1107.615
    PTS 1107.107
    PHF5A 1106.657
    NIPA2 1106.208
    LOC728564 1105.495
    LOC728666 1105.495
    CCM2 1104.829
    PLDN 1104.368
    TMEM189 1103.863
    LOC647302 1103.405
    ACOT9 1102.912
    EHD1 1102.392
    TMEM88 1101.651
    RIOK3 1101.651
    CHMP2A 1100.961
    LOC729495 1100.251
    LOC100129211 1100.251
    C11orf74 1099.23
    DHRS4 1099.23
    ALDH7A1 1098.483
    RYBP 1098.015
    CISD1 1097.296
    NCBP2 1097.296
    PLCG1 1096.586
    FBXW11 1096.097
    LAMP2 1095.627
    C11orf67 1095.123
    TXLNA 1094.675
    ST13 1094.182
    ASNSD1 1093.703
    THAP11 1093.181
    SCYL1 1092.677
    C20orf20 1092.201
    ANKRD11 1091.664
    KIAA0494 1090.984
    ATP6V1D 1090.984
    MAGED1 1090.273
    TIA1 1089.785
    HPRT1 1089.321
    C1orf128 1088.849
    STIP1 1088.359
    LAPTM4B 1087.642
    MED16 1087.642
    SLC16A3 1086.895
    EFCAB4A 1086.425
    ERAL1 1085.952
    AKR1B1 1085.496
    GLIPR2 1085.028
    SNRPC 1084.522
    SLC41A3 1084.067
    C12orf35 1083.54
    SMTN 1083.064
    SCAP 1082.62
    UBAC1 1082.171
    CLINT1 1081.704
    TNFRSF25 1081.222
    MRPL45 1080.789
    C4orf32 1080.313
    LOC100128196 1079.856
    CHFR 1079.43
    FUBP3 1078.945
    FLJ35390 1078.417
    ARSD 1077.77
    IMPDH1 1077.77
    TSPAN6 1077.097
    GLB1 1076.624
    SFRS2IP 1076.142
    RHOJ 1075.704
    TIMM22 1075.007
    ARAP3 1075.007
    MYC 1074.047
    PRICKLE1 1074.047
    LOC728620 1073.366
    ZNF467 1072.903
    FAM160B1 1072.44
    PRPF31 1071.962
    LOC100129742 1071.49
    FOXO1 1071.02
    P4HA2 1070.563
    C19orf2 1070.108
    XLKD1 1069.626
    NOS3 1069.22
    FXYD5 1068.745
    RAB32 1068.311
    CPNE1 1067.883
    ARPC1B 1067.446
    LOC729406 1067.012
    LSM3 1066.554
    MYOF 1066.106
    POGK 1065.674
    SFRS2B 1065.215
    GPR137 1064.777
    FAM189B 1064.313
    TOMM40 1063.822
    CRK 1063.404
    DSTN 1062.951
    UGP2 1062.271
    ORMDL1 1062.271
    LOC653566 1061.591
    RBMX 1061.134
    ASAP1 1060.679
    CDC25B 1059.996
    UTP11L 1059.996
    U2AF2 1059.358
    ARF5 1058.938
    ERCC1 1058.498
    VGLL4 1058.016
    CREB3L2 1057.327
    NSL1 1057.327
    AKR1A1 1056.706
    NUDT5 1056.246
    UBQLN1 1055.836
    VPS41 1055.356
    PDIA6 1054.718
    LOC100133516 1054.718
    PELO 1054.057
    LACTB 1053.587
    XRCC2 1053.146
    HIGD1A 1052.714
    SEC22C 1052.285
    CARD8 1051.832
    MAP1B 1051.378
    DRG1 1050.899
    STMN1 1050.489
    LOC440345 1050.027
    DCAF7 1049.575
    BOLA3 1049.18
    APRT 1048.483
    ZDHHC9 1048.483
    SFT2D1 1047.81
    ZNF207 1047.343
    FLJ36131 1046.928
    C6orf125 1046.454
    YIPF3 1045.998
    LOC729992 1045.539
    IGF2BP3 1045.127
    TM9SF2 1044.687
    DCTN6 1044.225
    FXR1 1043.764
    RPL34 1043.313
    AP2M1 1042.912
    LOC644330 1042.479
    TXNDC12 1042.033
    BEX1 1041.572
    PGM1 1041.145
    NRBP2 1040.679
    IRF2BP2 1040.249
    ITFG1 1039.802
    MRPL20 1039.355
    MRPS17 1038.91
    FAM3A 1038.477
    MAN2B2 1037.795
    S100A13 1037.795
    PTPN11 1037.141
    NPEPL1 1036.503
    DPAGT1 1036.503
    STUB1 1035.88
    CDK5RAP3 1035.203
    LOC100128266 1035.203
    LRRC41 1034.576
    RPS21 1034.106
    PIAS4 1033.669
    CHP 1033.24
    CPSF4 1032.79
    FRMD4A 1032.13
    CDK10 1032.13
    LOC650157 1031.44
    LOC100132717 1030.745
    G6PD 1030.745
    UROS 1030.116
    BC035081 1029.698
    LOC730255 1029.265
    ENPP2 1028.837
    CNN2 1028.433
    OSBPL9 1027.99
    TRPT1 1027.561
    RN7SK 1027.133
    COPS7A 1026.742
    NHP2 1026.284
    PAPSS1 1025.863
    MACF1 1025.215
    ACOT7 1025.215
    SERINC3 1024.519
    LAMA4 1024.067
    MRPS15 1023.652
    TM9SF4 1023.053
    ACAT2 1023.053
    LOC645166 1022.43
    NCOA7 1022.016
    TBC1D4 1021.59
    RHOQ 1021.206
    FAM39DP 1020.8
    TNFRSF1A 1020.394
    FKBP5 1019.963
    FAM120B 1019.554
    LCMT1 1019.166
    CCDC59 1018.55
    AK022936 1018.55
    RPUSD4 1017.934
    IGFBP3 1017.538
    SLC35E1 1017.154
    CCDC125 1016.336
    RIN2 1016.336
    MBTPS1 1016.336
    TMEM126B 1015.509
    GPR177 1015.087
    LOC728661 1014.67
    XPO1 1014.219
    ATG4B 1013.813
    DAP3 1013.397
    CISD1 1012.933
    STK19 1012.524
    AES 1011.867
    NDUFA13 1011.867
    NDRG4 1011.251
    FIBP 1010.835
    VHL 1010.439
    RNF38 1009.799
    PRMT1 1009.799
    VPS4B 1009.162
    SHMT2 1008.756
    MRPL34 1008.353
    OCIAD2 1007.943
    PSMD1 1007.326
    HSD17B4 1007.326
    VBP1 1006.484
    MCRS1 1006.484
    TNFAIP1 1005.872
    TNRC6B 1005.446
    COASY 1005.033
    ST3GAL1 1004.617
    RHBDD2 1003.993
    SURF4 1003.993
    KLHDC8B 1003.384
    TSPAN4 1002.961
    KDM5B 1002.552
    STK4 1002.151
    LPHN2 1001.764
    POLR2A 1001.35
    CD59 1000.938
    DNAL4 1000.492
    RHBDF2 1000.062
  • The 30-MV2-6 cells were maintained in EGM-MV2 media (PromoCell) plus TGFβ inhibitor (SB43154) (Cayman Chemical Co., Ann Arbor, Mich.) in a 5% CO2, 5% O2 humidified cell culture incubator. The 30-MV2-6 cells were seeded at a density of 40 k/cm2. The culture media was removed and after two washes with phosphate buffered saline (PBS), (PBS) was added at 0.1 ml/cm2 to produce conditioned medium from which exosomes were isolated. Alternatively, basal EGM-MV2 medium (PromoCell, Heidelberg, Germany) without fetal calf serum or growth factor additives was substituted for PBS. The media was conditioned by the cells in a humidified tissue culture incubator for 16 hours at 37° C. at 5% CO2 and 1% O2. The conditioned medium was collected and 0.5 volumes of Total Exosome Isolation Reagent (Life Technologies) was added and mixed well by vortexing until there was a homogenous solution. Alternatively, a solution of 15% polyethylene glycol (Hampton Research, Aliso Viejo, Calif.), 1.5 M NaCl (Sigma, St Louis, Mo.) was substituted for the Total Exosome Isolation Reagent. The sample was incubated at 4° C. for at least 16 hours to precipitate the exosomes, followed by centrifugation at 10,000×g for 1 hour at 4° C. The supernatant was removed and the pellet is resuspended in 0.01 volume of PBS.
  • Exosome particle size and concentration were measured using nanoparticle tracking analysis (NTA; Nanosight, Malvern Instrument, Ltd, Malvern Worcestershire, UK) and by ELISA. The experiments were repeated using commercially available HT1080 cells (ATCC) as a comparison. HT1080 cells are a human fibrosarcoma cell line known to form exosomes with vesicle forming ability (see, e.g. Kim et al. (2002) Cancer Res. 62:6312).
  • The results of the Nanosight NTA (triplicates) for exosome preparations derived from 30-MV2-6 and HT1080 cells (ATCC, Manassas, Va.) are shown in FIG. 1 . The results indicate that particles prepared from 30-MV2-6 are from 80 to 110 nm with predominant peak at 88 nm+/−2.9 nm. The particles prepared from a HT1080 human fibrosarcoma cells were larger by comparison with a mode of 120 nm+/−7.4 nm. The concentration of exosomes bearing the exosome marker CD63 was measured by ELISA, using samples of known concentrations of HT1080 exosomes as a standard curve. Samples were adsorbed to the ELISA plate by incubation overnight in PBS. The PBS was removed and wells were washed 3 times in ELISA wash buffer (Thermo Scientific, Waltham, Mass.) followed by incubation with primary anti-CD63 antibody (BD Pharmingen, Franklin Lakes, N.J.) for 1 hour at room temperature. The primary antibody was removed followed by washing 3 times in wash buffer and incubation with secondary antibody (HRP conjugated anti-mouse) (Invitrogen, Grand Island, N.Y.)) at 1:3000 dilution for 1 hour at room temperature. The wells were washed 3 additional times with wash buffer and incubated in Super sensitive TMB ELISA substrate (Sigma, St Louis, Mo.) for 0.5 hour followed by addition of ELISA stop solution (InVitrogen, Grand Island, N.Y.). The concentration of exosomes was determined by reading optical density in a standard plate reader at wavelength of 450 nm.
  • Example 2: Angiogenic Activity of Exosomes Prepared from a Human Embryonic Progenitor Cell Line
  • Angiogenic activity of exosomes was assayed using an in-vitro endothelial tube forming assay. The assay was performed in triplicate in a μ well slide (Ibidi, Verona, Wis.) or in single wells of a 96-well plate. The wells were coated with reduced growth factor Matrigel (BD, Franklin Lakes, N.J.). Human umbilical cord vascular endothelial cells (HUVEC) that were grown to 70-80% confluence were plated at 5000-7000 cells per well in a μ well slide in 50 ul of EGM-MV2 basal medium (Promocell, Heidelberg, Germany) (no supplements) containing up to 10 μl of exosomes in PBS or equivalent volume of PBS without exosomes as a negative control or in 50 μl of complete EGM-MV2 medium with growth factor supplements as a positive control. Alternatively, the assay was performed in a 96-well plate using 60,000 to 90,000 cells per well in 280 μl of medium and 20 μl of exosomes or PBS. The cells are incubated at 37° C. in a 5% CO2 incubator for 16-18 hours. The cells were photographed under phase contrast at low power or stained with calcein and photographed using a fluorescence microscope. The images were scored for cell covered area, total tube length, number of branch points, and number of loops using Wimasis image analysis (Ibidi, Verona, Wis.). At least 3 random images were quantified per well.
  • FIG. 2A shows an increase in HUVEC endothelial tube formation when grown in the presence of 30-MV2-6 derived exosomes (in PBS) compared to basal medium with an equivalent amount of PBS (with no exosomes) added (negative control). The quantified results (FIG. 2B) indicate that total tube length, cell covered area, branch points and the number of loops were all increased by the addition of exosomes compared to basal medium indicating that the 30-MV2-6 exosomes are angiogenic.
  • Angiogenic activity of exosomes was also assessed by their ability to stimulate in vitro tube formation using human embryonic stem (hES) cell derived perivascular embryonic progenitor cells (PEPCs) (also called 017-PC-A) cells bearing pericyte and stemness markers (CD146, CD133, Podoplanin)(U.S. patent application Ser. No. 14/625,621, filed on Feb. 18, 2015). The assay was performed as described for HUVECs except that hES cell derived PEPCs were used instead of HUVECs. The assay was performed in triplicate using μ well slides (Ibidi, Verona, Wis.). The hES pericytes that were grown in defined medium differ from HUVECs in their response to complete medium. HUVECs respond to complete EGM-MV2 medium in the tube forming assay with robust tube formation (FIG. 2A). In contrast, hES PEPCs migrate to form foci consisting of cellular aggregates (FIG. 3A) when grown on Matrigel in complete EGM-MV2 medium and thus exhibited reduced tube formation. The hES PEPCs grown in defined medium form incomplete tubes (FIG. 3B) similar to HUVEC in basal medium (FIG. 2A). Like HUVECs, the hES PEPCs grown in the presence of 30-MV2-6 exosomes displayed an increase in tube formation as shown in FIG. 3C-3E. The tube formation was dose responsive and quantitative analysis indicated an increase in all 4 tube formation parameters (FIG. 3F-3I). Unlike HUVECs which respond to both exosomes and complete medium with increased tube formation, hES derived PEPCs respond to exosomes with increased tube formation but respond to complete medium with reduced tube formation compared to basal medium. Thus, 30-MV2-6 exosomes induce angiogenesis in a non-angiogenic cell type that does not respond to the angiogenic factors present in EGM-MV2 complete medium. These data indicate that 30-MV2-6 derived exosomes do not simply mimic the factors in complete medium but instead are capable of stimulating hES derived PEPC tube formation by a mechanism that is distinct from the action of complete medium on these cells.
  • Example 3: Comparison of Angiogenic Activity of Exosomes Derived from a Human Embryonic Progenitor Cell Line and Exosomes Derived from Adult Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs)
  • Exosomes were prepared from an embryonic stem cell derived PureStem® cell line, 30-MV2-6, and from adult bone marrow-derived mesenchymal stem cells (BM-MSCs) from two different commercial sources (Lonza and Promocell), according to methods described in Example 1. The angiogenic activity was assessed using the in vitro endothelial tube formation assay described in Example 2. Briefly, the exosomes (2×108 particles/50 μl) were incubated with human umbilical cord vascular endothelial (HUVEC) cells for 12-16 hours on low growth factor Matrigel using a μ-well slide (Ibidi, Verona, Wis.). Tube length was assessed by image capture and analyzed using Angiogenesis Analyzer in ImageJ (http://rsb.info.nih.gov/ij/) image processing program. Total tube length formed per image/μ-well was calculated relative to total tube length formed for HUVEC in complete EGM-MV2 medium (CM) containing angiogenic growth factors (VEGF and FGF2) and fetal bovine serum (FBS).
  • Exosomes derived from the 30-MV2-6 cell line were compared to early passage BM-MSCs from the two different sources, Promocell (Heidelberg, Germany) (FIG. 4 , panel A) and from Lonza (Basel, Switzerland) (FIG. 4 , panel B). In both cases the angiogenic activity of 30-MV2-6 derived exosomes was greater than the angiogenic activity of BM-MSC derived exosomes. The total tube length of HUVECs incubated in basal medium with 30-MV2-6 derived exosomes (in PBS) was similar to HUVECs incubated in complete EGM-MV2 medium and significantly greater than BM-MSC derived exosomes (from either source) or HUVEC incubated in basal EGM-MV2 medium and PBS alone. The total tube length resulting from BM-MSC derived exosomes was on average slightly higher than PBS in basal medium (BM) but the difference was not statistically significant (p<0.05). These results indicate that embryonic endothelial progenitor stem cell line derived exosomes are advantageous over adult BM-MSC derived exosomes for inducing angiogenic activity in endothelial cells.
  • BM-MSC and 30-MV2-6 derived exosomes were further compared in a dose response experiment to determine differences in their potency. Exosomes were prepared and their concentration determined using nanoparticle tracking analysis (NTA; Nanosight, Malvern Instruments Ltd, Malvern, Worcestershire, UK). ELISA was used to confirm the presence of transpanins CD63, CD81 and CD9 that are typically expressed on exosomes. The 30-MV2-6 exosomes were tested at doses ranging from 50 million to 400 million exosomes per well. The BM-MSC exosomes were tested at doses ranging from 400 to 1200 million exosomes per well, because no significant activity was observed at 200 million exosomes per well. The results, shown in FIG. 5 indicate that the angiogenic response of HUVEC cells to 30-MV2-6 exosomes is dose responsive starting at 50 million per well and saturating at doses of ≥200 million exosomes per well (FIG. 5 , panel A). In contrast, the BM-MSC exosomes showed a dose response of increasing angiogenic activity at doses from 400 million to 1200 million exosomes per well (FIG. 5 , panel B). The potency of 30-MV2-6 derived exosomes was at least 6-fold greater than that of BM-MSC derived exosomes, having the equivalent activity at 200 million exosomes per well as BM-MSC derived exosomes at 1200 million exosomes per well (FIG. 5 , panel B).
  • Example 4: Comparison of miRNA Content in Exosomes Derived from a Human Embryonic Progenitor Cell Line Versus Exosomes Derived from Adult BM-MSCs
  • The miRNA content of the 30-MV2-6 derived exosomes was analyzed and compared to the miRNA content of the less angiogenic BM-MSC exosomes. RNA was extracted and purified from exosomes using the miRNeasy mini kit according to the manufacturer's recommended protocol (Qiagen, Hilden, Germany). The exosome RNA was quantified using a Nanodrop spectrophotometer and cDNA was prepared from the RNA using the miScript II RT kit (Qiagen, Hilden, Germany) according to the manufacturer's recommended protocol. The exosome cDNA was amplified by polymerase chain reaction (PCR) using the miScript pre-AMP PCR kit (Qiagen) and miScript pre-AMP pathway primer mix (human miFinder MBHS-001Z; Qiagen) according to the manufacturer's recommended protocol. Relative miRNA levels were assessed for 84 human miRNAs by quantitative PCR using the human miFinder miScript miRNA PCR array (#331221; Qiagen) according to the manufacturer's recommended protocol. The results were analyzed using the ΔΔCT method of relative quantitation available at (http://perdataanalysis.sabiosciences.com/mirna).
  • There were substantial differences in the miRNA content of the 30-MV2-6 derived exosomes and BM-MSC derived exosomes. The miRNAs with greater than 6-fold difference between 30-MV2-6 exosomes and BM-MSC exosomes are shown in FIG. 6A scatter plot. Table 2 lists miRNAs that are more than 2-fold overexpressed in 30-MV2-6 exosomes relative to BM-MSC exosomes and Table 3 lists miRNAs that are more than 2-fold underexpressed in 30-MV2-6 exosomes relative to BM-MSC exosomes.
  • TABLE 2
    miRNAs overexpressed in 30-MV2-6 exosomes
    compared to BM-MSC exosomes
    Fold
    miRNA Mature ID Difference
    hsa-miR-155-5p 3.98
    hsa-miR-18a-5p 2.54
    hsa-miR-374a-5p 2.69
    hsa-miR-126-3p 77.60
  • TABLE 3
    miRNAs underexpressed in 30-MV2-6 exosomes
    compared to BM-MSC exosomes
    Fold
    miRNA Mature ID Difference
    hsa-miR-142-5p −56.29
    hsa-miR-9-5p −14.69
    hsa-miR-27b-3p −6.92
    hsa-miR-101-3p −4.82
    hsa-let-7d-5p −3.12
    hsa-miR-16-5p −10.56
    hsa-let-7g-5p −7.60
    hsa-miR-30c-5p −3.26
    hsa-miR-96-5p −11.14
    hsa-miR-185-5p −3.44
    hsa-miR-142-3p −17.38
    hsa-miR-24-3p −9.55
    hsa-miR-181b-5p −2.03
    hsa-miR-302b-3p −42.52
    hsa-miR-30b-5p −3.34
    hsa-miR-21-5p −16.16
    hsa-miR-15b-5p −4.08
    hsa-miR-223-3p −18.36
    hsa-miR-194-5p −2.69
    hsa-miR-15a-5p −4.25
    hsa-miR-125b-5p −38.36
    hsa-miR-99a-5p −10.43
    hsa-miR-29b-3p −15.10
    hsa-miR-29a-3p −35.08
    hsa-miR-141-3p −4.30
    hsa-let-7a-5p −4.03
    hsa-miR-124-3p −13.56
    hsa-miR-92a-3p −2.28
    hsa-miR-23a-3p −5.74
    hsa-miR-25-3p −3.16
    hsa-let-7e-5p −2.35
    hsa-miR-376c-3p −752.81
    hsa-miR-144-3p −57.26
    hsa-miR-195-5p −10.03
    hsa-miR-143-3p −82.38
    hsa-miR-191-5p −4.88
    hsa-let-7i-5p −9.62
    hsa-miR-302a-3p −17.39
    hsa-miR-222-3p −2.31
    hsa-let-7b-5p −35.56
    hsa-miR-186-5p −3.40
    hsa-miR-196b-5p −71.33
    hsa-miR-27a-3p −3.42
    hsa-miR-22-3p −4.58
    hsa-miR-130a-3p −2.68
    hsa-let-7c-5p −10.92
    hsa-miR-29c-3p −24.99
    hsa-miR-140-3p −2.98
    hsa-miR-128-3p −2.77
    hsa-let-7f-5p −2.89
    hsa-miR-122-5p −9.20
    hsa-miR-100-5p −10.00
    hsa-miR-302c-3p −144.85
  • The miRNA with the highest relative expression in 30-MV2-6 exosomes compared to BM-MSC exosomes is miR-126-3p (77.6-fold difference, Table 2). MiR-126 is a known angiogenic miRNA (“angiomiR”) that is endothelial cell-specific and has been shown to regulate both vascular integrity and developmental angiogenesis. Fish et al. (2008) Dev. Cell 15(2):272; Zou et al. (2011) Circ. Res. 108 (2):201; Jakob and Landmesser (2012) Cardiovasc. Res. 93(4):614; Nicoli et al. (2010) Nature 464(7292): 1196. Induction of miR-126 in endothelial cells and transport of miR-126 via exosomes has been shown to be important for the effective treatment of myocardial infarction (MI) using transplanted cardiosphere derived cells in a mouse model. Ong et al. (2014) Circulation 130 (11 Suppl 1):S60.
  • Accordingly, the clonal human embryonic progenitor cell line derived, miR-126-containing exosomes of the instant invention can be used in in vitro angiogenesis studies as well as in treatment of myocardial infarction and other ischemic conditions, either by themselves or in combination with transplanted cells.
  • RNA from 30-MV2-6 exosomes was also used to compare the miRNA content of angiogenic versus non-angiogenic exosomes. Exosomes derived from HT1080 cells (a human sarcoma cell line) are not angiogenic in the HUVEC in vitro angiogenesis assay at 2.0×108 exosomes, a dose at which 30-MV2-6 exosomes show maximum angiogenic activity. HT1080 exosome RNA was analyzed on the miFinder miScript PCR array of 84 human miRNAs as described above and compared to 30-MV2-6 and BM-MSC exosome RNA (Table 4).
  • TABLE 4
    Exosomal miRNAs in BM-MSC and 30-
    MV2-6 Relative to HT1080 exosomes
    BM-MSC/ 30-MV2-6/
    HT1080 HT1080
    Fold Fold
    miRNA Mature ID Difference Difference
    hsa-miR-142-5p 13.3253 0.2367
    hsa-miR-9-5p 19.9437 1.3579
    hsa-miR-150-5p 4.3783 3.3457
    hsa-miR-27b-3p 100.911 14.5862
    hsa-miR-101-3p 14.0298 2.9096
    hsa-let-7d-5p 26.3588 8.441
    hsa-miR-103a-3p 4.5649 5.7031
    hsa-miR-16-5p 38.0552 3.605
    hsa-miR-26a-5p 41.3044 24.3186
    hsa-miR-32-5p 15.9192 14.3894
    hsa-miR-26b-5p 52.2677 31.9112
    hsa-let-7g-5p 76.0374 10.0027
    hsa-miR-30c-5p 17.5335 5.3714
    hsa-miR-96-5p 1.1148 0.1
    hsa-miR-185-5p 29.375 8.5466
    hsa-miR-142-3p 0.5468 0.0315
    hsa-miR-24-3p 40.061 4.1927
    hsa-miR-155-5p 1.0346 4.118
    hsa-miR-146a-5p 1.4501 1.0803
    hsa-miR-425-5p 3.3003 2.8192
    hsa-miR-181b-5p 16.2276 8.0058
    hsa-miR-302b-3p 45.0196 1.0589
    hsa-miR-30b-5p 19.9194 5.9589
    hsa-miR-21-5p 52.4899 3.2475
    hsa-miR-30e-5p 4.4078 3.3143
    hsa-miR-200c-3p 2.2547 1.3309
    hsa-miR-15b-5p 22.3822 5.4881
    hsa-miR-223-3p 202.2804 11.0146
    hsa-miR-194-5p 8.5348 3.1718
    hsa-miR-210-3p 1.8074 0.957
    hsa-miR-15a-5p 17.2702 4.0614
    hsa-miR-181a-5p 28.6581 16.7792
    hsa-miR-125b-5p 31.2942 0.8158
    hsa-miR-99a-5p 13.0061 1.2466
    hsa-miR-28-5p 11.1092 8.1182
    hsa-miR-320a 17.2154 13.7088
    hsa-miR-125a-5p 13.0238 8.8833
    hsa-miR-29b-3p 9.9121 0.6563
    hsa-miR-29a-3p 51.1831 1.4592
    hsa-miR-141-3p 7.9244 1.841
    hsa-miR-19a-3p 4.5159 3.3341
    hsa-miR-18a-5p 7.7906 19.7947
    hsa-miR-374a-5p 45.91 123.3183
    hsa-miR-423-5p 20.4698 18.6725
    hsa-let-7a-5p 25.3493 6.294
    hsa-miR-124-3p 20.2226 1.4916
    hsa-miR-92a-3p 14.4072 6.3169
    hsa-miR-23a-3p 52.5584 9.1492
    hsa-miR-25-3p 23.7331 7.5106
    hsa-let-7e-5p 22.9181 9.7439
    hsa-miR-376c-3p 2411.7739 3.2037
    hsa-miR-126-3p 123.9456 9618.223
    hsa-miR-144-3p 114.87 2.0061
    hsa-miR-424-5p 57.34 92.8881
    hsa-miR-30a-5p 3.966 3.4279
    hsa-miR-23b-3p 19.0703 13.0079
    hsa-miR-151a-5p 12.8604 25.4071
    hsa-miR-195-5p 40.2617 4.0149
    hsa-miR-143-3p 273.1733 3.316
    hsa-miR-30d-5p 3.9436 4.0508
    hsa-miR-191-5p 21.2256 4.3493
    hsa-let-7i-5p 62.9552 6.545
    hsa-miR-302a-3p 37.209 2.14
    hsa-miR-222-3p 22.8182 9.8699
    hsa-let-7b-5p 129.1842 3.6327
    hsa-miR-19b-3p 5.758 3.3451
    hsa-miR-17-5p 4.6186 7.3351
    hsa-miR-93-5p 16.2937 16.7172
    hsa-miR-186-5p 14.8015 4.3526
    hsa-miR-196b-5p 15.3129 0.2147
    hsa-miR-27a-3p 48.4243 14.148
    hsa-miR-22-3p 13.8229 3.0157
    hsa-miR-130a-3p 11.5632 4.3074
    hsa-let-7c-5p 93.5201 8.5615
    hsa-miR-29c-3p 39.8245 1.5935
    hsa-miR-140-3p 9.8899 3.3169
    hsa-miR-128-3p 21.7273 7.8472
    hsa-let-7f-5p 30.5392 10.5782
    hsa-miR-122-5p 71.7561 7.8014
    hsa-miR-20a-5p 4.6119 9.1362
    hsa-miR-106b-5p 5.093 8.4218
    hsa-miR-7-5p 4.8173 6.4945
    hsa-miR-100-5p 14.7509 1.4748
    hsa-miR-302c-3p 120.5214 0.832
  • The data in Table 4 and FIG. 6B illustrate the dramatic differences in the miRNA content of the three types of exosomes. These data show that HT1080 exosomes contain the lowest amount of miRNA for all miRNAs tested, except for miR-96-5p and miR-142-3p, which are lowest in 30-MV2-6 exosomes. The miRNA miR-142-3p is expressed at highest levels in HT1080 exosomes and is known to repress several inhibitors of oncogenic transformation. It is mimicked by Kaposi sarcoma viral miRNA, miR-K10a. Forte et al. (2015) J Virol 89(4): 2333. The miR-96-5p miRNA is present in highest levels in BM-MSC exosomes and is higher in HT1080 exosomes than 30-MV2-6 exosomes. This miRNA is thought to be involved in osteogenic and adipogenic differentiation in BM-MSCs (Laine et al. (2012) J Cell Biochem. 113(8):2687) but is also involved in tumor cell proliferation (Lin et al. (2010) Plos One 5(12):e15797; Haflidadottir et al. (2013) Plos One 8(8):e72400). Strikingly the miRNA with the highest levels in 30-MV2-6 exosomes relative to HT1080 exosomes is miR-126-3p. This known angiogenic miRNA is present at a 9618-fold higher level in 30-MV2-6 exosomes than HT1080 exosomes. MiR-155 is 4-fold higher in 30-MV2-6 exosomes than BM-MSC or HT1080 exosomes and is anti-angiogenic but pro-arteriogenic. Pankrtaz et al. (2015) Circulation 131(18):1575. Of the 9 remaining miRNAs that are highest in 30-MV2-6 exosomes, 6 are known to be involved in angiogenesis. The miRNAs miR-18a-5p, miR-20a-5p, miR-424-5p, miR-17-5p, and miR-7-5p miRNAs have anti-angiogenic activity. However, none of these anti-angiogenic miRNAs are more than 2.5-fold enriched in 30-MV2-6 exosomes compared to BM-MSC exosomes. The pro-angiogenic miR-106b is 9-fold enriched in 30-MV2-6 but only 4-fold enriched in BM-MSC exosomes compared to HT1080 exosomes. It is needed for neovascularization after hind limb ischemia. Semo et al. (2014) Eur Heart J. 35(45):3212.
  • Notably, several anti-angiogenic miRNAs, including miR-143-3p (Climent et al. (2015) Circ Res. 116(11):1753), miR-223-3p (Dai et al. (2014) Plos One 9(10):e108468), miR-222-3p (Suarez and Sessa (2009) Circ Res. 104(4):442), miR-15a, miR-15b and miR-16 (Spinetti et al. (2013) Circ Res. 112(2):335; Liu et al. (2012) Cell Physiol Biochem. 29(5-6):851)) were enriched for and/or present at highest level in the BM-MSC-derived exosomes.
  • Example 5: Comparison of Angiogenic Activity of Exosomes Derived from Various Clonal Embryonic Stem Cell Lines and Exosomes Derived from the Parental Pluriopotent Stem Cell Lines
  • Clonal embryonic progenitor cell lines were previously established from human pluripotent stem (hPS) cell lines using methods previously described. West et al. (2008) Regen Med. 3(3):287. The resulting cell lines are not immortalized but have higher replicative potential than primary cell lines because of their long telomere length that is near that of the parental hPS cell line from which they are derived. A wide diversity of cell types was produced by exposing hPS cells to an array of cell culture medium, cell matrix, and growth conditions followed by selective pressure for clonal growth and scalability. Over 140 such cell types have been determined to be distinct by analysis of total transcribed RNA using standard Illumina microarrays. The in vitro angiogenesis assay (described in detail in Example 2) was used to screen clonal embryonic progenitor cells for production of angiogenic exosomes. As shown in Table 5, most embryonic endothelial progenitor cell-derived exosomes have angiogenic activity in the range of 30-MV2-6 derived exosomes (+; relative tube length (RTL)>0.75 and <1.25). The 30-MV2-9 exosomes scored highest (++; RTL>1.25). Two endothelial progenitor lines scored negative (−; RTL<0.75). Exosomes from an osteochondral line, primary fibroblasts (BJ), BM-MSCs, and a human sarcoma cell line (HT1080) were also negative. The two clonal smooth muscle cell progenitor cell lines and one clonal pericyte line tested were positive in the in vitro vascular tube formation assay. Exosomes prepared from conditioned medium of the parental human embryonic stem cell lines H9 (WA09) and ESI-017 were also positive in the in vitro vascular tube formation assay.
  • TABLE 5
    TABLE 5: Angiogenic activity of PureStem vascular
    progenitors and other cell lines.
    Relative tube Angiogenic
    Cell Line Source Cell Type length Index
    30-MV2-9 PureStem Endothelial 1.67 ++
    30-MV2-6 PureStem Endothelial 1.07 +
    30-MV2-7 PureStem Endothelial 0.94 +
    30-MV2-17 PureStem Endothelial 0.76 +
    30-MV2-19 PureStem Endothelial 0.84 +
    RP1-MV2-8 PureStem Endothelial 1.07 +
    30-MV2-14 PureStem Endothelial 1.00 +
    30-MV2-15 PureStem Endothelial 1.00 +
    RP1-MV2-8 PureStem Endothelial 1.07 +
    30-MV2-24 PureStem Endothelial 0.41
    30-MV2-4 PureStem Endothelial 0.72
    W10 PureStem Smooth Muscle 0.83 +
    Z11 PureStem Smooth Muscle 0.92 +
    E164 PureStem Pericyte 0.86 +
    PC-M hES Pericyte 0.47
    BJ Primary Foreskin 0.61
    Fibroblast
    SM30 PureStem MSC-like 0.71
    MSCs Primary Adult 0.68
    BM-MSC
    HT1080 Transformed Sarcoma 0.71
    H9 Embryonic Pluripotent 1.06 +
    ESI-017 Embryonic Pluripotent 1.27 +
  • Example 6: In Vivo Angiogenic Activity of Embryonic Progenitor Stem Cell Derived Exosomes
  • Angiogenic activity of exosomes was assessed in vivo using the Matrigel plug assay in mice as previously described. Sahoo et al. (2011) Circ Res. 109(7):724. Immunocompromised mice (Female Nu/J mice aged 6-8 weeks; 2 plugs/mouse; 2 mice/group) were injected subcutaneously with approximately 300 μl of Matrigel containing PBS, 4×108/ml exosomes, or 150 ng/ml bFGF plus 60 ng/ml VEGF (positive control). The plugs were removed at day 14 after implant followed by fixation and paraffin embedding. The sections were stained with hematoxylin and Eosin (H&E) for histological examination and stained with von Willabrand factor antibody for detection of endothelial cells.
  • The data indicate that 30-MV2-6 exosomes are angiogenic in the Matrigel plug assay (FIG. 7 ). The exosome containing plugs show regions of infiltration of cells into the plug with vessel formation (FIG. 7 , panels A and C). The positive control plugs containing growth factors have regions of vessel formation (not shown) Immunostaining with antibody against von Willabrand factor (FIG. 7 , panels B and D) confirmed the endothelial identity of cells lining the vessel structures observed by H&E staining. The PBS control plugs show less cell infiltration and no vessel formation (FIG. 7 , panel E).
  • Example 7: Scale-up of Clonal Embryonic Progenitor Stem Cells for Exosome Production
  • Clonal embryonic progenitor cell lines described here are advantageous over other sources of biologically active exosomes because of their scalability. The parental pluripotent stem cell line to 30-MV2-6 which also produces angiogenic exosomes is costly to scale up because of the requirements for specialized medium and cell matrix (e.g. Matrigel). Primary endothelial stem cells or mesenchymal stromal cells rapidly lose differentiation and proliferative capacity upon culture in vitro. Typically MSCs begin to senesce in culture after 7-12 passages (approximately 10 population doublings) and show multiple changes including altered surface marker expression and increased autofluorescence. Wagner et al. (2008) Plos One 3(5):e2213. In contrast, human embryonic clonal progenitor lines such as the cell lines of the instant invention are grown under standard tissue culture conditions and medium and are highly scalable with typical replicative lifespans of 60 to 100 population doublings.
  • The Terumo Quantum Cell Expansion system (the bioreactor used in the instant example) is an automated hollow fiber cell culture platform designed for GMP compatible production of cells for use in cell therapy. The bioreactor was seeded at a density of approximately 900 cells/cm2 with approximately 4.0×107 30-MV2-6 cells (passage 9) and the cells were cultured for 13 days under their standard growth conditions of EGM-MV2 medium and 5% oxygen. The exosomes were collected by exchanging the complete medium for conditioning medium (basal EGM-MV2 medium without serum added; alternatively PBS may be also used). The conditioning medium was left in the bioreactor for 16 hours and collected for exosome purification. The cells were harvested by exchanging medium with a 0.25% trypsin solution to remove cells for the reactor, tested for viability and counted. Cells were scaled over 10-fold from the initial 40 million to approximately 440 million. The purified exosomes were quantified using CD63 detection ELISA (alternatively, nanoparticle tracking analysis as described in Example 1 may be used to quantify the exosomes). The yield of exosomes from one bioreactor run is at least 2.3×1010, which is equivalent to the approximate exosome yield from 72 T-225 flasks of 30-MV2-6 cells.
  • The purified exosomes were tested for angiogenic activity at a dose of 2.0×106 exosomes per well in the in vitro tube formation assay (described in detail in Example 2). As shown in FIG. 8 , the angiogenic activity of exosomes prepared from media conditioned by 30-MV2-6 cells grown in T-flasks was equivalent to the angiogenic activity of exosomes prepared from medium conditioned by 30-MV2-6 cells grown in the Quantum Cell Expansion system.
  • Example 8: Effect of Oxygen Concentration and Conditioning Medium on Exosome Activity
  • Hypoxia has been reported to increase exosome production from mammalian cells (Tadokoro et al. (2013) J Biol Chem. 288(48):34343; King et al. (2012) BMC Cancer 12:241). Furthermore, clonal embryonic progenitor cell lines are derived and maintained under low oxygen (5%). West et al. (2008) Regen Med. 3(3): 287. Therefore, 1% oxygen was tested for exosome production to determine if increasing hypoxia will increase exosome production or angiogenic activity.
  • Other stress conditions can also have an effect on exosome yield or activity. Serum starvation is used to induce exosome production. Nutrient deprivation was tested by using PBS as the conditioning medium. The use of PBS versus basal EGM-MV2 medium for conditioning the cells was also tested.
  • The results shown in FIG. 9 indicate that there is no significant difference in angiogenic activity of isolated exosomes when the medium was conditioned by cells incubated in 1% or 5% oxygen. These data also indicate the exosome angiogenic activity is not significantly different when PBS is used as the conditioning media compared to when the basal medium is used as the conditioning medium, although there is a trend toward higher activity when PBS is used.
  • Example 9: Quantitation of Exosome Concentration by ELISA Detection of CD63 on Intact Exosomes
  • There is a need for simple and convenient method to measure the concentration of exosome particles in a purified preparation of exosomes. Currently available ELISA kits for measuring exosome concentration (System Biosciences, Inc., Mountain View, Calif.) require lysing exosomes and have a lower limit of detection of approximately 2.0×108 exosomes. It is advantageous to measure low concentration samples directly without diluting the sample in a lysis buffer. Moreover, lysing the exosomes releases other proteins and nucleic acids that potentially interfere with the assay. The method described herein takes advantage of markers commonly presented on the surface of exosomes, such as transpanins CD63, CD9 and CD81 and allows for quantitation of intact exosomes.
  • A standard curve is prepared from exosome samples of known concentration (ranging from 5×108 to 8×107 exosomes/mL). The unknown samples are prepared in PBS or a buffer exchanged into PBS. Samples of intact exosomes are bound to 96 well ELISA plate wells in PBS at 50 μl/well for at least 16 hours at 37° C. The wells are washed 3×5 minutes in wash buffer (e.g. TBS-Tween). The wells are incubated with a mouse monoclonal antibody prepared in a suitable blocking buffer (e.g. PBS containing exosome depleted FBS and 0.05% Tween 20) that recognizes the extracellular domain of CD63 on intact exosomes for 1 hour at room temperature. The wells are washed again 3×5 minutes at room temperature. The wells are incubated with a suitable secondary antibody in a blocking buffer for detection of mouse anti-CD63 antibody bound to exosomes on the plate surface (e.g. HRP conjugated goat anti-mouse IgG) for 1 hour at room temperature. The wells are washed again 3×5 minutes at room temperature and the wells incubated with 50 μl of HRP substrate (e.g. Supersensitive TMB ELISA substrate) for 30 minutes at room temperature. The wells are washed 3×5 minutes at room temperature and 50 μl of stop buffer (0.16M sulfuric acid) is added to provide a fixed endpoint. The concentration of exosomes is quantitated by measuring the absorbance of each well at 450 nm.
  • An example of a standard curve and quantitation of samples is shown in FIG. 10 .

Claims (11)

1.-10. (canceled)
11. A method of inducing or enhancing a cell's ability to form vascular tube like structures comprising contacting a cell capable of making vascular tube like structures with an exosome isolated from a progenitor cell thereby inducing or enhancing a cell's ability to form vascular tube like structures.
12. The method of claim 11, wherein the cell capable of making vascular tube like structures is an endothelial cell.
13. The method of claim 12, wherein the endothelial cell is a HUVEC.
14. A method of providing or sustaining vasculature in a natural or an artificial organ for transplantation, the method comprising contacting the natural or artificial organ with an exosome isolated from a clonal progenitor cell.
15. The method of claim 14, wherein the clonal progenitor cell is positive for the expression of PCDHB2.
16. The method of claim 14, wherein the exosome comprises CD63.
17. A method of treating a degenerative disease associated with vascular aging in a subject in need of treatment, the method comprising administering to the subject an exosome isolated from a clonal progenitor cell.
18. The method of claim 17, wherein the degenerative disease associated with vascular aging is sarcopenia, dementia, chronic wounds, osteoporosis, cardiovascular disease, heart failure, infarction, chronic wounds, ulcer, clogged vessels or arteries, damaged vessels, stenotic vessels, arteriosclerosis, angina, peripheral vascular disease, Alzheimer's disease, ischemia, diabetes, cancer, a disease associated with cell replacement transplant or therapy, a disease associated with tissue and cell regenerative therapy, Parkinson's disease, stroke, or any combination thereof.
19. The method of claim 17, wherein the clonal progenitor cell line is positive for the expression of PCDHB2.
20. The method of claim 17, wherein the exosome comprises CD63.
US17/592,200 2014-07-03 2022-02-03 Exosomes from clonal progenitor cells Pending US20220403341A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/592,200 US20220403341A1 (en) 2014-07-03 2022-02-03 Exosomes from clonal progenitor cells

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462020869P 2014-07-03 2014-07-03
US14/748,215 US10240127B2 (en) 2014-07-03 2015-06-23 Exosomes from clonal progenitor cells
US16/270,295 US11274281B2 (en) 2014-07-03 2019-02-07 Exosomes from clonal progenitor cells
US17/592,200 US20220403341A1 (en) 2014-07-03 2022-02-03 Exosomes from clonal progenitor cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/270,295 Continuation US11274281B2 (en) 2014-07-03 2019-02-07 Exosomes from clonal progenitor cells

Publications (1)

Publication Number Publication Date
US20220403341A1 true US20220403341A1 (en) 2022-12-22

Family

ID=55748556

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/748,215 Active US10240127B2 (en) 2014-07-03 2015-06-23 Exosomes from clonal progenitor cells
US16/270,295 Active US11274281B2 (en) 2014-07-03 2019-02-07 Exosomes from clonal progenitor cells
US17/592,200 Pending US20220403341A1 (en) 2014-07-03 2022-02-03 Exosomes from clonal progenitor cells

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/748,215 Active US10240127B2 (en) 2014-07-03 2015-06-23 Exosomes from clonal progenitor cells
US16/270,295 Active US11274281B2 (en) 2014-07-03 2019-02-07 Exosomes from clonal progenitor cells

Country Status (1)

Country Link
US (3) US10240127B2 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2914615C (en) 2013-06-05 2023-10-17 Biotime, Inc. Compositions and methods for induced tissue regeneration in mammalian species
US11078462B2 (en) 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
US20170298352A1 (en) 2014-09-30 2017-10-19 Research Institute at Nationwide Children's Hospit al Compositions and methods for treating hepatic fibrosis
EP3356553A1 (en) 2015-09-29 2018-08-08 The Research Institute at Nationwide Children's Hospital Methods for detecting hepatic fibrosis and responsiveness to therapy
EP3362087A1 (en) * 2015-10-15 2018-08-22 The Scripps Research Institute BINDING ACTIVITY OF AMINOACYL-tRNA SYNTHETASE IN CHARCOT-MARIE-TOOTH (CMT) NEUROPATHY AND CMT-RELATED NEUROLOGICAL DISEASES
CA3004450A1 (en) 2015-11-18 2017-05-26 University Of Georgia Research Foundation, Inc. Neural cell extracellular vesicles
CA3019357A1 (en) 2016-04-04 2017-10-12 Biotime, Inc. Pluripotent stem cell-derived 3d retinal tissue and uses thereof
CN105925719B (en) * 2016-07-06 2019-09-27 北京泱深生物信息技术有限公司 Gene relevant to liver cancer differentiation and its application
US11718878B2 (en) 2017-08-21 2023-08-08 Seattle Children's Research Hospital Exosome profiling for diagnosis and monitoring of vasculitis and vasculopathies including Kawasaki disease
CN107702957A (en) * 2017-09-22 2018-02-16 上海华盈生物医药科技有限公司 The method and reagent of excretion body are extracted in a kind of serum
CN107899002B (en) * 2017-11-28 2020-02-18 首都医科大学宣武医院 Application of osteocalcin in regulation of endothelial progenitor cell exosomes
KR20200098600A (en) * 2017-12-14 2020-08-20 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 Purified exosome product, manufacturing method and method of use
US11938154B2 (en) * 2018-02-27 2024-03-26 Musc Foundation For Research Development Compositions and methods for treating and/or preventing sepsis and/or inflammatory conditions
CN108753682B (en) 2018-05-03 2019-08-02 中国医学科学院微循环研究所 Promote endothelial cell into the excretion body active ingredient and its preparation method and application of blood vessel
CN108823144B (en) 2018-05-03 2019-08-02 中国医学科学院微循环研究所 Inhibit the excretion body active ingredient and its preparation method and application of endothelial cell migration
CN108611374B (en) * 2018-05-15 2021-05-18 四川大学 miR-9 high expression tumor and treatment and specific marker characterization thereof
CA3135200A1 (en) * 2019-03-28 2020-10-01 Agex Therapeutics, Inc. Induced tissue regeneration using extracellular vesicles
JP7257277B2 (en) * 2019-07-12 2023-04-13 株式会社日立製作所 Cell culture monitoring device and cell culture system
CN112641803A (en) * 2019-09-25 2021-04-13 深圳光彩生命工程技术有限公司 Stem cell exosome preparation and preparation method thereof
CN110772483B (en) * 2019-11-13 2021-03-26 山东大学 Application of hydrogen sulfide modified mesenchymal stem cell outer vesicle serving as miRNA delivery vector in hypoxic-ischemic brain injury
EP4093408A4 (en) * 2020-01-22 2024-04-24 Agex Therapeutics Inc Therapeutic exosomes and method of producing them
WO2022114724A1 (en) * 2020-11-27 2022-06-02 (주)엑셀세라퓨틱스 Medium composition for producing exosome with high efficacy and high purity
CN114591955B (en) * 2022-02-25 2024-04-26 潍坊医学院 Cyclic RNA for promoting transformation of endothelial progenitor cells into mesenchymal cells and application thereof

Family Cites Families (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2197915B (en) 1986-11-19 1990-11-14 Rolls Royce Plc Improvements in or relating to fluid bearings
US5489508A (en) 1992-05-13 1996-02-06 University Of Texas System Board Of Regents Therapy and diagnosis of conditions related to telomere length and/or telomerase activity
US5639613A (en) 1992-05-13 1997-06-17 Board Of Regents, University Of Texas System Methods for cancer diagnosis and prognosis
US5686306A (en) 1992-05-13 1997-11-11 Board Of Regents, The University Of Texas System Methods and reagents for lengthening telomeres
US5922601A (en) 1995-01-19 1999-07-13 Biotransplant, Inc. High efficiency gene trap selection of regulated genetic loci
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6824993B2 (en) * 1995-06-06 2004-11-30 Human Genome Sciences, Inc. Antibodies that bind human prostate specific G-protein receptor HPRAJ70
US7585622B1 (en) 1996-10-01 2009-09-08 Geron Corporation Increasing the proliferative capacity of cells using telomerase reverse transcriptase
EP0986635A4 (en) 1997-01-10 2001-11-07 Life Technologies Inc Embryonic stem cell serum replacement
NO971997D0 (en) * 1997-04-29 1997-04-29 Kjetil Tasken Use of immunomodulatory agents
FR2766205B1 (en) 1997-07-16 2002-08-30 Inst Nat Sante Rech Med NOVEL METHOD FOR SENSITIZING ANTIGEN PRESENTING CELLS AND NOVEL MEANS FOR IMPLEMENTING THE METHOD
US5986020A (en) 1997-08-05 1999-11-16 Campbell; J. David Process for producing hyperbranched polymers
CA2307807C (en) 1997-10-23 2008-09-02 Andrea G. Bodnar Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
US20020051762A1 (en) 1998-01-23 2002-05-02 Shahin Rafii Purified populations of endothelial progenitor cells
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
US6248934B1 (en) 1998-10-26 2001-06-19 The Regents Of The University Of California Gene trap vectors
JP2002541253A (en) 1999-04-09 2002-12-03 スミスクライン・ビーチャム・コーポレイション Triarylimidazole
US20040199935A1 (en) 1999-06-30 2004-10-07 Chapman Karen B. Cytoplasmic transfer to de-differentiate recipient cells
IL147179A0 (en) 1999-06-30 2002-08-14 Advanced Cell Tech Inc Cytoplasmic transfer to de-differentiate recipient cells
US20030161817A1 (en) 2001-03-28 2003-08-28 Young Henry E. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
AU782846B2 (en) 1999-10-28 2005-09-01 University Of Massachusetts Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
AR029803A1 (en) 2000-02-21 2003-07-16 Smithkline Beecham Plc IMIDAZOLS REPLACED WITH PIRIDILE AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM
US6602711B1 (en) 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells
US7005252B1 (en) 2000-03-09 2006-02-28 Wisconsin Alumni Research Foundation Serum free cultivation of primate embryonic stem cells
GB0007405D0 (en) 2000-03-27 2000-05-17 Smithkline Beecham Corp Compounds
GB0027987D0 (en) 2000-11-16 2001-01-03 Smithkline Beecham Plc Compounds
EP1349851A4 (en) 2000-11-16 2004-09-08 Smithkline Beecham Corp Compounds
US20020069484A1 (en) 2000-11-28 2002-06-13 Creel Greg S. Rotatable handle structure for hand held tool
US20020142397A1 (en) 2000-12-22 2002-10-03 Philippe Collas Methods for altering cell fate
EP1356035B1 (en) 2000-12-22 2011-03-16 Kyowa Hakko Kirin Co., Ltd. Methods for cloning non-human mammals using reprogrammed donor chromatin or donor cells
GB0102668D0 (en) 2001-02-02 2001-03-21 Glaxo Group Ltd Compounds
US7514261B2 (en) 2001-08-09 2009-04-07 Cornell Research Foundation, Inc. Platelet-derived growth factor protection of cardiac myocardium
US6887706B2 (en) 2001-10-03 2005-05-03 Wisconsin Alumni Research Foundation Method of in vitro differentiation of transplantable neural precursor cells from primate embryonic stem cells
LU91074B1 (en) 2001-11-02 2004-04-30 Wisconsin Alumni Res Found Endothelial cells derived from primate embryonic stem cells
GB0127433D0 (en) 2001-11-15 2002-01-09 Smithkline Beecham Corp Compounds
JP4330995B2 (en) 2001-11-15 2009-09-16 チルドレンズ メディカル センター コーポレーション Methods for isolating, proliferating, and differentiating fetal stem cells from chorionic villi, amniotic fluid, and placenta, and methods for their therapeutic use
GB0127430D0 (en) 2001-11-15 2002-01-09 Smithkline Beecham Corp Compounds
JP2005510232A (en) 2001-11-26 2005-04-21 アドバンスド セル テクノロジー、インク. Production and use of reprogrammed human somatic cell nuclei and autologous and syngeneic human stem cells
US20030180268A1 (en) 2002-02-05 2003-09-25 Anthony Atala Tissue engineered construct for supplementing or replacing a damaged organ
EP1432724A4 (en) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
AU2003214566A1 (en) 2002-03-14 2003-09-22 Anosys, Inc. Functionalization of T cell derived vesicles and use thereof for the preparation of immunogenic pharmaceutical compositions
JP2005530800A (en) 2002-05-15 2005-10-13 スミスクライン・ビーチャム・コーポレイション Benzoxazine and benzoxazinone substituted triazoles
US20060003446A1 (en) 2002-05-17 2006-01-05 Gordon Keller Mesoderm and definitive endoderm cell populations
ES2449017T3 (en) 2002-06-21 2014-03-17 The University Of Utah Research Foundation Crosslinked compounds, and methods of preparation and use thereof
WO2004035748A2 (en) 2002-10-16 2004-04-29 Advanced Cell Technology, Inc. Methods using gene trapped stem cells for marking pathways of stem cell differentiation and making and isolating differentiated cells
EP1560931B1 (en) 2002-11-14 2011-07-27 Dharmacon, Inc. Functional and hyperfunctional sirna
US20040228847A1 (en) 2003-02-28 2004-11-18 Duke University Progenitor cells and methods of using same
PA8595001A1 (en) 2003-03-04 2004-09-28 Pfizer Prod Inc NEW CONDENSED HETEROAROMATIC COMPOUNDS THAT ARE INHIBITORS OF THE TRANSFORMING GROWTH FACTOR (TGF)
CN101137388A (en) 2003-05-15 2008-03-05 犹他卅大学研究基金会 Anti-adhesion composites and methods os use thereof
GB0326963D0 (en) 2003-11-19 2003-12-24 Glaxo Group Ltd Compounds
US7981871B2 (en) 2003-12-04 2011-07-19 University Of Utah Research Foundation Modified macromolescules and associated methods of synthesis and use
US20060112438A1 (en) 2004-01-02 2006-05-25 West Michael D Novel culture systems for ex vivo development
CA2556944C (en) 2004-03-05 2012-10-09 Taisho Pharmaceutical Co., Ltd. Thiazole derivative
JP2007528222A (en) 2004-03-09 2007-10-11 アブソルバー, アーベー Endothelial progenitor cells and methods of use thereof
WO2005090557A1 (en) 2004-03-23 2005-09-29 Daiichi Asubio Pharma Co., Ltd. Method of proliferating pluripotent stem cell
EP1766053A4 (en) 2004-06-02 2007-12-12 Sourcepharm Inc Rna-containing microvesicles and methods therefor
US8021847B2 (en) 2004-06-02 2011-09-20 Proxy Life Science Holdings, Inc. Microvesicle-based compositions and methods
ITRM20040568A1 (en) 2004-11-18 2005-02-18 Uni Degli Studi Di Roma Tor Vergata USE OF THE "PHAGE DISPLAY" TECHNIQUE FOR THE IDENTIFICATION OF PEPTIDES WITH CAPACITY OF STAMIN CELLS / PROGENITOR, PEPTIDES SO OBTAINED AND THEIR USES.
CA2597757C (en) 2005-02-10 2016-06-28 Regents Of The University Of Minnesota Vascular/lymphatic endothelial cells
US8492148B2 (en) 2005-02-23 2013-07-23 Foundation For Biomedical Research & Innovation Method for amplification of endothelial progenitor cell in vitro
AU2006252690A1 (en) 2005-05-27 2006-12-07 Advanced Cell Technology, Inc. Methods for identifying ligands for stem cells and cells derived therefrom
AU2006256650A1 (en) 2005-06-06 2006-12-14 Glykos Finland Oy Method of profiling a cell population
US20100158872A1 (en) 2005-06-23 2010-06-24 Mount Sinai School Of Medicine Of New York Univers Cardiomyocyte Cell Populations
EP1984487B1 (en) 2005-08-03 2022-10-12 Astellas Institute for Regenerative Medicine Improved methods of reprogramming animal somatic cells
US20090271335A1 (en) 2005-10-20 2009-10-29 Advanced Cell Technology, Inc. Totipotent, Nearly Totipotent or Pluripotent Mammalian Cells Homozygous or Hemizygous for One or More Histocompatibility Antigent Genes
US20080070303A1 (en) 2005-11-21 2008-03-20 West Michael D Methods to accelerate the isolation of novel cell strains from pluripotent stem cells and cells obtained thereby
WO2007062198A1 (en) 2005-11-21 2007-05-31 Advanced Cell Technology, Inc. Methods to accelerate the isolation of novel cell strains from pluripotent stem cells and cells obtained thereby
WO2007058671A1 (en) 2005-11-21 2007-05-24 West Michael D Novel uses of cells with prenatal patterns of gene expression
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
EP2091557A2 (en) 2006-11-15 2009-08-26 Coda Therapeutics, INC. Improved methods and compositions for wound healing
NZ578728A (en) 2007-01-09 2012-01-12 Lead Pharma Cel Models Ip B V Provision of new cardiomyocyte progenitor cells and cardiomyocytes derived therefrom
US8465732B2 (en) 2007-01-19 2013-06-18 Cornell Research Foundation, Inc. Endothelial cells expressing adenovirus E4ORF1 and methods of use thereof
WO2008094597A2 (en) 2007-01-30 2008-08-07 University Of Georgia Research Foundation, Inc. Early mesoderm cells, a stable population of mesendoderm cells that has utility for generation of endoderm and mesoderm lineages and multipotent migratory cells (mmc)
KR101516833B1 (en) 2007-03-23 2015-05-07 위스콘신 얼럼나이 리서어치 화운데이션 Somatic cell reprogramming
US20100111914A1 (en) 2007-05-21 2010-05-06 Yuanyuan Zhang Stem cells from urine and methods for using the same
FI20075417A0 (en) 2007-06-05 2007-06-05 Marjo-Riitta Suuronen Formulations and methods for culturing embryonic stem cells
JP2008307007A (en) 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
US20100255580A1 (en) 2007-07-18 2010-10-07 Lifesccan, Inc. Differentiation of Human Embryonic Stem Cells
WO2009052211A1 (en) 2007-10-15 2009-04-23 Caritas St. Elizabeth Medical Center Of Boston, Inc. Compositions and methods for enhancing lymphangiogenesis
WO2009067245A2 (en) 2007-11-20 2009-05-28 University Of Florida Research Foundation, Inc. Compositions and methods for tissue repair
US20110003008A1 (en) 2008-02-22 2011-01-06 Agency For Science, Technology And Research (A*Star) Mesenchymal stem cell particles
JP5801187B2 (en) 2008-03-27 2015-10-28 マウント シナイ スクール オブ メディスン Human cardiovascular progenitor cells
DK2297307T3 (en) 2008-06-04 2016-07-25 Cellular Dynamics Int Inc PROCEDURES FOR THE MANUFACTURE OF IPS CELLS USING NON-VIRAL METHODS
CA2695590C (en) 2008-06-27 2018-01-09 Kyoto University Method of efficiently establishing induced pluripotent stem cells
US20130115673A1 (en) 2008-07-16 2013-05-09 Biotime, Inc. Methods of Screening Embryonic Progenitor Cell Lines
US20100184033A1 (en) 2008-07-16 2010-07-22 West Michael D Methods to accelerate the isolation of novel cell strains from pluripotent stem cells and cells obtained thereby
WO2010021993A1 (en) 2008-08-19 2010-02-25 Cytori Therapeutics, Inc. Methods of using adipose tissue-derived cells in the treatment of the lymphatic system and malignant disease
EP2350320A4 (en) * 2008-11-12 2012-11-14 Caris Life Sciences Luxembourg Holdings Methods and systems of using exosomes for determining phenotypes
US20100183620A1 (en) 2008-11-26 2010-07-22 Kaumudi Bhawe Compositions and methods for regulating collagen and smooth muscle actin expression by serpine2
US8609927B2 (en) 2009-05-28 2013-12-17 Trustees Of Dartmouth College Caveolin 1-reporter protein knock-in mouse
CN102844036B (en) 2009-07-16 2016-01-20 生物时代股份有限公司 For the soft osteogenetic method and composition of in vitro and in vivo
EP2467470A2 (en) 2009-08-17 2012-06-27 Technion Research & Development Foundation Ltd. Pericyte progenitor cells and methods of generating and using same
WO2011053257A2 (en) 2009-11-02 2011-05-05 Agency For Science, Technology And Research (A*Star) Methods of monitoring cellular states
US20110312881A1 (en) 2009-12-21 2011-12-22 Amunix, Inc. Bifunctional polypeptide compositions and methods for treatment of metabolic and cardiovascular diseases
US20120301443A1 (en) 2009-12-29 2012-11-29 Cornell University Methods for developing endothelial cells from pluripotent cells and endothelial cells derived
WO2011109612A2 (en) 2010-03-03 2011-09-09 The General Hospital Corporation Method for selecting an ips cell
WO2011127458A2 (en) 2010-04-09 2011-10-13 University Of Southern California Systems and methods of cell activated, controlled release delivery of growth factors for tissue repair and regeneration
CA2800616C (en) 2010-05-27 2023-01-17 Biotime Inc. Improved methods of screening embryonic progenitor cell lines
WO2012020308A2 (en) 2010-08-13 2012-02-16 The University Court Of The University Of Glasgow Cellular and molecular therapies
CA2810488A1 (en) 2010-09-07 2012-03-15 Technion Research & Development Foundation Limited Novel methods and culture media for culturing pluripotent stem cells
US20120093885A1 (en) 2010-10-18 2012-04-19 Northwestern University Therapeutic vesicles
WO2012065065A1 (en) 2010-11-12 2012-05-18 Follica, Inc. Methods and compositions for modulating hair growth, wound healing and scar revision
CA2829586C (en) 2011-03-11 2021-03-02 Children's Medical Center Corporation Methods and compositions relating to mesenchymal stem cell exosomes
US20120295347A1 (en) 2011-05-20 2012-11-22 Steven Kessler Methods and Compositions for Producing Endothelial Progenitor Cells from Pluripotent Stem Cells
EP2726603B1 (en) 2011-06-29 2020-04-01 Biorestorative Therapies, Inc. Brown fat cell compositions and methods
JP5522127B2 (en) * 2011-07-15 2014-06-18 コニカミノルタ株式会社 Image forming apparatus and power transmission mechanism
ITRM20110403A1 (en) 2011-07-28 2013-01-29 Ospedale Pediatrico Bambino Gesu MICROWELES ISOLATED BY MESENCHIMAL CELLS AS IMMUNOSOPPRESSORS.
US20140349396A1 (en) * 2011-09-09 2014-11-27 Michael West Compositions and Methods Relating to Clonal Progenitor Cells
US9645149B2 (en) * 2011-09-30 2017-05-09 The Regents Of The University Of Michigan System for detecting rare cells
US20130115609A1 (en) 2011-11-08 2013-05-09 National Cheng Kung University Methods and Kits for Detecting Circulating Cancer Stem Cells
US10238755B2 (en) 2011-11-30 2019-03-26 The Wistar Institute Of Anatomy And Biology Methods and compositions for regulation of cell aging, carcinogenesis and reprogramming
JP6274510B2 (en) 2012-01-27 2018-02-07 国立大学法人京都大学 Method for inducing myocardial differentiation of pluripotent stem cells
ES2691296T3 (en) 2012-04-03 2018-11-26 Reneuron Limited Stem cell microparticles
GB201302468D0 (en) 2013-02-12 2013-03-27 Reneuron Ltd Stem cell product
SG11201407626QA (en) 2012-05-18 2014-12-30 Agency Science Tech & Res Umbilical cord mesenchymal stem cell exosomes
US20140010801A1 (en) 2012-06-08 2014-01-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Compositions and Methods for Restoring or Rejuvenating Stem/Progenitor Cell Function
WO2014013258A1 (en) 2012-07-19 2014-01-23 Reneuron Limited Stem cell microparticles
US20150216892A1 (en) 2012-08-03 2015-08-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
US9828603B2 (en) 2012-08-13 2017-11-28 Cedars Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9175263B2 (en) 2012-08-22 2015-11-03 Biotime, Inc. Methods and compositions for targeting progenitor cell lines
US20140178988A1 (en) 2012-10-08 2014-06-26 Biotime, Inc. Differentiated Progeny of Clonal Progenitor Cell Lines
WO2014091373A1 (en) 2012-12-11 2014-06-19 The University Court Of The University Of Glasgow Cellular and molecular therapies for peripheral vascular disease
SG11201506284RA (en) 2013-02-12 2015-09-29 Reneuron Ltd Method of producing microparticles
JP6142076B2 (en) 2013-05-03 2017-06-07 マイクロバイオティックス・インコーポレーテッド Antimicrobial enhancer
CA2914615C (en) 2013-06-05 2023-10-17 Biotime, Inc. Compositions and methods for induced tissue regeneration in mammalian species
US20160193252A1 (en) * 2013-08-14 2016-07-07 Reneuron Limited STEM CELL MICROPARTICLES AND miRNA
GB201317887D0 (en) 2013-10-09 2013-11-20 Reneuron Ltd Product
GB201317889D0 (en) 2013-10-09 2013-11-20 Reneuron Ltd Product and use
US20150275177A1 (en) 2013-11-25 2015-10-01 Biotime, Inc., Methods For Generating Pluripotent Stem Cell-Derived Brown Fat Cells
US11078462B2 (en) * 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
CA3202332A1 (en) 2015-12-07 2017-06-15 Agex Therapeutics, Inc. Methods for the re-derivation of diverse pluripotent stem cell-derived brown fat cells
AU2017278931B2 (en) 2016-06-07 2023-06-29 Agex Therapeutics, Inc. Improved methods for detecting and modulating the embryonic-fetal transition in mammalian species
US20190151372A1 (en) 2017-06-19 2019-05-23 Agex Therapeutics, Inc. Pericyte cell exosomes

Also Published As

Publication number Publication date
US11274281B2 (en) 2022-03-15
US20160108368A1 (en) 2016-04-21
US10240127B2 (en) 2019-03-26
US20190241873A1 (en) 2019-08-08

Similar Documents

Publication Publication Date Title
US20220403341A1 (en) Exosomes from clonal progenitor cells
US11918687B2 (en) SDC-2 exosome compositions and methods of isolation and use
De Simone et al. Identification of a Kupffer cell subset capable of reverting the T cell dysfunction induced by hepatocellular priming
EP3303636B1 (en) Companion methods for il-2-based therapies and mesenchymal stem cell-based therapies
AU2014217615B2 (en) Method of producing microparticles
AU2013245406B2 (en) Stem cell microparticles
EP2877187B1 (en) Stem cell microparticles
AU2014217614B2 (en) Stem cell microparticles and miRNA
JP7174758B2 (en) CD39 Stromal Stem Cell Isolation Methods and Uses
WO2020095044A1 (en) Monocytes for cancer targeting
US20240108689A1 (en) Modulation of a pathogenic phenotype in th1 cells
US20240156901A1 (en) Gal3bp polypeptide compositions and methods for treatment of cancer and determining treatment responsiveness
Ferguson Composition-Activity Analyses and Nucleic Acid Loading Strategies for Therapeutic Extracellular Vesicles
CA3224898A1 (en) Enhancement of hematopoietic stem cell and hematopoietic progenitor cell expansion with agents that activate tam receptors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: RECYTE THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LAROCCA, DANA;REEL/FRAME:061105/0458

Effective date: 20220802

Owner name: RECYTE THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HASSANIPOUR, MOHAMMAD;BIGNONE, PAOLA A.;SIGNING DATES FROM 20160105 TO 20160107;REEL/FRAME:061105/0385

AS Assignment

Owner name: JUVENESCENCE LIMITED, GREAT BRITAIN

Free format text: SECURITY INTEREST;ASSIGNORS:AGEX THERAPEUTICS, INC.;RECYTE THERAPEUTICS, INC.;REVERSE BIOENGINEERING, INC.;REEL/FRAME:065529/0594

Effective date: 20231109