WO2013148121A1 - Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling - Google Patents

Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling Download PDF

Info

Publication number
WO2013148121A1
WO2013148121A1 PCT/US2013/030080 US2013030080W WO2013148121A1 WO 2013148121 A1 WO2013148121 A1 WO 2013148121A1 US 2013030080 W US2013030080 W US 2013030080W WO 2013148121 A1 WO2013148121 A1 WO 2013148121A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
stem cells
pluripotent stem
subject
induced pluripotent
Prior art date
Application number
PCT/US2013/030080
Other languages
French (fr)
Inventor
Dinendar SINGLA
Original Assignee
University Of Central Florida Research Foundation, 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 University Of Central Florida Research Foundation, Inc. filed Critical University Of Central Florida Research Foundation, Inc.
Priority to CA2869163A priority Critical patent/CA2869163A1/en
Priority to US14/389,402 priority patent/US20150056172A1/en
Priority to EP13770363.3A priority patent/EP2830639B1/en
Publication of WO2013148121A1 publication Critical patent/WO2013148121A1/en

Links

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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0657Cardiomyocytes; Heart 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
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • 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
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells

Definitions

  • MI myocardial infarction
  • a method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof comprising (i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) administering to the subject fibroblast growth factor-8; and/or (iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9, and determining regeneration in the heart.
  • a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof comprising (i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) administering to the subject fibroblast growth factor-8; and/or (iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9, and determining a decrease in cardiac remodeling.
  • compositions for enhancing cardiac tissue/cell regeneration in a subject comprising (i) fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • compositions for attenuating adverse cardiac tissue/cell remodeling in a subject comprising (i) fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) fibroblast growth factor-8 and fibroblast growth factor-9.
  • a method of generating cardiac induced pluripotent stem cells comprising (i) inserting one or more nucleic acid constructs capable of expressing stem-cell like factors into a cardiac cell type, wherein the stem-cell like factors comprise Oct3/4, KIf4, Sox2, and c-Myc, or a combination thereof; and (ii) obtaining the cardiac induced pluripotent stem cells stably expressing the stem-cell like factors in the cardiac cell type.
  • Disclosed herein is a method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising administering conditioned medium from induced pluripotent stem cells, wherein the conditioned medium is administered with or without FGF-8.
  • a method of identifying a myocardial injury or infarcted myocardial tissue comprising measuring the amount of miR-486, wherein the amount of miR-486 decreases after myocardial injury or myocardial infarction, and wherein the amount of miR-486 increases after transplantation of induced pluripotent stem cells.
  • Disclosed herein is a method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising administering FGF-8.
  • Figure 1 shows indicia of stably transfected iPS cells.
  • Figure 2 shows that iPS cells generated from embryoid bodies differentiate into cardiac myocytes, smooth muscles ells, and endothelial cells following FGF-8 treatment.
  • Figure 3 shows the effects of FGF-8 on beating cardiac myocytes derived from the iPS-EB cell system.
  • Figure 4 shows the effects of both iPS-CM and FGF-8 on H9c2 cells following H 2 0 2 - induced apoptosis.
  • Figure 5 shows that transplanted RFP-iPS or ES cells differentiate into cardiac myocytes with electrical coupling.
  • Figure 6 shows that transplanted iPS cells inhibit apoptosis at 2 weeks post-MI.
  • Figure 7 shows MMP-2 expression in ES and iPS cell transplanted hearts.
  • Figure 8 shows reduced fibrosis and improved cardiac function following iPS cell transplantation.
  • Figure 9 shows the effect of FGF-8 treatment on cardiac myocyte apoptosis following MI and the effect of FGF-8 treatment on Bax and Bcl-2 expression.
  • Figure 10 shows a decrease in levels of miR-486 following MI and the effect of iPS cell transplantation.
  • Figure 11 shows the effect of treatment with ES and iPS cells following MI on p-Akt and PTEN expression as well as the effect of FGF-8 treatment on p-Akt and PTEN expression.
  • Figure 12 shows expression data of c-kit and notchl following sham treatment and myocardial infarction both with and without FGF-8 treatment.
  • Figure 13 shows histological and quantifiable data regarding the effect of sham-MI, MI, and MI+FGF-8 treatment on interstitial fibrosis and vascular fibrosis / vascular area (VF/VA).
  • Figure 14 shows various cardiac performance measures following sham-MI, MI, and MI with FGF-8 treatment.
  • Induced pluripotent stem (iPS) cells could be a potential cell type of choice to treat heart diseases but their basic mechanisms of cardiac repair and regeneration are largely unknown.
  • iPS cells were generated from H9c2 cells (originally isolated from embryonic rat heart ventricular tissue). These cells have the potential to differentiate into cardiac myocytes in vitro and in vivo. Following myocardial infarction, transplanted cardiac iPS cells inhibited post-MI remodeling and demonstrated cardiac regeneration.
  • FGF family members play an important role in various cell signal transduction pathways associated with embryo development (Dyer et al., 2009; Yun et al, 2010). Specifically, FGF-8 has a unique role in cell differentiation, proliferation, and specification. (Dyer et al, 2009). FGF-8 mutant mice demonstrate abnormalities in the heart, suggesting its role as a key growth factor required for cardiac development (Dyer et al, 2009). The role of FGF-8 in the injured adult heart or iPS cells is previously unexplored. The data presented herein demonstrate that FGF-8 contains both pro-cardiac and anti-apoptotic properties and that FGF-8 inhibits cardiac myocyte apoptosis in vitro and in vivo.
  • compositions useful in a method of enhancing cardiac tissue and/or cell regeneration are disclosed herein.
  • compositions useful in a method of attenuating cardiac tissue and/or cell remodeling are disclosed herein.
  • compositions useful in a method of inhibiting apoptosis and/or apoptosis-related mechanisms are disclosed herein.
  • compositions disclosed herein enhance cardiac tissue/cell regeneration.
  • a composition for enhancing cardiac tissue/cell regeneration in a subject comprising: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and/or (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed composition for enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9).
  • FGF-8 primed iPS cells fibroblast growth factor-8 primed induced pluripotent stem cells
  • CM of FGF-8 primed iPS cells conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells
  • FGF-8 fibroblast growth factor-8
  • FGF-9 fibroblast growth factor-9
  • a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed composition protects myocardium including, but not limited to human myocardium, when administered with or without iPS cells.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject enhances cells engraftment.
  • a disclosed composition enhances cell proliferation.
  • a disclosed composition enhances cell differentiation.
  • a disclosed composition can perform any combination of the following: enhancing cell engraftment, enhancing cell proliferation, and/or enhancing cell differentiation.
  • the induced pluripotent stem cells differentiate into cardiac myocytes.
  • the induced pluripotent stem cells are cardiac -committed.
  • a disclosed composition enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, and any other types of stem cells known to be effective by those of skill in the art.
  • a disclosed composition enhances c-kit positive cells or any other endogenous heart stem cells.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject improves angiogenesis in the subject.
  • a disclosed composition enhances cardiac function in the subject.
  • a disclosed composition both improves angiogenesis and enhances cardiac function in the subject.
  • enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
  • the conditioned media of a disclosed composition comprises one or more anti-apoptotic and anti-fibrotic factors.
  • the one or more anti-apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix metalloproteinase- 1 (TIMP-1).
  • the subject has experienced a cardiac ischemia/reperfusion event.
  • the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
  • the subject has a congenital heart defect.
  • the congenital heart defect is hypoplasia or pentalogy of Cantrell.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered to the subject prior to a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during a cardiac
  • a disclosed composition is administered to the subject following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject within 10, 15, 20, 25, 30, or more minutes following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered within 1, 2, 6, 12, 18, 24, or more hour following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject one or more times. For example, in an aspect, a disclosed composition is administered to the subject prior to and during a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during and following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject prior to and following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject prior to, during, and following a cardiac ischemia/reperfusion event.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject comprises between 5,000 and 500,000 induced pluripotent stem cells (iPS cells). In an aspect, a composition comprises approximately 100,000 iPS cells. In an aspect, a composition comprises less than 100,000 iPS cells. In an aspect, a composition comprises more than 100,000 iPS cells.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered to the subject in one intramyocardial injection. In an aspect, a disclosed composition is administered in two or more intramyocardial injections.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered into a peri-infarct zone of the injured myocardium. In an aspect, disclosed composition is administered into an infarcted zone of the injured myocardium. In an aspect, a disclosed composition is administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
  • a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject further comprises one or more immunosuppressive drugs.
  • Immunosuppressive drugs are known in the art.
  • the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • the one or more immunosuppressive drugs is cyclosporine A.
  • the induced pluripotent stem cells of a disclosed composition are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources.
  • the induced pluripotent stem cells of a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject are obtained from fibroblast cells.
  • the induced pluripotent stem cells are obtained from H9c2 cells.
  • the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor.
  • the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
  • the sternness factors are c-myc, oct 3/4, Klf4, and Sox2.
  • a disclosed composition enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
  • the subject is a mammal.
  • the mammal is a primate.
  • the mammal is a human.
  • the human is a patient.
  • compositions disclosed herein attenuate adverse cardiac tissue/cell remodeling.
  • a composition for attenuating adverse cardiac tissue/cell remodeling in a subject comprising: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and/or (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed composition for enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9).
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject comprises any one of the following:
  • a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed composition protects myocardium including, but not limited to human myocardium, when administered with or without iPS cells.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject improves angiogenesis in the subject.
  • a disclosed composition enhances cardiac function in the subject.
  • a disclosed composition improves angiogenesis and enhances cardiac function in the subject.
  • a disclosed composition for attenuating adverse cardiac remodeling inhibits fibrosis and/or fibrosis-related mechanisms.
  • a disclosed composition decreases the expression level or activity level of at least one matrix metalloproteinase.
  • the at least one matrix metalloproteinase is MMP-2.
  • the at least one matrix metalloproteinase is MMP-9.
  • the at least one matrix metalloproteinase comprises MMP-2 and MMP-9.
  • a disclosed composition for attenuating adverse cardiac tissue and/or cell remodeling in a subject inhibits or decreases necrosis.
  • a disclosed composition inhibits or decreases apoptosis and/or apoptosis-related mechanisms.
  • the apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells.
  • the apoptosis and/or apoptosis-related mechanism occurs in cardiac tissue of the subject.
  • a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, or decreases FOXOla expression.
  • a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression. In an aspect, a disclosed composition performs any combination of the following: increases miR-486 expression, increases Akt expression, decreases PTEN expression, and/or decreases FOXOla expression.
  • the induced pluripotent stem cells differentiate into cardiac myocytes.
  • the induced pluripotent stem cells are cardiac -committed.
  • a disclosed composition enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc.
  • a disclosed composition enhances c-kit positive cells or any other endogenous heart stem cells.
  • enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises each of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
  • the conditioned media of a disclosed composition comprises one or more anti-apoptotic and anti-fibrotic factors.
  • the one or more anti-apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix metalloproteinase- 1 (TIMP-1).
  • the subject has experienced a cardiac ischemia/reperfusion event.
  • the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
  • the subject has a congenital heart defect.
  • the congenital heart defect is hypoplasia or pentalogy of Cantrell.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered to the subject prior to a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject during a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject within 10, 15, 20, 25, 30, or more minutes following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered within 1, 2, 6, 12, 18, 24, or more hour following a cardiac ischemia/reperfusion event.
  • a disclosed composition is administered to the subject one or more times. For example, in an aspect, a disclosed composition is administered to the subject prior to and during a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during and following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject prior to and following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject prior to, during, and/or following a cardiac ischemia/reperfusion event.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject comprises between 5,000 and 500,000 induced pluripotent stem cells (iPS cells). In an aspect, a disclosed composition comprises approximately 100,000 iPS cells. In an aspect, a disclosed comprises less than 100,000 iPS cells. In an aspect, a disclosed composition comprises more than 100,000 iPS cells.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered to the subject in one intramycardial injection. In an aspect, a disclosed composition is administered in two or more intramycardial injections.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered into a peri-infarct zone of the injured myocardium. In an aspect, a disclosed composition is administered into an infarcted zone of the injured myocardium. In an aspect, a disclosed composition is administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
  • a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject further comprises one or more immunosuppressive drugs.
  • immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • the one or more immunosuppressive drugs is cyclosporine A.
  • the induced pluripotent stem cells of a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject are obtained from an autologous source.
  • the induced pluripotent stem cells are obtained from an allogeneic source.
  • the induced pluripotent stem cells are obtained from a syngeneic source.
  • the induced pluripotent stem cells are obtained from a combination of sources.
  • the induced pluripotent stem cells of a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject are obtained from fibroblast cells.
  • the induced pluripotent stem cells are obtained from H9c2 cells.
  • the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor.
  • the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
  • the sternness factors are c-myc, oct 3/4, Klf4, and Sox2.
  • a disclosed composition enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to,
  • mesenchymal stem cells mesenchymal stem cells, adipocyte stems cells, etc.
  • the subject is a mammal.
  • the mammal is a primate.
  • the mammal is a human.
  • the human is a patient. 3. PHARMACEUTICAL COMPOSITIONS
  • the invention relates to pharmaceutical compositions comprising a disclosed compositions for enhancing cardiac tissue and/or cell regeneration.
  • the invention relates to pharmaceutical compositions comprising a disclosed composition for attenuating adverse cardiac tissue and/or cell remodeling. That is, a pharmaceutical composition can be provided comprising a therapeutically effective amount of at least one disclosed compositions and a pharmaceutically acceptable carrier.
  • the invention relates to a method of enhancing cardiac tissue and/or cell regeneration in a subject.
  • a method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof comprising: (A) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (B) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) administering to the subject fibroblast growth factor-8; and/or (D) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9; and determining regeneration in the heart.
  • a disclosed method of enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9).
  • FGF-8 primed iPS cells fibroblast growth factor-8 primed induced pluripotent stem cells
  • CM of FGF-8 primed iPS cells conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells
  • FGF-8 fibroblast growth factor-8
  • FGF-9 fibroblast growth factor-9
  • FGF-8 primed i.e., CM of FGF-8 (i.e., FGF-8 &
  • a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed method protects myocardium including, but not limited to, human myocardium, with or without the administration of iPS cells.
  • fibroblast growth factor-8 (FGF-8) is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells).
  • FGF-8 is administered to the subject during the administration of the iPS cells.
  • FGF-8 is administered to the subject following the administration of the iPS cells.
  • FGF-8 is administered to the subject prior to and during the administration of the iPS cells.
  • FGF-8 is administered to the subject prior to and following the administration of the iPS cells.
  • FGF-8 is administered to the subject during and following the administration of the iPS cells.
  • a composition comprising fibroblast growth factor-8 (FGF-8) and fibroblast growth factor-9 (FGF-9) is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells).
  • a composition comprising FGF-8 and FGF-9 is administered to the subject during the administration of the iPS cells.
  • a composition comprising FGF-8 and FGF-9 is administered to the subject following the administration of the iPS cells.
  • a composition comprising FGF-8 and FGF-9 is administered to the subject prior to and during the administration of the iPS cells.
  • a composition comprising FGF-8 and FGF-9 is administered to the subject prior to and following the administration of the iPS cells. In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject during and following the administration of the iPS cells.
  • the conditioned media is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells).
  • the conditioned media is administered to the subject during the administration of the iPS cells.
  • the conditioned media is administered to the subject following the administration of the iPS cells.
  • the conditioned media is administered to the subject prior to and during the administration of the iPS cells.
  • the conditioned media is administered to the subject prior to and following the administration of the iPS cells.
  • the conditioned media is administered to the subject during and following the administration of the iPS cells.
  • the administration of induced pluripotent stem cells to the subject is repeated.
  • the administration of fibroblast growth factor-8 to the subject is repeated.
  • the administration of a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 to the subject is repeated.
  • the administration of the conditioned media to the subject is repeated.
  • the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to cardiac dysfunction.
  • the iPS cells are administered to the subject, during cardiac dysfunction.
  • the iPS cells administered to the subject following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to and during cardiac dysfunction.
  • the iPS cells are administered to the subject during and following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to and following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
  • the induced pluripotent stem cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction. In an aspect, the induced pluripotent stem cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction. [0084] In an aspect of a disclosed method, enhancing cardiac tissue and/or cell regeneration comprises enhancing cell engraftment. In an aspect, enhancing cardiac tissue and/or cell regeneration comprises enhancing cell differentiation. In an aspect of a disclosed method, enhancing cardiac tissue and/or cell regeneration comprises both enhancing cell engraftment and enhancing cell differentiation. In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac-committed.
  • a disclosed method comprises enhancing cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
  • a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof further comprises improving angiogenesis in the subject.
  • a disclosed method further comprises enhancing cardiac function in the subject.
  • a disclosed method further comprises improving both angiogenesis and enhancing cardiac function in the subject.
  • enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
  • the conditioned media of a method disclosed herein comprises one or more anti-apoptotic and anti-fibrotic factors.
  • the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix
  • TRIP-1 metalloproteinase-1
  • iPS cells induced pluripotent stem cells
  • approximately 100,000 iPS cells are administered to the subject.
  • less than 100,000 iPS cells are administered to the subject.
  • more than 100,000 iPS cells are administered to the subject.
  • the induced pluripotent stem cells can be administered in one intramycardial injection.
  • the induced pluripotent stem cells are administered in two or more intramycardial injections.
  • the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium.
  • the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium.
  • the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
  • a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof further comprises administering to the subject one or more immunosuppressive drugs.
  • Immunosuppressive drugs are known in the art.
  • the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • the one or more immunosuppressive drugs is cyclosporine A.
  • the one or more immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject prior to the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during and following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and following the administration of the induced pluripotent stem cells.
  • the cardiac dysfunction of a disclosed method is a cardiac
  • the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia,
  • the subject has a congenital heart defect.
  • the congenital heart defect is hypoplasia or pentalogy of Cantrell.
  • the induced pluripotent stem cells of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof are obtained from an autologous source.
  • the induced pluripotent stem cells are obtained from an allogeneic source.
  • the induced pluripotent stem cells are obtained from a syngeneic source.
  • the induced pluripotent stem cells are obtained from a combination of sources.
  • the induced pluripotent stem cells of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof are obtained from fibroblast cells.
  • the induced pluripotent stem cells are obtained from H9c2 cells.
  • the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one stemness factor.
  • the at least one stemness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
  • the stemness factors are c-myc, oct 3/4, Klf4, and Sox2.
  • a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, and other types of stem cells known to be effective by those of skill in the art.
  • the subject is a mammal.
  • the mammal is a primate.
  • the mammal is a human.
  • the human is a patient.
  • the invention relates to a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject.
  • a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof comprising: (A) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (B) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) administering to the subject fibroblast growth factor-8; and/or (D) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor- 9; and determining a decrease in cardiac remodeling.
  • a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof comprises any one of the following:
  • a disclosed method comprises administering one or two or three or more or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • a disclosed method protects myocardium including, but not limited to human myocardium, with or without the administration of iPS cells.
  • FGF-8 fibroblast growth factor-8
  • iPS cells induced pluripotent stem cells
  • FGF-8 is administered to the subject during the administration of the iPS cells.
  • FGF-8 is administered to the subject following the administration of the iPS cells.
  • FGF-8 is administered to the subject prior to and during the administration of the iPS cells.
  • FGF-8 is administered to the subject prior to and following the administration of the iPS cells.
  • FGF-8 is administered to the subject during and following the administration of the iPS cells.
  • a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells).
  • a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject during the administration of the iPS cells.
  • a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject following the administration of the iPS cells.
  • a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to and during the administration of the iPS cells.
  • a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to and following the administration of the iPS cells. In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject during and following the administration of the iPS cells.
  • the conditioned media is administered to the subject prior to the
  • the conditioned media is administered to the subject during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject following the administration of the iPS cells. In an aspect the conditioned media is administered to the subject prior to and during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject prior to and following the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject during and following the administration of the iPS cells.
  • the administration of induced pluripotent stem cells to the subject is repeated.
  • the administration of fibroblast growth factor-8 to the subject is repeated.
  • the administration of a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 to the subject is repeated.
  • the administration of the conditioned media to the subject is repeated.
  • the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject prior to cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject, during cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject or following cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject prior to and during cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject during and following cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject prior to and following cardiac dysfunction.
  • the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
  • the induced pluripotent stem cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction.
  • the induced pluripotent stem cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction.
  • Attenuating adverse cardiac remodeling comprises inhibiting fibrosis and/or fibrosis-related mechanisms.
  • inhibiting fibrosis and/or fibrosis-related mechanisms comprises decreasing the expression level or activity level of at least one matrix metalloproteinase.
  • the at least one matrix metalloproteinase is MMP-2.
  • the at least one matrix metalloproteinase is MMP- 9.
  • the at least one matrix metalloproteinase comprises MMP-2 and MMP-9.
  • the induced pluripotent stem cells differentiate into cardiac myocytes.
  • the induced pluripotent stem cells are cardiac -committed.
  • a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc.
  • a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
  • a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprises improving angiogenesis in the subject.
  • a disclosed method further comprises enhancing cardiac function in the subject.
  • a disclosed method further comprises improving angiogenesis and enhancing cardiac function in the subject.
  • enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry.
  • enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
  • a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprise inhibiting or decreasing necrosis.
  • a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprise inhibiting or decreasing apoptosis and/or apoptosis-related mechanisms.
  • the apoptosis and/or apoptosis-related mechanism occurs in the induced pluripotent stem cells.
  • the apoptosis and/or apoptosis-related mechanism occurs in cardiac tissue of the subject.
  • the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and/or a decrease in FOXOla expression.
  • the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression.
  • the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by any combination of the following: an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and/or a decrease in FOXOla expression.
  • the conditioned media of a method disclosed herein comprises one or more anti-apoptotic and anti-fibrotic factors.
  • the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix
  • TRIP-1 metalloproteinase-1
  • iPS cells induced pluripotent stem cells
  • approximately 100,000 iPS cells are administered to the subject.
  • less than 100,000 iPS cells are administered to the subject.
  • more than 100,000 iPS cells are administered to the subject.
  • the induced pluripotent stem cells can be administered in one intramycardial injection. In an aspect, the induced pluripotent stem cells are administered in two or more intramycardial injections.
  • the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium. In an aspect of a disclosed method, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
  • a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprises administering to the subject one or more immunosuppressive drugs.
  • the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, antiproliferatives, and mTOR inhibitors.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • immunosuppressive drugs is cyclosporine A.
  • the one or more of the immunosuppressive drugs is cyclosporine A.
  • immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject prior to the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject during the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject following the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject prior to and during the administration of the induced pluripotent stem cells.
  • the one or more immunosuppressive drugs can be administered to the subject during and following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and following the administration of the induced pluripotent stem cells.
  • the cardiac dysfunction of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof is a cardiac ischemia/reperfusion event.
  • the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
  • the subject has a congenital heart defect.
  • the congenital heart defect is hypoplasia or pentalogy of Cantrell.
  • the induced pluripotent stem cells of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof are obtained from an autologous source.
  • the induced pluripotent stem cells are obtained from an allogeneic source.
  • the induced pluripotent stem cells are obtained from a syngeneic source.
  • the induced pluripotent stem cells are obtained from a combination of sources.
  • the induced pluripotent stem cells of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof are obtained from fibroblast cells.
  • the induced pluripotent stem cells are obtained from H9c2 cells.
  • the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor.
  • the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
  • the sternness factors are c-myc, oct 3/4, Klf4, and Sox2.
  • a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
  • the subject is a mammal.
  • the mammal is a primate.
  • the mammal is a human.
  • the human is a patient.
  • a method of generating cardiac induced pluripotent stem cells comprises (i) inserting one or more nucleic acid constructs capable of expressing stem-cell like factors into a cardiac cell type, and (ii) obtaining the cardiac induced pluripotent stem cells stably expressing the stem-cell like factors in the cardiac cell type.
  • the stem-cell like factors comprise Oct3/4, KIf4, Sox2, and c-Myc, or a combination thereof.
  • the induced pluripotent stem cells differentiate into cardiac myocytes.
  • the induced pluripotent stem cells are cardiac-committed.
  • a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
  • the induced pluripotent stem cells are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In yet another aspect, the induced pluripotent stem cells are obtained from fibroblast cells. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor.
  • a disclosed induced pluripotent stem cells are used in a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject.
  • a disclosed induced pluripotent stem cells are used in a method of enhancing cardiac tissue and/or cell regeneration in a subject.
  • the subject is a mammal.
  • the mammal is a primate.
  • the mammal is a human.
  • the human is a patient.
  • a disclosed induced pluripotent stem cells are primed with fibroblast growth factor-8.
  • the induced pluripotent stem cells are administered with one or more of the following: conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • the timing of the administration of the induced pluripotent stem cells varies.
  • the induced pluripotent stem cells are administered prior to administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • the induced pluripotent stem cells are administered during the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • the induced pluripotent stem cells are administered following the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • the administration of induced pluripotent stem cells to the subject is repeated.
  • the induced pluripotent stem cells are administered both prior to and during, or prior to and following, or during and following, the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
  • the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to cardiac dysfunction.
  • the iPS cells are administered to the subject, during cardiac dysfunction.
  • the iPS cells are administered to the subject or following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to and during cardiac dysfunction.
  • the iPS cells are administered to the subject during and following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to and following cardiac dysfunction.
  • the iPS cells are administered to the subject prior to, during, and following cardiac dysfunction.
  • the iPS cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction.
  • the iPS cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction.
  • a disclosed induced pluripotent stem cells improve angiogenesis in a subject.
  • a disclosed induced pluripotent stem cells enhance cardiac function in a subject.
  • a disclosed induced pluripotent stem cells improve angiogenesis and enhance cardiac function in the subject.
  • enhancing cardiac function comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
  • a disclosed induced pluripotent stem cells inhibit or decrease necrosis. In an aspect, a disclosed induced pluripotent stem cells inhibit or decrease apoptosis and/or apoptosis-related mechanisms.
  • iPS cells induced pluripotent stem cells
  • approximately 100,000 iPS cells are administered to a subject.
  • less than 100,000 iPS cells are administered to a subject.
  • more than 100,000 iPS cells are administered to a subject.
  • the induced pluripotent stem cells are administered in one intramycardial injection. In an aspect, the induced pluripotent stem cells are administered in two or more intramycardial injections. In an aspect, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
  • a disclosed induced pluripotent stem cells are administered with one or more immunosuppressive drugs.
  • the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
  • the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs.
  • the one or more immunosuppressive drugs is cyclosporine A.
  • the one or more immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells. 4. IDENTIFYING A MYOCARDIAL INJURY
  • a method of identifying a myocardial injury or infarcted myocardial tissue comprises measuring the amount of miR-486.
  • the amount of miR-486 decreases after myocardial injury or myocardial infarction.
  • the amount of miR-486 increases after treatment of induced pluripotent stem cells.
  • a disclosed method of identifying a myocardial injury or infracted myocardial tissue further comprises measuring the amount of Akt expression, PTEN expression, and/or FOXOla expression.
  • a disclosed method of identifying a myocardial injury or infarcted myocardial tissue further comprises measuring apoptosis and/or apoptosis-related mechanisms.
  • the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression.
  • the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by any combination of the following: an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression.
  • a method of inhibiting apoptosis and/or apoptosis-related mechanisms comprises administering conditioned medium from induced pluripotent stem cells.
  • a method of inhibiting apoptosis and/or apoptosis-related mechanisms comprises administering FGF-8.
  • the conditioned media or the FGF-8 is administered to a subject.
  • the subject has experienced a cardiac ischemia/reperfusion event.
  • the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
  • the subject has a congenital heart defect.
  • the congenital heart defect is hypoplasia or pentalogy of Cantrell.
  • the conditioned media of a disclosed method comprises one or more anti-apoptotic and anti-fibrotic factors.
  • the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, and tissue inhibitor of matrix metalloproteinase- 1.
  • the conditioned medium is administered with or without fibroblast growth factor-8 (FGF-8).
  • the conditioned medium is administered with or without fibroblast growth factor-9 (FGF-9).
  • the conditioned medium is administered with or without interleukin-10 (IL-10).
  • the conditioned medium is administered with or without matrix metalloproteinase- 1 (MMP-1).
  • the conditioned medium is administered with one or more of the following: fibroblast growth factor-9, fibroblast growth factor-8, interleukin- 10, and tissue inhibitor of matrix metalloproteinase- 1.
  • the conditioned medium is administered with any combination of the following: fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, and tissue inhibitor of matrix metalloproteinase- 1, such as, for example, the combination of FGF-8 and FGF-9.
  • a disclosed method protects myocardium including, but not limited to human myocardium, with or without the administration of iPS cells.
  • a disclosed method of inhibiting apoptosis and/or apoptosis- related mechanisms further comprises administering induced pluripotent stem cells.
  • the induced pluripotent stem cells differentiate into cardiac myocytes.
  • the induced pluripotent stem cells are cardiac -committed.
  • a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc.
  • a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
  • the conditioned medium is administered to the subject prior to the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered during the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered following the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered the conditioned medium prior to and during the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered the conditioned medium prior to and following the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered to the subject during and following the
  • the apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells. In an aspect, the apoptosis and/or apoptosis-related mechanisms occurs in cardiac tissue of the subject.
  • a disclosed method increases miR-486 expression, increases Akt expression, decreases PTEN expression, and/or decreases FOXOla expression. In an aspect, a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression. In an aspect, a disclosed composition performs any combination of the following: increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression.
  • a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
  • compositions as investigational and/or research tools in the development and standardization of in vitro and in vivo test systems for evaluation in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic approaches for the enhancement of cardiac tissue and/or cell regeneration as well as the evaluation of the attenuation of adverse cardiac tissue and/or cell remodeling.
  • these approaches apply to subjects with cardiac dysfunction.
  • Ranges can be expressed herein as from “about” one particular value, and/or to "about” another particular value. When such a range is expressed, a further aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about,” it will be understood that the particular value forms a further aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as "about” that particular value in addition to the value itself. For example, if the value "10” is disclosed, then “about 10" is also disclosed. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
  • amino acid abbreviations are conventional one letter codes for the amino acids and are expressed as follows: A, alanine; B, asparagine or aspartic acid; C, cysteine; D aspartic acid; E, glutamate, glutamic acid; F, phenylalanine; G, glycine; H histidine; I isoleucine; K, lysine; L, leucine; M, methionine; N, asparagine; P, proline; Q, glutamine; R, arginine; S, serine; T, threonine; V, valine; W, tryptophan; Y, tyrosine; Z, glutamine or glutamic acid.
  • Peptide refers to any peptide, oligopeptide, polypeptide, gene product, expression product, or protein.
  • a peptide can be an enzyme.
  • a peptide is comprised of consecutive amino acids.
  • the term "peptide" encompasses naturally occurring or synthetic molecules.
  • the biological activity or biological action of a peptide refers to any function exhibited or performed by the peptide that is ascribed to the naturally occurring form of the peptide as measured or observed in vivo (i.e., in the natural physiological environment of the protein) or in vitro (i.e., under laboratory conditions).
  • a biological activity of caspase includes caspase enzymatic activity.
  • enzyme refers to any peptide that catalyzes a chemical reaction of other substances without itself being destroyed or altered upon completion of the reaction.
  • a peptide having enzymatic activity catalyzes the formation of one or more products from one or more substrates.
  • Such peptides can have any type of enzymatic activity including, without limitation, the enzymatic activity or enzymatic activities associated with enzymes such as those disclosed herein.
  • references in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed.
  • X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
  • a weight percent (wt. %) of a component is based on the total weight of the formulation or composition in which the component is included.
  • the terms "transformation” and "trans fection” mean the introduction of a nucleic acid, e.g., an expression vector, into a recipient cell including introduction of a nucleic acid to the chromosomal DNA of said cell.
  • the art is familiar with various compositions, methods, techniques, etc. used to effect the introduction of a nucleic acid into a recipient cell.
  • the art is familiar with such compositions, methods, techniques, etc for both eukaryotic and prokaryotic cells.
  • the art is familiar with such compositions, methods, techniques, etc. for the optimization of the introduction and expression of a nucleic acid into and within a recipient cell.
  • the term "subject" refers to the target of administration, e.g., an animal.
  • the subject of the herein disclosed methods can be a vertebrate, such as a mammal, a fish, a bird, a reptile, or an amphibian.
  • the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent.
  • the term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered.
  • the subject is a mammal.
  • a patient refers to a subject afflicted with a disease or disorder.
  • the term "patient” includes human and veterinary subjects.
  • the subject has been diagnosed with a need for treatment for cardiac dysfunction, such as, for example, a cardiac ischemia/reperfusion event prior to the administering step.
  • treatment refers to the medical management of a subject or a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder, such as, for example, an injured myocardium.
  • This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder.
  • this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.
  • the term covers any treatment of a subject, including a mammal (e.g., a human), and includes: (i) preventing the disease from occurring in a subject that can be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease, i.e., arresting its development; or (iii) relieving the disease, i.e., causing regression of the disease.
  • the disease, pathological condition, or disorder is cardiac dysfunction, such as, for example, a cardiac
  • prevent refers to precluding, averting, obviating, forestalling, stopping, or hindering something from happening, especially by advance action. It is understood that where reduce, inhibit or prevent are used herein, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed.
  • diagnosisd means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by compositions or methods disclosed herein.
  • diagnosis with cardiac dysfunction means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by a compound or composition that alleviates or ameliorates cardiac dysfunction.
  • diagnosis refers to having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition characterized by cardiac dysfunction and/or cardiac impairment and/or cardiac cell death, wherein improving or enhancing cardiac regeneration and/or cardiac function would be beneficial to the subject.
  • a diagnosis can be in reference to a disorder, such as cardiac dysfunction, and the like, as discussed herein.
  • the phrase "identified to be in need of treatment for a disorder," or the like, refers to selection of a subject based upon need for treatment of the disorder.
  • a subject can be identified as having a need for treatment of a disorder (e.g., a disorder related to cardiac dysfunction or myocardial infarction) based upon an earlier diagnosis by a person of skill and thereafter subjected to treatment for the disorder.
  • the identification can, in one aspect, be performed by a person different from the person making the diagnosis.
  • the administration can be performed by one who subsequently performed the administration.
  • administering and “administration” refer to any method of providing a pharmaceutical preparation to a subject. Such methods are well known to those skilled in the art and include, but are not limited to, intracardiac administration, oral administration, transdermal administration, administration by inhalation, nasal
  • a preparation can be administered therapeutically; that is, administered to treat an existing disease or condition.
  • a preparation can be administered prophylactically; that is, administered for prevention of a disease or condition.
  • contacting refers to bringing a disclosed compound and a cell, target receptor, or other biological entity together in such a manner that the compound can affect the activity of the target (e.g., receptor, transcription factor, cell, etc.), either directly; i.e., by interacting with the target itself, or indirectly; i.e., by interacting with another molecule, co-factor, factor, or protein on which the activity of the target is dependent.
  • the target e.g., receptor, transcription factor, cell, etc.
  • determining can refer to measuring or ascertaining a quantity or an amount or a change in expression and/or activity level, e.g., of a nucleotide or transcript or polypeptide.
  • determining the amount of a disclosed transcript or polypeptide in a sample as used herein can refer to the steps that the skilled person would take to measure or ascertain some quantifiable value of the transcript or polypeptide in the sample.
  • the art is familiar with the ways to measure an amount of the disclosed nucleotides, transcripts, polypeptides, etc.
  • the term "level" refers to the amount of a target molecule in a sample, e.g., a sample from a subject.
  • the amount of the molecule can be determined by any method known in the art and will depend in part on the nature of the molecule (i.e., gene, mRNA, cDNA, protein, enzyme, etc.). The art is familiar with quantification methods for nucleotides (e.g., genes, cDNA, mRNA, etc) as well as proteins, polypeptides, enzymes, etc. It is understood that the amount or level of a molecule in a sample need not be determined in absolute terms, but can be determined in relative terms (e.g., when compare to a control or a sham or an untreated sample).
  • the terms “effective amount” and “amount effective” refer to an amount that is sufficient to achieve the desired result or to have an effect on an undesired condition.
  • a “therapeutically effective amount” refers to an amount that is sufficient to achieve the desired therapeutic result or to have an effect on undesired symptoms, but is generally insufficient to cause adverse side affects.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed and like factors well known in the medical arts.
  • module is meant to alter, by increase or decrease.
  • modulator can mean a composition that can either increase or decrease the expression level or activity level of a gene or gene product such as a peptide. Modulation in expression or activity does not have to be complete. For example, expression or activity can be modulated by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression or activity of a gene or gene product has not been modulated by a composition.
  • EC5 0 is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% enhancement or activation of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc.
  • EC5 0 also refers to the concentration or dose of a substance that is required for 50% enhancement or activation in vivo, as further defined elsewhere herein.
  • EC50 can refer to the concentration or dose of compound that provokes a response halfway between the baseline and maximum response. The response can be measured in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest.
  • the response can be measured in vitro using cultured cardiac cells or in an ex vivo organ culture system with isolated cardiac cells, e.g., cardiomyocytes, vascular smooth muscle cells, endothelial cells, etc).
  • the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats.
  • the mouse or rat can be an inbred strain with phenotypic characteristics of interest such as, for example, obesity or diabetes.
  • the response can be measured in a transgenic or knockout mouse or rat wherein a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.
  • IC 50 is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% inhibition or diminution of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. IC 50 also refers to the concentration or dose of a substance that is required for 50% inhibition or diminution in vivo, as further defined elsewhere herein.
  • a substance e.g., a compound or a drug
  • IC 50 also refers to the half maximal (50%) inhibitory concentration (IC) or inhibitory dose of a substance.
  • the response can be measured in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest.
  • the response can be measured in vitro using cultured cardiac cells or in an ex vivo organ culture system with isolated cardiac cells (e.g., cardiomyocytes, vascular smooth muscle cells, endothelial cells, etc).
  • the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats.
  • the mouse or rat can be an inbred strain with phenotypic characteristics of interest such as, for example, obesity or diabetes.
  • the response can be measured in a transgenic or knockout mouse or rat wherein the a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.
  • pharmaceutically acceptable describes a material that is not biologically or otherwise undesirable, i.e., without causing an unacceptable level of undesirable biological effects or interacting in a deleterious manner.
  • pharmaceutically acceptable carrier refers to sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use.
  • aqueous and nonaqueous carriers, diluents, solvents or vehicles examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol and the like
  • carboxymethylcellulose and suitable mixtures thereof such as vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid and the like. It can also be desirable to include isotonic agents such as sugars, sodium chloride and the like.
  • Prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents, such as aluminum monostearate and gelatin, which delay absorption.
  • Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide, poly(orthoesters) and poly(anhydrides). Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use.
  • Suitable inert carriers can include sugars such as lactose.
  • at least 95% by weight of the particles of the active ingredient have an effective particle size in the range of 0.01 to 10 micrometers.
  • compositions of the invention Disclosed are the components to be used to prepare a composition of the invention as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds can not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular compound is disclosed and discussed and a number of modifications that can be made to a number of molecules including the compounds are discussed, specifically contemplated is each and every combination and permutation of the compound and the modifications that are possible unless specifically indicated to the contrary.
  • iPS cells expressed pluripotency markers, such as alkaline phosphatase and Oct3/4, was next determined.
  • pluripotency markers such as alkaline phosphatase and Oct3/4.
  • growing iPS cells were positive for the undifferentiated pluripotent marker Oct3/4 (which was compared with ES cells as a positive control).
  • Figure ID also shows RFP expressing ES and iPS cells (b and f); DAPI staining for ES and iPS cells (c and g), and a merged image Oct3/4, RFP, and DAPI of ES and iPS cells (d and h).
  • iPS cell lines retained the capability in vitro to differentiate into all three major heart cell types (i.e., cardiomyocytes, vascular smooth muscle cells and endothelial cells) present and still express the desired reporter protein was evaluated.
  • major heart cell types i.e., cardiomyocytes, vascular smooth muscle cells and endothelial cells
  • FIG. 2B shows that iPS cells form embryoid bodies (EBs) and differentiate into cardiac myocytes, smooth muscle cells and endothelial cells.
  • EBs were differentiated for 17 days.
  • a beating area i.e., cardiac myocytes
  • the merged image (2B-C) shows co-expression of myosin and RFP.
  • Smooth muscle cells were stained with anti-smooth muscle actin/Alexa 488 (2B-D), anti- RFP/Alexa 568 (2B-E), and DAPI (2B-F).
  • the merged image (2B-F) shows overlap of expression of smooth muscle a-actin and RFP. Endothelial cells that have formed a capillarylike structure were stained with anti-von Willebrand Factor/Alexa 488 (2B-G), anti- RFP/Alexa 568 (2B-H), and DAPI (2B-I).
  • the merged image (2B-I) shows co-expression of von Willebrand Factor and eGFP.
  • Figure 2C (top panel) shows the image of a western blot using anti-a-actin and anti- -actin following FGF-8 treatment (0 ng per mL and 4 ng per mL).
  • Figure 2C (bottom panel) shows the quantification of the western blot data.
  • FGF-8 significantly increased EB proliferation by 20-30% on D5 as compared to the controls (p ⁇ 0.05).
  • Figure 3A the results are plotted as a percentage increase in beating EBs at D0-D17.
  • the percentage of rhythmically beating EBs after treatment with FGF-8 (4 ng) was significantly greater compared with controls (37% versus 20%, at D17 after plating, *p ⁇ 0.05).
  • the data are from the sets of three-five independent experiments.
  • Figure 3B shows the staining of iPS cells treated with FGF-8 (top panel) and iPS cells not treated without FGF-8 with anti-a-actin and anti-RFP antibodies. 29.2% of cells that stained positively for a-actin from beating area.
  • H9c2 cells treated with 50-400 ⁇ of 3 ⁇ 4(3 ⁇ 4 demonstrated significant decrease in cell survival at the tested concentrations.
  • FGF-8 independently inhibited H2O2 induced apoptosis in this model was determined.
  • Cell were treated with 4 ng of FGF-8 following H2O2 treatment.
  • FGF-8 alone significantly reduced apoptosis.
  • Treatment with both iPS-CMs generated a more robust reduction in apoptosis then treatment with FGF-8 alone ( Figure 4).
  • Data are from the set of 8-10 independent experiments. This data indicated that FGF-8 as well as iPS-CMs prepared from iPS cells with or without FGF-8 treatment act as an anti- apoptotic factor as well.
  • FGF-8 demonstrated significant (p ⁇ 0.05) inhibition of H2O2 induced apoptosis in H9c2 cells. This was confirmed by apoptotic ELISA ( Figure 4A). The inhibition of apoptosis by iPS-CM as determined with apoptotic ELISA kit was significantly lower (p ⁇ 0.05) than the amount inhibited by FGF-8 alone ( Figure 4). The apoptotic ELISA data indicate the presence of factors other than FGF-8 that are also released into iPS-CMs and contribute in the inhibition of H2O2 induced apoptosis in H9c2 cells.
  • Figure 4B shows cell death detection (4B-i), caspase activity (4B-ii), tunnel % apoptotic nuclei (4B-iii), and trypan % apoptotic cells (4B-iv) following treatment with H2O2 (H); or H2O2 + conditioned medium (HICM); or H2O2 + conditioned medium + 4 ng of FGF- 8 (HICM4ng); or H 2 0 2 + 4 ng of FGF-8 (H4ng). Control is indicated as C.
  • cytokine and/or growth factors present in the iPS-CMs that can provide protection from FLCVinduced apoptosis were identified. 69 cytokine and/or growth factors were analyzed with the use of Luminex technology (Singla et al, 2007).
  • Cytoprotective proteins released into both iPS-CM and FGF-8-iPS-CM were present at higher levels as compared with H9c2-CM, ES-CM, or cell culture media used as a control. Three major proteins known to inhibit apoptosis and enhance cardiac cell proliferation in the embryonic hearts were identified (Table 5).
  • Myocardial infarctions were produced in C57BL/6 mice and iPS, ES,
  • H9c2 cells, or cell culture medium were transplanted post-MI.
  • a et al. 201 1; Fatma et al, 2010; Kumar et al, 2005
  • two intramyocardial injections of 10 ⁇ ⁇ of medium ⁇ 2.5 x 104 iPS cells were delivered into infarct border zones.
  • MI groups were transplanted with cell culture medium as a control. Hearts were removed and examined at 2 weeks after cell transplantation.
  • Cardiac myocyte specific anti-sarcomeric a-actin antibody was used to identify cardiac myocytes ( Figure 5A; panels a, e, I, and m), anti-RFP antibody was used to identify donor cells (Figure 5A; panel b, f, j, and n), and DAPI staining identified nuclei ( Figure 5A; panels c, g, k, and o). Merged images of all three stainings are shown in Figure 5A (panels d, h, 1, and p).
  • TUNEL staining TUNEL staining, caspase-3 immunostaining, and caspase-3 activity.
  • FIG. 6A TUNEL staining data showed a reduced amount of red apoptotic nuclei in MI + iPS or ES cell groups as compared to the wide spread red apoptotic nuclei in MI and MI + H9c2 cell groups ( Figure 6A).
  • DAPI stained total nuclei Figure 6A; panels b, e, h, and k
  • merged images are visible ( Figure 6A; panels c, f, i and 1).
  • the scale bar 50 ⁇ .
  • Figure 6B shows anti-sarcomeric a-actin staining ( Figure 6B; panels a and f),
  • caspase-3 activity was significantly (p ⁇ 0.05) reduced as compared to MI, and that H9c2 cells demonstrated no decrease in caspase-3 activity (Figure 6D).
  • the caspase-3 activity data coupled with the TUNEL apoptotic nuclei staining confirmed apoptosis in the heart.
  • MMP-2 and MMP-9 both of which are well-documented MMPs involved in the
  • FIG. 7 shows that hearts transplanted with ES and iPS cells demonstrated significantly reduced MMP-2 concentration as compared to MI and H9c2 cell controls.
  • Figure 7 shows that hearts transplanted with ES and iPS cells demonstrated significantly reduced MMP-9 concentration as compared to MI and H9c2 cell controls. (*p ⁇ 0.05 vs. sham, #p ⁇ 0.05 vs. MI and MI + H9c2).
  • Figures 13A-E show histological staining (trichome staining) demonstrating the effect of sham-MI, MI, and MI with FGF-8 treatment.
  • Figure 13G shows the volume of interstitial fibrotic area.
  • Figure 13H shows the comparison of vascular fibrosis (VF) vs. vascular area (VA) (in %) following sham-MI, MI, and MI with FGF-8 treatment.
  • VF vascular fibrosis
  • VA vascular area
  • FIG. 8A shows representative photomicrographs from Masson's trichrome stained heart sections from the various treatment groups. LV remodeling was determined by measuring total interstitial fibrotic area in mm 2 in all mice groups transplanted with or without stem cells.
  • Heart sections obtained from infarcted hearts transplanted with iPS and ES cells showed a significant (p ⁇ 0.05) decrease in fibrosis (0.25 ⁇ 0.09 mm 2 for MI + iPS cells and 0.21 ⁇ .05 mm 2 for MI+ES cells) as compared to the large fibrotic areas in MI and H9c2 cell transplanted hearts (0.94 ⁇ 0.03 mm 2 for MI and 0.9 ⁇ 07 mm 2 for MI + H9c2 cells, Figure 8B).
  • M-mode echocardiography was used to determine the effect of cell transplantation on LV size and function in mice two weeks post-MI.
  • Fractional shortening (FS) was improved (*p ⁇ 0.05) by H9c2 cell transplantation, ES cells, and cardiac iPS cells.
  • FS Fractional shortening
  • iPS cells showed a robust increase in FS as compared to H9c2 cells (*p ⁇ 0.05 vs. H9c2 and ES cells) and as compared to MI + cell culture medium (#p ⁇ 0.001 vs. MI) ( Figure 8C and 8D).
  • Figure 14A-F shows the results of various outcome measures including fractional shortening and % ejection fraction following sham-MI, MI, and MI with FGF-8 treatment.
  • LVIDd left ventricular internal diameter in diastole
  • LVIDs left ventricular internal diameter in systole
  • EDV end-diastolic volume
  • ESV end-systolic volume
  • FGF-8 is a novel anti-apoptotic factor
  • FIG 4 Two intramyocardial injections of 20 ⁇ ⁇ FGF-8 containing 50 ng/niL were performed. Hearts were removed and examined at 2 weeks post-MI. As shown in Figure 9A and Figure 9B, TU EL staining showed that apoptosis post-MI was significantly reduced in the FGF-8 treated MI hearts as compared to non-FGF-8 treated MI hearts (*p ⁇ 0.05). Figure 9C shows the casapse-3 activity under the same experimental conditions.
  • Figure 9D and Figure 9E shows the effect of FGF-8 treatment on the expression of Bcl-2 and Bax.
  • the Bcl-2 family consists of more than twenty anti- and pro-apoptotic members that modulate the balance between life and death.
  • Figure 9D shows western blotting of sham, MI, and MI+FGF-8 treated samples with anti-Bcl-2 and anti -beta-actin (control) antibodies.
  • Figure 9E (top panel) shows western blotting of sham, MI, and MI+FGF-8 treated samples with anti-Bax and anti- beta-actin (control) antibodies.
  • the MI heart data indicated a significant increase in cardiac apoptosis. This increase was blunted following iPS cell transplantation (see Figure 6).
  • a miRNA signature panel of 365 different miRNAs (rodent taqman array) was used to determine up- or down- regulation of various miRNAs with and without cell transplantation post-MI. As shown in Figure 10, the levels of miR-486 were significantly decreased following MI and this decrease was inhibited following iPS cell transplantation), indicating that miR-486 is a cell survival miRNA. (*p ⁇ 0.001 vs. sham, #p ⁇ 0.05 vs. MI).
  • FIG. 11C shows a western blot of sham, MI, and MI+FGF-8 utilizing anti-PTEN, anti-P- PTEN, and anti-Beta-actin antibodies.
  • Figure 1 ID shows the quantitation of these western blot experiments.
  • Figure 1 IE shows p-AKT expression in following sham treatment (sham), myocardial infarction (MI), and myocardial infarction with FGF-8 (MI+FGF8).
  • Figure 1 IF shows lipid peroxidase expression following sham treatment (sham), myocardial infarction (MI), and myocardial infarction with FGF-8 treatment (MI+FGF8).
  • FGF-8 is a predominant cardiac growth factor, which plays a role in the development of heart. (Ilagan et al., 2006; Marques et al, 2008; Reifers et al, 2000). FGF-8 is a growth factor which plays a role in heart development as explained above. (Ilagan et al, 2006). To date, the role of FGF-8 in adult heart regeneration or with iPS cell treatment has never been explored. However, as demonstrated herein, FGF-8 (i) acts as an anti-apoptotic factor in the adult infarcted heart, and (ii) activates iPS cells and stimulates differentiation into cardiac myocytes.
  • BrdU 50 mg/kg body weight, i.p. (Beltrami et al, 2003), which identifies nuclei in S phase in order to label actively dividing cells, is administered to the subject. Active cell growth of engrafted cells is identified using double-label immunostaining for RFP and BrdU (Dako). RFP identifies donor engrafted cells and BrdU identifies DNA synthesizing cells. To determine how many cells have started cell division at any given time, Ki-67 is utilized.
  • Ki-67 is a nuclear antigen that is associated with cell division and that labels proliferating cells (G1-, S-, G2 -phase and mitosis), but does not label quiescent or resting cells (GO-phase). Double-label immunostaining is used to detect proliferating cells using Ki-67 (Dako) antibodies and donor engrafted cells using RFP antibodies. Slides are analyzed using fluorescence (Olympus) and confocal microscopy.
  • Cell differentiation is determined using double-label immunostaining to detect donor cells (RFP antibody) and differentiated cell types using cell specific antibodies.
  • Cardiac-specific antibodies such as cardiac myosin heavy chain (MF-20 antibody, hybridoma bank), sarcomeric a-actin (Sigma), a-actinin, cardiac troponin I (Spectral Diagnostic), and connexin-43 (Chemicon) are used to detect cardiac myocytes and gap junctions. Endothelial cells are detected with factor VIII (Sigma) and Griffonia simplicifolia lectin (Sigma) antibodies. For smooth muscle cells, a-smooth muscle actin antibody (sigma) is used. To determine the newly formed progenitor cardiac myocytes, sections are immunostained with cardiac specific transcriptional factors such as GATA-4, Nkx2.5, and MEF2 (Santa Cruz Biotechnology). Sections are then stained with respective FITC or rhodamine conjugated secondary antibodies and counterstained with DAPI for nuclear visualization. Slides are analyzed using Olympus fluorescence and confocal microscopy.
  • ELISA Densitometry is used to measure band density.
  • ELISA is performed with a kit (R & D, Minneapolis) to measure MMP-2 and MMP-9 in the LV tissue homogenates.
  • Echocardiography is an accurate noninvasive tool for the determination of quantitative characterization of post- MI heart remodeling in the mouse model. (Singla et al, 201 1; Fatma et al, 2010). Echocardiography is done on all groups post-MI hearts at short- term and long-term time points. M-mode images (Singla et al, 201 1 ; Fatma et al., 2010) in a short axis view is performed to measure LV mass, LV mass to body ratio, LV anterior and posterior wall thickness, and fractional shortening.
  • Both human and mouse ES cells form complex teratomas when engrafted into an immune deficient host, a characteristic of ES cells resulting from their pluripotential capacity (Damjanov et al, 2004; Thomson et al, 1998). Teratoma formation is considered a major potential limitation in the therapeutic use of ES cells, especially when the transplanted undifferentiated number of cells is higher than 100,000 (Nussbaum et al, 2007). It has been suggested that intramyocardial transplantation of lxlO 6 ES or iPS (generated from fibroblasts) cells in the mouse heart causes teratomas (Nelson et al, 2009; Nussbaum et al, 2007).
  • iPS cells from various sources may vary in their epigenetic alterations (Kim et al, 2010).
  • iPS cells are generated from H9c2 cells using plasmid transfection. This method generation, the number of injected cells, and the location of injection are completely different than those studies reporting
  • C57BL/6 mice are treated with cyclosporine A (250-300 mg/kg) for 5 days before the cell transplantation (Menard et al, 2005; Giralt et al, 1997).
  • the cyclosporine A dose can be altered based on the desire for an average cyclosporine serum concentration of 300-400 ng/L (Menard et al, 2005; Giralt et al, 1997).
  • Animals are killed humanely at short- term (1, 14, and 28 days) and long-term (8, 12, and 18 weeks) time points after cell transplantation. Tissues are harvested for histological analysis and preparation of tissue homogenates.
  • FCC-iPS are treated as follows; FGF-8 (4 ng/mL) is added to the iPS cells in the differentiation medium (a medium which does not contain iPS cell self-renewal growth factors such as activin A, leukemia inhibitory factor and mouse embryonic fibroblast condition medium) for 48 hours. These iPS cells are considered as FGF-8 primed differentiated iPS cells. Control iPS or ES cells are cultured for 48 hours in a differentiation medium without FGF-8.
  • FGF-8 4 ng/mL
  • the differentiation medium a medium which does not contain iPS cell self-renewal growth factors such as activin A, leukemia inhibitory factor and mouse embryonic fibroblast condition medium
  • any infiltration of T-cells are identified using anti-CD3 and anti-CD4 antibodies (Zhao et al, 2011). H&E stained sections are used to identify the presence of any inflammatory cells.
  • Heart sections are prepared (Singla et al, 2011) for immunostaining to determine apoptosis and cell proliferation. Sections are stained with a RFP or GFP antibody along with a Oct3/4 (undifferentiated cell marker on iPS or ES cells) antibody to determine total number of engrafted cells. TU EL staining are combined with a caspase-3 antibody to establish the extent of cell death at each time point. (Singla et al, 2011 ; Singla et al, 2006; Singla et al, 2007). Sections are counter-stained with DAPI (Vectashield) to localize the nucleus.
  • DAPI Vectashield
  • engrafted cell apoptosis sections are stained with RFP, Oct3/4, and TUNEL.
  • Cell specific apoptosis within the host myocardium is determined using double- label immunostaining to detect apoptotic cells (TUNEL, Bax, and caspases) and native heart cell types.
  • Cardiac myocytes are detected with antibodies against cardiac myosin heavy chain (MF-20 antibody, hybridoma bank), sarcomeric a-actin (Sigma), a-actinin, and cardiac troponin I (Spectral Diagnostic). (Singla et al, 2011 ; Singla et al, 2006).
  • Endothelial cells are detected with factor VIII (Sigma) and Griffonia simplicifolia lectin (Sigma) antibodies.
  • factor VIII Griffonia simplicifolia lectin
  • For smooth muscle (SM) cells SM a-actin, or SM myosin heavy chain antibodies (Sigma) are used.
  • An antibody against fibronectin (Sigma) is used to detect fibroblasts. Sections will then be stained with FITC or rhodamine conjugated secondary antibodies and counterstained with DAPI for nuclear visualization. Olympus fluorescence and confocal microscopy are used to assess apoptosis.
  • Heart sections are stained with Mason's trichrome and Sirius red F3BA or
  • H&E, Masson's Trichrome, and immunostained sections are used to quantify fibrosis, apoptosis, and distribution of heart cell types in the left ventricular (LV) region of the heart.
  • LV left ventricular
  • IMA Integrated morphometry analysis
  • NIH image J processing software are employed for these analyses.
  • Acute MI is associated with irreversible cell loss due to apoptosis and necrosis (Anversa et al, 1997; Anversa et al, 1998; Kumar et al., 2002).
  • Adult or embryonic stem cells have the potential to form new cardiac cells in the injured myocardium (Singla et al, 2009).
  • Some studies suggest that an immediate release of autocrine or paracrine factors from donor stem cells decrease host myocardium remodeling (Singla et al, 2009; Gnecchi et al, 2005; Singla et al, 2010).
  • iPS-CM which can contain numerous cytoprotective factors, does not pose a threat of teratoma formation, such an injection offers the possibility of beneficial effects on cell transplantation.
  • iPS-CM from iPS-cells primed with FGF-8 and without FGF-8 is utilized
  • a myocardial infarction (MI) model for C57BL/6 mice having received or not having received cyclosporine A is utilized. If cyclosporine A generates no significant difference in cardiac remodeling, the use of cyclosporine A can be discontinued.
  • FCC-iPS-CM 15x
  • a cocktail of FGF-8 20 ⁇ .
  • the levels of released factor are compared to the controls and increases are confirmed with western blotting.
  • Heart sections are prepared for immunostaining and the degrees of apoptosis and fibrosis are evaluated. H&E, Masson's Trichrome, and Sirius red F3BA or PSR staining are used to analyze the effect of iPS cells on cell morphology and gross interstitial and perivascular fibrosis.
  • SDS-PAGE and Western blot analyses are utilized to determine the protein expression of collagen types I and III (Santa Cruz Biotechnology), MMP-2 and MMP-9 (Santa Cruz Biotechnology/Cell Signaling), and TIMP-1 (Santa Cruz Biotechnology).
  • ELISA Densitometry is used to measure band density.
  • ELISA is performed with a kit (R & D, Minneapolis) to measure MMP-2 and MMP-9 in the LV tissue homogenates.
  • Echocardiography is an accurate noninvasive tool for the determination of quantitative characterization of post- MI heart remodeling in the mouse model. (Singla et al, 2011; Fatma et al, 2010). Echocardiography is done on all groups post-MI hearts at short- term and long-term time points. M-mode images (Singla et al, 2011; Fatma et al., 2010) in a short axis view is performed to measure LV mass, LV mass to body ratio, LV anterior and posterior wall thickness, and fractional shortening.
  • miRNAs are small non-coding RNAs about 20-24 nucleotides in length that play a major role in the regulation of a variety of cell processes including apoptosis, hypertrophy, fibrosis, and cardiac differentiation (Care et al, 2007; Catalucci et al, 2008; Chen et al, 2006; Cimmino et al, 2005). Certain miRNAs such as miR-21, miR-133 and miR- 1 are involved in various pro- and anti-apoptotic and anti-fibrotic activities in the heart (Care et al, 2007; Catalucci et al, 2008; Chen et al., 2006; Cimmino et al, 2005).
  • miR-486 was down-regulated following ML, and that iPS cell transplantation significantly increased the levels of miR-486 following MI. While there are a few published papers describing miR-486 induction of myoblast differentiation, miR-486 involvement in cell survival in Sezary syndrome, and increased levels of miR-486 in the postnatal cardiac growth (Dey et al, 201 1; Narducci et al, 201 1; Small et al, 2010), there is no data regarding miR-486 levels and apoptosis following iPS cell transplantation.
  • FCC-iPS is treated as follows; FGF-8 (4 ng/mL) is added to the iPS cells in the differentiation medium (a medium which does not contain iPS cell self-renewal growth factors such as activin A, leukemia inhibitory factor and mouse embryonic fibroblast condition medium) for 48 hours. These iPS cells are considered as FGF-8 primed differentiated iPS cells.
  • FGF-8 4 ng/mL
  • iPS-CM from iPS-cells primed with FGF-8 and without FGF-8 is utilized.
  • LV are removed and miRNA is isolated using miRVANA kit (ABI).
  • miR-486 the levels of miR-486 are ascertained. miR-486 is also confirmed with northern blotting (Dey et al, 2011 ; Narducci et al, 201 1 ; Small et al, 2010). To determine the role of miR-486 on cardiac apoptosis, animals are injected with a selective inhibitor of miR-486 (locked nucleic acid, LNA-antagomir-486, 80 mg/kg) (Hatley et al, 2010; Patrick et al., 2010). To identify expression of miR-486 in the infarcted, non-infarcted, and peri-infarcted region post-MI, heart sections are stained using in situ-hybridization for miR-486.
  • a selective inhibitor of miR-486 locked nucleic acid, LNA-antagomir-486, 80 mg/kg
  • In situ- hybridization employing LNA-miR-486 specific probes (Exiqon) (Hosoda et al, 201 1) is utilized.
  • the expression of miR-486 in major heart cell types is characterized using double label immunostaining with cell specific antibodies.
  • sections are co-labeled with heart cell specific antibodies such as cardiac a-actin for cardiac myocytes, CD31 for endothelial cells and smooth muscle, a-actin for VSM cells as described above. Sections are counter-stained with DAPI to identify cell nuclei.
  • PTEN/FOXOIa targets and associated Akt are affected at the translational level proteins.
  • SDS-PAGE and Western blot analyses are performed to determine the protein expression of total and phosphorylated PTEN, FOXOla, and Akt (Cell Signaling). Densitometry is used to measure band density. Additionally, ELISA is performed with a kit (Ray Biotech Inc.) to measure total and phosphorylated PTEN, FOXOla, and Akt.
  • luciferase reporter vectors To confirm whether miR-486 directly binds to the 3 '-UTRs of its predicted target genes, PTEN and Foxola, luciferase reporter vectors are utilized.
  • the pCMV-Luc- 3UTR vector was constructed by sequential cloning the firefly luciferase gene (obtained from Promega) to the pcDNA 3.1 expression vector (Invitrogen) under the control of a CMV promoter.
  • luciferase reporter assay For the luciferase reporter assay, the 3 '-UTR sequences for mouse PTEN ( ⁇ 3.5kb in length) and Foxola ( ⁇ 1 kb in length) were generated by high-fidelity PCR and then cloned downstream of the firefly luciferase stop codon. HEK 293 cells are grown in 6-well plates until they reach 80% confluency. Luciferase reporter constructs are co-transfected to the cells with a negative control vector or miR-486 expression vector in the presence or absence of a specific synthetic antagomir against miR-486. The luciferase assay is performed 48 hours after transfection using a luciferase assay kit (Promega) according to the manufacturer' instructions. Luminometer is used to quantify luciferase activity.
  • Interleukin-6 is a component of human umbilical cord serum and stimulates hematopoiesis in embryonic stem cells in vitro. Exp. Hematol. 21, 774- 778 (1993).
  • Cimmino,A. et al. miR-15 and miR-16 induce apoptosis by targeting BCL2 9.
  • Ilagan,R. et al. FGF-8 is required for anterior heart field development 1.
  • Tissue inhibitor of matrix metalloproteinase 1 controls adipogenesis in obesity in mice and in humans.
  • Diabetologia 54, 1468-1479 (201 1).
  • CD34+CXCR4+ cells and non-selected mononuclear cells in patients with acute STEMI and reduced left ventricular ejection fraction results of randomized, multicentre Myocardial Regeneration by Intracoronary Infusion of Selected Population of Stem Cells in Acute Myocardial Infarction (REGENT) Trial 1. Eur. Heart J. 30, 1313-1321 (2009).

Abstract

In an aspect, the invention relates to compositions and methods for enhancing cardiac tissue and/or cell regeneration. In an aspect, the invention relates to compositions and methods for attenuating adverse cardiac tissue and/or cell remodeling. In an aspect, the invention relates to compositions and methods for inhibiting apoptosis. In an aspect, the invention relates to compositions and methods for identifying a myocardial injury. This abstract is intended as a scanning tool for purposes of searching in the particular art and is not intended to be limiting of the present invention.

Description

METHODS AND COMPOSITIONS USING FGF-8 TO ENHANCE CARDIAC REGENERATION AND ATTENUATE ADVERSE CARDIAC REMODELING
CROSS REFERENCE TO RELATED PATENT APPLICATIONS
[0001] This application claim priority to U.S. Provisional Patent Application No. 61/617,985 filed March 30, 2012, which is herein incorporated by reference in its entirety, and is related to U.S. Provisional Application No. 61/618,280 filed March 30, 2012, titled "Methods and Compositions Using FGF-9 to Enhance Neovascularization and Regeneration," by Dinendar Singla, which is herein incorporated by reference in its entirety.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
[0002] Parts of this invention were made with government support under 5R01HL090646-04 awarded by the National Institutes of Heath. The United States government has certain rights in the invention.
BACKGROUND
[0003] Myocardial infarction (MI) leads to heart failure implicated by complex mechanisms of cardiac myocyte cell death, hypertrophy, and fibrosis. The common therapeutic preferences to inhibit further deterioration of MI leading to end stage heart failure are very restricted. Heart transplantation required to manage end stage heart failure patients is not readily available. Therefore, new cellular therapies to repair and regenerate injured myocardium as well as to prevent the progression of adverse cardiac remodeling are under diligent examination.
[0004] Thus far, cell transplantation in the mouse, rat, and human infarcted hearts has been studied employing a wide variety of cell types such as skeletal myoblasts, embryonic stem (ES) cells, c-kit positive cardiac stem cells, CD45+ve bone marrow stem cells, mesenchymal stem cells (MSC), and hematopoietic stem cells. Indeed, adult stem cells in clinical trials have furnished significant insights into cell transplantation; however, their effectiveness is still in question. In contrast, ES cells have never reached clinical trials due to ethical and additional concerns. Thus, the optimal cell types for cardiac regeneration, repair and remodeling for transplantation in the injured myocardium has yet to be identified.
[0005] Despite advances in understanding the physiology and pathophysiology of cardiac dysfunction, including myocardial infarction, there is still a scarcity of compounds that are both potent, efficacious, and safe in the treatment of cardiac dysfunction. These needs and other needs are satisfied by the present invention. SUMMARY
[0006] Disclosed herein is a method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the method comprising (i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) administering to the subject fibroblast growth factor-8; and/or (iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9, and determining regeneration in the heart.
[0007] Disclosed herein is a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the method comprising (i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) administering to the subject fibroblast growth factor-8; and/or (iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9, and determining a decrease in cardiac remodeling.
[0008] Disclosed herein is a composition for enhancing cardiac tissue/cell regeneration in a subject, comprising (i) fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[0009] Disclosed herein is a composition for attenuating adverse cardiac tissue/cell remodeling in a subject, comprising (i) fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) fibroblast growth factor-8 and fibroblast growth factor-9.
[0010] Disclosed herein is a method of generating cardiac induced pluripotent stem cells, the method comprising (i) inserting one or more nucleic acid constructs capable of expressing stem-cell like factors into a cardiac cell type, wherein the stem-cell like factors comprise Oct3/4, KIf4, Sox2, and c-Myc, or a combination thereof; and (ii) obtaining the cardiac induced pluripotent stem cells stably expressing the stem-cell like factors in the cardiac cell type.
[0011] Disclosed herein is a method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising administering conditioned medium from induced pluripotent stem cells, wherein the conditioned medium is administered with or without FGF-8.
[0012] Disclosed herein is a method of identifying a myocardial injury or infarcted myocardial tissue, comprising measuring the amount of miR-486, wherein the amount of miR-486 decreases after myocardial injury or myocardial infarction, and wherein the amount of miR-486 increases after transplantation of induced pluripotent stem cells.
[0013] Disclosed herein is a method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising administering FGF-8.
BRIEF DESCRIPTION OF THE FIGURES
[0014] The accompanying figures, which are incorporated in and constitute a part of this specification, illustrate several aspects and together with the description serve to explain the principles of the invention.
[0015] Figure 1 shows indicia of stably transfected iPS cells.
[0016] Figure 2 shows that iPS cells generated from embryoid bodies differentiate into cardiac myocytes, smooth muscles ells, and endothelial cells following FGF-8 treatment.
[0017] Figure 3 shows the effects of FGF-8 on beating cardiac myocytes derived from the iPS-EB cell system.
[0018] Figure 4 shows the effects of both iPS-CM and FGF-8 on H9c2 cells following H202- induced apoptosis.
[0019] Figure 5 shows that transplanted RFP-iPS or ES cells differentiate into cardiac myocytes with electrical coupling.
[0020] Figure 6 shows that transplanted iPS cells inhibit apoptosis at 2 weeks post-MI.
[0021] Figure 7 shows MMP-2 expression in ES and iPS cell transplanted hearts.
[0022] Figure 8 shows reduced fibrosis and improved cardiac function following iPS cell transplantation.
[0023] Figure 9 shows the effect of FGF-8 treatment on cardiac myocyte apoptosis following MI and the effect of FGF-8 treatment on Bax and Bcl-2 expression.
[0024] Figure 10 shows a decrease in levels of miR-486 following MI and the effect of iPS cell transplantation.
[0025] Figure 11 shows the effect of treatment with ES and iPS cells following MI on p-Akt and PTEN expression as well as the effect of FGF-8 treatment on p-Akt and PTEN expression.
[0026] Figure 12 shows expression data of c-kit and notchl following sham treatment and myocardial infarction both with and without FGF-8 treatment. [0027] Figure 13 shows histological and quantifiable data regarding the effect of sham-MI, MI, and MI+FGF-8 treatment on interstitial fibrosis and vascular fibrosis / vascular area (VF/VA).
[0028] Figure 14 shows various cardiac performance measures following sham-MI, MI, and MI with FGF-8 treatment.
[0029] Additional advantages of the invention will be set forth in part in the description which follows, and in part will be obvious from the description, or can be learned by practice of the invention. The advantages of the invention will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed.
DETAILED DESCRIPTION
[0030] Induced pluripotent stem (iPS) cells could be a potential cell type of choice to treat heart diseases but their basic mechanisms of cardiac repair and regeneration are largely unknown. Using four (4) factors (Oct3/4, Sox2, Klf4, and c-Myc), iPS cells were generated from H9c2 cells (originally isolated from embryonic rat heart ventricular tissue). These cells have the potential to differentiate into cardiac myocytes in vitro and in vivo. Following myocardial infarction, transplanted cardiac iPS cells inhibited post-MI remodeling and demonstrated cardiac regeneration.
[0031] Data indicate that FGF family members play an important role in various cell signal transduction pathways associated with embryo development (Dyer et al., 2009; Yun et al, 2010). Specifically, FGF-8 has a unique role in cell differentiation, proliferation, and specification. (Dyer et al, 2009). FGF-8 mutant mice demonstrate abnormalities in the heart, suggesting its role as a key growth factor required for cardiac development (Dyer et al, 2009). The role of FGF-8 in the injured adult heart or iPS cells is previously unexplored. The data presented herein demonstrate that FGF-8 contains both pro-cardiac and anti-apoptotic properties and that FGF-8 inhibits cardiac myocyte apoptosis in vitro and in vivo.
A. COMPOSITIONS
[0032] Disclosed herein are compositions useful in a method of enhancing cardiac tissue and/or cell regeneration. Disclosed herein are compositions useful in a method of attenuating cardiac tissue and/or cell remodeling. Disclosed herein are compositions useful in a method of inhibiting apoptosis and/or apoptosis-related mechanisms. 1. COMPOSITIONS FOR ENHANCEMENT OF CARDIAC TISSUE AND/OR CELL
REGENERATION
[0033] Compositions disclosed herein enhance cardiac tissue/cell regeneration. Disclosed herein is a composition for enhancing cardiac tissue/cell regeneration in a subject, comprising: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and/or (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9. For example, a disclosed composition for enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9).
[0034] Table 1 - Listing of Disclosed Compositions
Figure imgf000007_0001
[0035] Thus, as detailed in the above table, a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[0036] In an aspect, a disclosed composition protects myocardium including, but not limited to human myocardium, when administered with or without iPS cells. [0037] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject enhances cells engraftment. In an aspect, a disclosed composition enhances cell proliferation. In an aspect, a disclosed composition enhances cell differentiation. In an aspect, a disclosed composition can perform any combination of the following: enhancing cell engraftment, enhancing cell proliferation, and/or enhancing cell differentiation.
[0038] In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac -committed. In an aspect, a disclosed composition enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, and any other types of stem cells known to be effective by those of skill in the art. In an aspect, a disclosed composition enhances c-kit positive cells or any other endogenous heart stem cells.
[0039] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject improves angiogenesis in the subject. In an aspect, a disclosed composition enhances cardiac function in the subject. In an aspect, a disclosed composition both improves angiogenesis and enhances cardiac function in the subject.
[0040] In an aspect, enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
[0041] In an aspect, the conditioned media of a disclosed composition comprises one or more anti-apoptotic and anti-fibrotic factors. In a further aspect, the one or more anti-apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix metalloproteinase- 1 (TIMP-1).
[0042] In an aspect, the subject has experienced a cardiac ischemia/reperfusion event. In an aspect, the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant. In an aspect, the subject has a congenital heart defect. In an aspect, the congenital heart defect is hypoplasia or pentalogy of Cantrell.
[0043] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered to the subject prior to a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during a cardiac
ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject within 10, 15, 20, 25, 30, or more minutes following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered within 1, 2, 6, 12, 18, 24, or more hour following a cardiac ischemia/reperfusion event.
[0044] In an aspect, a disclosed composition is administered to the subject one or more times. For example, in an aspect, a disclosed composition is administered to the subject prior to and during a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during and following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject prior to and following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject prior to, during, and following a cardiac ischemia/reperfusion event.
[0045] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject comprises between 5,000 and 500,000 induced pluripotent stem cells (iPS cells). In an aspect, a composition comprises approximately 100,000 iPS cells. In an aspect, a composition comprises less than 100,000 iPS cells. In an aspect, a composition comprises more than 100,000 iPS cells.
[0046] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered to the subject in one intramyocardial injection. In an aspect, a disclosed composition is administered in two or more intramyocardial injections.
[0047] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject is administered into a peri-infarct zone of the injured myocardium. In an aspect, disclosed composition is administered into an infarcted zone of the injured myocardium. In an aspect, a disclosed composition is administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
[0048] In an aspect, a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject further comprises one or more immunosuppressive drugs. Immunosuppressive drugs are known in the art. In an aspect, the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors. In an aspect, the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs is cyclosporine A.
[0049] In an aspect, the induced pluripotent stem cells of a disclosed composition are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources.
[0050] In yet another aspect, the induced pluripotent stem cells of a disclosed composition for enhancing cardiac tissue/cell regeneration in a subject are obtained from fibroblast cells. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor. In an aspect, the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof. For example, in an aspect, the sternness factors are c-myc, oct 3/4, Klf4, and Sox2.
[0051] In an aspect, a disclosed composition enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
[0052] In an aspect, the subject is a mammal. In an aspect, the mammal is a primate. In an aspect, the mammal is a human. In an aspect, the human is a patient.
2. COMPOSITIONS FOR ATTENUATION OF ADVERSE CARDIAC TISSUE/CELL
REMODELING
[0053] Compositions disclosed herein attenuate adverse cardiac tissue/cell remodeling. Disclosed herein is a composition for attenuating adverse cardiac tissue/cell remodeling in a subject, comprising: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and/or (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9. For example, a disclosed composition for enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9). For example, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject comprises any one of the following:
[0054] Table 2 - Listing of Disclosed Compositions
Figure imgf000011_0001
[0055] Thus, as detailed in the above table, a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[0056] In an aspect, a disclosed composition protects myocardium including, but not limited to human myocardium, when administered with or without iPS cells.
[0057] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject improves angiogenesis in the subject. In an aspect, a disclosed composition enhances cardiac function in the subject. In an aspect, a disclosed composition improves angiogenesis and enhances cardiac function in the subject.
[0058] In an aspect, a disclosed composition for attenuating adverse cardiac remodeling inhibits fibrosis and/or fibrosis-related mechanisms. In an aspect, a disclosed composition decreases the expression level or activity level of at least one matrix metalloproteinase. In an aspect, the at least one matrix metalloproteinase is MMP-2. In an aspect, the at least one matrix metalloproteinase is MMP-9. In an aspect, the at least one matrix metalloproteinase comprises MMP-2 and MMP-9.
[0059] In an aspect, a disclosed composition for attenuating adverse cardiac tissue and/or cell remodeling in a subject inhibits or decreases necrosis. In an aspect, a disclosed composition inhibits or decreases apoptosis and/or apoptosis-related mechanisms. In an aspect, the apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells. In an aspect, the apoptosis and/or apoptosis-related mechanism occurs in cardiac tissue of the subject. In an aspect, a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, or decreases FOXOla expression. In an aspect, a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression. In an aspect, a disclosed composition performs any combination of the following: increases miR-486 expression, increases Akt expression, decreases PTEN expression, and/or decreases FOXOla expression.
[0060] In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac -committed. In an aspect, a disclosed composition enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed composition enhances c-kit positive cells or any other endogenous heart stem cells.
[0061] In an aspect, enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises each of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
[0062] In an aspect, the conditioned media of a disclosed composition comprises one or more anti-apoptotic and anti-fibrotic factors. In a further aspect, the one or more anti-apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix metalloproteinase- 1 (TIMP-1).
[0063] In an aspect, the subject has experienced a cardiac ischemia/reperfusion event. In an aspect, the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant. In an aspect, the subject has a congenital heart defect. In an aspect, the congenital heart defect is hypoplasia or pentalogy of Cantrell.
[0064] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered to the subject prior to a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject within 10, 15, 20, 25, 30, or more minutes following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered within 1, 2, 6, 12, 18, 24, or more hour following a cardiac ischemia/reperfusion event.
[0065] In an aspect, a disclosed composition is administered to the subject one or more times. For example, in an aspect, a disclosed composition is administered to the subject prior to and during a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject during and following a cardiac ischemia/reperfusion event. For example, in an aspect, a disclosed composition is administered to the subject prior to and following a cardiac ischemia/reperfusion event. In an aspect, a disclosed composition is administered to the subject prior to, during, and/or following a cardiac ischemia/reperfusion event.
[0066] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject comprises between 5,000 and 500,000 induced pluripotent stem cells (iPS cells). In an aspect, a disclosed composition comprises approximately 100,000 iPS cells. In an aspect, a disclosed comprises less than 100,000 iPS cells. In an aspect, a disclosed composition comprises more than 100,000 iPS cells.
[0067] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered to the subject in one intramycardial injection. In an aspect, a disclosed composition is administered in two or more intramycardial injections.
[0068] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject is administered into a peri-infarct zone of the injured myocardium. In an aspect, a disclosed composition is administered into an infarcted zone of the injured myocardium. In an aspect, a disclosed composition is administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
[0069] In an aspect, a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject further comprises one or more immunosuppressive drugs.
Immunosuppressive drugs are known in the art. In an aspect, the one or more
immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors. In an aspect, the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs is cyclosporine A.
[0070] In an aspect, the induced pluripotent stem cells of a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources.
[0071] In yet another aspect, the induced pluripotent stem cells of a disclosed composition for attenuating adverse cardiac tissue/cell remodeling in a subject are obtained from fibroblast cells. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor. In an aspect, the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof. For example, in an aspect, the sternness factors are c-myc, oct 3/4, Klf4, and Sox2.
[0072] In an aspect, a disclosed composition enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to,
mesenchymal stem cells, adipocyte stems cells, etc.
[0073] In an aspect, the subject is a mammal. In an aspect, the mammal is a primate. In an aspect, the mammal is a human. In an aspect, the human is a patient. 3. PHARMACEUTICAL COMPOSITIONS
[0074] In an aspect, the invention relates to pharmaceutical compositions comprising a disclosed compositions for enhancing cardiac tissue and/or cell regeneration. In an aspect, the invention relates to pharmaceutical compositions comprising a disclosed composition for attenuating adverse cardiac tissue and/or cell remodeling. That is, a pharmaceutical composition can be provided comprising a therapeutically effective amount of at least one disclosed compositions and a pharmaceutically acceptable carrier.
B. METHODS COMPRISING A DISCLOSED COMPOSITION
1. ENHANCING CARDIAC TISSUE AND/OR CELL REGENERATION
[0075] In one aspect, the invention relates to a method of enhancing cardiac tissue and/or cell regeneration in a subject. Disclosed herein is a method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the method comprising: (A) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (B) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) administering to the subject fibroblast growth factor-8; and/or (D) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9; and determining regeneration in the heart. For example, a disclosed method of enhancing cardiac tissue and/or cell regeneration in a subject comprises any one of the following combinations of fibroblast growth factor-8 primed induced pluripotent stem cells (FGF-8 primed iPS cells); conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells (CM of FGF-8 primed iPS cells); fibroblast growth factor-8 (FGF-8); and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 (FGF-8 and FGF-9).
[0076] Table 3 - Listing of Various Components of Disclosed Compositions used in
Disclosed Methods
Figure imgf000015_0001
Listing of Various Components of Disclosed Compositions used in Disclosed Methods
Comprising A Comprising B Comprising D
Comprising C
(i.e., FGF-8 primed (i.e., CM of FGF-8 (i.e., FGF-8 &
(i.e., FGF-8)
iPS cells) iPS cells) FGF-9)
A+B+C B+A+C C+A+B D+A+B
A+B+D B+A+D C+A+D D+A+C
A+C+D B+C+D C+B+D D+B+C
A+B+C+D B+A+C+D C+A+B+D D+A+B+C
[0077] Thus, as detailed in the above table, a disclosed composition comprises one or two or three or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[0078] In an aspect, a disclosed method protects myocardium including, but not limited to, human myocardium, with or without the administration of iPS cells.
[0079] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell regeneration, fibroblast growth factor-8 (FGF-8) is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells). In an aspect, FGF-8 is administered to the subject during the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject following the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject prior to and during the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject prior to and following the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject during and following the administration of the iPS cells.
[0080] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell regeneration, a composition comprising fibroblast growth factor-8 (FGF-8) and fibroblast growth factor-9 (FGF-9) is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells). In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject during the administration of the iPS cells. In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject following the administration of the iPS cells. In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject prior to and during the administration of the iPS cells. In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject prior to and following the administration of the iPS cells. In an aspect, a composition comprising FGF-8 and FGF-9 is administered to the subject during and following the administration of the iPS cells.
[0081] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell
regeneration, the conditioned media is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells). In an aspect, the conditioned media is administered to the subject during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject following the administration of the iPS cells. In an aspect the conditioned media is administered to the subject prior to and during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject prior to and following the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject during and following the administration of the iPS cells.
[0082] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell
regeneration, the administration of induced pluripotent stem cells to the subject is repeated. In an aspect, the administration of fibroblast growth factor-8 to the subject is repeated. In an aspect, the administration of a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 to the subject is repeated. In an aspect, the administration of the conditioned media to the subject is repeated.
[0083] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell
regeneration following cardiac dysfunction in a subject in need therefore, the induced pluripotent stem cells (iPS cells) are administered to the subject prior to, during, and/or following cardiac dysfunction. For example, in an aspect, the iPS cells are administered to the subject prior to cardiac dysfunction. In an aspect, the iPS cells are administered to the subject, during cardiac dysfunction. In an aspect, the iPS cells administered to the subject following cardiac dysfunction. For example, in an aspect, the iPS cells are administered to the subject prior to and during cardiac dysfunction. In an aspect, the iPS cells are administered to the subject during and following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to and following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to, during, and/or following cardiac dysfunction. In an aspect, the induced pluripotent stem cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction. In an aspect, the induced pluripotent stem cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction. [0084] In an aspect of a disclosed method, enhancing cardiac tissue and/or cell regeneration comprises enhancing cell engraftment. In an aspect, enhancing cardiac tissue and/or cell regeneration comprises enhancing cell differentiation. In an aspect of a disclosed method, enhancing cardiac tissue and/or cell regeneration comprises both enhancing cell engraftment and enhancing cell differentiation. In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac-committed. In an aspect, a disclosed method comprises enhancing cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
[0085] In an aspect, a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof further comprises improving angiogenesis in the subject. In an aspect, a disclosed method further comprises enhancing cardiac function in the subject. In an aspect, a disclosed method further comprises improving both angiogenesis and enhancing cardiac function in the subject.
[0086] In an aspect, enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
[0087] In an aspect, the conditioned media of a method disclosed herein comprises one or more anti-apoptotic and anti-fibrotic factors. In a further aspect, the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix
metalloproteinase-1 (TIMP-1).
[0088] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, between 5,000 and 500,000 induced pluripotent stem cells (iPS cells) are administered to the subject. In an aspect, approximately 100,000 iPS cells are administered to the subject. In an aspect, less than 100,000 iPS cells are administered to the subject. In an aspect, more than 100,000 iPS cells are administered to the subject.
[0089] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the induced pluripotent stem cells can be administered in one intramycardial injection. In an aspect, the induced pluripotent stem cells are administered in two or more intramycardial injections.
[0090] In an aspect of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
[0091] In an aspect, a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof further comprises administering to the subject one or more immunosuppressive drugs. Immunosuppressive drugs are known in the art. In an aspect, the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors. In an aspect, the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs is cyclosporine A. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during and following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and following the administration of the induced pluripotent stem cells.
[0092] In an aspect, the cardiac dysfunction of a disclosed method is a cardiac
ischemia/reperfusion event. In an aspect, the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia,
Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant. In an aspect, the subject has a congenital heart defect. In an aspect, the congenital heart defect is hypoplasia or pentalogy of Cantrell.
[0093] In an aspect, the induced pluripotent stem cells of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources.
[0094] In yet another aspect, the induced pluripotent stem cells of a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof are obtained from fibroblast cells. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one stemness factor. In an aspect, the at least one stemness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof. For example, in an aspect, the stemness factors are c-myc, oct 3/4, Klf4, and Sox2.
[0095] In an aspect, a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, and other types of stem cells known to be effective by those of skill in the art.
[0096] In an aspect of a disclosed method enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the subject is a mammal. In an aspect, the mammal is a primate. In an aspect, the mammal is a human. In an aspect, the human is a patient. 2. ATTENUATING ADVERSE CARDIAC TISSUE AND/OR CELL REMODELING
[0097] In one aspect, the invention relates to a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject. Disclosed herein is a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the method comprising: (A) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells; (B) administering to the subject conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) administering to the subject fibroblast growth factor-8; and/or (D) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor- 9; and determining a decrease in cardiac remodeling. For example, a disclosed method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, comprises any one of the following:
[0098] Table 4 - Listing of Various Components of Disclosed Compositions used in Disclosed Methods
Figure imgf000021_0001
[0099] Thus, as detailed in the table, a disclosed method comprises administering one or two or three or more or all four of the following: (A) fibroblast growth factor-8 primed induced pluripotent stem cells; (B) conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells; (C) fibroblast growth factor-8; and (D) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[00100] In an aspect, a disclosed method protects myocardium including, but not limited to human myocardium, with or without the administration of iPS cells. [00101] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling, fibroblast growth factor-8 (FGF-8) is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells). In an aspect, FGF-8 is administered to the subject during the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject following the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject prior to and during the administration of the iPS cells. In an aspect, FGF-8is administered to the subject prior to and following the administration of the iPS cells. In an aspect, FGF-8 is administered to the subject during and following the administration of the iPS cells.
[00102] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to the administration of the induced pluripotent stem cells (iPS cells). In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject during the administration of the iPS cells. In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject following the administration of the iPS cells. In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to and during the administration of the iPS cells. In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject prior to and following the administration of the iPS cells. In an aspect, a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 is administered to the subject during and following the administration of the iPS cells.
[00103] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling, the conditioned media is administered to the subject prior to the
administration of the induced pluripotent stem cells (iPS cells). In an aspect, the conditioned media is administered to the subject during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject following the administration of the iPS cells. In an aspect the conditioned media is administered to the subject prior to and during the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject prior to and following the administration of the iPS cells. In an aspect, the conditioned media is administered to the subject during and following the administration of the iPS cells.
[00104] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling, the administration of induced pluripotent stem cells to the subject is repeated. In an aspect, the administration of fibroblast growth factor-8 to the subject is repeated. In an aspect, the administration of a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9 to the subject is repeated. In an aspect, the administration of the conditioned media to the subject is repeated.
[00105] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject prior to cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject, during cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject or following cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject prior to and during cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject during and following cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject prior to and following cardiac dysfunction. For example, in an aspect, the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction. In an aspect, the induced pluripotent stem cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction. In an aspect, the induced pluripotent stem cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction.
[00106] In an aspect of a disclosed method, attenuating adverse cardiac remodeling comprises inhibiting fibrosis and/or fibrosis-related mechanisms. In an aspect, inhibiting fibrosis and/or fibrosis-related mechanisms comprises decreasing the expression level or activity level of at least one matrix metalloproteinase. In an aspect, the at least one matrix metalloproteinase is MMP-2. In an aspect, the at least one matrix metalloproteinase is MMP- 9. In an aspect, the at least one matrix metalloproteinase comprises MMP-2 and MMP-9.
[00107] In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac -committed. In an aspect, a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
[00108] In an aspect, a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprises improving angiogenesis in the subject. In an aspect, a disclosed method further comprises enhancing cardiac function in the subject. In an aspect, a disclosed method further comprises improving angiogenesis and enhancing cardiac function in the subject.
[00109] In an aspect, enhancing cardiac function in a disclosed method comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises all of the following: (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry. In an aspect, enhancing cardiac function in a disclosed method comprises any combination of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
[00110] In an aspect, a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprise inhibiting or decreasing necrosis.
[00111] In an aspect, a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprise inhibiting or decreasing apoptosis and/or apoptosis-related mechanisms. In an aspect, the apoptosis and/or apoptosis-related mechanism occurs in the induced pluripotent stem cells. In an aspect, the apoptosis and/or apoptosis-related mechanism occurs in cardiac tissue of the subject. In an aspect, the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and/or a decrease in FOXOla expression. In an aspect, the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression. In an aspect, the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by any combination of the following: an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and/or a decrease in FOXOla expression.
[00112] In an aspect, the conditioned media of a method disclosed herein comprises one or more anti-apoptotic and anti-fibrotic factors. In a further aspect, the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-8 (FGF-8), fibroblast growth factor-9 (FGF-9), interleukin-10 (IL-10), and tissue inhibitor of matrix
metalloproteinase-1 (TIMP-1).
[00113] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, between 5,000 and 500,000 induced pluripotent stem cells (iPS cells) are administered to the subject. In an aspect, approximately 100,000 iPS cells are administered to the subject. In an aspect, less than 100,000 iPS cells are administered to the subject. In an aspect, more than 100,000 iPS cells are administered to the subject.
[00114] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the induced pluripotent stem cells can be administered in one intramycardial injection. In an aspect, the induced pluripotent stem cells are administered in two or more intramycardial injections.
[00115] In an aspect of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium. In an aspect of a disclosed method, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
[00116] In an aspect, a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof further comprises administering to the subject one or more immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, antiproliferatives, and mTOR inhibitors. In an aspect, the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs. In an aspect, the one or more
immunosuppressive drugs is cyclosporine A. In an aspect, the one or more
immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells. For example, in an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and during the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject during and following the administration of the induced pluripotent stem cells. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to and following the administration of the induced pluripotent stem cells.
[00117] In an aspect, the cardiac dysfunction of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof is a cardiac ischemia/reperfusion event. In an aspect, the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant. In an aspect, the subject has a congenital heart defect. In an aspect, the congenital heart defect is hypoplasia or pentalogy of Cantrell.
[00118] In an aspect, the induced pluripotent stem cells of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources.
[00119] In yet another aspect, the induced pluripotent stem cells of a disclosed method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof are obtained from fibroblast cells. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor. In an aspect, the at least one sternness factor comprises c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof. For example, in an aspect, the sternness factors are c-myc, oct 3/4, Klf4, and Sox2. [00120] In an aspect, a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
[00121] In an aspect of a disclosed method enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the subject is a mammal. In an aspect, the mammal is a primate. In an aspect, the mammal is a human. In an aspect, the human is a patient.
3. GENERATING INDUCED PLURIPOTENT STEM CELLS
[00122] Disclosed herein is a method of generating cardiac induced pluripotent stem cells. In an aspect, a method of generating cardiac induced pluripotent stem cells comprises (i) inserting one or more nucleic acid constructs capable of expressing stem-cell like factors into a cardiac cell type, and (ii) obtaining the cardiac induced pluripotent stem cells stably expressing the stem-cell like factors in the cardiac cell type. In an aspect, the stem-cell like factors comprise Oct3/4, KIf4, Sox2, and c-Myc, or a combination thereof. In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac-committed. In an aspect, a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells.
[00123] In an aspect, the induced pluripotent stem cells are obtained from an autologous source. In an aspect, the induced pluripotent stem cells are obtained from an allogeneic source. In an aspect, the induced pluripotent stem cells are obtained from a syngeneic source. In yet another aspect, the induced pluripotent stem cells are obtained from fibroblast cells. In an aspect, the induced pluripotent stem cells are obtained from a combination of sources. In a further aspect, the induced pluripotent stem cells are obtained from H9c2 cells. In an aspect, the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one sternness factor.
[00124] In an aspect, a disclosed induced pluripotent stem cells are used in a method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject. In an aspect, a disclosed induced pluripotent stem cells are used in a method of enhancing cardiac tissue and/or cell regeneration in a subject. In an aspect, the subject is a mammal. In an aspect, the mammal is a primate. In an aspect, the mammal is a human. In an aspect, the human is a patient.
[00125] In an aspect, a disclosed induced pluripotent stem cells are primed with fibroblast growth factor-8. In an aspect, the induced pluripotent stem cells are administered with one or more of the following: conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[00126] In an aspect, the timing of the administration of the induced pluripotent stem cells varies. For example, in an aspect, the induced pluripotent stem cells are administered prior to administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9. In an aspect, the induced pluripotent stem cells are administered during the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9. In an aspect, the induced pluripotent stem cells are administered following the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9. In an aspect, the administration of induced pluripotent stem cells to the subject is repeated. For example, in an aspect, the induced pluripotent stem cells are administered both prior to and during, or prior to and following, or during and following, the administration of conditioned media of fibroblast growth factor-8 primed induced pluripotent stem cells, fibroblast growth factor-8, and/or a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
[00127] In an aspect, the induced pluripotent stem cells (iPS cells) are administered to the subject prior to, during, and/or following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to cardiac dysfunction. In an aspect, the iPS cells are administered to the subject, during cardiac dysfunction. In an aspect, the iPS cells are administered to the subject or following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to and during cardiac dysfunction. In an aspect, the iPS cells are administered to the subject during and following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to and following cardiac dysfunction. In an aspect, the iPS cells are administered to the subject prior to, during, and following cardiac dysfunction. In an aspect, the iPS cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction. In an aspect, the iPS cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction.
[00128] In an aspect, a disclosed induced pluripotent stem cells improve angiogenesis in a subject. In an aspect, a disclosed induced pluripotent stem cells enhance cardiac function in a subject. In an aspect, a disclosed induced pluripotent stem cells improve angiogenesis and enhance cardiac function in the subject. In an aspect, enhancing cardiac function comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and/or (vi) normalizing of heart geometry.
[00129] In an aspect, a disclosed induced pluripotent stem cells inhibit or decrease necrosis. In an aspect, a disclosed induced pluripotent stem cells inhibit or decrease apoptosis and/or apoptosis-related mechanisms.
[00130] In an aspect, between 5,000 and 500,000 induced pluripotent stem cells (iPS cells) are administered to a subject. In an aspect, approximately 100,000 iPS cells are administered to a subject. In an aspect, less than 100,000 iPS cells are administered to a subject. In an aspect, more than 100,000 iPS cells are administered to a subject.
[00131] In an aspect, the induced pluripotent stem cells are administered in one intramycardial injection. In an aspect, the induced pluripotent stem cells are administered in two or more intramycardial injections. In an aspect, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into an infarcted zone of the injured myocardium. In an aspect, the induced pluripotent stem cells are administered into a peri-infarct zone of the injured myocardium and into an infarcted zone of the injured myocardium.
[00132] In an aspect, a disclosed induced pluripotent stem cells are administered with one or more immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors. In an aspect, the one or more immunosuppressive drugs can be a combination of immunosuppressive drugs. In an aspect, the one or more immunosuppressive drugs is cyclosporine A. In an aspect, the one or more immunosuppressive drugs can be administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells. 4. IDENTIFYING A MYOCARDIAL INJURY
[00133] Disclosed herein is a method of identifying a myocardial injury or infarcted myocardial tissue. In an aspect, a method of identifying a myocardial injury or infarcted myocardial tissue comprises measuring the amount of miR-486. In an aspect, the amount of miR-486 decreases after myocardial injury or myocardial infarction. In an aspect, wherein the amount of miR-486 increases after treatment of induced pluripotent stem cells. In an aspect, a disclosed method of identifying a myocardial injury or infracted myocardial tissue further comprises measuring the amount of Akt expression, PTEN expression, and/or FOXOla expression.
[00134] In an aspect, a disclosed method of identifying a myocardial injury or infarcted myocardial tissue further comprises measuring apoptosis and/or apoptosis-related mechanisms. In an aspect, the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression. In an aspect, the inhibiting or decreasing of apoptosis and/or apoptosis-related mechanism is accompanied by any combination of the following: an increase in miR-486 expression, an increase in Akt expression, a decrease in PTEN expression, and a decrease in FOXOla expression.
5. INHIBITING APOPTOSIS AND/OR APOPTOSIS-RELATED MECHANISMS
[00135] Disclosed herein are methods of inhibiting apoptosis and/or apoptosis-related mechanisms. In an aspect, a method of inhibiting apoptosis and/or apoptosis-related mechanisms comprises administering conditioned medium from induced pluripotent stem cells. In an aspect, a method of inhibiting apoptosis and/or apoptosis-related mechanisms comprises administering FGF-8. In an aspect, the conditioned media or the FGF-8 is administered to a subject.
[00136] In an aspect, the subject has experienced a cardiac ischemia/reperfusion event.
In an aspect, the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant. In an aspect, the subject has a congenital heart defect. In an aspect, the congenital heart defect is hypoplasia or pentalogy of Cantrell.
[00137] In an aspect, the conditioned media of a disclosed method comprises one or more anti-apoptotic and anti-fibrotic factors. In a further aspect, the one or more anti- apoptotic and anti-fibrotic factors comprise fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, and tissue inhibitor of matrix metalloproteinase- 1. In an aspect, the conditioned medium is administered with or without fibroblast growth factor-8 (FGF-8). In an aspect, the conditioned medium is administered with or without fibroblast growth factor-9 (FGF-9). In an aspect, the conditioned medium is administered with or without interleukin-10 (IL-10). In an aspect, the conditioned medium is administered with or without matrix metalloproteinase- 1 (MMP-1). In an aspect, the conditioned medium is administered with one or more of the following: fibroblast growth factor-9, fibroblast growth factor-8, interleukin- 10, and tissue inhibitor of matrix metalloproteinase- 1. In an aspect, the conditioned medium is administered with any combination of the following: fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, and tissue inhibitor of matrix metalloproteinase- 1, such as, for example, the combination of FGF-8 and FGF-9.
[00138] In an aspect, a disclosed method protects myocardium including, but not limited to human myocardium, with or without the administration of iPS cells.
[00139] In an aspect, a disclosed method of inhibiting apoptosis and/or apoptosis- related mechanisms further comprises administering induced pluripotent stem cells. In an aspect, the induced pluripotent stem cells differentiate into cardiac myocytes. In a further aspect, the induced pluripotent stem cells are cardiac -committed. In an aspect, a disclosed method enhances cardiac myocyte differentiation in iPS cells or any other source of stem cells in regenerative medicine including, but not limited to, mesenchymal stem cells, adipocyte stems cells, etc. In an aspect, a disclosed method enhances c-kit positive cells or any other endogenous heart stem cells. In an aspect, the conditioned medium is administered to the subject prior to the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered during the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered following the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered the conditioned medium prior to and during the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered the conditioned medium prior to and following the administration of the induced pluripotent stem cells. In an aspect, the conditioned medium is administered to the subject during and following the
administration of the induced pluripotent stem cells.
[00140] In an aspect, the apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells. In an aspect, the apoptosis and/or apoptosis-related mechanisms occurs in cardiac tissue of the subject. In an aspect, a disclosed method increases miR-486 expression, increases Akt expression, decreases PTEN expression, and/or decreases FOXOla expression. In an aspect, a disclosed composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression. In an aspect, a disclosed composition performs any combination of the following: increases miR-486 expression, increases Akt expression, decreases PTEN expression, and decreases FOXOla expression.
[00141] In an aspect, a disclosed method enhances neovascularization in iPS cells and/or any other stem cells used in regenerative medicine including, but not limited to mesenchymal stem cells, adipocyte stems cells, etc.
6. NON-MEDICAL USES
[00142] Also disclosed herein are uses of a disclosed compositions as investigational and/or research tools in the development and standardization of in vitro and in vivo test systems for evaluation in laboratory animals such as cats, dogs, rabbits, monkeys, rats and mice, as part of the search for new therapeutic approaches for the enhancement of cardiac tissue and/or cell regeneration as well as the evaluation of the attenuation of adverse cardiac tissue and/or cell remodeling. In an aspect, these approaches apply to subjects with cardiac dysfunction.
7. DEFINITIONS
[00143] Unless otherwise expressly stated, it is in no way intended that any method or aspect set forth herein be construed as requiring that its steps be performed in a specific order. Accordingly, where a method claim does not specifically state in the claims or descriptions that the steps are to be limited to a specific order, it is no way intended that an order be inferred, in any respect. This holds for any possible non-express basis for interpretation, including matters of logic with respect to arrangement of steps or operational flow, plain meaning derived from grammatical organization or punctuation, or the number or type of aspects described in the specification.
[00144] As used in the specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the context clearly dictates otherwise.
[00145] The word "or" as used herein means any one member of a particular list and also includes any combination of members of that list.
[00146] Ranges can be expressed herein as from "about" one particular value, and/or to "about" another particular value. When such a range is expressed, a further aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent "about," it will be understood that the particular value forms a further aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. It is also understood that there are a number of values disclosed herein, and that each value is also herein disclosed as "about" that particular value in addition to the value itself. For example, if the value "10" is disclosed, then "about 10" is also disclosed. It is also understood that each unit between two particular units are also disclosed. For example, if 10 and 15 are disclosed, then 11, 12, 13, and 14 are also disclosed.
[00147] As used herein, the amino acid abbreviations are conventional one letter codes for the amino acids and are expressed as follows: A, alanine; B, asparagine or aspartic acid; C, cysteine; D aspartic acid; E, glutamate, glutamic acid; F, phenylalanine; G, glycine; H histidine; I isoleucine; K, lysine; L, leucine; M, methionine; N, asparagine; P, proline; Q, glutamine; R, arginine; S, serine; T, threonine; V, valine; W, tryptophan; Y, tyrosine; Z, glutamine or glutamic acid.
[00148] "Peptide" as used herein refers to any peptide, oligopeptide, polypeptide, gene product, expression product, or protein. For example, a peptide can be an enzyme. A peptide is comprised of consecutive amino acids. The term "peptide" encompasses naturally occurring or synthetic molecules.
[00149] In general, the biological activity or biological action of a peptide refers to any function exhibited or performed by the peptide that is ascribed to the naturally occurring form of the peptide as measured or observed in vivo (i.e., in the natural physiological environment of the protein) or in vitro (i.e., under laboratory conditions). For example, a biological activity of caspase includes caspase enzymatic activity.
[00150] The term "enzyme" as used herein refers to any peptide that catalyzes a chemical reaction of other substances without itself being destroyed or altered upon completion of the reaction. Typically, a peptide having enzymatic activity catalyzes the formation of one or more products from one or more substrates. Such peptides can have any type of enzymatic activity including, without limitation, the enzymatic activity or enzymatic activities associated with enzymes such as those disclosed herein.
[00151] References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a compound containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the compound.
[00152] A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.
[00153] As used herein, the terms "optional" or "optionally" means that the subsequently described event or circumstance can or can not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
[00154] As used herein, the terms "transformation" and "trans fection" mean the introduction of a nucleic acid, e.g., an expression vector, into a recipient cell including introduction of a nucleic acid to the chromosomal DNA of said cell. The art is familiar with various compositions, methods, techniques, etc. used to effect the introduction of a nucleic acid into a recipient cell. The art is familiar with such compositions, methods, techniques, etc for both eukaryotic and prokaryotic cells. The art is familiar with such compositions, methods, techniques, etc. for the optimization of the introduction and expression of a nucleic acid into and within a recipient cell.
[00155] As used herein, the term "subject" refers to the target of administration, e.g., an animal. Thus, the subject of the herein disclosed methods can be a vertebrate, such as a mammal, a fish, a bird, a reptile, or an amphibian. Alternatively, the subject of the herein disclosed methods can be a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, cat, guinea pig or rodent. The term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be covered. In one aspect, the subject is a mammal. A patient refers to a subject afflicted with a disease or disorder. The term "patient" includes human and veterinary subjects. In some aspects of the disclosed methods, the subject has been diagnosed with a need for treatment for cardiac dysfunction, such as, for example, a cardiac ischemia/reperfusion event prior to the administering step.
[00156] As used herein, the term "treatment" refers to the medical management of a subject or a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder, such as, for example, an injured myocardium. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. In various aspects, the term covers any treatment of a subject, including a mammal (e.g., a human), and includes: (i) preventing the disease from occurring in a subject that can be predisposed to the disease but has not yet been diagnosed as having it; (ii) inhibiting the disease, i.e., arresting its development; or (iii) relieving the disease, i.e., causing regression of the disease. In an aspect, the disease, pathological condition, or disorder is cardiac dysfunction, such as, for example, a cardiac
ischemia/reperfusion event.
[00157] As used herein, the term "prevent" or "preventing" refers to precluding, averting, obviating, forestalling, stopping, or hindering something from happening, especially by advance action. It is understood that where reduce, inhibit or prevent are used herein, unless specifically indicated otherwise, the use of the other two words is also expressly disclosed.
[00158] As used herein, the term "diagnosed" means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by compositions or methods disclosed herein. For example, "diagnosed with cardiac dysfunction" means having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition that can be diagnosed or treated by a compound or composition that alleviates or ameliorates cardiac dysfunction. As a further example, "diagnosed with a need for improving or enhancing cardiac regeneration and/or cardiac function" refers to having been subjected to a physical examination by a person of skill, for example, a physician, and found to have a condition characterized by cardiac dysfunction and/or cardiac impairment and/or cardiac cell death, wherein improving or enhancing cardiac regeneration and/or cardiac function would be beneficial to the subject. Such a diagnosis can be in reference to a disorder, such as cardiac dysfunction, and the like, as discussed herein.
[00159] As used herein, the phrase "identified to be in need of treatment for a disorder," or the like, refers to selection of a subject based upon need for treatment of the disorder. For example, a subject can be identified as having a need for treatment of a disorder (e.g., a disorder related to cardiac dysfunction or myocardial infarction) based upon an earlier diagnosis by a person of skill and thereafter subjected to treatment for the disorder. It is contemplated that the identification can, in one aspect, be performed by a person different from the person making the diagnosis. It is also contemplated, in a further aspect, that the administration can be performed by one who subsequently performed the administration.
[00160] As used herein, the terms "administering" and "administration" refer to any method of providing a pharmaceutical preparation to a subject. Such methods are well known to those skilled in the art and include, but are not limited to, intracardiac administration, oral administration, transdermal administration, administration by inhalation, nasal
administration, topical administration, intravaginal administration, ophthalmic
administration, intraaural administration, intracerebral administration, rectal administration, sublingual administration, buccal administration, and parenteral administration, including injectable such as intravenous administration, intra-arterial administration, intramuscular administration, and subcutaneous administration. Administration can be continuous or intermittent. In various aspects, a preparation can be administered therapeutically; that is, administered to treat an existing disease or condition. In further various aspects, a preparation can be administered prophylactically; that is, administered for prevention of a disease or condition.
[00161] The term "contacting" as used herein refers to bringing a disclosed compound and a cell, target receptor, or other biological entity together in such a manner that the compound can affect the activity of the target (e.g., receptor, transcription factor, cell, etc.), either directly; i.e., by interacting with the target itself, or indirectly; i.e., by interacting with another molecule, co-factor, factor, or protein on which the activity of the target is dependent.
[00162] As used herein, the term "determining" can refer to measuring or ascertaining a quantity or an amount or a change in expression and/or activity level, e.g., of a nucleotide or transcript or polypeptide. For example, determining the amount of a disclosed transcript or polypeptide in a sample as used herein can refer to the steps that the skilled person would take to measure or ascertain some quantifiable value of the transcript or polypeptide in the sample. The art is familiar with the ways to measure an amount of the disclosed nucleotides, transcripts, polypeptides, etc.
[00163] As used herein, the term "level" refers to the amount of a target molecule in a sample, e.g., a sample from a subject. The amount of the molecule can be determined by any method known in the art and will depend in part on the nature of the molecule (i.e., gene, mRNA, cDNA, protein, enzyme, etc.). The art is familiar with quantification methods for nucleotides (e.g., genes, cDNA, mRNA, etc) as well as proteins, polypeptides, enzymes, etc. It is understood that the amount or level of a molecule in a sample need not be determined in absolute terms, but can be determined in relative terms (e.g., when compare to a control or a sham or an untreated sample).
[00164] As used herein, the terms "effective amount" and "amount effective" refer to an amount that is sufficient to achieve the desired result or to have an effect on an undesired condition. For example, a "therapeutically effective amount" refers to an amount that is sufficient to achieve the desired therapeutic result or to have an effect on undesired symptoms, but is generally insufficient to cause adverse side affects. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration; the route of administration; the rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed and like factors well known in the medical arts.
[00165] By "modulate" is meant to alter, by increase or decrease. As used herein, a
"modulator" can mean a composition that can either increase or decrease the expression level or activity level of a gene or gene product such as a peptide. Modulation in expression or activity does not have to be complete. For example, expression or activity can be modulated by about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, 100% or any percentage in between as compared to a control cell wherein the expression or activity of a gene or gene product has not been modulated by a composition.
[00166] As used herein, "EC50," is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% enhancement or activation of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. EC50 also refers to the concentration or dose of a substance that is required for 50% enhancement or activation in vivo, as further defined elsewhere herein. Alternatively, EC50 can refer to the concentration or dose of compound that provokes a response halfway between the baseline and maximum response. The response can be measured in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest. For example, the response can be measured in vitro using cultured cardiac cells or in an ex vivo organ culture system with isolated cardiac cells, e.g., cardiomyocytes, vascular smooth muscle cells, endothelial cells, etc). Alternatively, the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats. The mouse or rat can be an inbred strain with phenotypic characteristics of interest such as, for example, obesity or diabetes. As appropriate, the response can be measured in a transgenic or knockout mouse or rat wherein a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.
[00167] As used herein, "IC50," is intended to refer to the concentration or dose of a substance (e.g., a compound or a drug) that is required for 50% inhibition or diminution of a biological process, or component of a process, including a protein, subunit, organelle, ribonucleoprotein, etc. IC50 also refers to the concentration or dose of a substance that is required for 50% inhibition or diminution in vivo, as further defined elsewhere herein.
Alternatively, IC50 also refers to the half maximal (50%) inhibitory concentration (IC) or inhibitory dose of a substance. The response can be measured in a in vitro or in vivo system as is convenient and appropriate for the biological response of interest. For example, the response can be measured in vitro using cultured cardiac cells or in an ex vivo organ culture system with isolated cardiac cells (e.g., cardiomyocytes, vascular smooth muscle cells, endothelial cells, etc). Alternatively, the response can be measured in vivo using an appropriate research model such as rodent, including mice and rats. The mouse or rat can be an inbred strain with phenotypic characteristics of interest such as, for example, obesity or diabetes. As appropriate, the response can be measured in a transgenic or knockout mouse or rat wherein the a gene or genes has been introduced or knocked-out, as appropriate, to replicate a disease process.
[00168] The term "pharmaceutically acceptable" describes a material that is not biologically or otherwise undesirable, i.e., without causing an unacceptable level of undesirable biological effects or interacting in a deleterious manner. As used herein, the term "pharmaceutically acceptable carrier" refers to sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents such as paraben, chlorobutanol, phenol, sorbic acid and the like. It can also be desirable to include isotonic agents such as sugars, sodium chloride and the like.
Prolonged absorption of the injectable pharmaceutical form can be brought about by the inclusion of agents, such as aluminum monostearate and gelatin, which delay absorption. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide, poly(orthoesters) and poly(anhydrides). Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues. The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable media just prior to use. Suitable inert carriers can include sugars such as lactose. Desirably, at least 95% by weight of the particles of the active ingredient have an effective particle size in the range of 0.01 to 10 micrometers.
[00169] Disclosed are the components to be used to prepare a composition of the invention as well as the compositions themselves to be used within the methods disclosed herein. These and other materials are disclosed herein, and it is understood that when combinations, subsets, interactions, groups, etc. of these materials are disclosed that while specific reference of each various individual and collective combinations and permutation of these compounds can not be explicitly disclosed, each is specifically contemplated and described herein. For example, if a particular compound is disclosed and discussed and a number of modifications that can be made to a number of molecules including the compounds are discussed, specifically contemplated is each and every combination and permutation of the compound and the modifications that are possible unless specifically indicated to the contrary. Thus, if a class of molecules A, B, and C are disclosed as well as a class of molecules D, E, and F and an example of a combination molecule, A-D is disclosed, then even if each is not individually recited each is individually and collectively
contemplated meaning combinations, A-E, A-F, B-D, B-E, B-F, C-D, C-E, and C-F are considered disclosed. Likewise, any subset or combination of these is also disclosed. Thus, for example, the sub-group of A-E, B-F, and C-E would be considered disclosed. This concept applies to all aspects of this application including, but not limited to, steps in methods of making and using the compositions of the invention. Thus, if there are a variety of additional steps that can be performed it is understood that each of these additional steps can be performed with any specific embodiment or combination of embodiments of the methods of the invention.
[00170] The present invention can be understood more readily by reference to the following detailed description of the invention and the Examples included therein.
[00171] All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided herein can be different from the actual publication dates, which can require independent confirmation.
C. EXPERIMENTAL
[00172] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
[00173] Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in °C or is at ambient temperature, and pressure is at or near atmospheric. 1. GENERAL EXPERIMENTS
a. ESTABLISHMENT OF IPS CELL LINES AND DIFFERENTIATION OF CARDIAC CELL TYPE
[00174] Stably transfected iPS cell lines expressing red fluorescence protein (RFP) were generated. (Singla et al, 201 1). H9c2 cells were transfected with an expression vector expressing mouse sternness four factors (i.e., the 4F expression vector). The four sternness factors were c-Myc, Oct 3/4, Klf4 and Sox2. Figure 1A shows photomicrographs of untransduced H9c2 cells and shows transduced H9c2 cells forming ES cell like colony (right panel) (scale bar = 50 μιη). Untransduced H9c2 cells appear elongated, but following transduction with the 4F expression vector, these H9c2 cells appeared morphologically distinct from the untransduced H9c2 cells (Figure 1A, left panel vs. right panel).
[00175] To determine whether these four factors were present in the generated iPS cells, western blot analysis was utilized to show expression for these four sternness factors in the iPS cells. As shown in Figure IB, western blot analysis did not show the presence of any one of the four factors (c-Myc, Oct3/4, Klf4 and Sox2) in the untransfected H9c2 cells, but increased expression of four factors was detected in the transduced H9c2 cells.
[00176] Next, whether generated iPS cells expressed pluripotency markers, such as alkaline phosphatase and Oct3/4, was next determined. As shown in Figure 1C, iPS cells expressed alkaline phosphatase in 100% of growing cells (left panel). Similar expression was observed in ES cells, which were used as a positive control (middle panel). In contrast, untransduced H9c2 cells were negative for alkaline phosphatase staining (right panel) (scale bar = 250 μιη). Moreover, as seen in Figure ID, growing iPS cells were positive for the undifferentiated pluripotent marker Oct3/4 (which was compared with ES cells as a positive control). Figure ID also shows RFP expressing ES and iPS cells (b and f); DAPI staining for ES and iPS cells (c and g), and a merged image Oct3/4, RFP, and DAPI of ES and iPS cells (d and h). These data indicate that generated iPS cells were reprogrammed and expressed pluripotent markers, (scale bar = 100 μιη). (Singla et al, 2011).
[00177] Whether the iPS cell lines retained the capability in vitro to differentiate into all three major heart cell types (i.e., cardiomyocytes, vascular smooth muscle cells and endothelial cells) present and still express the desired reporter protein was evaluated. b. FGF-8 ENHANCEMENT OF CARDIAC MYOCYTE DIFFERENTIATION FROM
IPS CELLS
[00178] The ability of iPS cell-derived EB cell culture system to produce beating cardiac myocytes was confirmed. Whether direct treatment of iPS cells with FGF-8 promoted differentiation into beating cardiac myocytes. EBs derived from iPS cells were treated with 0, 2, 4, and 8 ng per mL of the bio-active form of recombinant human FGF-8 and examined by microscopy from day 0 (DO) through day 17 (D17) after plating. Figure 2 A (top panel) shows the size of EBs in inches following treatment with FGF-8. Figure 2A (bottom panel) shows the number of beating EBs following the same course of FGF-8 treatment. Figure 2B shows that iPS cells form embryoid bodies (EBs) and differentiate into cardiac myocytes, smooth muscle cells and endothelial cells. EBs were differentiated for 17 days. A beating area (i.e., cardiac myocytes) was stained with anti-myosin/Alexa 488 (2B-A), anti-RFP/Alexa 568 (2B- B), and DAPI (2B-C). The merged image (2B-C) shows co-expression of myosin and RFP. Smooth muscle cells were stained with anti-smooth muscle actin/Alexa 488 (2B-D), anti- RFP/Alexa 568 (2B-E), and DAPI (2B-F). The merged image (2B-F) shows overlap of expression of smooth muscle a-actin and RFP. Endothelial cells that have formed a capillarylike structure were stained with anti-von Willebrand Factor/Alexa 488 (2B-G), anti- RFP/Alexa 568 (2B-H), and DAPI (2B-I). The merged image (2B-I) shows co-expression of von Willebrand Factor and eGFP. Figure 2C (top panel) shows the image of a western blot using anti-a-actin and anti- -actin following FGF-8 treatment (0 ng per mL and 4 ng per mL). Figure 2C (bottom panel) shows the quantification of the western blot data.
[00179] FGF-8 significantly increased EB proliferation by 20-30% on D5 as compared to the controls (p < 0.05). In Figure 3A, the results are plotted as a percentage increase in beating EBs at D0-D17. The percentage of rhythmically beating EBs after treatment with FGF-8 (4 ng) was significantly greater compared with controls (37% versus 20%, at D17 after plating, *p < 0.05). The data are from the sets of three-five independent experiments. Figure 3B shows the staining of iPS cells treated with FGF-8 (top panel) and iPS cells not treated without FGF-8 with anti-a-actin and anti-RFP antibodies. 29.2% of cells that stained positively for a-actin from beating area.
c. ESTABLISHMENT OF APOPTOTIC CELL CULTURE MODEL AND DETERMINATION OF EFFECTS OF IPS-CM AND FGF-8 ON APOPTOSIS
[00180] The effects of iPS-CM on induced apoptosis cells by H2O2 were examined.
The concentrations of H2O2 required for induction of apoptosis in H9c2 cells was determined. H9c2 cells treated with 50-400 μηι of ¾(¾ demonstrated significant decrease in cell survival at the tested concentrations. The observed maximum decrease in cell survival (58%) occurred at 400 μιη H202 (p < 0.05) (Singla et al., 2007; Singla et al, 2008).
[00181] Using quantitative apoptotic ELISA method (Singla et al., 2007; Singla et al, 2008), the effect of iPS-CM prepared using both (i) with or (ii) without FGF-8 treated iPS cells on H2O2 induced apoptosis in H9c2 cells. The histograms of Figure 4A shows a significant increase in DNA fragmentation as measured by apoptotic ELISA (Singla et al, 2007) in H2O2 treatment when compared with untreated control group (p <0.05). The amount of cell death induced by H2O2 was significantly reduced (p < 0.05) following treatment with both iPS-CMs (Figure 4). Next, whether FGF-8 independently inhibited H2O2 induced apoptosis in this model was determined. Cell were treated with 4 ng of FGF-8 following H2O2 treatment. FGF-8 alone significantly reduced apoptosis. Treatment with both iPS-CMs generated a more robust reduction in apoptosis then treatment with FGF-8 alone (Figure 4). Data are from the set of 8-10 independent experiments. This data indicated that FGF-8 as well as iPS-CMs prepared from iPS cells with or without FGF-8 treatment act as an anti- apoptotic factor as well.
[00182] The role of FGF-8 in H2O2 induced apoptosis in H9c2 cells was evaluated.
FGF-8 demonstrated significant (p < 0.05) inhibition of H2O2 induced apoptosis in H9c2 cells. This was confirmed by apoptotic ELISA (Figure 4A). The inhibition of apoptosis by iPS-CM as determined with apoptotic ELISA kit was significantly lower (p < 0.05) than the amount inhibited by FGF-8 alone (Figure 4). The apoptotic ELISA data indicate the presence of factors other than FGF-8 that are also released into iPS-CMs and contribute in the inhibition of H2O2 induced apoptosis in H9c2 cells.
[00183] Figure 4B shows cell death detection (4B-i), caspase activity (4B-ii), tunnel % apoptotic nuclei (4B-iii), and trypan % apoptotic cells (4B-iv) following treatment with H2O2 (H); or H2O2 + conditioned medium (HICM); or H2O2 + conditioned medium + 4 ng of FGF- 8 (HICM4ng); or H202 + 4 ng of FGF-8 (H4ng). Control is indicated as C.
[00184] Those cytokines and/or growth factors present in the iPS-CMs that can provide protection from FLCVinduced apoptosis were identified. 69 cytokine and/or growth factors were analyzed with the use of Luminex technology (Singla et al, 2007).
Cytoprotective proteins released into both iPS-CM and FGF-8-iPS-CM were present at higher levels as compared with H9c2-CM, ES-CM, or cell culture media used as a control. Three major proteins known to inhibit apoptosis and enhance cardiac cell proliferation in the embryonic hearts were identified (Table 5).
[00185] Table 5. Identification of Factors Released From iPS-CM
Figure imgf000044_0001
d. ESTABLISHMENT OF THE MYOCARDIAL INFARCTION MODEL AND CARDIAC REGENERATION
[00186] Myocardial infarctions (Mis) were produced in C57BL/6 mice and iPS, ES,
H9c2 cells, or cell culture medium were transplanted post-MI. (Singla et al., 201 1; Fatma et al, 2010; Kumar et al, 2005) Immediately after coronary ligation, two intramyocardial injections of 10 μϊ^ of medium ± 2.5 x 104 iPS cells were delivered into infarct border zones. (Singla et al, 201 1; Fatma et al, 2010). LV of MI + cell culture medium (n = 8 in each group) group were compared with MI + iPS cells, MI + ES cells, and MI + H9c2 cells. MI groups were transplanted with cell culture medium as a control. Hearts were removed and examined at 2 weeks after cell transplantation.
[00187] Cardiac myocyte specific anti-sarcomeric a-actin antibody was used to identify cardiac myocytes (Figure 5A; panels a, e, I, and m), anti-RFP antibody was used to identify donor cells (Figure 5A; panel b, f, j, and n), and DAPI staining identified nuclei (Figure 5A; panels c, g, k, and o). Merged images of all three stainings are shown in Figure 5A (panels d, h, 1, and p). Heart sections stained with an anti-RFP antibody demonstrated that the transplanted iPS cells were present in the peri-infarct and infarct regions of the myocardium in C57BL/6 mice (Figure 5A; panel n). There were no stained cells in H9c2 cell transplanted hearts (Figure 5A; panel f), MI + ES or iPS cell transplanted hearts stained with an anti-RFP antibody demonstrated patchy areas of engrafted cells in the peri-infarct and infarct region of the heart (Figure 5A; panels j and n). In Figure 5 A, the scale bar = 50 μιη.
[00188] To confirm whether transplanted cells differentiated into cardiac myocytes, all four groups were co-labeled with a sarcomeric a-actin antibody. Colocalization of cardiac actin and RFP was demonstrated in both cell transplanted sections, which indicated that the transplanted stem cells differentiate into new cardiac myocytes (Figure 5A; panels d, h, 1, and p). Newly generated cardiac myocytes were rarely found in the H9c2 cell transplanted group. Quantitative data shows that there were 1.92 ± 0.3% newly formed cardiac myocytes in ES cell transplanted hearts as compared to 2.37 ± 0.3% in iPS cells (Figure 5B). The percent positive differentiated cardiac myocytes in iPS and ES groups were significantly (p < 0.05) different compared to the H9c2 cells and MI groups (*p < 0.001 vs. MI and H9c2 cells). There was no statistical difference between iPS and ES cell groups (Figure 5B).
[00189] To confirm whether newly generated cardiac myocytes demonstrate electrical coupling in the native myocardium, heart sections were triple immunolabeled (i.e., RFP to detect donor iPS cells (panels b and g), sarcomeric a-actin to detect cardiac myocytes (panels a and f), and connexin-43 to identify gap junctions (panels c and h). DAPI staining to identify nuclei is shown in Figure 5C (panels d and i) and the merged image is shown in panels e and j. Figure 5C shows newly generated cardiac myocytes form gap junctions, were electrically coupled, and integrated into the myocardium. (Singla et al, 201 1). In Figure 5C (panels a-e), the scale bar scale bar = 50 μιη, and for panels (f-j), the scale bar = 20 μιη.
e. INHIBITION OF APOPTOSIS BY THE TRANSPLANTATION OF IPS CELLS
[00190] Effects of transplanted iPS cells on cardiac apoptosis were determined by
TUNEL staining, caspase-3 immunostaining, and caspase-3 activity. (Singla et al, 201 1). TUNEL staining data showed a reduced amount of red apoptotic nuclei in MI + iPS or ES cell groups as compared to the wide spread red apoptotic nuclei in MI and MI + H9c2 cell groups (Figure 6A). DAPI stained total nuclei (Figure 6A; panels b, e, h, and k) and merged images are visible (Figure 6A; panels c, f, i and 1). In Figure 6A, the scale bar = 50 μιη.
[00191] Figure 6B shows anti-sarcomeric a-actin staining (Figure 6B; panels a and f),
TUNEL (Figure 6A; panels b and g), caspase-3 immunolabeling, (Figure 6B; panels c and h), and DAPI (Figure 6B; panels d and i). Merged images are shown in Figure 6B (panels e and j). In panels a-e, the scale bar = 50 μιη and in panels f-j, the scale bar = 20 μιη.
[00192] Quantitative TUNEL apoptotic nuclei showed transplanted iPS cells significantly (p < 0.05) reduced apoptotic nuclei (0.6 ± .07%) as compared to the MI or MI + H9c2 cell groups (1.2 ± 0.1% apoptotic nuclei/section for MI; 1 ± 0.2% apoptotic nuclei/section for MI ± H9c2 cells; Figure 6C). Transplanted ES cells reduced apoptosis comparable to the iPS cell group. TU EL stained heart sections were also combined with caspase-3 staining to confirm cell death was apoptotic in nature.
[00193] Some of the sections were also co-stained with a third antibody, sarcomeric a- actin, to confirm apoptosis occurred in the cardiomyocytes. The data in Figure 6B confirms apoptotic nuclei were stained with TUNEL, were co-labeled with caspase-3 antibody, and were positive with sarcomeric a-actin, which indicated that apoptosis occurred in cardiac myocytes. Next, caspase-3 activity, which is considered a hallmark of apoptosis, was measured. The data showed that following iPS or ES cell transplantation, caspase-3 activity was significantly (p < 0.05) reduced as compared to MI, and that H9c2 cells demonstrated no decrease in caspase-3 activity (Figure 6D). The caspase-3 activity data coupled with the TUNEL apoptotic nuclei staining confirmed apoptosis in the heart.
f. INHIBITION OF FIBROSIS BY TRANSPLANTED IPS CELLS
[00194] To determine whether inhibition of fibrosis was associated with changes in
MMP-2 and MMP-9, both of which are well-documented MMPs involved in the
pathogenesis of MI, the levels of MMP-2 and MMP-9 were quantified using an enzyme- linked immunoassay. As shown in Figure 7 (top panel), hearts transplanted with ES and iPS cells demonstrated significantly reduced MMP-2 concentration as compared to MI and H9c2 cell controls (*p < 0.05 vs. sham, #p < 0.05 vs. MI and MI + H9c2). Figure 7 (bottom panel) shows that hearts transplanted with ES and iPS cells demonstrated significantly reduced MMP-9 concentration as compared to MI and H9c2 cell controls. (*p < 0.05 vs. sham, #p < 0.05 vs. MI and MI + H9c2).
[00195] Also, Figures 13A-E show histological staining (trichome staining) demonstrating the effect of sham-MI, MI, and MI with FGF-8 treatment. Figure 13G shows the volume of interstitial fibrotic area. Figure 13H shows the comparison of vascular fibrosis (VF) vs. vascular area (VA) (in %) following sham-MI, MI, and MI with FGF-8 treatment.
[00196] Histological examinations were then performed to determine whether transplanted iPS or ES cells inhibited adverse cardiac remodeling (Singla et al, 201 1). Figure 8A shows representative photomicrographs from Masson's trichrome stained heart sections from the various treatment groups. LV remodeling was determined by measuring total interstitial fibrotic area in mm2 in all mice groups transplanted with or without stem cells. Heart sections obtained from infarcted hearts transplanted with iPS and ES cells showed a significant (p < 0.05) decrease in fibrosis (0.25 ± 0.09 mm2 for MI + iPS cells and 0.21 ± .05 mm2 for MI+ES cells) as compared to the large fibrotic areas in MI and H9c2 cell transplanted hearts (0.94 ± 0.03 mm2 for MI and 0.9 ± 07 mm2 for MI + H9c2 cells, Figure 8B).
g. ECHOCARDIOGRAPHY EVIDENCE OF IMPROVEMENT IN LV FUNCTION WITH CELL TRANSPLANTATION
[00197] M-mode echocardiography was used to determine the effect of cell transplantation on LV size and function in mice two weeks post-MI. Fractional shortening (FS) was improved (*p < 0.05) by H9c2 cell transplantation, ES cells, and cardiac iPS cells. (Singla et al, 201 1). iPS cells showed a robust increase in FS as compared to H9c2 cells (*p < 0.05 vs. H9c2 and ES cells) and as compared to MI + cell culture medium (#p < 0.001 vs. MI) (Figure 8C and 8D).
[00198] Moreover, Figure 14A-F shows the results of various outcome measures including fractional shortening and % ejection fraction following sham-MI, MI, and MI with FGF-8 treatment. (LVIDd = left ventricular internal diameter in diastole; LVIDs = left ventricular internal diameter in systole; EDV = end-diastolic volume; ESV = end-systolic volume). These data are quantified in Table 6
[00199] Table 6. Cardiac Function
Figure imgf000047_0001
h. INHIBITION OF APOPTOSIS VIA TRANSPLANTED FGF-8
[00200] The cell culture data indicated that FGF-8 is a novel anti-apoptotic factor (see
Figure 4). Two intramyocardial injections of 20 μϊ^ FGF-8 containing 50 ng/niL were performed. Hearts were removed and examined at 2 weeks post-MI. As shown in Figure 9A and Figure 9B, TU EL staining showed that apoptosis post-MI was significantly reduced in the FGF-8 treated MI hearts as compared to non-FGF-8 treated MI hearts (*p < 0.05). Figure 9C shows the casapse-3 activity under the same experimental conditions.
[00201] Furthermore, Figure 9D and Figure 9E shows the effect of FGF-8 treatment on the expression of Bcl-2 and Bax. Briefly, the Bcl-2 family consists of more than twenty anti- and pro-apoptotic members that modulate the balance between life and death. Figure 9D (top panel) shows western blotting of sham, MI, and MI+FGF-8 treated samples with anti-Bcl-2 and anti -beta-actin (control) antibodies. Figure 9D (bottom panel) shows the quantitative results of the western blotting experiments involving Bcl-2 (n = 5). Figure 9E (top panel) shows western blotting of sham, MI, and MI+FGF-8 treated samples with anti-Bax and anti- beta-actin (control) antibodies. Figure 9E (bottom panel) shows the quantitative results of the western blotting experiments involving Bax (n = 3).
i. EFFECT OF TRANSPLANTED STEM CELLS ON MIR-486 EXPRESSION
[00202] The MI heart data indicated a significant increase in cardiac apoptosis. This increase was blunted following iPS cell transplantation (see Figure 6). A miRNA signature panel of 365 different miRNAs (rodent taqman array) was used to determine up- or down- regulation of various miRNAs with and without cell transplantation post-MI. As shown in Figure 10, the levels of miR-486 were significantly decreased following MI and this decrease was inhibited following iPS cell transplantation), indicating that miR-486 is a cell survival miRNA. (*p < 0.001 vs. sham, #p < 0.05 vs. MI).
j . EFFECT OF TRANSPLANTED IPS CELLS ON AKT AND PTEN PATHWAYS
[00203] Direct and indirect mechanisms of apoptotic inhibition can occurs through activation of various cell survival cascades such as adenosine triphosphate-dependent tyrosine kinase (Akt). Using a phosho-Akt ELISA, it was determined that hearts transplanted with ES and iPS cells had significant activation of p-Akt as compared to hearts transplanted with H9c2 cells and cell culture medium (Figure 1 1A, *p < 0.05 vs. sham, #p < 0.05 vs. MI and MI + H9c2). To evaluate whether increased activation of p-Akt was attributable to inhibition of phosphoinositide-3 -kinase (PI3K)/Akt signaling, levels of phosphatase and tensin homolog (PTEN), an inhibitor of PI3K phosphorylation, were quantified. This data showed a significant reduction in PTEN concentration in hearts transplanted with ES and iPS cells as compared to hearts transplanted with H9c2 cells or cell culture medium (Figure 1 IB, *p < 0.05 vs. sham, #p < 0.05 vs. MI and MI + H9c2).
[00204] The effect of FGF-8 treatment on PTEN and P-PTEN expression in heart homogenates two weeks after either sham-treatment (sham) or myocardial infarction (MI) or myocardial infarction with FGF-8 treatment (MI+FGF8) is shown in Figures 11C and 1 ID. Figure 11C shows a western blot of sham, MI, and MI+FGF-8 utilizing anti-PTEN, anti-P- PTEN, and anti-Beta-actin antibodies. Figure 1 ID (top and bottom panels) shows the quantitation of these western blot experiments. Figure 1 IE shows p-AKT expression in following sham treatment (sham), myocardial infarction (MI), and myocardial infarction with FGF-8 (MI+FGF8). Figure 1 IF shows lipid peroxidase expression following sham treatment (sham), myocardial infarction (MI), and myocardial infarction with FGF-8 treatment (MI+FGF8).
2. DETERMINATION OF WHETHER TRANSPLANTATION OF IPS CELLS PRIMED WITH FGF-8 ENHANCES CARDIAC REPAIR AND REGENERATION FOLLOWING MI
a. GENERAL METHODS
[00205] Cell therapy is currently being examined for angiogenesis, cardiac
regeneration, and repair in MI. (Singla et al, 201 1 ; Fatma et al., 2010; Behfar et al, 2002; Balsam et al., 2004; Beltrami et al, 2003; Shujia et al, 2008). Success in cell transplantation requires an understanding of the molecular mechanisms of improved cardiac function and identification of appropriate growth factor or factors to enhance engraftment and proliferation following cell transplantation. Certain growth factors such as granulocyte-colony stimulating factor and stem cell factor have been shown to enhance myocardial regeneration. (Orlic et al, 2001). Mouse ES cells in the presence of TGFP2 have been shown to enhance cardiac myocyte differentiation. (Singla et al, 2005).
[00206] FGF-8 is a predominant cardiac growth factor, which plays a role in the development of heart. (Ilagan et al., 2006; Marques et al, 2008; Reifers et al, 2000). FGF-8 is a growth factor which plays a role in heart development as explained above. (Ilagan et al, 2006). To date, the role of FGF-8 in adult heart regeneration or with iPS cell treatment has never been explored. However, as demonstrated herein, FGF-8 (i) acts as an anti-apoptotic factor in the adult infarcted heart, and (ii) activates iPS cells and stimulates differentiation into cardiac myocytes.
(1) DETERMINATION OF IPS CELL PROLIFERATION AND CELL DIVISION
POST-MI
[00207] The determination of the number of engrafted iPS cells which have started proliferating is as follows: BrdU (50 mg/kg body weight, i.p. (Beltrami et al, 2003), which identifies nuclei in S phase in order to label actively dividing cells, is administered to the subject. Active cell growth of engrafted cells is identified using double-label immunostaining for RFP and BrdU (Dako). RFP identifies donor engrafted cells and BrdU identifies DNA synthesizing cells. To determine how many cells have started cell division at any given time, Ki-67 is utilized. Ki-67 is a nuclear antigen that is associated with cell division and that labels proliferating cells (G1-, S-, G2 -phase and mitosis), but does not label quiescent or resting cells (GO-phase). Double-label immunostaining is used to detect proliferating cells using Ki-67 (Dako) antibodies and donor engrafted cells using RFP antibodies. Slides are analyzed using fluorescence (Olympus) and confocal microscopy.
(2) IDENTIFICATION OF CELL TYPE DIFFERENTIATION POST-MI
[00208] Cell differentiation is determined using double-label immunostaining to detect donor cells (RFP antibody) and differentiated cell types using cell specific antibodies.
Cardiac-specific antibodies such as cardiac myosin heavy chain (MF-20 antibody, hybridoma bank), sarcomeric a-actin (Sigma), a-actinin, cardiac troponin I (Spectral Diagnostic), and connexin-43 (Chemicon) are used to detect cardiac myocytes and gap junctions. Endothelial cells are detected with factor VIII (Sigma) and Griffonia simplicifolia lectin (Sigma) antibodies. For smooth muscle cells, a-smooth muscle actin antibody (sigma) is used. To determine the newly formed progenitor cardiac myocytes, sections are immunostained with cardiac specific transcriptional factors such as GATA-4, Nkx2.5, and MEF2 (Santa Cruz Biotechnology). Sections are then stained with respective FITC or rhodamine conjugated secondary antibodies and counterstained with DAPI for nuclear visualization. Slides are analyzed using Olympus fluorescence and confocal microscopy.
(3) IDENTIFICATION OF MMPS ASSOCIATED WITH FIBROSIS
[00209] SDS-PAGE and Western blot analyses are utilized to determine the protein expression of collagen types I and III (Santa Cruz Biotechnology), MMP-2 and MMP-9 (Santa Cruz Biotechnology/Cell Signaling), and TIMP-1 (Santa Cruz Biotechnology).
Densitometry is used to measure band density. ELISA is performed with a kit (R & D, Minneapolis) to measure MMP-2 and MMP-9 in the LV tissue homogenates.
(4) FUNCTIONAL ASSESSMENT OF THE REGENERATED HEART
[00210] Echocardiography is an accurate noninvasive tool for the determination of quantitative characterization of post- MI heart remodeling in the mouse model. (Singla et al, 201 1; Fatma et al, 2010). Echocardiography is done on all groups post-MI hearts at short- term and long-term time points. M-mode images (Singla et al, 201 1 ; Fatma et al., 2010) in a short axis view is performed to measure LV mass, LV mass to body ratio, LV anterior and posterior wall thickness, and fractional shortening.
(5) AVOIDANCE OF TERATOMA FORMATION
[00211] Both human and mouse ES cells form complex teratomas when engrafted into an immune deficient host, a characteristic of ES cells resulting from their pluripotential capacity (Damjanov et al, 2004; Thomson et al, 1998). Teratoma formation is considered a major potential limitation in the therapeutic use of ES cells, especially when the transplanted undifferentiated number of cells is higher than 100,000 (Nussbaum et al, 2007). It has been suggested that intramyocardial transplantation of lxlO6 ES or iPS (generated from fibroblasts) cells in the mouse heart causes teratomas (Nelson et al, 2009; Nussbaum et al, 2007). However, no teratoma formation was observed with transplantation of 300,000 cells (Behfar et al. 2002). The transplantation of 3xl04 undifferentiated ES cells did not result in the formation of teratomas after 2 (Singla et al, 2006) and 4 weeks. No teratoma formation in the infarcted heart following H9c2 cell-induced iPS cell transplantation (5xl04
undifferentiated iPS cells) was observed. (Singla et al, 201 1). This number of transplanted iPS cells was less than that in studies reporting teratoma formation (Nussbaum et al, 2007) (range varies from 100,000 to one million undifferentiated ES or iPS cells). In an effort to avoid teratoma formation, the experiments discussed below utilize 1 x 105 differentiated iPS cardiac committed (FGF-8 primed) cells. If a larger number of transplanted iPS cells are required by methods disclosed herein, then teratoma detection studies are utilized.
(6) EXAMINATION OF IMMUNOGENICITY OF IPS CELLS
[00212] iPS cells from various sources may vary in their epigenetic alterations (Kim et al, 2010). One study suggested that iPS cells generated from fibroblasts using retroviral and episomal approaches are immunogenic in nature. (Zhao et al, 2011). However, generated iPS cells by the episomal approach were less immunogenic compared with the cells generated by the retroviral approach (Zhao et al, 201 1). As reported herein, iPS cells are generated from H9c2 cells using plasmid transfection. This method generation, the number of injected cells, and the location of injection are completely different than those studies reporting
immunogenicity of iPS cells (Zhao et al, 2011). Methods described herein utilize experimental groups that receive and do not receive cyclosporine.
b. DETERMINATION OF THE EFFECTS OF INTRAMYOCARDIAL INJECTION OF DIFFERENTIATED FCC-IPS CELLS
[00213] Using an established myocardial infarction (MI) model (Singla et al, 2011), RFP-labeled differentiated FCC-iPS cells (total of 1 x 105 cells) are injected in two different intramyocardial injections in the peri-infarct zone of the infarcted heart. (Singla et al, 2011). Using the same established MI model, differentiated iPS, ES cells, fibroblast reprogrammed iPS cells, H9c2 cells, FGF-8, and cell culture medium (as a control) are also injected. The effects of these injections on cardiac remodeling and regeneration are quantified. [00214] C57BL/6 mice are treated with cyclosporine A (250-300 mg/kg) for 5 days before the cell transplantation (Menard et al, 2005; Giralt et al, 1997). The cyclosporine A dose can be altered based on the desire for an average cyclosporine serum concentration of 300-400 ng/L (Menard et al, 2005; Giralt et al, 1997). Animals are killed humanely at short- term (1, 14, and 28 days) and long-term (8, 12, and 18 weeks) time points after cell transplantation. Tissues are harvested for histological analysis and preparation of tissue homogenates.
[00215] FCC-iPS are treated as follows; FGF-8 (4 ng/mL) is added to the iPS cells in the differentiation medium (a medium which does not contain iPS cell self-renewal growth factors such as activin A, leukemia inhibitory factor and mouse embryonic fibroblast condition medium) for 48 hours. These iPS cells are considered as FGF-8 primed differentiated iPS cells. Control iPS or ES cells are cultured for 48 hours in a differentiation medium without FGF-8.
[00216] With respect to immunogenicity, any infiltration of T-cells are identified using anti-CD3 and anti-CD4 antibodies (Zhao et al, 2011). H&E stained sections are used to identify the presence of any inflammatory cells.
c. IDENTIFICATION OF APOPTOSIS AND FIBROSIS
[00217] Heart sections are prepared (Singla et al, 2011) for immunostaining to determine apoptosis and cell proliferation. Sections are stained with a RFP or GFP antibody along with a Oct3/4 (undifferentiated cell marker on iPS or ES cells) antibody to determine total number of engrafted cells. TU EL staining are combined with a caspase-3 antibody to establish the extent of cell death at each time point. (Singla et al, 2011 ; Singla et al, 2006; Singla et al, 2007). Sections are counter-stained with DAPI (Vectashield) to localize the nucleus. To determine engrafted cell apoptosis, sections are stained with RFP, Oct3/4, and TUNEL. Cell specific apoptosis within the host myocardium is determined using double- label immunostaining to detect apoptotic cells (TUNEL, Bax, and caspases) and native heart cell types. Cardiac myocytes are detected with antibodies against cardiac myosin heavy chain (MF-20 antibody, hybridoma bank), sarcomeric a-actin (Sigma), a-actinin, and cardiac troponin I (Spectral Diagnostic). (Singla et al, 2011 ; Singla et al, 2006). Endothelial cells are detected with factor VIII (Sigma) and Griffonia simplicifolia lectin (Sigma) antibodies. For smooth muscle (SM) cells, SM a-actin, or SM myosin heavy chain antibodies (Sigma) are used. An antibody against fibronectin (Sigma) is used to detect fibroblasts. Sections will then be stained with FITC or rhodamine conjugated secondary antibodies and counterstained with DAPI for nuclear visualization. Olympus fluorescence and confocal microscopy are used to assess apoptosis.
[00218] Heart sections are stained with Mason's trichrome and Sirius red F3BA or
PSR to analyze the effect of iPS cells on gross interstitial and perivascular fibrosis.
Specifically, H&E, Masson's Trichrome, and immunostained sections are used to quantify fibrosis, apoptosis, and distribution of heart cell types in the left ventricular (LV) region of the heart. (Singla et al, 2011; Singla et al, 2007). Integrated morphometry analysis (IMA) and NIH image J processing software are employed for these analyses.
3. DETERMINATION OF WHETHER POST-MI TRANSPLANTATION OF IPS-CM ENHANCES ATTENUATION OF ADVERSE CARDIAC REMODELING AS COMPARED TO A GROWTH FACTOR COCKTAIL
[00219] Acute MI is associated with irreversible cell loss due to apoptosis and necrosis (Anversa et al, 1997; Anversa et al, 1998; Kumar et al., 2002). Adult or embryonic stem cells have the potential to form new cardiac cells in the injured myocardium (Singla et al, 2009). Some studies suggest that an immediate release of autocrine or paracrine factors from donor stem cells decrease host myocardium remodeling (Singla et al, 2009; Gnecchi et al, 2005; Singla et al, 2010). However, there is no data available that determines the effects of secreted autocrine or paracrine factors from iPS cells in a MI model.
[00220] The data presented herein indicate that factors released from iPS cells are distinct and include anti-apoptotic and anti-fibrotic factors such as FGF-9, IL-10 and ΤΓΜΡ- 1. Since injection of iPS-CM, which can contain numerous cytoprotective factors, does not pose a threat of teratoma formation, such an injection offers the possibility of beneficial effects on cell transplantation.
a. EXAMINATION OF EFFECTS OF INTRAMYOCARDIAL INJECTION OF IPS- CM ON CARDIAC REMODELING POST-MI
[00221] iPS-CM from iPS-cells primed with FGF-8 and without FGF-8 is utilized
(Singla et al, 2007; Singla et al, 2008). A myocardial infarction (MI) model for C57BL/6 mice having received or not having received cyclosporine A is utilized. If cyclosporine A generates no significant difference in cardiac remodeling, the use of cyclosporine A can be discontinued. Using two intramyocardial injections totaling 20 μϊ^ volume are administered post-MI for (i) FCC-iPS-CM (15x) (Singla et al, 2011; Fatma et al, 2010), (ii) a cocktail of FGF-8 (20 μΐ. of 50 ng/mL) + FGF-9 (20 μί, 50 ng/mL) + IL-10 (20 of 100 ng/mL (Kobbe et al., 2009)) + TIMP-1 (20 μΐ, of 100 ng/mL (Meissbruger et al., 2011), or (iii) cell culture medium (as a control). Additional cell specific CM such ES-CM and H9c2-CM are used for comparison purposes. Subsequently, the effects on cardiac remodeling are quantified. Heart homogenates are taken and released factors are examined using Luminex technology, cytokine array, and ELISA methods (Singla et al, 2007; Singla et al, 2008; Singla et al, 2011). The levels of released factor are compared to the controls and increases are confirmed with western blotting. Heart sections are prepared for immunostaining and the degrees of apoptosis and fibrosis are evaluated. H&E, Masson's Trichrome, and Sirius red F3BA or PSR staining are used to analyze the effect of iPS cells on cell morphology and gross interstitial and perivascular fibrosis.
b. IDENTIFICATION OF MMPS ASSOCIATED WITH FIBROSIS
[00222] SDS-PAGE and Western blot analyses are utilized to determine the protein expression of collagen types I and III (Santa Cruz Biotechnology), MMP-2 and MMP-9 (Santa Cruz Biotechnology/Cell Signaling), and TIMP-1 (Santa Cruz Biotechnology).
Densitometry is used to measure band density. ELISA is performed with a kit (R & D, Minneapolis) to measure MMP-2 and MMP-9 in the LV tissue homogenates.
c. FUNCTIONAL ASSESSMENT OF THE REGENERATED HEART
[00223] Echocardiography is an accurate noninvasive tool for the determination of quantitative characterization of post- MI heart remodeling in the mouse model. (Singla et al, 2011; Fatma et al, 2010). Echocardiography is done on all groups post-MI hearts at short- term and long-term time points. M-mode images (Singla et al, 2011; Fatma et al., 2010) in a short axis view is performed to measure LV mass, LV mass to body ratio, LV anterior and posterior wall thickness, and fractional shortening.
4. DETERMINATION OF WHETHER ATTENUATION OF APOPTOSIS IS MEDIATED VIA MIR-486 AND ITS PTEN AND FOXOIA TARGETS AND ASSOCIATED AKT PATHWAY.
[00224] miRNAs are small non-coding RNAs about 20-24 nucleotides in length that play a major role in the regulation of a variety of cell processes including apoptosis, hypertrophy, fibrosis, and cardiac differentiation (Care et al, 2007; Catalucci et al, 2008; Chen et al, 2006; Cimmino et al, 2005). Certain miRNAs such as miR-21, miR-133 and miR- 1 are involved in various pro- and anti-apoptotic and anti-fibrotic activities in the heart (Care et al, 2007; Catalucci et al, 2008; Chen et al., 2006; Cimmino et al, 2005). In the studies described herein, miR-486 was down-regulated following ML, and that iPS cell transplantation significantly increased the levels of miR-486 following MI. While there are a few published papers describing miR-486 induction of myoblast differentiation, miR-486 involvement in cell survival in Sezary syndrome, and increased levels of miR-486 in the postnatal cardiac growth (Dey et al, 201 1; Narducci et al, 201 1; Small et al, 2010), there is no data regarding miR-486 levels and apoptosis following iPS cell transplantation.
a. EXAMINATION OF MIR-486 LEVELS AND ITS TARGETS (PTEN/FOXOIA
AND ASSOCIATED AKT PATHWAY)
[00225] MI hearts transplanted with primed iPS cells or CM are utilized as described above. Briefly, FCC-iPS is treated as follows; FGF-8 (4 ng/mL) is added to the iPS cells in the differentiation medium (a medium which does not contain iPS cell self-renewal growth factors such as activin A, leukemia inhibitory factor and mouse embryonic fibroblast condition medium) for 48 hours. These iPS cells are considered as FGF-8 primed differentiated iPS cells. iPS-CM from iPS-cells primed with FGF-8 and without FGF-8 is utilized.
[00226] LV are removed and miRNA is isolated using miRVANA kit (ABI).Using
RT-PCR, the levels of miR-486 are ascertained. miR-486 is also confirmed with northern blotting (Dey et al, 2011 ; Narducci et al, 201 1 ; Small et al, 2010). To determine the role of miR-486 on cardiac apoptosis, animals are injected with a selective inhibitor of miR-486 (locked nucleic acid, LNA-antagomir-486, 80 mg/kg) (Hatley et al, 2010; Patrick et al., 2010). To identify expression of miR-486 in the infarcted, non-infarcted, and peri-infarcted region post-MI, heart sections are stained using in situ-hybridization for miR-486. In situ- hybridization employing LNA-miR-486 specific probes (Exiqon) (Hosoda et al, 201 1) is utilized. The expression of miR-486 in major heart cell types is characterized using double label immunostaining with cell specific antibodies. In brief, after in situ hybridization, sections are co-labeled with heart cell specific antibodies such as cardiac a-actin for cardiac myocytes, CD31 for endothelial cells and smooth muscle, a-actin for VSM cells as described above. Sections are counter-stained with DAPI to identify cell nuclei.
[00227] To determine whether PTEN/FOXOIa targets and associated Akt are affected at the translational level proteins are prepared from LV heart homogenates. SDS-PAGE and Western blot analyses are performed to determine the protein expression of total and phosphorylated PTEN, FOXOla, and Akt (Cell Signaling). Densitometry is used to measure band density. Additionally, ELISA is performed with a kit (Ray Biotech Inc.) to measure total and phosphorylated PTEN, FOXOla, and Akt.
b. UTILIZATION OF LUCIFERASE REPORTER ASSAY FOR MIR-486 TARGETS
[00228] To confirm whether miR-486 directly binds to the 3 '-UTRs of its predicted target genes, PTEN and Foxola, luciferase reporter vectors are utilized. The pCMV-Luc- 3UTR vector was constructed by sequential cloning the firefly luciferase gene (obtained from Promega) to the pcDNA 3.1 expression vector (Invitrogen) under the control of a CMV promoter. For the luciferase reporter assay, the 3 '-UTR sequences for mouse PTEN (~3.5kb in length) and Foxola (~1 kb in length) were generated by high-fidelity PCR and then cloned downstream of the firefly luciferase stop codon. HEK 293 cells are grown in 6-well plates until they reach 80% confluency. Luciferase reporter constructs are co-transfected to the cells with a negative control vector or miR-486 expression vector in the presence or absence of a specific synthetic antagomir against miR-486. The luciferase assay is performed 48 hours after transfection using a luciferase assay kit (Promega) according to the manufacturer' instructions. Luminometer is used to quantify luciferase activity.
[00229] All of the compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention.
[00230] More specifically, certain agents which are both chemically and
physiologically related can be substituted for the agents described herein while the same or similar results can be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
[00231] Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.
D. REFERENCES
[00232] The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
[00233] Anversa,P. et al. Apoptosis and myocardial infarction. Basic Res. Cardiol. 93 Suppl 3, 8-12 (1998).
[00234] Anversa,P. et al. Myocyte cell death and ventricular remodeling. Curr. Opin. Nephrol. Hypertens. 6, 169-176 (1997).
[00235] Balsam,L.B. et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 428, 668-673 (2004).
[00236] Behfar,A. et al. Stem cell differentiation requires a paracrine pathway in the heart. FASEB J. 16, 1558-1566 (2002).
[00237] Beltrami,A.P. et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 1 14, 763-776 (2003).
[00238] Biesecker,L.G. et al. Interleukin-6 is a component of human umbilical cord serum and stimulates hematopoiesis in embryonic stem cells in vitro. Exp. Hematol. 21, 774- 778 (1993).
[00239] Care, A. et al. MicroRNA-133 controls cardiac hypertrophy. Nat. Med. 13,
613-618 (2007).
[00240] Catalucci,D. et al. MicroRNAs control gene expression: importance for cardiac development and pathophysiology 25. Ann. N. Y. Acad. Sci. 1 123, 20-29 (2008).
[00241] Chang,M.Y. et al. Direct reprogramming of rat neural precursor cells and fibroblasts into pluripotent stem cells 1. PLoS. One. 5, e9838 (2010).
[00242] Chen,J.F. et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat. Genet. 38, 228-233 (2006).
[00243] Cimmino,A. et al. miR-15 and miR-16 induce apoptosis by targeting BCL2 9.
Proc. Natl. Acad. Sci. U. S. A 102, 13944-13949 (2005).
[00244] DamjanoVjI. From stem cells to germ cell tumors and back. Verh. Dtsch. Ges.
Pathol. 88, 39-44 (2004).
[00245] Dey,B.K. et al. miR-206 and -486 induce myoblast differentiation by downregulating Pax7. Mol. Cell Biol. 31, 203-214 (2011).
[00246] Dhingra,S. et al. p38 and ERK1/2 MAPKs mediate the interplay of TNF-alpha and IL-10 in regulating oxidative stress and cardiac myocyte apoptosis. Am. J. Physiol Heart Circ. Physiol 293, H3524-H3531 (2007).
[00247] Dyer,L.A. et al. The role of secondary heart field in cardiac development.
Dev. Biol. 336, 137-144 (2009).
[00248] Fatma,S. et al. Factors Released from Embryonic Stem Cells Stimulate c-kit-
FLK-l(+ve) Progenitor Cells and Enhance Neovascularization. Antioxid. Redox. Signal. 13, 1857-1865 (2010).
[00249] Foo, et al. Death begets failure in the heart. J Clin Invest 2005;115:565-571).
[00250] Frontini,M.J. et al. Fibroblast growth factor 9 delivery during angiogenesis produces durable, vasoresponsive microvessels wrapped by smooth muscle cells. Nat. Biotechnol. 29, 421-427 (201 1).
[00251] Giralt,S. et al. Phase I trial of cyclosporine-induced autologous graft- versus- host disease in patients with multiple myeloma undergoing high-dose chemotherapy with autologous stem-cell rescue. J. Clin. Oncol. 15, 667-673 (1997).
[00252] Gnecchi,M. et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat. Med. 11, 367-368 (2005).
[00253] Haider & Ashraf. Bone marrow cell transplantation in clinical perspective. J
Mol Cell Cardiol 2005;38:225-235.
[00254] Hatley,M.E. et al. Modulation of K-Ras -dependent lung tumorigenesis by
MicroRNA-21. Cancer Cell 18, 282-293 (2010).
[00255] Hosoda,T. et al. Human cardiac stem cell differentiation is regulated by a mircrine mechanism. Circulation 123, 1287-1296 (2011).
[00256] Huang, J. Y. et al. Fibroblast growth factor 9 upregulates heme oxygenase- 1 and gamma-glutamylcysteine synthetase expression to protect neurons from 1 -methyl-4- phenylpyridinium toxicity. Free Radic. Biol. Med. 49, 1099-1 108 (2010).
[00257] Ilagan,R. et al. FGF-8 is required for anterior heart field development 1.
Development 133, 2435-2445 (2006).
[00258] Kajstura, et al. Bone marrow cells differentiate in cardiac cell lineages after infarction independently of cell fusion. Circ Res 2005;96: 127-137
[00259] Kim,K. et al. Epigenetic memory in induced pluripotent stem cells. Nature
467, 285-290 (2010).
[00260] Kobbe,P. et al. IL-10 administration attenuates pulmonary neutrophil infiltration and alters pulmonary iNOS activation following hemorrhagic shock. Inflamm. Res. 58, 170-174 (2009).
[00261] KofidiSjT. et al. Stimulation of paracrine pathways with growth factors enhances embryonic stem cell engraftment and host-specific differentiation in the heart after ischemic myocardial injury. Circulation 1 11, 2486-2493 (2005).
[00262] Kumar,D. et al. Distinct mouse coronary anatomy and myocardial infarction consequent to ligation. Coron. Artery Dis. 16, 41-44 (2005).
[00263] Kumar,D. et al. Oxidative stress and apoptosis in heart dysfunction. Herz 27,
662-668 (2002).
[00264] Kuzmenkin,A. et al. Functional characterization of cardiomyocytes derived from murine induced pluripotent stem cells in vitro 1. FASEB J. 23, 4168-4180 (2009). [00265] Lunde,K. et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N. Engl. J. Med. 355, 1199-1209 (2006).
[00266] Marques,S.R. et al. Reiterative roles for FGF signaling in the establishment of size and proportion of the zebrafish heart . Dev. Biol. 321, 397-406 (2008).
[00267] Meissburger,B. et al. Tissue inhibitor of matrix metalloproteinase 1 (TIMP1) controls adipogenesis in obesity in mice and in humans. Diabetologia 54, 1468-1479 (201 1).
[00268] MenardjC. et al. Transplantation of cardiac-committed mouse embryonic stem cells to infarcted sheep myocardium: a preclinical study. Lancet 366, 1005-1012 (2005).
[00269] Narducci,M.G. et al. MicroRNA profiling reveals that miR-21, miR486 and miR-214 are upregulated and involved in cell survival in Sezary syndrome. Cell Death. Dis. 2, el51 (201 1).
[00270] Nelson,T.J. et al. Repair of acute myocardial infarction by human sternness factors induced pluripotent stem cells. Circulation 120, 408-416 (2009).
[00271] Nussbaum,J. et al. Transplantation of undifferentiated murine embryonic stem cells in the heart: teratoma formation and immune response. FASEB J. 21, 1345-1357 (2007).
[00272] Orlic et al, Bone marrow cells regenerate infarcted myocardium. Nature
200;410:701-705
[00273] Orlic,D. et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc. Natl. Acad. Sci. U. S. A 98, 10344-10349 (2001).
[00274] Park,I.H. et al. Disease-specific induced pluripotent stem cells 1. Cell 134,
877-886 (2008).
[00275] Park,I.H. et al. Generation of human-induced pluripotent stem cells 4. Nat.
Protoc. 3, 1180-1 186 (2008).
[00276] Patrick,D.M. et al. Stress-dependent cardiac remodeling occurs in the absence of microRNA-21 in mice. J. Clin. Invest 120, 3912-3916 (2010).
[00277] Reifers,F. et al. Induction and differentiation of the zebrafish heart requires fibroblast growth factor 8 (FGF-8/acerebellar) 1. Development 127, 225-235 (2000).
[00278] Shujia,J. et al. Stable therapeutic effects of mesenchymal stem cell-based multiple gene delivery for cardiac repair. Cardiovasc. Res. 77, 525-533 (2008).
[00279] Singla et al, Transplanted embryonic stem cells following mouse myocardial infarction inhibit apoptosis and cardiac remodeling. Am J Physiol Heart Circ Physiol
2007;293 :H1308-H1314).
[00280] Singla. Stem cells in the infarcted heart. J Cardiovasc Transl Res 2010;3 :73- 78)
[00281] Singla,D.K. et al. Factors released from embryonic stem cells inhibit apoptosis of H9c2 cells. Am. J. Physiol Heart Circ. Physiol 293, H1590-H1595 (2007).
[00282] Singla,D.K. et al. Enhancement by growth factors of cardiac myocyte differentiation from embryonic stem cells: A promising foundation for cardiac regeneration. Biochem. Biophys. Res. Commun. 335, 637-642 (2005).
[00283] Singla,D.K. et al. Transforming growth factor-beta2 enhances differentiation of cardiac myocytes from embryonic stem cells. Biochem. Biophys. Res. Commun. 332, 135- 141 (2005).
[00284] Singla,D.K. Embryonic stem cells in cardiac repair and regeneration.
Antioxid. Redox. Signal. 1 1, 1857-1863 (2009).
[00285] Singla,D.K. et al. Transplantation of embryonic stem cells into the infarcted mouse heart: formation of multiple cell types. J. Mol. Cell Cardiol. 40, 195-200 (2006).
[00286] Singla,D.K. Stem cells in the infarcted heart. J. Cardiovasc. Transl. Res. 3, 73-
78 (2010).
[00287] Singla,D.K. et al. iPS Cells Repair and Regenerate Infarcted Myocardium.
Mol. Pharm.(201 1).
[00288] Singla,D.K. et al. Transplanted embryonic stem cells following mouse myocardial infarction inhibit apoptosis and cardiac remodeling. Am. J. Physiol Heart Circ. Physiol 293, H1308-H1314 (2007).
[00289] Singla,D.K. et al. Factors released from embryonic stem cells inhibit apoptosis in H9c2 cells through PI3K/Akt but not ERK pathway. Am. J. Physiol Heart Circ. Physiol 295, H907-H913 (2008).
[00290] Singla,D.K. et al. TGF-beta2 treatment enhances cytoprotective factors released from embryonic stem cells and inhibits apoptosis in infarcted myocardium. Am. J. Physiol Heart Circ. Physiol 300, H1442-H1450 (2011).
[00291] Slager,H.G. et al. Transforming growth factor-beta in the early mouse embryo: implications for the regulation of muscle formation and implantation. Dev. Genet. 14, 212- 224 (1993).
[00292] Small,E.M. et al. Regulation of PI3-kinase/Akt signaling by muscle-enriched microR A-486. Proc. Natl. Acad. Sci. U. S. A 107, 4218-4223 (2010).
[00293] Takahashi,K. et al. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors 3. Cell 126, 663-676 (2006). [00294] Tendera,M. et al. Intracoronary infusion of bone marrow-derived selected
CD34+CXCR4+ cells and non-selected mononuclear cells in patients with acute STEMI and reduced left ventricular ejection fraction: results of randomized, multicentre Myocardial Regeneration by Intracoronary Infusion of Selected Population of Stem Cells in Acute Myocardial Infarction (REGENT) Trial 1. Eur. Heart J. 30, 1313-1321 (2009).
[00295] Thomson,J.A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1 147 (1998).
[00296] Wiles,M.V. et al. Multiple hematopoietic lineages develop from embryonic stem (ES) cells in culture. Development 1 11, 259-267 (1991).
[00297] Yun,Y.R. et al. Fibroblast growth factors: biology, function, and application for tissue regeneration. J. Tissue Eng 2010, 218142 (2010).
[00298] Zhang,J. et al. Functional cardiomyocytes derived from human induced pluripotent stem cells 2. Circ. Res. 104, e30-e41 (2009).
[00299] Zhao,T. et al. Immunogenicity of induced pluripotent stem cells.
Nature(2011).

Claims

s claimed is:
A method of enhancing cardiac tissue and/or cell regeneration following cardiac dysfunction in a subject in need thereof, the method comprising:
(i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells,
(ii) administering to the subject conditioned media of fibroblast growth factor-
8 primed induced pluripotent stem cells;
(iii) administering to the subject fibroblast growth factor-8; and/or
(iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9; and
determining regeneration in the heart.
A method of attenuating adverse cardiac tissue and/or cell remodeling following cardiac dysfunction in a subject in need thereof, the method comprising:
(i) administering to the subject fibroblast growth factor-8 primed induced pluripotent stem cells;
(ii) administering to the subject conditioned media of fibroblast growth factor-
8 primed induced pluripotent stem cells;
(iii) administering to the subject fibroblast growth factor-8; and/or
(iv) administering to the subject a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9; and
determining a decrease in cardiac remodeling.
The method of claim 1, wherein enhancing cardiac tissue and/or cell regeneration comprises enhancing cell engraftment.
The method of claim 1, wherein enhancing cardiac tissue and/or cell regeneration comprises enhancing cell proliferation.
The method of claim 1, wherein enhancing cardiac tissue and/or cell regeneration comprises enhancing cell differentiation.
The method of claim 1 or claim 2, wherein the induced pluripotent stem cells differentiate into cardiac myocytes.
The method of claim 2, wherein attenuating adverse cardiac remodeling comprises inhibiting fibrosis and/or fibrosis-related mechanisms.
8. The method of claim 7, wherein expression or activity of at least one matrix metalloproteinase is decreased.
9. The method of claim 8, wherein the at least one matrix metalloproteinase is MMP-2 or MMP-9.
10. The method of claim 1 or claim 2, further comprising improving angiogenesis in the subject.
1 1. The method of claim 1 or claim 2, further comprising enhancing cardiac function in the subject.
12. The method of claim 11, wherein enhancing cardiac function comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (v) normalizing of heart geometry, or (vi) a combination thereof.
13. The method of claim 1 or claim 2, wherein the conditioned media comprises anti- apoptotic and anti-fibrotic factors.
14. The method of claim 13, wherein anti-apoptotic and anti-fibrotic factors comprise fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, and tissue inhibitor of matrix metallopreinase-1.
15. The method of claim 2, further comprising inhibiting or decreasing necrosis.
16. The method of claim 2, further comprising inhibiting or decreasing apoptosis and/or apoptosis-related mechanisms.
17. The method of claim 16, wherein apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells.
18. The method of claim 16, wherein apoptosis and/or apoptosis-related mechanisms occurs in cardiac tissue of the subject.
19. The method of claim 16, wherein miR-486 expression increases, Akt expression increases, PTEN expression decreases, FOXOla expression decreases, and/or a combination thereof.
20. The method of claim 1 or claim 2, wherein the induced pluripotent stem cells are cardiac-committed.
21. The method of claim 1 or claim 2, wherein between 5,000 and 500,000 induced pluripotent stem cells are administered to the subject.
22. The method of claim 21, wherein approximately 100,000 induced pluripotent stem cells are administered to the subject.
23. The method of claim 21, wherein less than 100,000 induced pluripotent stem cells are administered to the subject.
24. The method of claim 21, wherein more than 100,000 induced pluripotent stem cells are administered to the subject.
25. The method of any one of claims 22-24, wherein the induced pluripotent stem cells are administered in one intramycardial injection.
26. The method of any one of claims 22-24, wherein the induced pluripotent stem cells are administered in two or more intramycardial injections.
27. The method of any one of claims 22-24, wherein the induced pluripotent stem cells are administered into a peri-infarct zone and/or an infarcted zone of the injured myocardium.
28. The method of claim 1 or claim 2, further comprising administering to the subject one or more immunosuppressive drugs.
29. The method of claim 28, wherein the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
30. The method of claim 28, wherein the one or more immunosuppressive drugs is cyclosporine A.
31. The method of claim 28, wherein the one or more immunosuppressive drugs is administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells.
32. The method of claim 1 or claim 2, further comprising administering fibroblast growth factor- 8 to the subject.
33. The method of claim 32, wherein the fibroblast growth factor-8 is administered to the subject prior to, during, and/or following the administration of the induced pluripotent stem cells.
34. The method of claim 1 or claim 2, further comprising repeating the administration of induced pluripotent stem cells to the subject.
35. The method of claim 32, further comprising repeating the administration of fibroblast growth factor-8 to the subject.
36. The method of claim 1 or claim 2, wherein the cardiac dysfunction is a cardiac ischemia/reperfusion event.
37. The method of claim 36, wherein the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
38. The method of claim 1 or claim 2, wherein the cardiac dysfunction is a congenital heart defect.
39. The method of claim 38, wherein the congenital heart defect is hypoplasia or pentalogy of Cantrell.
40. The method of claim 1 or claim 2, wherein the induced pluripotent stem cells are administered to the subject prior to, during, and/or following cardiac dysfunction.
41. The method of claim 40, wherein the induced pluripotent stem cells are administered within 10, 15, 20, 25, 30, or more minutes following cardiac dysfunction.
42. The method of claim 40, wherein the induced pluripotent stem cells are administered within 1, 2, 6, 12, 18, 24, or more hours following cardiac dysfunction.
43. The method of claim 1 or claim 2, wherein the induced pluripotent stem cells are obtained from an autologous, allogeneic, or syngeneic source.
44. The method of claim 1 or claim 2, wherein the induced pluripotent stem cells are obtained from fibroblast cells.
45. The method of claim 1 or claim 2, wherein the induced pluripotent stem cells are obtained from H9c2 ells.
46. The method of claim 44 or claim 45, wherein the fibroblast cells or the H9c2 cells are transfected with a vector comprising a nucleic acid molecule encoding at least one stemness factor.
47. The method of claim 46, wherein the at least one stemness factor comprise c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
48. A composition for enhancing cardiac tissue/cell regeneration in a subject, comprising:
(i) fibroblast growth factor-8 primed induced pluripotent stem cells; (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
49. A composition for attenuating adverse cardiac tissue/cell remodeling in a subject, comprising: (i) fibroblast growth factor-8 primed induced pluripotent stem cells, and/or (ii) conditioned medium of fibroblast growth factor-8 primed induced pluripotent stem cells; (iii) fibroblast growth factor-8; and/or (iv) a composition comprising fibroblast growth factor-8 and fibroblast growth factor-9.
50. The composition of claim 47 or claim 48, wherein the subject has experienced a cardiac ischemia/reperfusion event.
51. The composition of claim 50, wherein the ischemia/reperfusion event is myocardial infarction, myocardial ischemia, myocardial reperfusion, subendocardial ischemia, Takayasu's arteritis, atrial fibrillation, hemorrhagic stroke, an event that occurs during cardiac surgery where a heart lung machine is used such as coronary artery bypass, or an event that occurs during the preservation of an organ for transplant.
52. The composition of claim 47 or claim 48, wherein the subject has a congenital heart defect.
53. The composition of claim 52, wherein the congenital heart defect is hypoplasia or pentalogy of Cantrell.
54. The composition of claim 50, wherein the composition is administered within 10, 15, 20, 25, 30, or more minutes following the ischemia/reperfusion event.
55. The composition of claim 50, wherein the composition is administered within 1, 2, 6, 12, 18, 24, or more hour following the ischemia/reperfusion event.
56. The composition of claim 47, wherein the composition enhances cell engraftment.
57. The composition of claim 47, wherein the composition enhances cell proliferation.
58. The composition of claim 47, wherein the composition enhances cell differentiation.
59. The composition of claim 47 or claim 48, wherein the induced pluripotent stem cells differentiate into cardiac myocytes.
60. The composition of claim 48, wherein the composition inhibits fibrosis and/or fibrosis-related mechanisms.
61. The composition of claim 60, wherein the composition decreases expression or activity of at least one matrix metalloproteinase.
62. The composition of claim 61, wherein the at least one matrix metalloproteinase is MMP-2 or MMP-9.
63. The composition of claim 47 or claim 48, wherein the composition improves angiogenesis in the subject.
64. The composition of claim 47 or claim 48, wherein the composition enhances cardiac function in the subject.
65. The composition of claim 64, wherein enhancing cardiac function comprises one or more of (i) improving left ventricular function, (ii) improving fractional shortening, (iii) improving ejection fraction, (iv) reducing end-diastolic volume, (v) decreasing left ventricular mass, and (vi) normalizing of heart geometry, and (vii) a combination thereof.
66. The composition of claim 47 or claim 48, wherein the conditioned media comprises anti-apoptotic and anti-fibrotic factors.
67. The composition of claim 66, wherein anti-apoptotic and anti-fibrotic factors are fibroblast growth factor-9, fibroblast growth factor-8, interleukin-10, or tissue inhibitor of matrix metalloproteinase-1, or a combination thereof.
68. The composition of claim 48, wherein the composition inhibits or decreases necrosis.
69. The composition of claim 48, wherein the composition inhibits or decreases apoptosis and/or apoptosis-related mechanisms.
70. The composition of claim 69, wherein the apoptosis and/or apoptosis-related mechanisms occurs in the induced pluripotent stem cells.
71. The composition of claim 69, wherein the apoptosis and/or apoptosis-related mechanisms occurs in the cardiac tissue of the subject.
72. The composition of claim 69, wherein the composition increases miR-486 expression, increases Akt expression, decreases PTEN expression, decreases FOXOla expression, and/or a combination thereof.
73. The composition of claim 47 or claim 48, wherein the induced pluripotent stem cells are cardiac -committed.
74. The composition of claim 47 or claim 48, wherein the composition comprises 5,000 and 500,000 induced pluripotent stem cells.
75. The composition of claim 74, wherein the composition comprises approximately 100,000 induced pluripotent stem cells.
76. The composition of claim 74, wherein the composition comprises less than 100,000 induced pluripotent stem cells.
77. The composition of claim 74, wherein the composition comprises more than 100,000 induced pluripotent stem cells.
78. The composition of any one of claims 74-77, wherein the composition is administered to the subject in one intramycardial injection.
79. The composition of any one of claims 74-77, wherein the composition is administered to the subject in two or more intramycardial injections.
80. The composition of any one of claims 74-77, wherein the composition is administered into a peri-infarct zone and/or an infarcted zone of the injured myocardium.
81. The composition of claim 47 or claim 48, further comprising one or more immunosuppressive drugs.
82. The composition of claim 81, wherein the one or more immunosuppressive drugs comprise corticosteroids, calcineurin inhibitors, anti-proliferatives, and mTOR inhibitors.
83. The composition of claim 81, wherein the one or more immunosuppressive drugs is cyclosporine A.
84. The composition of claim 47 or claim 48, further comprising fibroblast growth factor- 8.
85. The composition of claim 47 or claim 48, wherein the induced pluripotent stem cells are obtained from an autologous, allogeneic, or syngeneic source.
86. The composition of claim 47 or claim 48, wherein the induced pluripotent stem cells are obtained from fibroblast cells.
87. The composition of claim 47 or claim 48, wherein the induced pluripotent stem cells are obtained from H9c2 cells.
88. The composition of claim 86 or claim 87, wherein the cells are transfected with at least one stemness factor.
89. The composition of claim 88, wherein the at least one stemness factors is c-myc, oct 3/4, Klf4, nanog, or Sox2, or a combination thereof.
90. The composition of 86 or claim 87, wherein the cells are transected with c-myc, oct 3/4, Klf4, or Sox2, or a combination thereof.
91. A method of generating cardiac induced pluripotent stem cells, the method
comprising:
(i) inserting one or more nucleic acid constructs capable of expressing stem- cell like factors into a cardiac cell type, wherein the stem-cell like factors comprise Oct3/4, KIf4, Sox2, and c-Myc, or a combination thereof; and
(ii) obtaining the cardiac induced pluripotent stem cells stably expressing the stem-cell like factors in the cardiac cell type.
92. A method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising: administering conditioned medium from induced pluripotent stem cells, wherein the conditioned medium is administered with or without FGF-8.
93. A method of identifying a myocardial injury or infarcted myocardial tissue,
comprising: measuring the amount of miR-486, wherein the amount of miR-486 decreases after myocardial injury or myocardial infarction, and wherein the amount of miR-486 increases after transplantation of induced pluripotent stem cells.
94. A method of inhibiting apoptosis and/or apoptosis-related mechanisms, comprising: administering FGF-8.
PCT/US2013/030080 2012-03-30 2013-03-09 Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling WO2013148121A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA2869163A CA2869163A1 (en) 2012-03-30 2013-03-09 Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling
US14/389,402 US20150056172A1 (en) 2012-03-30 2013-03-09 Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling
EP13770363.3A EP2830639B1 (en) 2012-03-30 2013-03-09 Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261617985P 2012-03-30 2012-03-30
US61/617,985 2012-03-30

Publications (1)

Publication Number Publication Date
WO2013148121A1 true WO2013148121A1 (en) 2013-10-03

Family

ID=49261001

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/030080 WO2013148121A1 (en) 2012-03-30 2013-03-09 Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling

Country Status (3)

Country Link
EP (1) EP2830639B1 (en)
CA (1) CA2869163A1 (en)
WO (1) WO2013148121A1 (en)

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020061837A1 (en) 1996-09-05 2002-05-23 Lough John W. Bone morphogenetic protein and fibroblast growth factor compositions and methods for the induction of cardiogenesis
US20020142457A1 (en) 1999-12-28 2002-10-03 Akihiro Umezawa Cell having the potentiality of differentiation into cardiomyocytes
WO2006081190A2 (en) 2005-01-25 2006-08-03 Five Prime Therapeutics, Inc. Compositions and methods for treating cardiac conditions
EP2098244A1 (en) 2008-03-04 2009-09-09 Medizinische Hochschule Hannover Pharmaceutical composition for the treatment of myocardial infarction
WO2009118928A1 (en) 2008-03-26 2009-10-01 Kyoto University Efficient production and use of highly cardiogenic progenitors and cardiomyocytes from embryonic and induced pluripotent stem cells
US20110097799A1 (en) 2009-10-19 2011-04-28 Casey Stankewicz Cardiomyocyte production
US20110200568A1 (en) * 2008-08-08 2011-08-18 Yasuhiro Ikeda Induced pluripotent stem cells
US20110250182A1 (en) * 2010-04-07 2011-10-13 Anthrogenesis Corporation Angiogenesis using placental stem cells
US20110260727A1 (en) * 2010-04-21 2011-10-27 Punchard William F B Shim insert for high-field MRI magnets
WO2011139357A1 (en) * 2010-05-03 2011-11-10 The Texas A & M University System Adult stem cells/progenitor cells and stem cell proteins for treatment of eye injuries and diseases

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020061837A1 (en) 1996-09-05 2002-05-23 Lough John W. Bone morphogenetic protein and fibroblast growth factor compositions and methods for the induction of cardiogenesis
US20020142457A1 (en) 1999-12-28 2002-10-03 Akihiro Umezawa Cell having the potentiality of differentiation into cardiomyocytes
WO2006081190A2 (en) 2005-01-25 2006-08-03 Five Prime Therapeutics, Inc. Compositions and methods for treating cardiac conditions
EP2098244A1 (en) 2008-03-04 2009-09-09 Medizinische Hochschule Hannover Pharmaceutical composition for the treatment of myocardial infarction
WO2009118928A1 (en) 2008-03-26 2009-10-01 Kyoto University Efficient production and use of highly cardiogenic progenitors and cardiomyocytes from embryonic and induced pluripotent stem cells
US20110200568A1 (en) * 2008-08-08 2011-08-18 Yasuhiro Ikeda Induced pluripotent stem cells
US20110097799A1 (en) 2009-10-19 2011-04-28 Casey Stankewicz Cardiomyocyte production
US20110250182A1 (en) * 2010-04-07 2011-10-13 Anthrogenesis Corporation Angiogenesis using placental stem cells
US20110260727A1 (en) * 2010-04-21 2011-10-27 Punchard William F B Shim insert for high-field MRI magnets
WO2011139357A1 (en) * 2010-05-03 2011-11-10 The Texas A & M University System Adult stem cells/progenitor cells and stem cell proteins for treatment of eye injuries and diseases

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
ANVERSA, P. ET AL.: "Apoptosis and myocardial infarction", BASIC RES. CARDIOL., vol. 93, no. 3, 1998, pages 8 - 12
ANVERSA, P. ET AL.: "Myocyte cell death and ventricular remodeling", CURR. OPIN. NEPHROL. HYPERTENS., vol. 6, 1997, pages 169 - 176
BALSAM, L.B. ET AL., HAEMATOPOIETIC STEM CELLS ADOPT MATURE HAEMATOPOIETIC
SINGLA ET AL., AM. J. PHYS. HEART CIRC. PHYSIOL., vol. 293, no. 3, 2007, pages H1590 - H1595
SINGLA ET AL., MOLECULAR PHARMACEUTICS, vol. 8, no. 5, 2011, pages 1573 - 1581
SINGLA.: "Embryonic Stem Cells in Cardiac Repair and Regeneration.", ANTIOXIDANTS & REDOX SIGNALING, vol. 11, no. 8, 2009, pages 1857 - 1863, XP055112853 *

Also Published As

Publication number Publication date
EP2830639A4 (en) 2016-01-20
CA2869163A1 (en) 2013-10-03
EP2830639B1 (en) 2019-05-08
EP2830639A1 (en) 2015-02-04

Similar Documents

Publication Publication Date Title
French et al. A naturally derived cardiac extracellular matrix enhances cardiac progenitor cell behavior in vitro
Drago et al. The stem cell secretome and its role in brain repair
Lü et al. Both the transplantation of somatic cell nuclear transfer-and fertilization-derived mouse embryonic stem cells with temperature-responsive chitosan hydrogel improve myocardial performance in infarcted rat hearts
US20110305673A1 (en) Compositions and methods for tissue repair
AU2017308738B2 (en) Method for generating mesoderm and/or endothelial colony forming cell-like cells having in vivo blood vessel forming capacity
CN102459576A (en) Compositions and methods for modulating stem cells and uses thereof
US11913020B2 (en) Method for improving stem cell migration using ethionamide
US9771555B2 (en) Canine amniotic membrane-derived multipotent stem cells
Van Laake et al. Cardiomyocytes derived from stem cells
Senthilkumar et al. Remarkable migration propensity of dental pulp stem cells towards neurodegenerative milieu: An in vitro analysis
US10443044B2 (en) Generating cardiac progenitor cells from pluripotent stem cells using isoxazole or isoxazole like compounds
US20210230551A1 (en) Enhancement of fibroblast plasticity for treatment of disc degeneration
US20200129433A1 (en) Stem cell-derived microvesicles with enhanced efficacy, use thereof, and method for enhancing efficacy
CN113710258A (en) Knockdown of PRPF31 gene expression enhances survival of human cells differentiated in vitro
Cho et al. Granulocyte colony-stimulating factor treatment enhances the efficacy of cellular cardiomyoplasty with transplantation of embryonic stem cell-derived cardiomyocytes in infarcted myocardium
JP5940147B2 (en) Cell preparation containing myocardial directional cells
US20230212570A1 (en) Cell-penetrating peptide-microrna conjugates for intracellular cell delivery
EP2830639B1 (en) Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling
de Lázaro et al. Non-viral induction of transient cell reprogramming in skeletal muscle to enhance tissue regeneration
WO2013148122A1 (en) Methods and compositions using fgf-9 to enchance neovascularization and regeneration
US20150056172A1 (en) Methods and compositions using fgf-8 to enhance cardiac regeneration and attenuate adverse cardiac remodeling
US9193954B2 (en) Methods and compositions for mesenchymal stem cell proliferation
WO2020190672A1 (en) Cardiomyocyte-derived exosomes inducing regeneration of damaged heart tissue
US20200190475A1 (en) Methods for identifying and isolating cardiac stem cells and methods for making and using them
US20230285353A1 (en) A chemical cocktail driving expansion of myogenic stem cells

Legal Events

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

Ref document number: 13770363

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2869163

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2013770363

Country of ref document: EP