Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Lab Invest ; 98(5): 682-691, 2018 05.
Article in English | MEDLINE | ID: mdl-29453401

ABSTRACT

Sonic Hedgehog (Shh) signaling induces neovascularization and angiogenesis. It is not known whether the hedgehog signaling pathway in endothelial cells is essential to angiogenesis. Smoothened (Smo) transduces hedgehog signaling across the cell membrane. This study assessed whether endothelial Smoothened-dependent Shh signaling is required for Shh-mediated angiogenesis and ischemic tissue repair. Endothelial-specific smoothened knockout mice, eSmoNull were created using Cre-lox recombination system. eSmoNull mice had no observable phenotype at baseline and showed normal cardiac function. Smoothened in CD31+ cells isolated from eSmoNull hearts was significantly reduced compared to CD31+ cells from eSmoWT littermate control hearts. Fluorescence immunostaining of eSmoNull heart sections showed Smo expression in endothelial cells was abolished. The hind-limb ischemia (HLI) model was used to assess the response to ischemic injury. Perfusion ratio, limb motor function, and limb necrosis were not significantly different after HLI between eSmoNull mice and eSmoWT. Capillary densities in the ischemic limb in eSmoNull mice were also similar to eSmoWT at 4 weeks after HLI. Next, response to exogenous Shh was assessed in the corneal angiogenesis model. There was no significant difference in corneal angiogenesis induced by administration of Shh pellets between eSmoWT and eSmoNull mice. Furthermore, in vitro experiments demonstrated that direct Shh had limited effects on endothelial cell proliferation and migration. However, conditioned media from Shh-treated fibroblasts had a more potent effect on endothelial cell proliferation and migration than non-treated conditioned media. Furthermore, Shh treatment of fibroblasts dramatically stimulated angiogenic growth factor expression, including PDGF-B, VEGF-A, HGF and IGF. PDGF-B was the most upregulated and may contribute to the large neo-vessels associated with Shh-induced angiogenesis. Taken together, these data demonstrate that Shh signaling via Smoothened in endothelial cells is not required for angiogenesis and ischemic tissue repair. Shh signaling via stromal cells likely mediates its angiogenic effects.


Subject(s)
Endothelial Cells/physiology , Hedgehog Proteins/physiology , Ischemia/physiopathology , Neovascularization, Physiologic , Signal Transduction/physiology , Smoothened Receptor/physiology , Animals , Cells, Cultured , Fibroblasts/physiology , Hindlimb/blood supply , Male , Mice
2.
Cell Rep ; 21(6): 1471-1480, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117554

ABSTRACT

Aging drives the occurrence of numerous diseases, including cardiovascular disease (CVD). Recent studies indicate that blood from young mice reduces age-associated pathologies. However, the "anti-aging" factors in juvenile circulation remain poorly identified. Here, we characterize the role of the apelinergic axis in mammalian aging and identify apelin as an anti-aging factor. The expression of apelin (apln) and its receptor (aplnr) exhibits an age-dependent decline in multiple organs. Reduced apln signaling perturbs organismal homeostasis; mice harboring genetic deficiency of aplnr or apln exhibit enhanced cardiovascular, renal, and reproductive aging. Genetic or pharmacological abrogation of apln signaling also induces cellular senescence mediated, in part, by the activation of senescence-promoting transcription factors. Conversely, restoration of apln in 15-month-old wild-type mice reduces cardiac hypertrophy and exercise-induced hypertensive response. Additionally, apln-restored mice exhibit enhanced vigor and rejuvenated behavioral and circadian phenotypes. Hence, a declining apelinergic axis promotes aging, whereas its restoration extends the murine healthspan.


Subject(s)
Aging/genetics , Apelin Receptors/genetics , Apelin/genetics , Down-Regulation , Animals , Apelin/deficiency , Apelin/metabolism , Apelin Receptors/deficiency , Apelin Receptors/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Line , Coronary Vessels/cytology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Hypertension/etiology , Hypertension/metabolism , Lentivirus/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
3.
J Am Coll Cardiol ; 66(20): 2214-2226, 2015 11 17.
Article in English | MEDLINE | ID: mdl-26564600

ABSTRACT

BACKGROUND: MicroRNA (miR) dysregulation in the myocardium has been implicated in cardiac remodeling after injury or stress. OBJECTIVES: The aim of this study was to explore the role of miR in human CD34(+) cell (hCD34(+)) dysfunction in vivo after transplantation into the myocardium under ischemia-reperfusion (I-R) conditions. METHODS: In response to inflammatory stimuli, the miR array profile of endothelial progenitor cells was analyzed using a polymerase chain reaction-based miR microarray. miR-377 expression was assessed in myocardial tissue from human patients with heart failure (HF). We investigated the effect of miR-377 inhibition on an hCD34(+) cell angiogenic proteome profile in vitro and on cardiac repair and function after I-R injury in immunodeficient mice. RESULTS: The miR array data from endothelial progenitor cells in response to inflammatory stimuli indicated changes in numerous miR, with a robust decrease in the levels of miR-377. Human cardiac biopsies from patients with HF showed significant increases in miR-377 expression compared with nonfailing control hearts. The proteome profile of hCD34(+) cells transfected with miR-377 mimics showed significant decrease in the levels of proangiogenic proteins versus nonspecific control-transfected cells. We also validated that serine/threonine kinase 35 is a target of miR-377 using a dual luciferase reporter assay. In a mouse model of myocardial I-R, intramyocardial transplantation of miR-377 silenced hCD34(+) cells in immunodeficient mice, promoting neovascularization (at 28 days, post-I-R) and lower interstitial fibrosis, leading to improved left ventricular function. CONCLUSIONS: These findings indicate that HF increased miR-377 expression in the myocardium, which is detrimental to stem cell function, and transplantation of miR-377 knockdown hCD34(+) cells into ischemic myocardium promoted their angiogenic ability, attenuating left ventricular remodeling and cardiac fibrosis.


Subject(s)
Endothelial Progenitor Cells/metabolism , Heart Failure/metabolism , MicroRNAs/metabolism , Myocardium/metabolism , Reperfusion Injury/metabolism , Adult , Animals , Antigens, CD34 , Female , Heart , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , Myocardium/pathology , Neovascularization, Physiologic/physiology , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction
4.
Stem Cells ; 33(12): 3519-29, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26235810

ABSTRACT

Poor survival and function of transplanted cells in ischemic and inflamed myocardium likely compromises the functional benefit of stem cell-based therapies. We have earlier reported that co-administration of interleukin (IL)-10 and BMPAC enhances cell survival and improves left ventricular (LV) functions after acute myocardial infarction (MI) in mice. We hypothesized that IL-10 regulates microRNA-375 (miR-375) signaling in BMPACs to enhance their survival and function in ischemic myocardium after MI and attenuates left ventricular dysfunction after MI. miR-375 expression is significantly upregulated in BMPACs upon exposure to inflammatory/hypoxic stimulus and also after MI. IL-10 knockout mice display significantly elevated miR-375 levels. We report that ex vivo miR-375 knockdown in BMPAC before transplantation in the ischemic myocardium after MI significantly improve the survival and retention of transplanted BMPACs and also BMPAC-mediated post-infarct repair, neovascularization, and LV functions. Our in vitro studies revealed that knockdown of miR-375-enhanced BMPAC proliferation and tube formation and inhibited apoptosis; over expression of miR-375 in BMPAC had opposite effects. Mechanistically, miR-375 negatively regulated 3-phosphoinositide-dependent protein kinase-1 (PDK-1) expression and PDK-1-mediated activation of PI3kinase/AKT signaling. Interestingly, BMPAC isolated from IL-10-deficient mice showed elevated basal levels of miR-375 and exhibited functional deficiencies, which were partly rescued by miR-375 knockdown, enhancing BMPAC function in vitro and in vivo. Taken together, our studies suggest that miR-375 is negatively associated with BMPAC function and survival and IL-10-mediated repression of miR-375 enhances BMPAC survival and function.


Subject(s)
Bone Marrow Cells/metabolism , Interleukin-10/metabolism , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Bone Marrow Cells/pathology , Gene Knockdown Techniques , Interleukin-10/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocardium/pathology , Stem Cells/pathology
5.
Vasc Cell ; 7: 6, 2015.
Article in English | MEDLINE | ID: mdl-26161255

ABSTRACT

BACKGROUND: Multiple studies demonstrated pro-angiogenic effects of microRNA (miR)-27b. Its targets include Notch ligand Dll4, Sprouty (Spry)-2, PPARγ and Semaphorin (SEMA) 6A. miR-27 effects in the heart are context-dependent: although it is necessary for ventricular maturation, targeted overexpression in cardiomyocytes causes hypertrophy and dysfunction during development. Despite significant recent advances, therapeutic potential of miR-27b in cardiovascular disease and its effects in adult heart remain unexplored. Here, we assessed the therapeutic potential of miR-27b mimics and inhibitors in rodent models of ischemic disease and cancer. METHODS: We have used a number of models to demonstrate the effects of miR-27b mimicry and inhibition in vivo, including subcutaneous Matrigel plug assay, mouse models of hind limb ischemia and myocardial infarction and subcutaneous Lewis Lung carcinoma. RESULTS: Using mouse model of myocardial infarction due to the coronary artery ligation, we showed that miR-27b mimic had overall beneficial effects, including increased vascularization, decreased fibrosis and increased ejection fraction. In mouse model of critical limb ischemia, miR-27b mimic also improved tissue re-vascularization and perfusion. In both models, miR-27b mimic clearly decreased macrophage recruitment to the site of hypoxic injury. In contrast, miR-27b increased the recruitment of bone marrow derived cells to the neovasculature, as was shown using mice reconstituted with fluorescence-tagged bone marrow. These effects were due, at least in part, to the decreased expression of Dll4, PPARγ and IL10. In contrast, blocking miR-27b significantly decreased vascularization and reduced growth of subcutaneous tumors and decreased BMDCs recruitment to the tumor vasculature. CONCLUSIONS: Our study demonstrates the utility of manipulating miR-27b levels in the treatment of cardiovascular disease and cancer.

6.
Exp Dermatol ; 24(10): 773-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26010430

ABSTRACT

Delayed wound healing is one of the major complications in diabetes and is characterized by chronic proinflammatory response, and abnormalities in angiogenesis and collagen deposition. Sirtuin family proteins regulate numerous pathophysiological processes, including those involved in promotion of longevity, DNA repair, glycolysis and inflammation. However, the role of sirtuin 6 (SIRT6), a NAD+-dependent nuclear deacetylase, in wound healing specifically under diabetic condition remains unclear. To analyse the role of SIRT6 in cutaneous wound healing, paired 6-mm stented wound was created in diabetic db/db mice and injected siRNA against SIRT6 in the wound margins (transfection agent alone and nonsense siRNA served as controls). Wound time to closure was assessed by digital planimetry, and wounds were harvested for histology, immunohistochemistry and Western blotting. SIRT6-siRNA-treated diabetic wound showed impaired healing, which was associated with reduced capillary density (CD31-staining vessels) when compared to control treatment. Interestingly, SIRT6 deficiency decreased vascular endothelial growth factor expression and proliferation markers in the wounds. Furthermore, SIRT6 ablation in diabetic wound promotes nuclear factor-κB (NF-κB) activation resulting in increased expression of proinflammatory markers (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, tumor necrosis factor-α and interleukin-1ß) and increased oxidative stress. Collectively, our findings demonstrate that loss of SIRT6 in cutaneous wound aggravates proinflammatory response by increasing NF-κB activation, oxidative stress and decrease in angiogenesis in the diabetic mice. Based on these findings, we speculate that the activation of SIRT6 signalling might be a potential therapeutic approach for promoting wound healing in diabetics.


Subject(s)
Diabetes Complications/physiopathology , Re-Epithelialization/genetics , Sirtuins/deficiency , Sirtuins/genetics , Skin/metabolism , Animals , Cell Proliferation/genetics , Gene Knockdown Techniques , Granulation Tissue/physiopathology , Intercellular Adhesion Molecule-1/analysis , Interleukin-1beta/metabolism , Male , Mice , NF-kappa B/metabolism , Neovascularization, Physiologic/genetics , Oxidative Stress/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Sirtuins/metabolism , Skin/chemistry , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/analysis
7.
Circ Res ; 117(1): 52-64, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25904597

ABSTRACT

RATIONALE: Embryonic stem cells (ESCs) hold great promise for cardiac regeneration but are susceptible to various concerns. Recently, salutary effects of stem cells have been connected to exosome secretion. ESCs have the ability to produce exosomes, however, their effect in the context of the heart is unknown. OBJECTIVE: Determine the effect of ESC-derived exosome for the repair of ischemic myocardium and whether c-kit(+) cardiac progenitor cells (CPCs) function can be enhanced with ESC exosomes. METHODS AND RESULTS: This study demonstrates that mouse ESC-derived exosomes (mES Ex) possess ability to augment function in infarcted hearts. mES Ex enhanced neovascularization, cardiomyocyte survival, and reduced fibrosis post infarction consistent with resurgence of cardiac proliferative response. Importantly, mES Ex augmented CPC survival, proliferation, and cardiac commitment concurrent with increased c-kit(+) CPCs in vivo 8 weeks after in vivo transfer along with formation of bonafide new cardiomyocytes in the ischemic heart. miRNA array revealed significant enrichment of miR290-295 cluster and particularly miR-294 in ESC exosomes. The underlying basis for the beneficial effect of mES Ex was tied to delivery of ESC specific miR-294 to CPCs promoting increased survival, cell cycle progression, and proliferation. CONCLUSIONS: mES Ex provide a novel cell-free system that uses the immense regenerative power of ES cells while avoiding the risks associated with direct ES or ES-derived cell transplantation and risk of teratomas. ESC exosomes possess cardiac regeneration ability and modulate both cardiomyocyte and CPC-based repair programs in the heart.


Subject(s)
Embryonic Stem Cells/physiology , Exosomes/physiology , Myocardial Infarction/therapy , Animals , Cell Survival , Cell-Free System , Collagen , Drug Combinations , Embryonic Stem Cells/ultrastructure , Fibroblasts/physiology , Fibroblasts/ultrastructure , Fibrosis , Gene Expression Regulation, Developmental , Heart Ventricles , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/ultrastructure , Injections , Laminin , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Morphogenesis , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Oxygen Consumption , Proteoglycans , Rats , Rats, Sprague-Dawley , Transfection , Ultrasonography
8.
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
9.
J Biol Chem ; 289(4): 2099-111, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24297175

ABSTRACT

The Kv7 family (Kv7.1-7.5) of voltage-activated potassium channels contributes to the maintenance of resting membrane potential in excitable cells. Previously, we provided pharmacological and electrophysiological evidence that Kv7.4 and Kv7.5 form predominantly heteromeric channels and that Kv7 activity is regulated by protein kinase C (PKC) in response to vasoconstrictors in vascular smooth muscle cells. Direct evidence for Kv7.4/7.5 heteromer formation, however, is lacking. Furthermore, it remains to be determined whether both subunits are regulated by PKC. Utilizing proximity ligation assays to visualize single molecule interactions, we now show that Kv7.4/Kv.7.5 heteromers are endogenously expressed in vascular smooth muscle cells. Introduction of dominant-negative Kv7.4 and Kv7.5 subunits in mesenteric artery myocytes reduced endogenous Kv7 currents by 84 and 76%, respectively. Expression of an inducible protein kinase Cα (PKCα) translocation system revealed that PKCα activation is sufficient to suppress endogenous Kv7 currents in A7r5 rat aortic and mesenteric artery smooth muscle cells. Arginine vasopressin (100 and 500 pm) and the PKC activator phorbol 12-myristate 13-acetate (1 nm) each inhibited human (h) Kv7.5 and hKv7.4/7.5, but not hKv7.4 channels expressed in A7r5 cells. A decrease in hKv7.5 and hKv7.4/7.5 current densities was associated with an increase in PKC-dependent phosphorylation of the channel proteins. These findings provide further evidence for a differential regulation of Kv7.4 and Kv7.5 channel subunits by PKC-dependent phosphorylation and new mechanistic insights into the role of heteromeric subunit assembly for regulation of vascular Kv7 channels.


Subject(s)
KCNQ Potassium Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Kinase C-alpha/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Arginine Vasopressin/pharmacology , Carcinogens/pharmacology , Cell Line , Humans , KCNQ Potassium Channels/genetics , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Mutation, Missense , Myocytes, Smooth Muscle/cytology , Protein Kinase C-alpha/genetics , Rats , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate/pharmacology , Vasoconstrictor Agents/pharmacology
10.
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
11.
PLoS One ; 8(4): e60161, 2013.
Article in English | MEDLINE | ID: mdl-23560074

ABSTRACT

Diabetes is associated with a higher incidence of myocardial infarction (MI) and increased risk for adverse vascular and fibrogenic events post-MI. Bone marrow-derived progenitor cell (BMPC) therapy has been shown to promote neovascularization, decrease infarct area and attenuate left ventricular (LV) dysfunction after MI. Unlike vascular effects, the anti-fibrosis mechanisms of BMPC, specifically under diabetic conditions, are poorly understood. We demonstrated that intramyocardial delivery of BMPCs in infarcted diabetic db/db mice significantly down-regulates profibrotic miRNA-155 in the myocardium and improves LV remodeling and function. Furthermore, inhibition of paracrine factor hepatocyte growth factor (HGF) signaling in vivo suppressed the BMPC-mediated inhibition of miR-155 expression and the associated protective effect on cardiac fibrosis and function. In vitro studies confirmed that the conditioned media of BMPC inhibited miR-155 expression and profibrotic signaling in mouse cardiac fibroblasts under diabetic conditions. However, neutralizing antibodies directed against HGF blocked these effects. Furthermore, miR-155 over-expression in mouse cardiac fibroblasts inhibited antifibrotic Sloan-Kettering Institute proto-oncogene (Ski) and Ski-related novel gene, non-Alu-containing (SnoN) signaling and abrogated antifibrogenic response of HGF. Together, our data demonstrates that paracrine regulation of cardiac miRNAs by transplanted BMPCs contributes to the antifibrotic effects of BMPC therapy. BMPCs release HGF, which inhibits miR-155-mediated profibrosis signaling, thereby preventing cardiac fibrosis. These data suggest that targeting miR-155 might serve as a potential therapy against cardiac fibrosis in the diabetic heart.


Subject(s)
Bone Marrow Transplantation , Diabetes Mellitus/therapy , Hematopoietic Stem Cell Transplantation , MicroRNAs/antagonists & inhibitors , Myocardial Infarction/therapy , Ventricular Dysfunction, Left/therapy , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics
12.
PLoS One ; 7(9): e45046, 2012.
Article in English | MEDLINE | ID: mdl-22984607

ABSTRACT

Embryonic Stem Cells not only hold a lot of potential for use in regenerative medicine, but also provide an elegant and efficient way to study specific developmental processes and pathways in mammals when whole animal gene knock out experiments fail. We have investigated a pathway through which HDAC1 affects cardiovascular and more specifically cardiomyocyte differentiation in ES cells by controlling expression of SOX17 and BMP2 during early differentiation. This data explains current discrepancies in the role of HDAC1 in cardiovascular differentiation and sheds light into a new pathway through which ES cells determine cardiovascular cell fate.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Cell Differentiation/genetics , HMGB Proteins/genetics , Histone Deacetylase 1/genetics , Myocytes, Cardiac/metabolism , SOXF Transcription Factors/genetics , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Line , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , HMGB Proteins/metabolism , Histone Deacetylase 1/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Myocytes, Cardiac/cytology , Reverse Transcriptase Polymerase Chain Reaction , SOXF Transcription Factors/metabolism , Signal Transduction/genetics , Time Factors
13.
Circulation ; 126(4): 418-29, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22705886

ABSTRACT

BACKGROUND: Inflammation plays a critical role in adverse cardiac remodeling and heart failure. Therefore, approaches geared toward inhibiting inflammation may provide therapeutic benefits. We tested the hypotheses that genetic deletion of interleukin-10 (IL-10), a potent antiinflammatory cytokine, exacerbates pressure overload-induced adverse cardiac remodeling and hypertrophy and that IL-10 therapy inhibits this pathology. METHODS AND RESULTS: Cardiac hypertrophy was induced in wild-type and IL-10 knockout mice by isoproterenol (ISO) infusion. ISO-induced left ventricular dysfunction and hypertrophic remodeling, including fibrosis and fetal gene expression, were further exaggerated in knockout mice compared with wild-type mice. Systemic recombinant mouse IL-10 administration markedly improved left ventricular function and not only inhibited but also reversed ISO-induced cardiac remodeling. Intriguingly, a very similar cardioprotective response of IL-10 was found in transverse aortic constriction-induced hypertrophy and heart failure models. In neonatal rat ventricular myocytes and H9c2 myoblasts, ISO activated nuclear factor-κB and inhibited signal transducers and activators of transcription 3 (STAT3) phosphorylation. Interestingly, IL-10 suppressed ISO-induced nuclear factor-κB activation and attenuated STAT3 inhibition. Moreover, pharmacological and genetic inhibition of STAT3 reversed the protective effects of IL-10, whereas ectopic expression of constitutively active STAT3 mimicked the IL-10 responses on the ISO effects, confirming that the IL-10-mediated inhibition of nuclear factor-κB is STAT3 dependent. CONCLUSION: Taken together, our results suggest IL-10 treatment as a potential therapeutic approach to limit the progression of pressure overload-induced adverse cardiac remodeling.


Subject(s)
Cardiomegaly/drug therapy , Interleukin-10/pharmacology , Interleukin-10/therapeutic use , NF-kappa B/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Ventricular Dysfunction, Left/drug therapy , Ventricular Remodeling/drug effects , Animals , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Disease Models, Animal , Disease Susceptibility , Fibrosis , Interleukin-10/genetics , Isoproterenol/adverse effects , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/physiology
14.
Circ Res ; 111(2): 180-90, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22589372

ABSTRACT

RATIONALE: Although bone marrow endothelial progenitor cell (EPC)-based therapies improve the symptoms in patients with ischemic heart disease, their limited plasticity and decreased function in patients with existing heart disease limit the full benefit of EPC therapy for cardiac regenerative medicine. OBJECTIVE: We hypothesized that reprogramming mouse or human EPCs, or both, using small molecules targeting key epigenetic repressive marks would lead to a global increase in active gene transcription, induce their cardiomyogenic potential, and enhance their inherent angiogenic potential. METHOD AND RESULTS: Mouse Lin-Sca1(+)CD31(+) EPCs and human CD34(+) cells were treated with inhibitors of DNA methyltransferases (5-Azacytidine), histone deacetylases (valproic acid), and G9a histone dimethyltransferase. A 48-hour treatment led to global increase in active transcriptome, including the reactivation of pluripotency-associated and cardiomyocyte-specific mRNA expression, whereas endothelial cell-specific genes were significantly upregulated. When cultured under appropriate differentiation conditions, reprogrammed EPCs showed efficient differentiation into cardiomyocytes. Treatment with epigenetic-modifying agents show marked increase in histone acetylation on cardiomyocyte and pluripotent cell-specific gene promoters. Intramyocardial transplantation of reprogrammed mouse and human EPCs in an acute myocardial infarction mouse model showed significant improvement in ventricular functions, which was histologically supported by their de novo cardiomyocyte differentiation and increased capillary density and reduced fibrosis. Importantly, cell transplantation was safe and did not form teratomas. CONCLUSIONS: Taken together, our results suggest that epigenetically reprogrammed EPCs display a safe, more plastic phenotype and improve postinfarct cardiac repair by both neocardiomyogenesis and neovascularization.


Subject(s)
Cell Differentiation/genetics , Endothelial Cells/physiology , Epigenesis, Genetic/genetics , Myocardial Ischemia/genetics , Myocytes, Cardiac/physiology , Stem Cell Transplantation/methods , Up-Regulation/genetics , Animals , Cells, Cultured , Endothelial Cells/pathology , Endothelial Cells/transplantation , Endothelium, Vascular/pathology , Endothelium, Vascular/physiology , Endothelium, Vascular/transplantation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Ischemia/pathology , Myocardial Ischemia/surgery , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/genetics , Stem Cells/pathology , Stem Cells/physiology , Treatment Outcome
15.
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
16.
Tex Heart Inst J ; 38(5): 474-85, 2011.
Article in English | MEDLINE | ID: mdl-22163120

ABSTRACT

Bone marrow-derived CD34(+) cells are a well-characterized population of stem cells that have traditionally been used clinically to reconstitute the hematopoietic system after radiation or chemotherapy. More recently, CD34(+) cells have also been shown to induce therapeutic angiogenesis in animal models of myocardial, peripheral, and cerebral ischemia. The mechanism by which CD34(+) cells promote therapeutic angiogenesis is not completely understood, although evidence supports both direct incorporation of the cells into the expanding vasculature and paracrine secretion of angiogenic growth factors that support the developing microvasculature. Phase I and phase II clinical trials have explored the usefulness of CD34(+) cells in the treatment of ischemic conditions in human patients. As the population of patients diagnosed with some form of ischemic cardiovascular disease expands, the need for more effective treatments also grows, especially in patients who are refractory to standard pharmacologic or revascularization treatment. As phase III trials begin, CD34(+) cells will be definitively tested as a novel treatment for myocardial and peripheral ischemia. This review will discuss what is known about the CD34 antigen and the cells that harbor it, the preclinical evidence supporting the therapeutic potential of CD34(+) cells in ischemic models, and, last, the current evidence for the clinical usefulness of CD34(+) cells in the treatment of human ischemic disease.


Subject(s)
Angiogenic Proteins/metabolism , Antigens, CD34/metabolism , Cardiovascular Diseases/surgery , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Clinical Trials as Topic , Evidence-Based Medicine , Humans , Neovascularization, Physiologic , Regeneration , Treatment Outcome
17.
Mol Pharmacol ; 79(1): 10-23, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20876743

ABSTRACT

KCNQ4 and KCNQ5 potassium channel subunits are expressed in vascular smooth muscle cells, although it remains uncertain how these subunits assemble to form functional channels. Using patch-clamp techniques, we compared the electrophysiological characteristics and effects of diclofenac, a known KCNQ channel activator, on human KCNQ4 and KCNQ5 channels expressed individually or together in A7r5 rat aortic smooth muscle cells. The conductance curves of the overexpressed channels were fitted by a single Boltzmann function in each case (V(0.5) values: -31, -44, and -38 mV for KCNQ4, KCNQ5, and KCNQ4/5, respectively). Diclofenac (100 µM) inhibited KCNQ5 channels, reducing maximum conductance by 53%, but increased maximum conductance of KCNQ4 channels by 38%. The opposite effects of diclofenac on KCNQ4 and KCNQ5 could not be attributed to the presence of a basic residue (lysine) in the voltage-sensing domain of KCNQ5, because mutation of this residue to neutral glycine (the residue present in KCNQ4) resulted in a more effective block of the channel. Differences in deactivation rates and distinct voltage-dependent effects of diclofenac on channel activation and deactivation observed with each of the subunit combinations (KCNQ4, KCNQ5, and KCNQ4/5) were used as diagnostic tools to evaluate native KCNQ currents in vascular smooth muscle cells. A7r5 cells express only KCNQ5 channels endogenously, and their responses to diclofenac closely resembled those of the overexpressed KCNQ5 currents. In contrast, mesenteric artery myocytes, which express both KCNQ4 and KCNQ5 channels, displayed whole-cell KCNQ currents with properties and diclofenac responses characteristic of overexpressed heteromeric KCNQ4/5 channels.


Subject(s)
Diclofenac/pharmacology , KCNQ Potassium Channels/agonists , KCNQ Potassium Channels/antagonists & inhibitors , KCNQ Potassium Channels/chemistry , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Humans , KCNQ Potassium Channels/biosynthesis , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Patch-Clamp Techniques , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Rats , Rats, Sprague-Dawley
18.
Mol Cell Pharmacol ; 2(1): 15-19, 2010.
Article in English | MEDLINE | ID: mdl-20689646

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) are commonly used medications for the treatment of both acute and chronic pain. Selective cyclooxygenase-2 (COX-2) inhibitors, such as celecoxib (Celebrex(®)), rofecoxib (Vioxx(®)), and diclofenac, have been among the most widely prescribed NSAIDs because they prevent the generation of prostaglandins involved in inflammation and pain, but avoid some of the gastrointestinal complications associated with less selective COX-1/COX-2 inhibitors. In 2004, rofecoxib (Vioxx(®)) was voluntarily withdrawn from the market because of adverse cardiovascular side effects. This led to an explosion of research into the cardiovascular effects of the 'coxibs', which revealed differential cardiovascular risk profiles among the members of this drug class. The differential risk profiles may relate to the tendency of some of the drugs to elevate blood pressure (BP). An important component of BP regulation is dependent on the contractile state of vascular smooth muscle cells (VSMCs), which is controlled to a large extent by the activities of KCNQ (Kv7 family) potassium channels and L-type calcium channels. Our recently published data indicate that celecoxib, but not rofecoxib or diclofenac, at therapeutically relevant concentrations, acts as a Kv7 potassium channel activator and a calcium channel blocker, causing relaxation of VSMCs and decreasing vascular tone. These vasorelaxant ion channel effects may account for the differential cardiovascular risk profiles among the different COX-2 inhibitors. We further speculate that these properties may be exploited for therapeutic benefit in the treatment of cardiovascular diseases or other medical conditions.

19.
Mol Pharmacol ; 76(5): 1053-61, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19605525

ABSTRACT

Celecoxib, rofecoxib, and diclofenac are clinically used cyclooxygenase-2 (COX-2) inhibitors, which have been under intense scrutiny because long-term rofecoxib (Vioxx; Merck, Whitehouse Station, NJ) treatment was found to increase the risk of adverse cardiovascular events. A differential risk profile for these drugs has emerged, but the underlying mechanisms have not been fully elucidated. We investigated the effects of celecoxib, rofecoxib, and diclofenac on ionic currents and calcium signaling in vascular smooth muscle cells (VSMCs) using patch-clamp techniques and fura-2 fluorescence and on arterial constriction using pressure myography. Celecoxib, but not rofecoxib or diclofenac, dramatically enhanced KCNQ (K(v)7) potassium currents and suppressed L-type voltage-sensitive calcium currents in A7r5 rat aortic smooth muscle cells (native KCNQ currents or overexpressed human KCNQ5 currents) and freshly isolated rat mesenteric artery myocytes. The effects of celecoxib were concentration-dependent within the therapeutic concentration range, and were reversed on washout. Celecoxib, but not rofecoxib, also inhibited calcium responses to vasopressin in A7r5 cells and dilated intact or endothelium-denuded rat mesenteric arteries. A celecoxib analog, 2,5-dimethyl-celecoxib, which does not inhibit COX-2, mimicked celecoxib in its enhancement of vascular KCNQ5 currents, suppression of L-type calcium currents, and vasodilation. We conclude that celecoxib inhibits calcium responses in VSMCs by enhancing KCNQ5 currents and suppressing L-type calcium currents, which ultimately reduces vascular tone. These effects are independent of its COX-2 inhibitory actions and may explain the differential risk of cardiovascular events in patients taking different drugs of this class.


Subject(s)
Cardiovascular Agents/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Ion Channels/physiology , Muscle, Smooth, Vascular/drug effects , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cardiovascular Agents/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Cells, Cultured , Cyclooxygenase 2 Inhibitors/adverse effects , Humans , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/physiology , Rats , Rats, Sprague-Dawley , Risk Factors , Vascular Resistance/drug effects , Vascular Resistance/physiology
20.
Mol Pharmacol ; 74(5): 1171-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18684841

ABSTRACT

Potassium channels play an important role in electrical signaling of excitable cells such as neurons, cardiac myocytes, and vascular smooth muscle cells (VSMCs). In particular, the KCNQ (Kv7) family of voltage-activated K(+) channels functions to stabilize negative resting membrane potentials and thereby opposes electrical excitability. Of the five known members of the mammalian Kv7 family, Kv7.1 was originally recognized for its role in cardiac myocytes, where it contributes to repolarization of the cardiac action potential. Kv7.2 to Kv7.5 were first discovered in neurons, in which they play a well characterized role in neurotransmitter-stimulated action potential firing. Over the past 5 years, important new roles for Kv7 channels have been identified. Kv7 channels have been found to be expressed in VSMCs from several vascular beds where they contribute to the regulation of vascular tone. There is evidence that Kv7.5 channels in VSMCs are targeted by the hormone vasopressin to mediate its physiological vasoconstrictor actions and evidence that neuronal Kv7 channels in the baroreceptors of the aortic arch adjust the sensitivity of the mechanosensitive neurons to changes in arterial blood pressure. These newly identified physiological roles for Kv7 channels in the cardiovascular system warrant increased attention because pharmacological modulators of this family of channels are being used clinically to treat a variety of neurological disorders. This raises questions about the cardiovascular side effects associated with existing therapies, but there is also obvious potential to capitalize on the established and evolving pharmacology of these channels to develop new therapies for cardiovascular diseases.


Subject(s)
Cardiovascular System/metabolism , KCNQ1 Potassium Channel/physiology , Action Potentials/drug effects , Humans , KCNQ1 Potassium Channel/drug effects , KCNQ1 Potassium Channel/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...