Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Circ Res ; 120(9): 1466-1476, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28298297

ABSTRACT

RATIONALE: Paracrine secretions seem to mediate therapeutic effects of human CD34+ stem cells locally transplanted in patients with myocardial and critical limb ischemia and in animal models. Earlier, we had discovered that paracrine secretion from human CD34+ cells contains proangiogenic, membrane-bound nanovesicles called exosomes (CD34Exo). OBJECTIVE: Here, we investigated the mechanisms of CD34Exo-mediated ischemic tissue repair and therapeutic angiogenesis by studying their miRNA content and uptake. METHODS AND RESULTS: When injected into mouse ischemic hindlimb tissue, CD34Exo, but not the CD34Exo-depleted conditioned media, mimicked the beneficial activity of their parent cells by improving ischemic limb perfusion, capillary density, motor function, and their amputation. CD34Exo were found to be enriched with proangiogenic miRNAs such as miR-126-3p. Knocking down miR-126-3p from CD34Exo abolished their angiogenic activity and beneficial function both in vitro and in vivo. Interestingly, injection of CD34Exo increased miR-126-3p levels in mouse ischemic limb but did not affect the endogenous synthesis of miR-126-3p, suggesting a direct transfer of stable and functional exosomal miR-126-3p. miR-126-3p enhanced angiogenesis by suppressing the expression of its known target, SPRED1, simultaneously modulating the expression of genes involved in angiogenic pathways such as VEGF (vascular endothelial growth factor), ANG1 (angiopoietin 1), ANG2 (angiopoietin 2), MMP9 (matrix metallopeptidase 9), TSP1 (thrombospondin 1), etc. Interestingly, CD34Exo, when treated to ischemic hindlimbs, were most efficiently internalized by endothelial cells relative to smooth muscle cells and fibroblasts, demonstrating a direct role of stem cell-derived exosomes on mouse endothelium at the cellular level. CONCLUSIONS: Collectively, our results have demonstrated a novel mechanism by which cell-free CD34Exo mediates ischemic tissue repair via beneficial angiogenesis. Exosome-shuttled proangiogenic miRNAs may signify amplification of stem cell function and may explain the angiogenic and therapeutic benefits associated with CD34+ stem cell therapy.


Subject(s)
Angiogenic Proteins/metabolism , Antigens, CD34/metabolism , Endothelial Progenitor Cells/transplantation , Exosomes/transplantation , Ischemia/surgery , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Angiogenic Proteins/genetics , Animals , Biomarkers/metabolism , Cells, Cultured , Culture Media, Conditioned/metabolism , Disease Models, Animal , Endothelial Progenitor Cells/metabolism , Exosomes/metabolism , Gene Expression Regulation , Hindlimb , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Motor Activity , Paracrine Communication , Phenotype , RNA Interference , Recovery of Function , Regional Blood Flow , Signal Transduction , Time Factors , Transfection
2.
PLoS One ; 11(1): e0147615, 2016.
Article in English | MEDLINE | ID: mdl-26808574

ABSTRACT

The role of inflammation on atherosclerosis and restenosis is well established. Restenosis is thought to be a complex response to injury, which includes early thrombus formation, acute inflammation and neo-intimal growth. Inflammatory cells are likely contributors in the host response to vascular injury, via cytokines and chemokines secretion, including TNF-alpha (TNF). We have previously shown that IL-10 inhibits TNF and other inflammatory mediators produced in response to cardiovascular injuries. The specific effect of IL-10 on endothelial cell (ECs) biology is not well elucidated. Here we report that in a mouse model of carotid denudation, IL-10 knock-out mice (IL-10KO) displayed significantly delayed Re-endothelialization and enhanced neo-intimal growth compared to their WT counterparts. Exogenous recombinant IL-10 treatment dramatically blunted the neo-intimal thickening while significantly accelerating the recovery of the injured endothelium in WT mice. In vitro, IL-10 inhibited negative effects of TNF on ECs proliferation, ECs cell cycle, ECs-monocyte adhesion and ECs apoptosis. Furthermore, IL-10 treatment attenuated TNF-induced smooth muscle cells proliferation. Our data suggest that IL-10 differentially regulate endothelial and vascular smooth cells proliferation and function and thus inhibits neo-intimal hyperplasia. Thus, these results may provide insights necessary to develop new therapeutic strategies to limit vascular restenosis during percutaneous coronary intervention (PCI) in the clinics.


Subject(s)
Carotid Arteries/pathology , Endothelium, Vascular/pathology , Interleukin-10/physiology , Tunica Intima/pathology , Animals , Cell Adhesion/physiology , Cell Cycle Checkpoints/physiology , Cell Proliferation , Hyperplasia , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/physiology
3.
Cardiovasc Res ; 104(3): 412-22, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25341896

ABSTRACT

AIMS: The E2F transcription factors are best characterized for their roles in cell-cycle regulation, cell growth, and cell death. Here we investigated the potential role of E2F1 in cardiac neovascularization. METHODS AND RESULTS: We induced myocardial infarction (MI) by ligating the left anterior descending artery in wild-type (WT) and E2F1(-/-) mice. E2F1(-/-) mice demonstrated a significantly better cardiac function and smaller infarct sizes than WT mice. At infarct border zone, capillary density and endothelial cell (EC) proliferation were greater, apoptotic ECs were fewer, levels of VEGF and placental growth factor (PlGF) were higher, and p53 level was lower in E2F1(-/-) than in WT mice. Blockade of VEGF receptor 2 (VEGFR2) signalling with the selective inhibitor SU5416 or with the VEGFR2-blocking antibody DC101 abolished the differences between E2F1(-/-) mice and WT mice in cardiac function, infarct size, capillary density, EC proliferation, and EC apoptosis. In vitro, hypoxia-induced VEGF and PlGF up-regulation was significantly greater in E2F1(-/-) than in WT cardiac fibroblasts, and E2F1 overexpression suppressed PlGF up-regulation in both WT and p53(-/-) cells; however, VEGF up-regulation was suppressed only in WT cells. E2F1 interacted with and stabilized p53 under hypoxic conditions, and both E2F1 : p53 binding and the E2F1-induced suppression of VEGF promoter activity were absent in cells that expressed an N-terminally truncated E2F1 mutant. CONCLUSION: E2F1 limits cardiac neovascularization and functional recovery after MI by suppressing VEGF and PlGF up-regulation through p53-dependent and -independent mechanisms, respectively.


Subject(s)
Coronary Vessels/physiology , E2F1 Transcription Factor/metabolism , Neovascularization, Physiologic , Pregnancy Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Fibroblasts/metabolism , Gene Expression Regulation , Heart/physiology , Hypoxia/metabolism , Male , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Placenta Growth Factor , Proteasome Endopeptidase Complex/metabolism , Recovery of Function , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
J Mol Cell Cardiol ; 74: 231-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25009075

ABSTRACT

The translation of cell-based therapies for ischemic tissue repair remains limited by several factors, including poor cell survival and limited target site retention. Advances in nanotechnology enable the development of specifically designed delivery matrices to address these limitations and thereby improve the efficacy of cell-based therapies. Given the relevance of integrin signaling for cellular homeostasis, we developed an injectable, bioactive peptide-based nanofiber matrix that presents an integrin-binding epitope derived from fibronectin, and evaluated its feasibility as a supportive artificial matrix for bone marrow-derived pro-angiogenic cells (BMPACs) used as a therapy in ischemic tissue repair. Incubation of BMPACs with these peptide nanofibers in vitro significantly attenuated apoptosis while enhancing proliferation and adhesion. Pro-angiogenic function was enhanced, as cells readily formed tubes. These effects were, in part, mediated via p38, and p44/p42 MAP kinases, which are downstream pathways of focal adhesion kinase. In a murine model of hind limb ischemia, an intramuscular injection of BMPACs within this bioactive peptide nanofiber matrix resulted in greater retention of cells, enhanced capillary density, increased limb perfusion, reduced necrosis/amputation, and preserved function of the ischemic limb compared to treatment with cells alone. This self-assembling, bioactive peptide nanofiber matrix presenting an integrin-binding domain of fibronectin improves regenerative efficacy of cell-based strategies in ischemic tissue by enhancing cell survival, retention, and reparative functions.


Subject(s)
Bone Marrow Cells/cytology , Epitopes/metabolism , Fibronectins/metabolism , Ischemia/therapy , Nanofibers/administration & dosage , Peptides/administration & dosage , Animals , Biocompatible Materials , Bone Marrow Cells/metabolism , Cell Survival , Cell- and Tissue-Based Therapy/methods , Epitopes/chemistry , Fibronectins/chemistry , Gene Expression , Hindlimb/blood supply , Hindlimb/drug effects , Hindlimb/injuries , Integrins/metabolism , Ischemia/pathology , Male , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nanofibers/chemistry , Neovascularization, Physiologic , Peptides/chemical synthesis , Peptides/metabolism , Protein Binding , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
5.
J Biol Chem ; 288(25): 18022-34, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23645678

ABSTRACT

We have shown previously that estrogen (estradiol, E2) supplementation enhances voluntary alcohol consumption in ovariectomized female rodents and that increased alcohol consumption impairs ischemic hind limb vascular repair. However, the effect of E2-induced alcohol consumption on post-infarct myocardial repair and on the phenotypic/functional properties of endothelial progenitor cells (EPCs) is not known. Additionally, the molecular signaling of alcohol-estrogen interactions remains to be elucidated. This study examined the effect of E2-induced increases in ethanol consumption on post-infarct myocardial function/repair. Ovariectomized female mice, implanted with 17ß-E2 or placebo pellets were given access to alcohol for 6 weeks and subjected to acute myocardial infarction. Left ventricular functions were consistently depressed in mice consuming ethanol compared with those receiving only E2. Alcohol-consuming mice also displayed significantly increased infarct size and reduced capillary density. Ethanol consumption also reduced E2-induced mobilization and homing of EPCs to injured myocardium compared with the E2-alone group. In vitro, exposure of EPCs to ethanol suppressed E2-induced proliferation, survival, and migration and markedly altered E2-induced estrogen receptor-dependent cell survival signaling and gene expression. Furthermore, ethanol-mediated suppression of EPC biology was endothelial nitric oxide synthase-dependent because endothelial nitric oxide synthase-null mice displayed an exaggerated response to post-acute myocardial infarction left ventricular functions. These data suggest that E2 modulation of alcohol consumption, and the ensuing EPC dysfunction, may negatively compete with the beneficial effects of estrogen on post-infarct myocardial repair.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/drug effects , Estradiol/pharmacology , Ethanol/pharmacology , Myocardium/metabolism , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Estradiol/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Ovariectomy , Protein Binding/drug effects , Receptors, Estrogen/metabolism , Stem Cells/metabolism , Stem Cells/physiology
6.
Angiogenesis ; 16(1): 45-58, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22941227

ABSTRACT

Neural stem cells (NSCs) differentiate into endothelial cells (ECs) and neuronal cells. Estradiol (E2) is known to exhibit proangiogenic effects on ischemic tissues via EC activation. Therefore, we hypothesized that E2 can promote the therapeutic potential of NSC transplantation for injured nerve repair via the differentiation of NSCs into ECs during neovascularization. NSCs isolated from newborn mouse brains were transplanted into injured sciatic nerves with (NSC/E2 group) or without E2-conjugated gelatin hydrogel (E2 group). The NSC/E2 group exhibited the greatest recovery in motor nerve conduction velocity, voltage amplitude, and exercise tolerance. Histological analyses revealed increased intraneural vascularity and blood perfusion as well as striking NSC recruitment to the neovasculature in the injured nerves in the NSC/E2 group. In vitro, E2 enhanced the NSC migration and proliferation inhibiting apoptosis. Fluorescence-activated cell sorting analysis also revealed that E2 significantly increased the percentage of CD31 in NSCs, and the effect of E2 was completely neutralized by the estrogen receptor antagonist ICI. The combination of E2 administration and NSC transplantation cooperatively improved the functional recovery of injured peripheral nerves, at least in part, via E2-associated NSC differentiation into ECs. These findings provide a novel mechanistic insight into both NSC biology and the biological effects of endogenous E2.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Endothelial Cells/cytology , Estradiol/pharmacology , Neovascularization, Physiologic/drug effects , Neural Stem Cells/cytology , Peripheral Nerve Injuries/therapy , Animals , Cell Proliferation/drug effects , Combined Modality Therapy , Endothelial Cells/drug effects , Estradiol/blood , Estradiol/therapeutic use , Female , Lactic Acid/administration & dosage , Lactic Acid/pharmacology , Mice , Mice, Inbred C57BL , Nerve Crush , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Peripheral Nerve Injuries/blood , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/physiopathology , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Recovery of Function/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Stem Cell Transplantation
7.
Circulation ; 127(1): 63-73, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23204107

ABSTRACT

BACKGROUND: CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. METHODS AND RESULTS: Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4(+) cells were mobilized for just 1 day in both groups, but the mobilization of sca1(+)/flk1(+) cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1(+)/flk1(+) cell mobilization without altering the mobilization of CXCR4(+) cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. CONCLUSIONS: AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Subject(s)
Heterocyclic Compounds/pharmacology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines , Bone Marrow Transplantation , Cardiotonic Agents/pharmacology , Cyclams , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic/drug effects , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Nitric Oxide Synthase Type III/genetics , Recovery of Function/drug effects , Signal Transduction/drug effects
8.
Circ Cardiovasc Interv ; 5(6): 821-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23192920

ABSTRACT

BACKGROUND: Critical limb ischemia portends a risk of major amputation of 25% to 35% within 1 year of diagnosis. Preclinical studies provide evidence that intramuscular injection of autologous CD34+ cells improves limb perfusion and reduces amputation risk. In this randomized, double-blind, placebo-controlled pilot study, we evaluated the safety and efficacy of intramuscular injections of autologous CD34+ cells in subjects with moderate or high-risk critical limb ischemia, who were poor or noncandidates for surgical or percutaneous revascularization (ACT34-CLI). METHODS AND RESULTS: Twenty-eight critical limb ischemia subjects were randomized and treated: 7 to 1 × 10(5) (low-dose) and 9 to 1 × 10(6) (high-dose) autologous CD34+ cells/kg; and 12 to placebo (control). Intramuscular injections were distributed into 8 sites within the ischemic lower extremity. At 6 months postinjection, 67% of control subjects experienced a major or minor amputation versus 43% of low-dose and 22% of high-dose cell-treated subjects (P=0.137). This trend continued at 12 months, with 75% of control subjects experiencing any amputation versus 43% of low-dose and 22% of high-dose cell-treated subjects (P=0.058). Amputation incidence was lower in the combined cell-treated groups compared with control group (6 months: P=0.125; 12 months: P=0.054), with the low-dose and high-dose groups individually showing trends toward improved amputation-free survival at 6 months and 12 months. No adverse safety signal was associated with cell administration. CONCLUSIONS: This study provides evidence that intramuscular administration of autologous CD34+ cells was safe in this patient population. Favorable trends toward reduced amputation rates in cell-treated versus control subjects were observed. These findings warrant further exploration in later-phase clinical trials. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00616980.


Subject(s)
Antigens, CD34/analysis , Ischemia/surgery , Lower Extremity/blood supply , Stem Cell Transplantation , Stem Cells/immunology , Aged , Aged, 80 and over , Amputation, Surgical , Analysis of Variance , Biomarkers/analysis , Critical Illness , Disease-Free Survival , Double-Blind Method , Female , Humans , Injections, Intramuscular , Ischemia/physiopathology , Limb Salvage , Male , Middle Aged , Pilot Projects , Prospective Studies , Quality of Life , Recovery of Function , Stem Cell Transplantation/adverse effects , Time Factors , Transplantation, Autologous , Treatment Outcome , United States , Wound Healing
9.
PLoS One ; 7(5): e38009, 2012.
Article in English | MEDLINE | ID: mdl-22662261

ABSTRACT

In many organisms early development is under control of the maternal genome and zygotic gene expression is delayed until the mid-blastula transition (MBT). As zygotic transcription initiates, cell cycle checkpoints become activated and the tempo of cell division slows. The mechanisms that activate zygotic transcription at the MBT are incompletely understood, but they are of interest because they may resemble mechanisms that cause stem cells to stop dividing and terminally differentiate. The unstable regulatory protein Geminin is thought to coordinate cell division with cell differentiation. Geminin is a bi-functional protein. It prevents a second round of DNA replication during S and G2 phase by binding and inhibiting the essential replication factor Cdt1. Geminin also binds and inhibits a number of transcription factors and chromatin remodeling proteins and is thought to keep dividing cells in an undifferentiated state. We previously found that the cells of Geminin-deficient Xenopus embryos arrest in G2 phase just after the MBT then disintegrate at the onset of gastrulation. Here we report that they also fail to express most zygotic genes. The gene expression defect is cell-autonomous and is reproduced by over-expressing Cdt1 or by incubating the embryos in hydroxyurea. Geminin deficient and hydroxyurea-treated blastomeres accumulate DNA damage in the form of double stranded breaks. Bypassing the Chk1 pathway overcomes the cell cycle arrest caused by Geminin depletion but does not restore zygotic gene expression. In fact, bypassing the Chk1 pathway by itself induces double stranded breaks and abolishes zygotic transcription. We did not find evidence that Geminin has a replication-independent effect on transcription. We conclude that Geminin is required to maintain genome integrity during the rapid cleavage divisions, and that DNA damage disrupts zygotic gene transcription at the MBT, probably through activation of DNA damage checkpoint pathways.


Subject(s)
Blastula/metabolism , Cell Cycle Proteins/genetics , Gene Expression Regulation, Developmental , Xenopus/embryology , Xenopus/genetics , Zygote/metabolism , Animals , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Female , Geminin , Gene Deletion , Hydroxyurea/pharmacology , Male , Mutation , Protein Binding , T-Box Domain Proteins/metabolism , Transcription, Genetic/drug effects , Xenopus Proteins/metabolism
10.
Circ Res ; 111(3): 312-21, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22581926

ABSTRACT

RATIONALE: Ischemic cardiovascular disease represents one of the largest epidemics currently facing the aging population. Current literature has illustrated the efficacy of autologous, stem cell therapies as novel strategies for treating these disorders. The CD34+ hematopoetic stem cell has shown significant promise in addressing myocardial ischemia by promoting angiogenesis that helps preserve the functionality of ischemic myocardium. Unfortunately, both viability and angiogenic quality of autologous CD34+ cells decline with advanced age and diminished cardiovascular health. OBJECTIVE: To offset age- and health-related angiogenic declines in CD34+ cells, we explored whether the therapeutic efficacy of human CD34+ cells could be enhanced by augmenting their secretion of the known angiogenic factor, sonic hedgehog (Shh). METHODS AND RESULTS: When injected into the border zone of mice after acute myocardial infarction, Shh-modified CD34+ cells (CD34(Shh)) protected against ventricular dilation and cardiac functional declines associated with acute myocardial infarction. Treatment with CD34(Shh) also reduced infarct size and increased border zone capillary density compared with unmodified CD34 cells or cells transfected with the empty vector. CD34(Shh) primarily store and secrete Shh protein in exosomes and this storage process appears to be cell-type specific. In vitro analysis of exosomes derived from CD34(Shh) revealed that (1) exosomes transfer Shh protein to other cell types, and (2) exosomal transfer of functional Shh elicits induction of the canonical Shh signaling pathway in recipient cells. CONCLUSIONS: Exosome-mediated delivery of Shh to ischemic myocardium represents a major mechanism explaining the observed preservation of cardiac function in mice treated with CD34(Shh) cells.


Subject(s)
Antigens, CD34/administration & dosage , Hedgehog Proteins/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Myocardial Infarction/surgery , Animals , Antigens, CD34/therapeutic use , Cells, Cultured , Hedgehog Proteins/therapeutic use , Humans , Male , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Myocardial Infarction/physiopathology , NIH 3T3 Cells , Ventricular Dysfunction/physiopathology , Ventricular Dysfunction/surgery
11.
Lab Invest ; 92(4): 532-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22330336

ABSTRACT

Both estradiol (E2) and Sonic Hedgehog (Shh) contribute to angiogenesis and nerve regeneration. Here, we investigated whether E2 improves the recovery of injured nerves by downregulating the Shh inhibitor hedgehog-interacting protein (HIP) and increasing Shh-induced angiogenesis. Mice were treated with local injections of E2 or placebo one week before nerve-crush injury; 28 days after injury, nerve conduction velocity, exercise duration, and vascularity were significantly greater in E2-treated mice than in placebo-treated mice. E2 treatment was also associated with higher mRNA levels of Shh, the Shh receptor Patched-1, and the Shh transcriptional target Gli1, but with lower levels of HIP. The E2-induced enhancement of nerve vascularity was abolished by the Shh inhibitor cyclopamine, and the effect of E2 treatment on Shh, Gli1, and HIP mRNA expression was abolished by the E2 inhibitor ICI. Gli-luciferase activity in human umbilical-vein endothelial cells (HUVECs) increased more after treatment with E2 and Shh than after treatment with E2 alone, and E2 treatment reduced HIP expression in HUVECs and Schwann cells without altering Shh expression. Collectively, these findings suggest that E2 improves nerve recovery, at least in part, by reducing HIP expression, which subsequently leads to an increase in Shh signaling and Shh-induced angiogenesis.


Subject(s)
Carrier Proteins/metabolism , Estradiol/metabolism , Hedgehog Proteins/metabolism , Membrane Glycoproteins/metabolism , Neovascularization, Physiologic , Nerve Regeneration , Peripheral Nerve Injuries/metabolism , Animals , Down-Regulation , Estradiol/administration & dosage , Female , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor , Peripheral Nerve Injuries/drug therapy , Receptors, Cell Surface/metabolism , Recovery of Function , Signal Transduction , Zinc Finger Protein GLI1
12.
J Invest Dermatol ; 132(3 Pt 1): 711-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22048734

ABSTRACT

The antagonism of CXC-chemokine receptor 4 (CXCR4) with AMD3100 improves cardiac performance after myocardial infarction by augmenting the recruitment of endothelial progenitor cells (EPCs) from the bone marrow to the regenerating vasculature. We investigated whether AMD3100 may accelerate diabetes-impaired wound healing through a similar mechanism. Skin wounds were made on the backs of leptin receptor-deficient mice and treated with AMD3100 or saline. Fourteen days after treatment, wound closure was significantly more complete in AMD3100-treated mice (AMD3100: 87.0 ± 2.6%, saline: 33.1 ± 1.8%; P<0.0001) and was accompanied by greater collagen fiber formation, capillary density, smooth muscle-containing vessel density, and monocyte/macrophage infiltration. On day 7 after treatment, AMD3100 was associated with higher circulating EPC and macrophage counts, and with significantly upregulated mRNA levels of stromal cell-derived factor 1 and platelet-derived growth factor B in the wound bed. AMD3100 also promoted macrophage proliferation and phagocytosis and the migration and proliferation of diabetic mouse primary dermal fibroblasts and 3T3 fibroblasts, which express very little CXCR4. In conclusion, a single topical application of AMD3100 promoted wound healing in diabetic mice by increasing cytokine production, mobilizing bone marrow EPCs, and enhancing the activity of fibroblasts and monocytes/macrophages, thereby increasing both angiogenesis and vasculogenesis. Not all of the AMD3100-mediated effects evolved through CXCR4 antagonism.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Heterocyclic Compounds/therapeutic use , Receptors, CXCR4/antagonists & inhibitors , Skin/drug effects , Skin/injuries , Wound Healing/drug effects , Animals , Benzylamines , Capillaries/growth & development , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/biosynthesis , Collagen/biosynthesis , Cyclams , Female , Fibroblasts/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Phagocytosis/drug effects , Proto-Oncogene Proteins c-sis/biosynthesis , Receptors, Leptin/deficiency , Skin/pathology , Stem Cells , Treatment Outcome
13.
Circ Res ; 109(7): 724-8, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21835908

ABSTRACT

RATIONALE: Transplantation of human CD34(+) stem cells to ischemic tissues has been associated with reduced angina, improved exercise time, and reduced amputation rates in phase 2 clinical trials and has been shown to induce neovascularization in preclinical models. Previous studies have suggested that paracrine factors secreted by these proangiogenic cells are responsible, at least in part, for the angiogenic effects induced by CD34(+) cell transplantation. OBJECTIVE: Our objective was to investigate the mechanism of CD34(+) stem cell-induced proangiogenic paracrine effects and to examine if exosomes, a component of paracrine secretion, are involved. METHODS AND RESULTS: Exosomes collected from the conditioned media of mobilized human CD34(+) cells had the characteristic size (40 to 90 nm; determined by dynamic light scattering), cup-shaped morphology (electron microscopy), expressed exosome-marker proteins CD63, phosphatidylserine (flow cytometry) and TSG101 (immunoblotting), besides expressing CD34(+) cell lineage marker protein, CD34. In vitro, CD34(+) exosomes replicated the angiogenic activity of CD34(+) cells by increasing endothelial cell viability, proliferation, and tube formation on Matrigel. In vivo, the CD34(+) exosomes stimulated angiogenesis in Matrigel plug and corneal assays. Interestingly, exosomes from CD34(+) cells but not from CD34(+) cell-depleted mononuclear cells had angiogenic activity. CONCLUSIONS: Our data demonstrate that human CD34(+) cells secrete exosomes that have independent angiogenic activity both in vitro and in vivo. CD34(+) exosomes may represent a significant component of the paracrine effect of progenitor cell transplantation for therapeutic angiogenesis.


Subject(s)
Angiogenic Proteins/metabolism , Antigens, CD34/metabolism , Corneal Neovascularization/metabolism , Endothelial Cells/metabolism , Exosomes/metabolism , Neovascularization, Physiologic , Paracrine Communication , Stem Cells/metabolism , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Corneal Neovascularization/immunology , Culture Media, Conditioned/metabolism , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Exosomes/immunology , Exosomes/ultrastructure , Flow Cytometry , Humans , Light , Male , Mice , Mice, Nude , Microscopy, Electron, Transmission , Particle Size , Phosphatidylserines/metabolism , Platelet Membrane Glycoproteins/metabolism , Scattering, Radiation , Stem Cells/immunology , Stem Cells/ultrastructure , Tetraspanin 30 , Transcription Factors/metabolism
14.
J Am Coll Cardiol ; 57(24): 2444-52, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21658566

ABSTRACT

OBJECTIVES: This study was designed to compare the effectiveness of Sonic hedgehog (Shh) gene transfer, AMD3100-induced progenitor-cell mobilization, and Shh-AMD3100 combination therapy for treatment of surgically induced myocardial infarction (MI) in mice. BACKGROUND: Shh gene transfer improves myocardial recovery by up-regulating angiogenic genes and enhancing the incorporation of bone marrow-derived progenitor cells (BMPCs) in infarcted myocardium. Here, we investigated whether the effectiveness of Shh gene therapy could be improved with AMD3100-induced progenitor-cell mobilization. METHODS: Gene expression and cell function were evaluated in cells cultured with medium collected from fibroblasts transfected with plasmids encoding human Shh (phShh). MI was induced in wild-type mice, in matrix metalloproteinase (MMP)-9 knockout mice, and in mice transplanted with bone marrow that expressed green-fluorescent protein. Mice were treated with 100 µg of phShh (administered intramyocardially), 5 mg/kg of AMD3100 (administered subcutaneously), or both; cardiac function was evaluated echocardiographically, and fibrosis, capillary density, and BMPC incorporation were evaluated immunohistochemically. RESULTS: phShh increased vascular endothelial growth factor and stromal cell-derived factor 1 expression in fibroblasts; the medium from phShh-transfected fibroblasts increased endothelial-cell migration and the migration, proliferation, and tube formation of BMPCs. Combination therapy enhanced cardiac functional recovery (i.e., left ventricular ejection fraction) in wild-type mice, but not in MMP-9 knockout mice, and was associated with less fibrosis, greater capillary density and smooth muscle-containing vessel density, and enhanced BMPC incorporation. CONCLUSIONS: Combination therapy consisting of intramyocardial Shh gene transfer and AMD3100-induced progenitor-cell mobilization improves cardiac functional recovery after MI and is superior to either individual treatment for promoting therapeutic neovascularization.


Subject(s)
Genetic Therapy/methods , Hedgehog Proteins/administration & dosage , Heterocyclic Compounds/pharmacology , Myocardial Infarction/therapy , Neovascularization, Physiologic/physiology , Stem Cells/metabolism , Ventricular Remodeling/physiology , Animals , Benzylamines , Cell Movement/drug effects , Cell Movement/physiology , Cyclams , Disease Models, Animal , Gene Transfer Techniques , Humans , Injections, Intralesional , Injections, Subcutaneous , Mice , Mice, Knockout , Myocardial Infarction/pathology , Random Allocation , Sensitivity and Specificity , Survival Rate , Treatment Outcome
16.
Circ Res ; 107(9): 1083-93, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20847314

ABSTRACT

RATIONALE: The mobilization of bone marrow (BM) progenitor cells (PCs) is largely governed by interactions between stromal cell-derived factor (SDF)-1 and CXC chemokine receptor (CXCR)4. Ischemic injury disrupts the SDF-1-CXCR4 interaction and releases BM PCs into the peripheral circulation, where the mobilized cells are recruited to the injured tissue and contribute to vessel growth. BM PCs can also be mobilized by the pharmacological CXCR4 antagonist AMD3100, but the other components of the SDF-1-CXCR4 signaling pathway are largely unknown. c-kit, a membrane-bound tyrosine kinase and the receptor for stem cell factor, has also been shown to play a critical role in BM PC mobilization and ischemic tissue repair. OBJECTIVE: To investigate the functional interaction between SDF-1-CXCR4 signaling and c-kit activity in BM PC mobilization. METHODS AND RESULTS: AMD3100 administration failed to mobilize BM PCs in mice defective in c-kit kinase activity or in mice transplanted with BM cells that expressed a constitutively active c-kit mutant. Furthermore, BM levels of phosphorylated (phospho)-c-kit declined after AMD3100 administration and after CXCR4 deletion. In cells adhering to culture plates coated with vascular cell adhesion molecule 1, SDF-1 and stem cell factor increased phospho-c-kit levels, and AMD3100 treatment suppressed SDF-1-induced, but not SCF-induced, c-kit phosphorylation. SDF-1-induced c-kit phosphorylation also required the activation of Src nonreceptor tyrosine kinase: pretreatment of cells with a selective Src inhibitor blocked both c-kit phosphorylation and the interaction between c-kit and phospho-Src. CONCLUSIONS: These findings indicate that the regulation of BM PC trafficking by SDF-1 and CXCR4 is dependent on Src-mediated c-kit phosphorylation.


Subject(s)
Bone Marrow Cells/physiology , Cell Movement/physiology , Proto-Oncogene Proteins c-kit/metabolism , Receptors, CXCR4/physiology , Stem Cells/physiology , Animals , Benzylamines , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Cell Line , Cell Movement/genetics , Chemokine CXCL12/physiology , Cyclams , Enzyme Activation/drug effects , Enzyme Activation/genetics , Heterocyclic Compounds/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics , Stem Cells/drug effects , Stem Cells/enzymology , src-Family Kinases/physiology
17.
J Oncol ; 2010: 928628, 2010.
Article in English | MEDLINE | ID: mdl-20652008

ABSTRACT

Telomere homolog oligonucleotides (T-oligos) activate an innate telomere-based program that leads to multiple anticancer effects. T-oligos act at telomeres to initiate signaling through the Werner protein and ATM kinase. We wanted to determine if T-oligos have antiangiogenic effects. We found that T-oligo-treated human melanoma (MM-AN) cells had decreased expression of vascular endothelial growth factor (VEGF), VEGF receptor 2, angiopoeitin-1 and -2 and decreased VEGF secretion. T-oligos activated the transcription factor E2F1 and inhibited the activity of the angiogenic transcription factor, HIF-1alpha. T-oligos inhibited EC tubulogenesis and total tumor microvascular density matrix invasion by MM-AN cells and ECs in vitro. In melanoma SCID xenografts, two systemic T-oligo injections decreased by 60% (P < .004) total tumor microvascular density and the functional vessels density by 80% (P < .002). These findings suggest that restriction of tumor angiogenesis is among the host's innate telomere-based anticancer responses and provide further evidence that T-oligos may offer a powerful new approach for melanoma treatment.

18.
Proc Natl Acad Sci U S A ; 107(24): 11008-13, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20534467

ABSTRACT

We hypothesized that a small molecule CXCR4 antagonist, AMD3100 (AMD), could augment the mobilization of bone marrow (BM)-derived endothelial progenitor cells (EPCs), thereby enhancing neovascularization and functional recovery after myocardial infarction. Single-dose AMD injection administered after the onset of myocardial infarction increased circulating EPC counts and myocardial vascularity, reduced fibrosis, and improved cardiac function and survival. In mice transplanted with traceable BM cells, AMD increased BM-derived cell incorporation in the ischemic border zone. In contrast, continuous infusion of AMD, although increasing EPCs in the circulation, worsened outcome by blocking EPC incorporation. In addition to its effects as a CXCR4 antagonist, AMD also up-regulated VEGF and matrix metalloproteinase 9 (MMP-9) expression, and the benefits of AMD were not observed in the absence of MMP-9 expression in the BM. These findings suggest that AMD3100 preserves cardiac function after myocardial infarction by enhancing BM-EPC-mediated neovascularization, and that these benefits require MMP-9 expression in the BM, but not in the ischemic region. Our results indicate that AMD3100 could be a potentially useful therapy for the treatment of myocardial infarction.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Receptors, CXCR4/antagonists & inhibitors , Animals , Base Sequence , Benzylamines , Blood Cell Count , Capillaries/drug effects , Capillaries/pathology , Cyclams , DNA Primers/genetics , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Heterocyclic Compounds/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/metabolism
19.
J Mol Cell Cardiol ; 49(3): 490-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20478312

ABSTRACT

The morphogen Sonic hedgehog (Shh) promotes neovascularization in adults by inducing pro-angiogenic cytokine expression in fibroblasts; however, the direct effects of Shh on endothelial cell (EC) function during angiogenesis are unknown. Our findings indicate that Shh promotes capillary morphogenesis (tube length on Matrigel increased to 271+/-50% of the length in untreated cells, p=0.00003), induces EC migration (modified Boyden chamber assay, 191+/-35% of migration in untreated cells, p=0.00009), and increases EC expression of matrix metalloproteinase 9 (MMP-9) and osteopontin (OPN) mRNA (real-time RT-PCR), which are essential for Shh-induced angiogenesis both in vitro and in vivo. Shh activity in ECs is mediated by Rho, rather than through the "classic" Shh signaling pathway, which involves the Gli transcription factors. The Rho dependence of Shh-induced EC angiogenic activity was documented both in vitro, with dominant-negative RhoA and Rho kinase (ROCK) constructs, and in vivo, with the ROCK inhibitor Y27632 in the mouse corneal angiogenesis model. Finally, experiments performed in MMP-9- and OPN-knockout mice confirmed the roles of the ROCK downstream targets MMP-9 and OPN in Shh-induced angiogenesis. Collectively, our results identify a "nonclassical" pathway by which Shh directly modulates EC phenotype and angiogenic activity.


Subject(s)
Aorta/metabolism , Corneal Neovascularization/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Neovascularization, Physiologic/physiology , rho-Associated Kinases/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Apoptosis , Blotting, Western , Cattle , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Corneal Neovascularization/pathology , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Hedgehog Proteins/genetics , Humans , Kruppel-Like Transcription Factors/physiology , Matrix Metalloproteinase 9/physiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Morphogenesis , Nerve Tissue Proteins/physiology , Osteopontin/physiology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zinc Finger Protein Gli3 , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
20.
FASEB J ; 24(7): 2484-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20219984

ABSTRACT

Prolonged inflammatory response is associated with left ventricular (LV) dysfunction and adverse remodeling following myocardial infarction (MI). IL-10 inhibits inflammation by suppressing HuR-mediated mRNA stabilization of proinflammatory cytokines. Here we report that following MI, IL-10(-/-) mice showed exaggerated LV dysfunction, fibrosis, and cardiomyocyte apoptosis. Short-hairpin RNA (shRNA)-mediated knockdown of HuR in the myocardium significantly reversed MI-induced LV dysfunctions and LV remodeling. HuR knockdown significantly reduced MI-induced cardiomyocyte apoptosis concomitant with reduced p53 expression. Moreover, HuR knockdown significantly reduced infarct size and fibrosis area, which in turn was associated with decreased TGF-beta expression. In vitro, stable knockdown of HuR in mouse macrophage cell line RAW 264.7 corroborated in vivo data and revealed reduced mRNA expression of TNF-alpha, TGF-beta, and p53 following LPS challenge, which was associated with a marked reduction in the mRNA stability of these genes. Taken together, our studies suggest that HuR is a direct target of IL-10, and HuR knockdown mimics anti-inflammatory effects of IL-10.


Subject(s)
Antigens, Surface/genetics , Inflammation/prevention & control , Interleukin-10/deficiency , Myocardial Infarction/pathology , Myocardium/pathology , RNA-Binding Proteins/genetics , Ventricular Dysfunction, Left/prevention & control , Animals , Antigens, Surface/physiology , Cell Line , ELAV Proteins , ELAV-Like Protein 1 , Inflammation/etiology , Interleukin-10/physiology , Macrophages , Mice , Mice, Knockout , Myocardium/metabolism , RNA Stability , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...