Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1849(9): 1198-208, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26187004

ABSTRACT

The human DMTF1 (DMP1) transcription factor, a DNA binding protein that interacts with cyclin D, is a positive regulator of the p14ARF (ARF) tumor suppressor. Our earlier studies have shown that three differentially spliced human DMP1 mRNAs, α, ß and γ, arise from the human gene. We now show that DMP1α, ß and γ isoforms differentially regulate ARF expression and promote distinct cellular functions. In contrast to DMP1α, DMP1ß and γ did not activate the ARF promoter, whereas only ß resulted in a dose-dependent inhibition of DMP1α-induced transactivation of the ARF promoter. Ectopic expression of DMP1ß reduced endogenous ARF mRNA levels in human fibroblasts. The DMP1ß- and γ-isoforms share domains necessary for the inhibitory function of the ß-isoform. That DMP1ß may interact with DMP1α to antagonize its function was shown in DNA binding assays and in cells by the close proximity of DMP1α/ß in the nucleus. Cells stably expressing DMP1ß, as well as shRNA targeting all DMP1 isoforms, disrupted cellular growth arrest induced by serum deprivation or in PMA-derived macrophages in the presence or absence of cellular p53. DMP1 mRNA levels in acute myeloid leukemia samples, as compared to granulocytes, were reduced. Treatment of acute promyelocytic leukemia patient samples with all-trans retinoic acid promoted differentiation to granulocytes and restored DMP1 transcripts to normal granulocyte levels. Our findings imply that DMP1α- and ß-ratios are tightly regulated in hematopoietic cells and DMP1ß antagonizes DMP1α transcriptional regulation of ARF resulting in the alteration of cellular control with a gain in proliferation.


Subject(s)
Cell Proliferation/physiology , Gene Expression Regulation/physiology , Protein Isoforms/physiology , Transcription Factors/physiology , Transcription, Genetic/physiology , Tumor Suppressor Protein p14ARF/genetics , Animals , Cell Line , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Protein Isoforms/genetics , RNA Splicing , RNA, Messenger/metabolism , Transcription Factors/genetics
2.
J Biol Chem ; 290(22): 13935-47, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25882843

ABSTRACT

Human cardiac progenitor cells (hCPC) improve heart function after autologous transfer in heart failure patients. Regenerative potential of hCPCs is severely limited with age, requiring genetic modification to enhance therapeutic potential. A legacy of work from our laboratory with Pim1 kinase reveals effects on proliferation, survival, metabolism, and rejuvenation of hCPCs in vitro and in vivo. We demonstrate that subcellular targeting of Pim1 bolsters the distinct cardioprotective effects of this kinase in hCPCs to increase proliferation and survival, and antagonize cellular senescence. Adult hCPCs isolated from patients undergoing left ventricular assist device implantation were engineered to overexpress Pim1 throughout the cell (PimWT) or targeted to either mitochondrial (Mito-Pim1) or nuclear (Nuc-Pim1) compartments. Nuc-Pim1 enhances stem cell youthfulness associated with decreased senescence-associated ß-galactosidase activity, preserved telomere length, reduced expression of p16 and p53, and up-regulation of nucleostemin relative to PimWT hCPCs. Alternately, Mito-Pim1 enhances survival by increasing expression of Bcl-2 and Bcl-XL and decreasing cell death after H2O2 treatment, thereby preserving mitochondrial integrity superior to PimWT. Mito-Pim1 increases the proliferation rate by up-regulation of cell cycle modulators Cyclin D, CDK4, and phospho-Rb. Optimal stem cell traits such as proliferation, survival, and increased youthful properties of aged hCPCs are enhanced after targeted Pim1 localization to mitochondrial or nuclear compartments. Targeted Pim1 overexpression in hCPCs allows for selection of the desired phenotypic properties to overcome patient variability and improve specific stem cell characteristics.


Subject(s)
Gene Expression Regulation , Heart/physiology , Proto-Oncogene Proteins c-pim-1/metabolism , Stem Cells/metabolism , Apoptosis , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Cellular Senescence , Green Fluorescent Proteins/metabolism , Heart Failure , Heart Ventricles/metabolism , Humans , Lentivirus/metabolism , Mitochondria/metabolism , Myocardium/cytology , Myocardium/metabolism , Phenotype , Regeneration , Stem Cells/cytology , Subcellular Fractions/metabolism , beta-Galactosidase/metabolism
3.
Cell Metab ; 20(4): 626-38, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295787

ABSTRACT

Macroautophagy (hereafter autophagy) is the major pathway by which macromolecules and organelles are degraded. Autophagy is regulated by the mTOR signaling pathway-the focal point for integration of metabolic information, with mTORC1 playing a central role in balancing biosynthesis and catabolism. Of the various inputs to mTORC1, the amino acid sensing pathway is among the most potent. Based upon transcriptome analysis of neurons subjected to nutrient deprivation, we identified let-7 microRNA as capable of promoting neuronal autophagy. We found that let-7 activates autophagy by coordinately downregulating the amino acid sensing pathway to prevent mTORC1 activation. Let-7 induced autophagy in the brain to eliminate protein aggregates, establishing its physiological relevance for in vivo autophagy modulation. Moreover, peripheral delivery of let-7 anti-miR repressed autophagy in muscle and white fat, suggesting that let-7 autophagy regulation extends beyond CNS. Hence, let-7 plays a central role in nutrient homeostasis and proteostasis regulation in higher organisms.


Subject(s)
Amino Acids/metabolism , Autophagy , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipose Tissue, White/metabolism , Animals , Base Sequence , Brain/metabolism , Cells, Cultured , HEK293 Cells , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Sequence Alignment , Signal Transduction
4.
Hum Mol Genet ; 22(20): 4127-35, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23740943

ABSTRACT

microRNAs (miRNAs) are dysregulated in a variety of disease states, suggesting that this newly discovered class of gene expression repressors may be viable therapeutic targets. A microarray of miRNA changes in ALS-model superoxide dismutase 1 (SOD1)(G93A) rodents identified 12 miRNAs as significantly changed. Six miRNAs tested in human ALS tissues were confirmed increased. Specifically, miR-155 was increased 5-fold in mice and 2-fold in human spinal cords. To test miRNA inhibition in the central nervous system (CNS) as a potential novel therapeutic, we developed oligonucleotide-based miRNA inhibitors (anti-miRs) that could inhibit miRNAs throughout the CNS and in the periphery. Anti-miR-155 caused global derepression of targets in peritoneal macrophages and, following intraventricular delivery, demonstrated widespread functional distribution in the brain and spinal cord. After treating SOD1(G93A) mice with anti-miR-155, we significantly extended survival by 10 days and disease duration by 15 days (38%) while a scrambled control anti-miR did not significantly improve survival or disease duration. Therefore, antisense oligonucleotides may be used to successfully inhibit miRNAs throughout the brain and spinal cord, and miR-155 is a promising new therapeutic target for human ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , MicroRNAs/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Adult , Aged , Amyotrophic Lateral Sclerosis/metabolism , Animals , Brain/drug effects , Brain/metabolism , Case-Control Studies , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Middle Aged , Molecular Targeted Therapy , Oligonucleotides, Antisense/metabolism , Rats , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxide Dismutase/genetics
5.
Genes Dev ; 27(12): 1365-77, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23788623

ABSTRACT

Myeloid cell leukemia-1 (MCL-1) is an anti-apoptotic BCL-2 protein that is up-regulated in several human cancers. MCL-1 is also highly expressed in myocardium, but its function in myocytes has not been investigated. We generated inducible, cardiomyocyte-specific Mcl-1 knockout mice and found that ablation of Mcl-1 in the adult heart led to rapid cardiomyopathy and death. Although MCL-1 is known to inhibit apoptosis, this process was not activated in MCL-1-deficient hearts. Ultrastructural analysis revealed disorganized sarcomeres and swollen mitochondria in myocytes. Mitochondria isolated from MCL-1-deficient hearts exhibited reduced respiration and limited Ca(2+)-mediated swelling, consistent with opening of the mitochondrial permeability transition pore (mPTP). Double-knockout mice lacking MCL-1 and cyclophilin D, an essential regulator of the mPTP, exhibited delayed progression to heart failure and extended survival. Autophagy is normally induced by myocardial stress, but induction of autophagy was impaired in MCL-1-deficient hearts. These data demonstrate that MCL-1 is essential for mitochondrial homeostasis and induction of autophagy in the heart. This study also raises concerns about potential cardiotoxicity for chemotherapeutics that target MCL-1.


Subject(s)
Autophagy/genetics , Heart Failure/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Cardiomegaly/genetics , Cell Respiration/genetics , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis/genetics , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Analysis
6.
J Am Coll Cardiol ; 60(14): 1278-87, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22841153

ABSTRACT

OBJECTIVES: The goal of this study was to demonstrate the enhancement of human cardiac progenitor cell (hCPC) reparative and regenerative potential by genetic modification for the treatment of myocardial infarction. BACKGROUND: Regenerative potential of stem cells to repair acute infarction is limited. Improved hCPC survival, proliferation, and differentiation into functional myocardium will increase efficacy and advance translational implementation of cardiac regeneration. METHODS: hCPCs isolated from the myocardium of heart failure patients undergoing left ventricular assist device implantation were engineered to express green fluorescent protein (hCPCe) or Pim-1-GFP (hCPCeP). Functional tests of hCPC regenerative potential were performed with immunocompromised mice by using intramyocardial adoptive transfer injection after infarction. Myocardial structure and function were monitored by echocardiographic and hemodynamic assessment for 20 weeks after delivery. hCPCe and hCPCeP expressing luciferase were observed by using bioluminescence imaging to noninvasively track persistence. RESULTS: hCPCeP exhibited augmentation of reparative potential relative to hCPCe control cells, as shown by significantly increased proliferation coupled with amelioration of infarction injury and increased hemodynamic performance at 20 weeks post-transplantation. Concurrent with enhanced cardiac structure and function, hCPCeP demonstrated increased cellular engraftment and differentiation with improved vasculature and reduced infarct size. Enhanced persistence of hCPCeP versus hCPCe was revealed by bioluminescence imaging at up to 8 weeks post-delivery. CONCLUSIONS: Genetic engineering of hCPCs with Pim-1 enhanced repair of damaged myocardium. Ex vivo gene delivery to modify stem cells has emerged as a viable option addressing current limitations in the field. This study demonstrates that efficacy of hCPCs from the failing myocardium can be safely and significantly enhanced through expression of Pim-1 kinase, setting the stage for use of engineered cells in pre-clinical settings.


Subject(s)
Genetic Engineering , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Cell Proliferation , Echocardiography , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hemodynamics , Humans , Luminescent Measurements , Mice , Myocytes, Cardiac/enzymology , Neovascularization, Pathologic , Proto-Oncogene Proteins c-pim-1/genetics , Stem Cell Transplantation , Stem Cells/enzymology
7.
Circ Res ; 111(1): 77-86, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22619278

ABSTRACT

RATIONALE: Bone marrow-derived cells to treat myocardial injury improve cardiac function and support beneficial cardiac remodeling. However, survival of stem cells is limited due to low proliferation of transferred cells. OBJECTIVE: To demonstrate long-term potential of c-kit(+) bone marrow stem cells (BMCs) enhanced with Pim-1 kinase to promote positive cardiac remodeling. METHODS AND RESULTS: Lentiviral modification of c-kit(+) BMCs to express Pim-1 (BMCeP) increases proliferation and expression of prosurvival proteins relative to BMCs expressing green fluorescent protein (BMCe). Intramyocardial delivery of BMCeP at time of infarction supports improvements in anterior wall dimensions and prevents left ventricle dilation compared with hearts treated with vehicle alone. Reduction of the akinetic left ventricular wall was observed in BMCeP-treated hearts at 4 and 12 weeks after infarction. Early recovery of cardiac function in BMCeP-injected hearts facilitated modest improvements in hemodynamic function up to 12 weeks after infarction between cell-treated groups. Persistence of BMCeP is improved relative to BMCe within the infarct together with increased recruitment of endogenous c-kit(+) cells. Delivery of BMC populations promotes cellular hypertrophy in the border and infarcted regions coupled with an upregulation of hypertrophic genes. Thus, BMCeP treatment yields improved structural remodeling of infarcted myocardium compared with control BMCs. CONCLUSIONS: Genetic modification of BMCs with Pim-1 may serve as a therapeutic approach to promote recovery of myocardial structure. Future approaches may take advantage of salutary BMC actions in conjunction with other stem cell types to increase efficacy of cellular therapy and improve myocardial performance in the injured myocardium.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Myocardial Infarction/surgery , Myocardium/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Regeneration , Tissue Engineering , Animals , Apoptosis , Bone Marrow Cells/pathology , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Lentivirus/genetics , Male , Mice , Myocardial Contraction , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Recovery of Function , Signal Transduction , Time Factors , Tissue Engineering/methods , Transduction, Genetic , Ultrasonography , Ventricular Function, Left , Ventricular Remodeling
8.
Physiol Rev ; 91(3): 1023-70, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21742795

ABSTRACT

One of the greatest examples of integrated signal transduction is revealed by examination of effects mediated by AKT kinase in myocardial biology. Positioned at the intersection of multiple afferent and efferent signals, AKT exemplifies a molecular sensing node that coordinates dynamic responses of the cell in literally every aspect of biological responses. The balanced and nuanced nature of homeostatic signaling is particularly essential within the myocardial context, where regulation of survival, energy production, contractility, and response to pathological stress all flow through the nexus of AKT activation or repression. Equally important, the loss of regulated AKT activity is primarily the cause or consequence of pathological conditions leading to remodeling of the heart and eventual decompensation. This review presents an overview compendium of the complex world of myocardial AKT biology gleaned from more than a decade of research. Summarization of the widespread influence that AKT exerts upon myocardial responses leaves no doubt that the participation of AKT in molecular signaling will need to be reckoned with as a seemingly omnipresent regulator of myocardial molecular biological responses.


Subject(s)
Myocardium/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Calcium Signaling/physiology , Cardiomyopathies/physiopathology , Cell Survival/physiology , Enzyme Activation , Humans , MicroRNAs/metabolism , Mitochondria/enzymology , Myocardial Contraction/physiology , Neovascularization, Physiologic/physiology , Protein Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Sex Characteristics , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 108(15): 6145-50, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21444791

ABSTRACT

Nucleolar stress, characterized by loss of nucleolar integrity, has not been described in the cardiac context. In addition to ribosome biogenesis, nucleoli are critical for control of cell proliferation and stress responses. Our group previously demonstrated induction of the nucleolar protein nucleostemin (NS) in response to cardiac pathological insult. NS interacts with nucleophosmin (NPM), a marker of nucleolar stress with cytoprotective properties. The dynamic behavior of NS and NPM reveal that nucleolar disruption is an early event associated with stress response in cardiac cells. Rapid translocation of NS and NPM to the nucleoplasm and suppression of new preribosomal RNA synthesis occurs in both neonatal rat cardiomyocytes (NRCM) and cardiac progenitor cells (CPC) upon exposure to doxorubicin or actinomycin D. Silencing of NS significantly increases cell death resulting from doxorubicin treatment in CPC, whereas NPM knockdown alone induces cell death. Overexpression of either NS or NPM significantly decreases caspase 8 activity in cultured cardiomyocytes challenged with doxorubicin. The presence of altered nucleolar structures resulting from myocardial infarction in mice supports the model of nucleolar stress as a general response to pathological injury. Collectively, these findings serve as the initial description of myocardial nucleolar stress and establish the postulate that nucleoli acts as sensors of stress, regulating the cellular response to pathological insults.


Subject(s)
Carrier Proteins/metabolism , Cell Nucleolus/metabolism , Myocardium/metabolism , Nuclear Proteins/metabolism , Stress, Physiological , Animals , Aorta/metabolism , Aorta/pathology , Apoptosis , Cell Nucleolus/pathology , Cells, Cultured , Constriction, Pathologic/metabolism , Constriction, Pathologic/pathology , GTP-Binding Proteins , Humans , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nucleophosmin , RNA, Ribosomal/biosynthesis , RNA-Binding Proteins , Rats
10.
Circ Res ; 108(8): 960-70, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21350213

ABSTRACT

RATIONALE: Stem cell therapies to regenerate damaged cardiac tissue represent a novel approach to treat heart disease. However, the majority of adoptively transferred stem cells delivered to damaged myocardium do not survive long enough to impart protective benefits, resulting in modest functional improvements. Strategies to improve survival and proliferation of stem cells show promise for significantly enhancing cardiac function and regeneration. OBJECTIVE: To determine whether injected cardiac progenitor cells (CPCs) genetically modified to overexpress nuclear Akt (CPCeA) increase structural and functional benefits to infarcted myocardium relative to control CPCs. METHODS AND RESULTS: CPCeA exhibit significantly increased proliferation and secretion of paracrine factors compared with CPCs. However, CPCeA exhibit impaired capacity for lineage commitment in vitro. Infarcted hearts receiving intramyocardial injection of CPCeA have increased recruitment of endogenous c-kit cells compared with CPCs, but neither population provides long-term functional and structural improvements compared with saline-injected controls. Pharmacological inhibition of Akt alleviated blockade of lineage commitment in CPCeA. CONCLUSIONS: Although overexpression of nuclear Akt promotes rapid proliferation and secretion of protective paracrine factors, the inability of CPCeA to undergo lineage commitment hinders their capacity to provide functional or structural benefits to infarcted hearts. Despite enhanced recruitment of endogenous CPCs, lack of functional improvement in CPCeA-treated hearts demonstrates CPC lineage commitment is essential to the regenerative response. Effective stem cell therapies must promote cellular survival and proliferation without inhibiting lineage commitment. Because CPCeA exhibit remarkable proliferative potential, an inducible system mediating nuclear Akt expression could be useful to augment cell therapy approaches.


Subject(s)
Cell Nucleus/enzymology , Gene Expression Regulation, Enzymologic , Growth Inhibitors/physiology , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Proto-Oncogene Proteins c-akt/biosynthesis , Stem Cells/enzymology , Animals , Cell Lineage/genetics , Cell Nucleus/pathology , Cell Proliferation , Cells, Cultured , Female , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Male , Mice , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardium/cytology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , Stem Cell Transplantation/methods , Stem Cells/cytology , Stem Cells/pathology , Structure-Activity Relationship
11.
J Mol Cell Cardiol ; 51(4): 554-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21255581

ABSTRACT

Stem cells mediate tissue repair throughout the lifespan of an organism. However, the ability of stem cells to mitigate catastrophic damage, such as that sustained after major myocardial infarction is inadequate to rebuild the heart and restore functional capacity. However, capitalizing on the ability of these cells to attenuate damage in the myocardium, various maneuvers that enhance repair mechanisms to improve cardiac structure and function after injury are being investigated. These studies have led to discovery of various factors that mediate cardioprotection and enhance endogenous repair by 1) salvaging surviving myocardium, 2) promoting homing of stem cells and 3) increasing survival and proliferation of stem cell populations at the site of injury. Herein we report upon a downstream target of Akt kinase, named Pim-1, which promotes cardioprotective signaling and enhances cardiac structure and function after pathological injury. The compilation of studies presented here supports use of Pim-1 to enhance long-term myocardial repair after pathological damage. This article is part of a special issue entitled "Key Signaling Molecules in Hypertrophy and Heart Failure."


Subject(s)
Proto-Oncogene Proteins c-pim-1/physiology , Signal Transduction , Animals , Cell Proliferation , Cell Survival , Heart/physiopathology , Humans , Mice , Mice, Transgenic , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/enzymology , Myocardium/pathology , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Regeneration , Stem Cell Transplantation , Stem Cells/enzymology , Stem Cells/physiology
12.
Eur Heart J ; 32(17): 2179-88, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21228009

ABSTRACT

AIMS: The cascade of events leading to compromised mitochondrial integrity in response to stress is mediated by various combinatorial interactions of pro- and anti-apoptotic molecules. Nur77, an immediate early gene that encodes a nuclear orphan receptor, translocates from the nucleus to mitochondria to induce cytochrome c release and apoptosis in cancer cells in response to various pro-apoptotic treatments. However, the role of Nur77 in the cardiac setting is still unclear. The objective of this study is to determine the physiological relevance and pathophysiological importance of Nur77 in cardiomyocytes. METHODS AND RESULTS: Myocardial Nur77 is upregulated following cardiomyopathic injury and, while expressed in the postnatal myocardium, declines in level within weeks after birth. Nur77 is localized predominantly in cardiomyocyte nuclei under normal conditions where it is not apoptotic, but translocates to mitochondria in response to oxidative stress both in vitro and in vivo. Mitochondrial localization of Nur77 induces cytochrome c release and typical morphological features of apoptosis, including chromatin condensation and DNA fragmentation. Knockdown of Nur77 rescued hydrogen peroxide-induced cardiomyocyte apoptosis. CONCLUSION: Translocation of Nur77 from the nucleus to the mitochondria in cardiomyocytes results in the loss of mitochondrial integrity and subsequent apoptosis in response to ischaemia/reperfusion injury. Our findings identify Nur77 as a novel mediator of cardiomyocyte apoptosis and warrants further investigation of mitochondrial Nur77 translocation as a mechanism to control cell death in the treatment of ischaemic heart diseases.


Subject(s)
Apoptosis/physiology , Mitochondria, Heart/physiology , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Animals , Constriction , Female , Male , Mice , Myocardial Reperfusion Injury/pathology , Rats , Rats, Sprague-Dawley , Transfection , Up-Regulation
13.
Circ Res ; 106(7): 1265-74, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20203306

ABSTRACT

RATIONALE: Cardioprotective signaling mediates antiapoptotic actions through multiple mechanisms including maintenance of mitochondrial integrity. Pim-1 kinase is an essential downstream effector of AKT-mediated cardioprotection but the mechanistic basis for maintenance of mitochondrial integrity by Pim-1 remains unexplored. This study details antiapoptotic actions responsible for enhanced cell survival in cardiomyocytes with elevated Pim-1 activity. OBJECTIVE: The purpose of this study is to demonstrate that the cardioprotective kinase Pim-1 acts to inhibit cell death by preserving mitochondrial integrity in cardiomyocytes. METHODS AND RESULTS: A combination of biochemical, molecular, and microscopic analyses demonstrate beneficial effects of Pim-1 on mitochondrial integrity. Pim-1 protein level increases in the mitochondrial fraction with a corresponding decrease in the cytosolic fraction of myocardial lysates from hearts subjected to 30 minutes of ischemia followed by 30 minutes of reperfusion. Cardiac-specific overexpression of Pim-1 results in higher levels of antiapoptotic Bcl-X(L) and Bcl-2 compared to samples from normal hearts. In response to oxidative stress challenge, Pim-1 preserves the inner mitochondrial membrane potential. Ultrastructure of the mitochondria is maintained by Pim-1 activity, which prevents swelling induced by calcium overload. Finally, mitochondria isolated from hearts created with cardiac-specific overexpression of Pim-1 show inhibition of cytochrome c release triggered by a truncated form of proapoptotic Bid. CONCLUSION: Cardioprotective action of Pim-1 kinase includes preservation of mitochondrial integrity during cardiomyopathic challenge conditions, thereby raising the potential for Pim-1 kinase activation as a therapeutic interventional approach to inhibit cell death by antagonizing proapoptotic Bcl-2 family members that regulate the intrinsic apoptotic pathway.


Subject(s)
Apoptosis , Mitochondria, Heart/enzymology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Animals, Newborn , BH3 Interacting Domain Death Agonist Protein/metabolism , Cell Survival , Cells, Cultured , Cytochromes c/metabolism , Disease Models, Animal , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Transgenic , Mitochondria, Heart/ultrastructure , Mitochondrial Swelling , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/ultrastructure , Oxidative Stress , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , bcl-X Protein/metabolism
14.
Circ Res ; 106(5): 891-901, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20075333

ABSTRACT

RATIONALE: Cardioprotective effects of Pim-1 kinase have been previously reported but the underlying mechanistic basis may involve a combination of cellular and molecular mechanisms that remain unresolved. The elucidation of the mechanistic basis for Pim-1 mediated cardioprotection provides important insights for designing therapeutic interventional strategies to treat heart disease. OBJECTIVE: Effects of cardiac-specific Pim-1 kinase expression on the cardiac progenitor cell (CPC) population were examined to determine whether Pim-1 mediates beneficial effects through augmenting CPC activity. METHODS AND RESULTS: Transgenic mice created with cardiac-specific Pim-1 overexpression (Pim-wt) exhibit enhanced Pim-1 expression in both cardiomyocytes and CPCs, both of which show increased proliferative activity assessed using 5-bromodeoxyuridine (BrdU), Ki-67, and c-Myc relative to nontransgenic controls. However, the total number of CPCs was not increased in the Pim-wt hearts during normal postnatal growth or after infarction challenge. These results suggest that Pim-1 overexpression leads to asymmetric division resulting in maintenance of the CPC population. Localization and quantitation of cell fate determinants Numb and alpha-adaptin by confocal microscopy were used to assess frequency of asymmetric division in the CPC population. Polarization of Numb in mitotic phospho-histone positive cells demonstrates asymmetric division in 65% of the CPC population in hearts of Pim-wt mice versus 26% in nontransgenic hearts after infarction challenge. Similarly, Pim-wt hearts had fewer cells with uniform alpha-adaptin staining indicative of symmetrically dividing CPCs, with 36% of the CPCs versus 73% in nontransgenic sections. CONCLUSIONS: These findings define a mechanistic basis for enhanced myocardial regeneration in transgenic mice overexpressing Pim-1 kinase.


Subject(s)
Cell Cycle , Cell Proliferation , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Regeneration , Stem Cells/enzymology , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Histones/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Mutation , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Nerve Tissue Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-pim-1/genetics , Stem Cells/pathology , Time Factors
15.
Circulation ; 120(21): 2077-87, 2009 Nov 24.
Article in English | MEDLINE | ID: mdl-19901187

ABSTRACT

BACKGROUND: Despite numerous studies demonstrating the efficacy of cellular adoptive transfer for therapeutic myocardial regeneration, problems remain for donated cells with regard to survival, persistence, engraftment, and long-term benefits. This study redresses these concerns by enhancing the regenerative potential of adoptively transferred cardiac progenitor cells (CPCs) via genetic engineering to overexpress Pim-1, a cardioprotective kinase that enhances cell survival and proliferation. METHODS AND RESULTS: Intramyocardial injections of CPCs overexpressing Pim-1 were given to infarcted female mice. Animals were monitored over 4, 12, and 32 weeks to assess cardiac function and engraftment of Pim-1 CPCs with echocardiography, in vivo hemodynamics, and confocal imagery. CPCs overexpressing Pim-1 showed increased proliferation and expression of markers consistent with cardiogenic lineage commitment after dexamethasone exposure in vitro. Animals that received CPCs overexpressing Pim-1 also produced greater levels of cellular engraftment, persistence, and functional improvement relative to control CPCs up to 32 weeks after delivery. Salutary effects include reduction of infarct size, greater number of c-kit(+) cells, and increased vasculature in the damaged region. CONCLUSIONS: Myocardial repair is significantly enhanced by genetic engineering of CPCs with Pim-1 kinase. Ex vivo gene delivery to enhance cellular survival, proliferation, and regeneration may overcome current limitations of stem cell-based therapeutic approaches.


Subject(s)
Genetic Engineering , Genetic Therapy , Myocardial Infarction/therapy , Myocardium/cytology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Female , Humans , Male , Mice , Myocardial Infarction/physiopathology , Proto-Oncogene Proteins c-kit/analysis
16.
Regen Med ; 4(6): 823-33, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903002

ABSTRACT

AIMS: Cardiac stem cells (CSCs) show potential as a cellular therapeutic approach to blunt tissue damage and facilitate reparative and regenerative processes after myocardial infarction. Despite multiple published reports of improvement, functional benefits remain modest using normal stem cells delivered by adoptive transfer into damaged myocardium. The goal of this study is to enhance survival and proliferation of CSCs that have undergone lineage commitment in early phases as evidenced by expression of proteins driven by the alpha-myosin heavy chain (alphaMHC) promoter. The early increased expression of survival kinases augments expansion of the cardiogenic CSC pool and subsequent daughter progeny. MATERIALS & METHODS: Normal CSCs engineered with fluorescent reporter protein constructs under control of the alphaMHC promoter show transgene protein expression, confirming activity of the promoter in CSCs. Cultured CSCs from both nontransgenic and cardiac-specific transgenic mice expressing survival kinases driven by the alphaMHC promoter were analyzed to characterize transgene expression following treatments to promote differentiation in culture. RESULTS & CONCLUSION: Therapeutic genes controlled by the alphaMHC promoter can be engineered into and expressed in CSCs and cardiomyocyte progeny with the goal of improving the efficacy of cardiac stem cell therapy.


Subject(s)
Genetic Engineering , Myocardium/cytology , Myocytes, Cardiac/cytology , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/genetics , Stem Cells/physiology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoenzyme Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-pim-1/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transgenes/physiology
17.
J Mol Cell Cardiol ; 47(1): 96-103, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19269295

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/phosphoinositide dependent kinase 1 (PDK1) signaling pathway exerts cardioprotective effects in the myocardium through activation of key proteins including Akt. Activated Akt accumulates in nuclei of cardiomyocytes suggesting that biologically relevant targets are located in that subcellular compartment. Nuclear Akt activity could be potentiated in both intensity and duration by the presence of a nuclear-associated PI3K/PDK1 signaling cascade as has been described in other non-myocyte cell types. PI3K/PDK1 distribution was determined in vitro and in vivo by immunostaining and nuclear extraction of cultured rat neonatal cardiomyocytes or transgenic mouse hearts. Results show that PI3K and PDK1 are present at a basal level in cardiomyocytes nuclei and that cardioprotective stimulation with atrial natriuretic peptide (ANP) increases their nuclear localization. In comparison, overexpression of nuclear-targeted Akt does not mediate increased translocation of either PI3K or PDK1 indicating that accumulation of Akt does not drive PI3K or PDK1 into the nuclear compartment. Furthermore, PI3K and phospho-Akt(473) show parallel temporal accumulation in the nucleus following (MI) infarction challenge. These findings demonstrate the presence of a dynamically regulated nuclear-associated signaling cascade involving PI3K and PDK that presumably influences nuclear Akt activation.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Animals, Newborn , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Cyclic GMP/pharmacology , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , Rats
18.
Proc Natl Acad Sci U S A ; 105(37): 13889-94, 2008 Sep 16.
Article in English | MEDLINE | ID: mdl-18784362

ABSTRACT

Pim-1 kinase exerts potent cardioprotective effects in the myocardium downstream of AKT, but the participation of Pim-1 in cardiac hypertrophy requires investigation. Cardiac-specific expression of Pim-1 (Pim-WT) or the dominant-negative mutant of Pim-1 (Pim-DN) in transgenic mice together with adenoviral-mediated overexpression of these Pim-1 constructs was used to delineate the role of Pim-1 in hypertrophy. Transgenic overexpression of Pim-1 protects mice from pressure-overload-induced hypertrophy relative to wild-type controls as evidenced by improved hemodynamic function, decreased apoptosis, increases in antihypertrophic proteins, smaller myocyte size, and inhibition of hypertrophic signaling after challenge. Similarly, Pim-1 overexpression in neonatal rat cardiomyocyte cultures inhibits hypertrophy induced by endothelin-1. On the cellular level, hearts of Pim-WT mice show enhanced incorporation of BrdU into myocytes and a hypercellular phenotype compared to wild-type controls after hypertrophic challenge. In comparison, transgenic overexpression of Pim-DN leads to dilated cardiomyopathy characterized by increased apoptosis, fibrosis, and severely depressed cardiac function. Furthermore, overexpression of Pim-DN leads to reduced contractility as evidenced by reduced Ca(2+) transient amplitude and decreased percentage of cell shortening in isolated myocytes. These data support a pivotal role for Pim-1 in modulation of hypertrophy by impacting responses on molecular, cellular, and organ levels.


Subject(s)
Cardiomegaly/enzymology , Cardiomegaly/pathology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Animals, Genetically Modified , Aorta/enzymology , Apoptosis , Cardiomegaly/chemically induced , Cardiomegaly/physiopathology , Cells, Cultured , Endothelin-1/pharmacology , Fibrosis , Muscle Contraction , Proto-Oncogene Proteins c-pim-1/genetics , Rats
19.
Circ Res ; 102(9): 1025-35, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18369158

ABSTRACT

The Notch network regulates multiple cellular processes, including cell fate determination, development, differentiation, proliferation, apoptosis, and regeneration. These processes are regulated via Notch-mediated activity that involves hepatocyte growth factor (HGF)/c-Met receptor and phosphatidylinositol 3-kinase/Akt signaling cascades. The impact of HGF on Notch signaling was assessed following myocardial infarction as well as in cultured cardiomyocytes. Notch1 is activated in border zone cardiomyocytes coincident with nuclear c-Met following infarction. Intramyocardial injection of HGF enhances Notch1 and Akt activation in adult mouse myocardium. Corroborating evidence in cultured cardiomyocytes shows treatment with HGF or insulin increases levels of Notch effector Hes1 in immunoblots, whereas overexpression of activated Notch intracellular domain prompts a 3-fold increase in phosphorylated Akt. Infarcted hearts injected with adenoviral vector expressing Notch intracellular domain treatment exhibit improved hemodynamic function in comparison with control mice after 4 weeks, implicating Notch signaling in a cardioprotective role following cardiac injury. These results indicate Notch activation in cardiomyocytes is mediated through c-Met and Akt survival signaling pathways, and Notch1 signaling in turn enhances Akt activity. This mutually supportive crosstalk suggests a positive survival feedback mechanism between Notch and Akt signaling in adult myocardium following injury.


Subject(s)
Myocardial Infarction/metabolism , Myocardium/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Feedback, Physiological , Female , Hemodynamics , Hepatocyte Growth Factor/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Time Factors , Transcription Factor HES-1 , Transduction, Genetic
20.
Nat Med ; 13(12): 1467-75, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18037896

ABSTRACT

The serine-threonine kinases Pim-1 and Akt regulate cellular proliferation and survival. Although Akt is known to be a crucial signaling protein in the myocardium, the role of Pim-1 has been overlooked. Pim-1 expression in the myocardium of mice decreased during postnatal development, re-emerged after acute pathological injury in mice and was increased in failing hearts of both mice and humans. Cardioprotective stimuli associated with Akt activation induced Pim-1 expression, but compensatory increases in Akt abundance and phosphorylation after pathological injury by infarction or pressure overload did not protect the myocardium in Pim-1-deficient mice. Transgenic expression of Pim-1 in the myocardium protected mice from infarction injury, and Pim-1 expression inhibited cardiomyocyte apoptosis with concomitant increases in Bcl-2 and Bcl-X(L) protein levels, as well as in Bad phosphorylation levels. Relative to nontransgenic controls, calcium dynamics were significantly enhanced in Pim-1-overexpressing transgenic hearts, associated with increased expression of SERCA2a, and were depressed in Pim-1-deficient hearts. Collectively, these data suggest that Pim-1 is a crucial facet of cardioprotection downstream of Akt.


Subject(s)
Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-pim-1/physiology , Animals , Apoptosis , Cell Nucleus/metabolism , Humans , Mice , Mice, Knockout , Myocardium/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-pim-1/biosynthesis , Rats , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...