Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Basic Res Cardiol ; 110(3): 29, 2015 May.
Article in English | MEDLINE | ID: mdl-25893875

ABSTRACT

Phase I clinical trials applying autologous progenitor cells to treat heart failure have yielded promising results; however, improvement in function is modest, indicating a need to enhance cardiac stem cell reparative capacity. Notch signaling plays a crucial role in cardiac development, guiding cell fate decisions that underlie myocyte and vessel differentiation. The Notch pathway is retained in the adult cardiac stem cell niche, where level and duration of Notch signal influence proliferation and differentiation of cardiac progenitors. In this study, Notch signaling promotes growth, survival and differentiation of cardiac progenitor cells into smooth muscle lineages in vitro. Cardiac progenitor cells expressing tamoxifen-regulated intracellular Notch1 (CPCeK) are significantly larger and proliferate more slowly than control cells, exhibit elevated mTORC1 and Akt signaling, and are resistant to oxidative stress. Vascular smooth muscle and cardiomyocyte markers increase in CPCeK and are augmented further upon ligand-mediated induction of Notch signal. Paracrine signals indicative of growth, survival and differentiation increase with Notch activity, while markers of senescence are decreased. Adoptive transfer of CPCeK into infarcted mouse myocardium enhances preservation of cardiac function and reduces infarct size relative to hearts receiving control cells. Greater capillary density and proportion of vascular smooth muscle tissue in CPCeK-treated hearts indicate improved vascularization. Finally, we report a previously undescribed signaling mechanism whereby Notch activation stimulates CPC growth, survival and differentiation via mTORC1 and paracrine factor expression. Taken together, these findings suggest that regulated Notch activation potentiates the reparative capacity of CPCs in the treatment of cardiac disease.


Subject(s)
Cell Differentiation/physiology , Cell- and Tissue-Based Therapy/methods , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Receptors, Notch/metabolism , Stem Cell Transplantation/methods , Adoptive Transfer , Animals , Cell Lineage , Disease Models, Animal , Immunoblotting , Immunohistochemistry , Mice , Myocytes, Cardiac/metabolism , Real-Time Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/metabolism
2.
Mol Ther ; 22(1): 178-85, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24002692

ABSTRACT

Stem cell survival and retention in myocardium after injury following adoptive transfer is low. Elevated catecholamine levels coinciding with myocardial injury adversely affect cardiac progenitor cell (CPC) survival. The G protein-coupled receptor kinase 2 (GRK2)-derived inhibitory peptide, ßARKct, enhance myocyte contractility, survival, and normalize the neurohormonal axis in failing heart, however salutary effects of ßARKct on CPC survival and proliferation are unknown. Herein, we investigated whether the protective effects of ßARKct expression seen in the failing heart relate to CPCs. Modified CPCs expressing ßARKct enhanced AKT/eNOS signaling through protective ß2-adrenergic receptors (ß2-ARs). In addition, to the actions of ßARKct expression on ß2- AR signaling, pharmacologic inhibition of GRK2 also increased ß2-AR signaling in nonengineered CPCs (lacking ßARKct) but had limited effects in ßARKct engineered CPCs providing evidence for the strength of the ßARKct in inhibiting GRK2 in these cells. Increased proliferation and metabolic activity were observed in ßARKct-engineered CPCs following catecholamine stimulation indicating improved adrenergic tolerance. ßARKct modification of CPCs increased survival and proliferation following adoptive transfer in an acute myocardial infarction model concomitant with increased expression of ß-AR. Thus, ßARKct engineering of CPCs promotes survival and proliferation of injected cells following myocardial infarction, which includes improved ß-adrenergic tolerance essential for stem cell survival.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/genetics , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Peptide Fragments/genetics , Animals , Catecholamines/pharmacology , Cell Proliferation , Cell Survival , Disease Models, Animal , G-Protein-Coupled Receptor Kinase 2/chemistry , Gene Expression , Heart/drug effects , Humans , Mice , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction/drug effects , Transduction, Genetic
3.
Mol Carcinog ; 52(7): 526-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22389255

ABSTRACT

Reactivation of the HMGA1 protoncogene is very frequent in human cancer, but still very little is known on the molecular mechanisms leading to this event. Prompted by the finding of putative E2F binding sites in the human HMGA1 promoter and by the frequent deregulation of the RB/E2F1 pathway in human carcinogenesis, we investigated whether E2F1 might contribute to the regulation of HMGA1 gene expression. Here we report that E2F1 induces HMGA1 by interacting with a 193 bp region of the HMGA1 promoter containing an E2F binding site surrounded by three putative Sp1 binding sites. Both gain and loss of function experiments indicate that Sp1 functionally interacts with E2F1 to promote HMGA1 expression. However, while Sp1 constitutively binds HMGA1 promoter, it is the balance between different E2F family members that tunes the levels of HMGA1 expression between quiescence and proliferation. Finally, we found increased HMGA1 expression in pituitary and thyroid tumors developed in Rb(+/-) mice, supporting the hypothesis that E2F1 is a novel important regulator of HMGA1 expression and that deregulation of the RB/E2F1 path might significantly contribute to HMGA1 deregulation in cancer.


Subject(s)
E2F1 Transcription Factor/metabolism , HMGA1a Protein/genetics , Pituitary Neoplasms/metabolism , Sp1 Transcription Factor/metabolism , Thyroid Neoplasms/metabolism , Animals , Base Sequence , Binding Sites , Blotting, Western , Chromatin Immunoprecipitation , E2F1 Transcription Factor/genetics , HMGA1a Protein/metabolism , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Pituitary Neoplasms/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retinoblastoma Protein/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Thyroid Neoplasms/genetics , Transcriptional Activation
4.
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
5.
Cell Transplant ; 21(9): 1997-2008, 2012.
Article in English | MEDLINE | ID: mdl-22469297

ABSTRACT

Presently, orthotopic liver transplant is the major therapeutic option for patients affected by primary liver diseases. This procedure is characterized by major invasive surgery, scarcity of donor organs, high costs, and lifelong immunosuppressive treatment. Transplant of hepatic precursor cells represents an attractive alternative. These cells could be used either for allogeneic transplantation or for autologous transplant after ex vivo genetic modification. We used stromal cells isolated from adipose tissue (AT-SCs) as platforms for autologous cell-mediated gene therapy. AT-SCs were transduced with lentiviral vectors expressing firefly luciferase, allowing for transplanted cell tracking by bioluminescent imaging (BLI). As a complementary approach, we followed circulating human α1-antitrypsin (hAAT) levels after infusion of AT-SCs overexpressing hAAT. Cells were transplanted into syngeneic mice after CCl(4)-induced hepatic injury. Luciferase bioluminescence signals and serum hAAT levels were measured at different time points after transplantation and demonstrate persistence of transplanted cells for up to 2 months after administration. These data, along with immunohistochemical analysis, suggest engraftment and repopulation of injured livers by transplanted AT-SCs. Moreover, by transcriptional targeting using cellular tissue-specific regulatory sequences, we confirmed that AT-SCs differentiate towards a hepatogenic-like phenotype in vitro and in vivo. Additionally, in transplanted cells reisolated from recipient animals' livers, we detected activation of the α-fetoprotein (AFP) promoter. This promoter is normally transcriptionally silenced in adult tissues but can be reactivated during liver regeneration, suggesting commitment towards hepatogenic-like differentiation of engrafted cells in vivo. Our data support AT-SC-mediated gene therapy as an innovative therapeutic option for disorders of liver metabolism.


Subject(s)
Adipose Tissue/cytology , Genetic Therapy/methods , Liver Diseases/surgery , Liver Regeneration/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Cell Differentiation , Humans , Liver Diseases/pathology , Mesenchymal Stem Cells/cytology , Mice , Tissue Distribution
6.
Am J Physiol Heart Circ Physiol ; 300(5): H1875-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21357510

ABSTRACT

The efficacy of cardiac repair by stem cell administration relies on a successful functional integration of injected cells into the host myocardium. Safety concerns have been raised about the possibility that stem cells may induce foci of arrhythmia in the ischemic myocardium. In a previous work (36), we showed that human cord blood CD34(+) cells, when cocultured on neonatal mouse cardiomyocytes, exhibit excitation-contraction coupling features similar to those of cardiomyocytes, even though no human genes were upregulated. The aims of the present work are to investigate whether human CD34(+) cells, isolated after 1 wk of coculture with neonatal ventricular myocytes, possess molecular and functional properties of cardiomyocytes and to discriminate, using a reporter gene system, whether cardiac differentiation derives from a (trans)differentiation or a cell fusion process. Umbilical cord blood CD34(+) cells were isolated by a magnetic cell sorting method, transduced with a lentiviral vector carrying the enhanced green fluorescent protein (EGFP) gene, and seeded onto primary cultures of spontaneously beating rat neonatal cardiomyocytes. Cocultured EGFP(+)/CD34(+)-derived cells were analyzed for their electrophysiological features at different time points. After 1 wk in coculture, EGFP(+) cells, in contact with cardiomyocytes, were spontaneously contracting and had a maximum diastolic potential (MDP) of -53.1 mV, while those that remained isolated from the surrounding myocytes did not contract and had a depolarized resting potential of -11.4 mV. Cells were then resuspended and cultured at low density to identify EGFP(+) progenitor cell derivatives. Under these conditions, we observed single EGFP(+) beating cells that had acquired an hyperpolarization-activated current typical of neonatal cardiomyocytes (EGFP(+) cells, -2.24 ± 0.89 pA/pF; myocytes, -1.99 ± 0.63 pA/pF, at -125 mV). To discriminate between cell autonomous differentiation and fusion, EGFP(+)/CD34(+) cells were cocultured with cardiac myocytes infected with a red fluorescence protein-lentiviral vector; under these conditions we found that 100% of EGFP(+) cells were also red fluorescent protein positive, suggesting cell fusion as the mechanism by which cardiac functional features are acquired.


Subject(s)
Antigens, CD34/metabolism , Cell Communication/physiology , Cell Fusion/methods , Fetal Blood/cytology , Myocytes, Cardiac/cytology , Stem Cells/cytology , Stem Cells/immunology , Animals , Antigens, CD34/genetics , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Cord Blood Stem Cell Transplantation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Models, Animal , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Rats , Stem Cells/physiology
7.
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
8.
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
9.
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
10.
Cardiovasc Res ; 86(2): 293-301, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20007689

ABSTRACT

AIMS: Acidification is associated with a variety of pathological and physiological conditions. In the present study, we aimed at investigating whether acidic pH may regulate endothelial cell (EC) functions via the chemokine receptor CXCR4, a key modulator of EC biological activities. METHODS AND RESULTS: Exposure of ECs to acidic pH reversibly inhibited mRNA and protein CXCR4 expression, CXCL12/stromal cell-derived factor (SDF)-1-driven EC chemotaxis in vitro, and CXCR4 expression and activation in vivo in a mouse model. Further, CXCR4 signalling impaired acidosis-induced rescue from apoptosis in ECs. The inhibition of CXCR4 expression occurred transcriptionally and was hypoxia-inducible factor (HIF)-1alpha-dependent as demonstrated by both HIF-1alpha and HIF-1alpha dominant negative overexpression, by HIF-1alpha silencing, and by targeted mutation of the -29 to -25 hypoxia response element (HRE) in the -357/-59 CXCR4 promoter fragment. Moreover, chromatin immunoprecipitation (ChIP) analysis showed endogenous HIF-1alpha binding to the CXCR4 promoter that was enhanced by acidification. CONCLUSION: The results of the present study identify CXCR4 as a key player in the EC response to acidic pH and show, for the first time, that HRE may function not only as an effector of hypoxia, but also as an acidosis response element, and raise the possibility that this may constitute a more general mechanism of transcriptional regulation at acidic pH.


Subject(s)
Acidosis/metabolism , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptors, CXCR4/metabolism , Acidosis/chemically induced , Acidosis/immunology , Acidosis/pathology , Ammonium Chloride , Animals , Apoptosis , Binding Sites , Cell Hypoxia , Cells, Cultured , Chemokine CXCL12/metabolism , Chemotaxis , Chromatin Immunoprecipitation , Disease Models, Animal , Down-Regulation , Endothelial Cells/immunology , Endothelial Cells/pathology , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mutation , Phosphorylation , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Receptors, CXCR4/genetics , Time Factors , Transcription, Genetic , Transfection
11.
Neoplasia ; 11(8): 732-42, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19649203

ABSTRACT

Cutaneous melanoma is the most aggressive skin cancer; it is highly metastatic and responds poorly to current therapies. The expression of platelet-derived growth factor receptors (PDGF-Rs) is reported to be reduced in metastatic melanoma compared with benign nevi or normal skin; we then hypothesized that PDGF-Ralpha may control growth of melanoma cells. We show here that melanoma cells overexpressing PDGF-Ralpha respond to serum with a significantly lower proliferation compared with that of controls. Apoptosis, cell cycle arrest, pRb dephosphorylation, and DNA synthesis inhibition were also observed in cells overexpressing PDGF-Ralpha. Proliferation was rescued by PDGF-Ralpha inhibitors, allowing to exclude nonspecific toxic effects and indicating that PDGF-Ralpha mediates autocrine antiproliferation signals in melanoma cells. Accordingly, PDGF-Ralpha was found to mediate staurosporine cytotoxicity. A protein array-based analysis of the mitogen-activated protein kinase pathway revealed that melanoma cells overexpressing PDGF-Ralpha show a strong reduction of c-Jun phosphorylated in serine 63 and of protein phosphatase 2A/Balpha and a marked increase of p38gamma, mitogen-activated protein kinase kinase 3, and signal regulatory protein alpha1 protein expression. In a mouse model of primary melanoma growth, infection with the Ad-vector overexpressing PDGF-Ralpha reached a significant 70% inhibition of primary melanoma growth (P < .001) and a similar inhibition of tumor angiogenesis. All together, these data demonstrate that PDGF-Ralpha strongly impairs melanoma growth likely through autocrine mechanisms and indicate a novel endogenous mechanism involved in melanoma control.


Subject(s)
Melanoma/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/physiology , Skin Neoplasms/metabolism , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Line, Tumor , Cell Proliferation , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , In Situ Nick-End Labeling , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , Melanoma/genetics , Mice , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 12/metabolism , Phosphorylation , Protein Array Analysis , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Skin Neoplasms/genetics , Transfection
12.
PLoS One ; 4(8): e6603, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19668373

ABSTRACT

BACKGROUND: In the cardiovascular system, laminar shear stress (SS) is one of the most important source of endothelial protecting signals. Physical and chemical agents, however, including ionising radiations and anticancer drugs, may injure endothelial cells determining an increase in oxidative stress and genotoxic damage. Whether the SS protective function remains intact in the presence of strong oxidants or DNA damage is currently unclear. METHODS AND RESULTS: To investigate this aspect a series of experiments were performed in which HUVEC were exposed to sub-lethal doses of the radio-mimetic compound Bleomycin (Bleo; 10 microg/ml) which generated free radicals (ROS) without significantly compromising cell survival. Remarkably, the application of a SS of 12 dyne/cm(2) did not protect endothelial cells but markedly accelerated apoptosis compared to controls kept in static culture and in the presence of Bleo. Experiments with the inducible nitric oxide synthase (iNOS) inhibitor GW274150 significantly reduced the SS-dependent apoptosis indicating that the production of NO was relevant for this effect. At molecular level, the ataxia-telangectasia-mutated (ATM) kinase, the homeodomain-interacting protein kinase-2 (HIPK2) and p53 were found activated along a pro-apoptotic signalling pathway while p21(waf1,cip1,sdi1) was prevented from its protective action. RNA interference experiments revealed that HIPK2 and p53 were both important for this process, however, only the forced expression p21(waf1,cip1,sdi1) fully restored the SS-dependent pro-survival function. CONCLUSIONS: This study provides the first evidence that, in the presence of genotoxic damage, laminar flow contributes to endothelial toxicity and death and identifies molecular targets potentially relevant in endothelial dysfunction and cardiovascular disease pathogenesis.


Subject(s)
Carrier Proteins/metabolism , Endothelium, Vascular/enzymology , Protein Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Base Sequence , Bleomycin/pharmacology , Blotting, Western , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , DNA Primers , Endothelium, Vascular/cytology , Enzyme Activation , Humans , Polymerase Chain Reaction , RNA Interference , Reactive Oxygen Species , Tumor Suppressor Protein p53/metabolism
13.
Neoplasia ; 10(8): 773-81, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18670638

ABSTRACT

HMGA1 is a member of a small family of architectural transcription factors involved in the coordinate assembly of multiprotein complexes referred to as enhanceosomes. In addition to their role in cell proliferation, differentiation, and development, high-mobility group proteins of the A type (HMGA) family members behave as transforming protoncogenes either in vitro or in animal models. Recent reports indicated that HMGA1 might counteract p53 pathway and provided an interesting hint on the mechanisms determining HMGA's transforming potential. HMGA1 expression is deregulated in a very large array of human tumors, including cervical cancer, but very limited information is available on the molecular mechanisms leading to HMGA1 deregulation in cancer cells. Here, we report that HMGA1 expression is sustained by human papilloma virus (HPV) E6/E7 proteins in cervical cancer, as demonstrated by either E6/E7 overexpression or by repression through RNA interference. Knocking down HMGA1 expression by means of RNA interference, we also showed that it is involved in cell proliferation and contributes to p53 inactivation in this type of neoplasia. Finally, we show that HMGA1 is necessary for the full expression of HPV18 E6 and E7 oncoproteins thus establishing a positive autoregulatory loop between HPV E6/E7 and HMGA1 expression.


Subject(s)
Cell Transformation, Viral/genetics , Gene Expression Regulation, Neoplastic , HMGA1a Protein/genetics , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , HMGA1a Protein/metabolism , HeLa Cells , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/virology , RNA, Messenger/genetics , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/pathology
14.
Mol Cancer Res ; 6(5): 751-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18458053

ABSTRACT

BRAF-activating mutations have been reported in several types of cancer, including melanoma ( approximately 70% of cases), thyroid (30-70%), ovarian (15-30%), and colorectal cancer (5-20%). Mutant BRAF has constitutive kinase activity and causes hyperactivation of the mitogen-activated protein kinase pathway. BRAF silencing induces regression of melanoma xenografts, indicating the essential role of BRAF for cell survival. We set up an inducible short hairpin RNA system to compare the role of oncogenic BRAF in thyroid carcinoma versus melanoma cells. Although BRAF knockdown led to apoptosis in the melanoma cell line A375, the anaplastic thyroid carcinoma cell ARO underwent growth arrest upon silencing, with little or no cell death. Reexpression of the thyroid differentiation marker, sodium iodide symporter, was induced after long-term silencing. The different outcome of BRAF down-regulation in the two cell lines was associated with an opposite regulation of p21(CIP1/WAF1) expression levels in response to the block of the BRAF mitogenic signal. These results were confirmed using a specific BRAF small-molecule inhibitor, PLX4032. Restoration of p21(CIP1/WAF1) expression rescued melanoma cells from death. Altogether, our data indicate that oncogenic BRAF inhibition can have a different effect on cell fate depending on the cellular type. Furthermore, we suggest that a BRAF-independent mechanism of cell survival exists in anaplastic thyroid cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Melanoma/drug therapy , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Inhibitory Concentration 50 , Models, Biological , RNA/chemistry , Symporters/metabolism
15.
J Biol Chem ; 280(15): 14790-8, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15687494

ABSTRACT

Telomere dysfunction contributes to reduced cell viability, altered differentiation, and impaired regenerative/proliferative responses. Recent advances indicate that telomerase activity confers a pro-angiogenic phenotype to endothelial cells and their precursors. We have investigated whether telomerase contributes to tissue regeneration following hind limb ischemia and vascular endothelial growth factor 165 (VEGF(165)) treatment. VEGF delivery induced angiogenesis and increased expression of the telomerase reverse transcriptase (TERT) and telomerase activity in skeletal muscles and satellite and endothelial cells. Adenovirus-mediated transfer of wild type TERT but not of a dominant negative mutant, TERTdn, significantly induced capillary but not arteriole formation. However, when co-delivered with VEGF, TERTdn abrogated VEGF-dependent angiogenesis, arteriogenesis, and blood flow increase. This effect was paralleled by in vitro evidence that telomerase inhibition by 3'-azido-3'-deoxythymidine in VEGF-treated endothelial cells strongly reduced capillary density and promoted apoptosis in the absence of serum. Similar results were obtained with adenovirus-mediated expression of TERTdn and AKTdn, both reducing endogenous TERT activity and angiogenesis on Matrigel. Mechanistically, neo-angiogenesis in our system involved: (i) VEGF-dependent activation of telomerase through the nitric oxide pathway and (ii) telomerase-dependent activation of endothelial cell differentiation and protection from apoptosis. Furthermore, detection of TERT in activated satellite cells identified them as VEGF targets during muscle regeneration. Because TERT behaves as an angiogenic factor and a downstream effector of VEGF signaling, telomerase activity appears required for VEGF-dependent remodeling of ischemic tissue at the capillaries and arterioles level.


Subject(s)
Ischemia , Telomerase/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Collagen/chemistry , DNA-Binding Proteins , Drug Combinations , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Extremities , Genetic Therapy/methods , Genetic Vectors , Humans , In Situ Nick-End Labeling , Laminin/chemistry , Mice , Microscopy, Fluorescence , Muscle, Skeletal/metabolism , Neovascularization, Pathologic , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Perfusion , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Proteoglycans/chemistry , Rats , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic , Transfection , Umbilical Veins/cytology , Up-Regulation
16.
Blood ; 104(12): 3472-82, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15284120

ABSTRACT

Chemokine stromal derived factor 1 (SDF-1) is involved in trafficking of hematopoietic stem cells (HSCs) from the bone marrow (BM) to peripheral blood (PB) and has been found to enhance postischemia angiogenesis. This study was aimed at investigating whether SDF-1 plays a role in differentiation of BM-derived c-kit(+) stem cells into endothelial progenitor cells (EPCs) and in ischemia-induced trafficking of stem cells from PB to ischemic tissues. We found that SDF-1 enhanced EPC number by promoting alpha(2), alpha(4), and alpha(5) integrin-mediated adhesion to fibronectin and collagen I. EPC differentiation was reduced in mitogen-stimulated c-kit(+) cells, while cytokine withdrawal or the overexpression of the cyclin-dependent kinase (CDK) inhibitor p16(INK4) restored such differentiation, suggesting a link between control of cell cycle and EPC differentiation. We also analyzed the time course of SDF-1 expression in a mouse model of hind-limb ischemia. Shortly after femoral artery dissection, plasma SDF-1 levels were up-regulated, while SDF-1 expression in the bone marrow was down-regulated in a timely fashion with the increase in the percentage of PB progenitor cells. An increase in ischemic tissue expression of SDF-1 at RNA and protein level was also observed. Finally, using an in vivo assay such as injection of matrigel plugs, we found that SDF-1 improves formation of tubulelike structures by coinjected c-kit(+) cells. Our findings unravel a function for SDF-1 in increase of EPC number and formation of vascular structures by bone marrow progenitor cells.


Subject(s)
Chemokines, CXC/physiology , Chemotaxis , Endothelial Cells/cytology , Hematopoietic Stem Cells/physiology , Ischemia/pathology , Animals , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Cycle/physiology , Cell Differentiation , Chemokine CXCL12 , Chemokines, CXC/blood , Chemokines, CXC/genetics , Endothelium, Vascular/cytology , Integrins , Ischemia/metabolism , Mice , Neovascularization, Pathologic , Phenotype , Proto-Oncogene Proteins c-kit
SELECTION OF CITATIONS
SEARCH DETAIL