Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 64(6): 661-73, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17380307

ABSTRACT

Regardless of erroneous claims by a minority of reports, adult cardiomyocytes are terminally differentiated cells which do not re-enter the cell-cycle under any known physiological or pathological circumstances. However, it has recently been shown that the adult heart has a robust myocardial regenerative potential, which challenges the accepted notions of cardiac cellular biology. The source of this regenerative potential is constituted by resident cardiac stem cells (CSCs). These CSCs, through both cell transplantation and in situ activation, have the capacity to regenerate significant segmental and diffuse myocyte losts, restoring anatomical integrity and ventricular function. Thus, CSC identification has started a brand new discipline of cardiac biology that could profoundly changed the outlook of cardiac physiology and the potential for treatment of cardiac failure. Nonetheless, the dawn of this new era should not be set back by premature attempts at clinical application before having accumulated the required scientifically reproducible data.


Subject(s)
Myocardium/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Fusion , Cell Lineage , Humans , Regeneration
2.
Diabetes ; 50(10): 2363-75, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11574421

ABSTRACT

To determine whether enzymatic p53 glycosylation leads to angiotensin II formation followed by p53 phosphorylation, prolonged activation of the renin-angiotensin system, and apoptosis, ventricular myocytes were exposed to levels of glucose mimicking diabetic hyperglycemia. At a high glucose concentration, O-glycosylation of p53 occurred between 10 and 20 min, reached its peak at 1 h, and then decreased with time. Angiotensin II synthesis increased at 45 min and 1 h, resulting in p38 mitogen-activated protein (MAP) kinase-driven p53 phosphorylation at Ser 390. p53 phosphorylation was absent at the early time points, becoming evident at 1 h, and increasing progressively from 3 h to 4 days. Phosphorylated p53 at Ser 18 and activated c-Jun NH(2)-terminal kinases were identified with hyperglycemia, whereas extracellular signal-regulated kinase was not phosphorylated. Upregulation of p53 was associated with an accumulation of angiotensinogen and AT(1) and enhanced production of angiotensin II. Bax quantity also increased. These multiple adaptations paralleled the concentrations of glucose in the medium and the duration of the culture. Myocyte death by apoptosis directly correlated with glucose and angiotensin II levels. Inhibition of O-glycosylation prevented the initial synthesis of angiotensin II, p53, and p38-MAP kinase (MAPK) phosphorylation and apoptosis. AT(1) blockade had no influence on O-glycosylation of p53, but it interfered with p53 phosphorylation; losartan also prevented phosphorylation of p38-MAPK by angiotensin II. Inhibition of p38-MAPK mimicked at a more distal level the consequences of losartan. In conclusion, these in vitro results support the notion that hyperglycemia with diabetes promotes myocyte apoptosis mediated by activation of p53 and effector responses involving the local renin-angiotensin system.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation/physiology , Hyperglycemia/genetics , JNK Mitogen-Activated Protein Kinases , Myocardium/cytology , Tumor Suppressor Protein p53/physiology , Angiotensin II/biosynthesis , Animals , Cells, Cultured , DNA/metabolism , Glycosylation , Heart Ventricles , Hydrogen-Ion Concentration , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Losartan/pharmacology , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases
3.
Circ Res ; 89(3): 279-86, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11485979

ABSTRACT

Cell death has been questioned as a mechanism of ventricular failure. In this report, we tested the hypothesis that apoptotic death of myocytes, endothelial cells, and fibroblasts is implicated in the development of the dilated myopathy induced by ventricular pacing. Accumulation of reactive oxygen products such as nitrotyrosine, potentiation of the oxidative stress response by p66(shc) expression, formation of p53 fragments, release of cytochrome c, and caspase activation were examined to establish whether these events were coupled with apoptotic cell death in the paced dog heart. Myocyte, endothelial cell, and fibroblast apoptosis was detected before indices of severe impairment of cardiac function became apparent. Cell death increased with the duration of pacing, and myocyte death exceeded endothelial cell and fibroblast death throughout. Nitrotyrosine formation and p66(shc) levels progressively increased with pacing and were associated with cell apoptosis. Similarly, p50 (DeltaN) fragments augmented paralleling the degree of cell death in the failing heart. Moreover, cytochrome c release and activation of caspase-9 and -3 increased from 1 to 4 weeks of pacing. In conclusion, cardiac cell death precedes ventricular decompensation and correlates with the time-dependent deterioration of function in this model. Oxidative stress may be critical for activation of apoptosis in the overloaded heart.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Apoptosis , Cardiomyopathy, Dilated/physiopathology , Oxidative Stress , Tyrosine/analogs & derivatives , Ventricular Dysfunction/etiology , Ventricular Dysfunction/physiopathology , Animals , Blotting, Western , Cardiac Pacing, Artificial , Cardiomyopathy, Dilated/pathology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cytochrome c Group/metabolism , Disease Models, Animal , Dogs , Enzyme Activation/physiology , Hemodynamics , Immunohistochemistry , In Situ Nick-End Labeling , Myocardium/metabolism , Myocardium/pathology , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins , Tumor Suppressor Protein p53/metabolism , Tyrosine/metabolism , Ventricular Dysfunction/pathology
4.
Proc Natl Acad Sci U S A ; 98(18): 10344-9, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504914

ABSTRACT

Attempts to repair myocardial infarcts by transplanting cardiomyocytes or skeletal myoblasts have failed to reconstitute healthy myocardium and coronary vessels integrated structurally and functionally with the remaining viable portion of the ventricular wall. The recently discovered growth and transdifferentiation potential of primitive bone marrow cells (BMC) prompted us, in an earlier study, to inject in the border zone of acute infarcts Lin(-) c-kit(POS) BMC from syngeneic animals. These BMC differentiated into myocytes and vascular structures, ameliorating the function of the infarcted heart. Two critical determinants seem to be required for the transdifferentiation of primitive BMC: tissue damage and a high level of pluripotent cells. On this basis, we hypothesized here that BMC, mobilized by stem cell factor and granulocyte-colony stimulating factor, would home to the infarcted region, replicate, differentiate, and ultimately promote myocardial repair. We report that, in the presence of an acute myocardial infarct, cytokine-mediated translocation of BMC resulted in a significant degree of tissue regeneration 27 days later. Cytokine-induced cardiac repair decreased mortality by 68%, infarct size by 40%, cavitary dilation by 26%, and diastolic stress by 70%. Ejection fraction progressively increased and hemodynamics significantly improved as a consequence of the formation of 15 x 10(6) new myocytes with arterioles and capillaries connected with the circulation of the unaffected ventricle. In conclusion, mobilization of primitive BMC by cytokines might offer a noninvasive therapeutic strategy for the regeneration of the myocardium lost as a result of ischemic heart disease and, perhaps, other forms of cardiac pathology.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Animals , Cell Differentiation , Cell Division , Granulocyte Colony-Stimulating Factor/pharmacology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hematopoietic Stem Cell Mobilization/methods , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Regeneration , Stem Cell Factor/pharmacology , Transplantation, Isogeneic
5.
Proc Natl Acad Sci U S A ; 98(15): 8626-31, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11447262

ABSTRACT

The role and even the existence of myocyte proliferation in the adult heart remain controversial. Documentation of cell cycle regulators, DNA synthesis, and mitotic images has not modified the view that myocardial growth can only occur from hypertrophy of an irreplaceable population of differentiated myocytes. To improve understanding the biology of the heart and obtain supportive evidence of myocyte replication, three indices of cell proliferation were analyzed in dogs affected by a progressive deterioration of cardiac performance and dilated cardiomyopathy. The magnitude of cycling myocytes was evaluated by the expression of Ki67 in nuclei. Ki67 labeling of left ventricular myocytes increased 5-fold, 12-fold, and 17-fold with the onset of moderate and severe ventricular dysfunction and overt failure, respectively. Telomerase activity in vivo is present only in multiplying cells; this enzyme increased 2.4-fold and 3.1-fold in the decompensated heart, preserving telomeric length in myocytes. The contribution of cycling myocytes to telomerase activity was determined by the colocalization of Ki67 and telomerase in myocyte nuclei. More than 50% of Ki67-positive cells expressed telomerase in the overloaded myocardium, suggesting that these myocytes were the morphological counterpart of the biochemical assay of enzyme activity. Moreover, we report that 20--30% of canine myocytes were telomerase competent, and this value was not changed by cardiac failure. In conclusion, the enhanced expression of Ki67 and telomerase activity, in combination with Ki67-telomerase labeling of myocyte nuclei, support the notion that myocyte proliferation contributes to cardiac hypertrophy of the diseased heart.


Subject(s)
Heart Failure/metabolism , Myocardium/metabolism , Telomerase/metabolism , Telomere/physiology , Animals , Cell Division , DNA-Binding Proteins , Dogs , Heart Failure/pathology , Humans , Ki-67 Antigen/analysis , Myocardium/cytology , Myocardium/enzymology , Telomerase/biosynthesis
6.
N Engl J Med ; 344(23): 1750-7, 2001 Jun 07.
Article in English | MEDLINE | ID: mdl-11396441

ABSTRACT

BACKGROUND: The scarring of the heart that results from myocardial infarction has been interpreted as evidence that the heart is composed of myocytes that are unable to divide. However, recent observations have provided evidence of proliferation of myocytes in the adult heart. Therefore, we studied the extent of mitosis among myocytes after myocardial infarction in humans. METHODS: Samples from the border of the infarct and from areas of the myocardium distant from the infarct were obtained from 13 patients who had died 4 to 12 days after infarction. Ten normal hearts were used as controls. Myocytes that had entered the cell cycle in preparation for cell division were measured by labeling of the nuclear antigen Ki-67, which is associated with cell division. The fraction of myocyte nuclei that were undergoing mitosis was determined, and the mitotic index (the ratio of the number of nuclei undergoing mitosis to the number not undergoing mitosis) was calculated. The presence of mitotic spindles, contractile rings, karyokinesis, and cytokinesis was also recorded. RESULTS: In the infarcted hearts, Ki-67 expression was detected in 4 percent of myocyte nuclei in the regions adjacent to the infarcts and in 1 percent of those in regions distant from the infarcts. The reentry of myocytes into the cell cycle resulted in mitotic indexes of 0.08 percent and 0.03 percent, respectively, in the zones adjacent to and distant from the infarcts. Events characteristic of cell division--the formation of the mitotic spindles, the formation of contractile rings, karyokinesis, and cytokinesis--were identified; these features demonstrated that there was myocyte proliferation after myocardial infarction. CONCLUSIONS: Our results challenge the dogma that the adult heart is a postmitotic organ and indicate that the regeneration of myocytes may be a critical component of the increase in muscle mass of the myocardium.


Subject(s)
Mitosis , Myocardial Infarction/pathology , Myocardium/cytology , Regeneration , Antibodies, Monoclonal , Case-Control Studies , Cell Division , Heart/physiology , Humans , Ki-67 Antigen/analysis , Ki-67 Antigen/immunology , Microscopy, Confocal , Mitotic Index , Myocardial Infarction/physiopathology , Myocardium/chemistry
7.
Rev Esp Cardiol ; 54(5): 543-50, 2001 May.
Article in Spanish | MEDLINE | ID: mdl-11412743

ABSTRACT

The classic dogma, still prevalent in cardiology, that the adult myocardium is a terminally differentiated tissue unable to produce new cardiomyocytes needs to be revised in light of recent results. In human and experimental animals there is now incontrovertible evidence that new myocytes are continuously generated throughout life in response to physiological and pathological stimuli. Moreover, the elucidation of mechanisms responsible for the hypertrophic response indicate similarity and overlap with the mechanisms involved in cell death by apoptosis as well as cell growth. During cardiac development, from birth to adulthood, there is a balance between the stimuli induce cell growth -by hypertrophy and hyperplasia- on one hand and those that induce programmed cell death on the other. In human and experimental animals it has been well documented that pathological conditions, such as diabetes and hypertension, can increase dramatically the rate of cell death. Moreover, high rates of cell death have been measured in normal adult human hearts and those of mice and rats. No surprisingly, these values increase significantly with age and high in senescence. By themselves, these high rates of normal cell death provide a very compelling argument in favor of cardiomyocyte regeneration. Without cell renewal, these rates of cell death would be incompatible with survival because the heart would disappear before early adulthood. As expected, direct measurement of rates of new cell formation in adult hearts demonstrate high rates of cell renewal that compensate for cell death. Thus, the heart is in continuous cellular turnover with new myocardial cells replacing the older ones. Experiments with fetal mouse cardiocytes shows that the retinoblastoma protein is responsible for the cardiocyte withdrawal from the cell cycle during development. The identification in the adult heart of a subpopulation of quiescent cells that have many of the characteristics of stem cells able to rapidly enter the cell cycle and generate new cardiocytes is yet another evidence that the heart continuously produces new cardiocytes to replace those that disappear either by apoptosis or necrosis.Surprisingly, stem cells other that those from the heart are able to produce new cardiocytes and repopulate the myocardium. We have used bone marrow stem cells injected into the border zone of post-coronary occlusion necrosis. Remarkably, these cells have proven to be very effective in generating new myocardium in the necrotic zone that is integrated to the rest of the muscle and irrigated by new vessels. These results demonstrate that stem cells provide a new avenue for the generation of new contractile tissue. This approach could prove useful in the treatment of chronic cardiac failure and post-ischemic necrosis.


Subject(s)
Heart Transplantation , Myocardium/cytology , Regeneration/physiology , Stem Cells , Adult , Humans
8.
Diabetes ; 50(6): 1414-24, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375343

ABSTRACT

Stimulation of the local renin-angiotensin system and apoptosis characterize the diabetic heart. Because IGF-1 reduces angiotensin (Ang) II and apoptosis, we tested whether streptozotocin-induced diabetic cardiomyopathy was attenuated in IGF-1 transgenic mice (TGM). Diabetes progressively depressed ventricular performance in wild-type mice (WTM) but had no hemodynamic effect on TGM. Myocyte apoptosis measured at 7 and 30 days after the onset of diabetes was twofold higher in WTM than in TGM. Myocyte necrosis was apparent only at 30 days and was more severe in WTM. Diabetic nontransgenic mice lost 24% of their ventricular myocytes and showed a 28% myocyte hypertrophy; both phenomena were prevented by IGF-1. In diabetic WTM, p53 was increased in myocytes, and this activation of p53 was characterized by upregulation of Bax, angiotensinogen, Ang type 1 (AT(1)) receptors, and Ang II. IGF-1 overexpression decreased these biochemical responses. In vivo accumulation of the reactive O(2) product nitrotyrosine and the in vitro formation of H(2)O(2)-(.)OH in myocytes were higher in diabetic WTM than TGM. Apoptosis in vitro was detected in myocytes exhibiting high H(2)O(2)-(.)OH fluorescence, and apoptosis in vivo was linked to the presence of nitrotyrosine. H(2)O(2)-(.)OH generation and myocyte apoptosis in vitro were inhibited by the AT(1) blocker losartan and the O(2) scavenger TIRON: In conclusion, IGF-1 interferes with the development of diabetic myopathy by attenuating p53 function and Ang II production and thus AT(1) activation. This latter event might be responsible for the decrease in oxidative stress and myocyte death by IGF-1.


Subject(s)
Angiotensin II/physiology , Cardiomyopathies/prevention & control , Diabetic Angiopathies/prevention & control , Insulin-Like Growth Factor I/pharmacology , Oxidative Stress/drug effects , Tyrosine/analogs & derivatives , Angiotensin II/pharmacology , Animals , Apoptosis/drug effects , Cardiomyopathies/physiopathology , DNA/metabolism , Diabetic Angiopathies/physiopathology , Insulin-Like Growth Factor I/genetics , Mice , Mice, Transgenic/genetics , Myocardium/cytology , Myocardium/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/physiology , Tissue Distribution , Tumor Suppressor Protein p53/genetics , Tyrosine/metabolism , Ventricular Function
9.
Nature ; 410(6829): 701-5, 2001 Apr 05.
Article in English | MEDLINE | ID: mdl-11287958

ABSTRACT

Myocardial infarction leads to loss of tissue and impairment of cardiac performance. The remaining myocytes are unable to reconstitute the necrotic tissue, and the post-infarcted heart deteriorates with time. Injury to a target organ is sensed by distant stem cells, which migrate to the site of damage and undergo alternate stem cell differentiation; these events promote structural and functional repair. This high degree of stem cell plasticity prompted us to test whether dead myocardium could be restored by transplanting bone marrow cells in infarcted mice. We sorted lineage-negative (Lin-) bone marrow cells from transgenic mice expressing enhanced green fluorescent protein by fluorescence-activated cell sorting on the basis of c-kit expression. Shortly after coronary ligation, Lin- c-kitPOS cells were injected in the contracting wall bordering the infarct. Here we report that newly formed myocardium occupied 68% of the infarcted portion of the ventricle 9 days after transplanting the bone marrow cells. The developing tissue comprised proliferating myocytes and vascular structures. Our studies indicate that locally delivered bone marrow cells can generate de novo myocardium, ameliorating the outcome of coronary artery disease.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Myocardium/pathology , Animals , Cell Differentiation , Connexin 43/metabolism , DNA-Binding Proteins/metabolism , Female , Green Fluorescent Proteins , Ki-67 Antigen/metabolism , Luminescent Proteins/metabolism , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/cytology , Myogenic Regulatory Factors , Proto-Oncogene Proteins c-kit/metabolism , Transcription Factors/metabolism
10.
Circ Res ; 88(3): 298-304, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11179197

ABSTRACT

Ventricular pacing leads to a dilated myopathy in which cell death and myocyte hypertrophy predominate. Because angiotensin II (Ang II) stimulates myocyte growth and triggers apoptosis, we tested whether canine myocytes express the components of the renin-angiotensin system (RAS) and whether the local RAS is upregulated with heart failure. p53 modulates transcription of angiotensinogen (Aogen) and AT(1) receptors in myocytes, raising the possibility that enhanced p53 function in the decompensated heart potentiates Ang II synthesis and Ang II-mediated responses. Therefore, the presence of mRNA transcripts for Aogen, renin, angiotensin-converting enzyme, chymase, and AT(1) and AT(2) receptors was evaluated by reverse transcriptase-polymerase chain reaction in myocytes. Changes in the protein expression of these genes were then determined by Western blot in myocytes from control dogs and dogs affected by congestive heart failure. p53 binding to the promoter of Aogen and AT(1) receptor was also determined. Ang II in myocytes was measured by ELISA and by immunocytochemistry and confocal microscopy. Myocytes expressed mRNAs for all the constituents of RAS, and heart failure was characterized by increased p53 DNA binding to Aogen and AT(1). Additionally, protein levels of Aogen, renin, cathepsin D, angiotensin-converting enzyme, and AT(1) were markedly increased in paced myocytes. Conversely, chymase and AT(2) proteins were not altered. Ang II quantity and labeling of myocytes increased significantly with cardiac decompensation. In conclusion, dog myocytes synthesize Ang II, and activation of p53 function with ventricular pacing upregulates the myocyte RAS and the generation and secretion of Ang II. Ang II may promote myocyte growth and death, contributing to the development of heart failure.


Subject(s)
Heart Failure/physiopathology , Renin-Angiotensin System/physiology , Ventricular Function , Actins/metabolism , Angiotensin II/metabolism , Animals , Binding, Competitive , Blotting, Western , Cardiac Pacing, Artificial , Cathepsin D/metabolism , Chymases , Dogs , Heart Ventricles/cytology , Heart Ventricles/metabolism , Immunohistochemistry , Microscopy, Confocal , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Receptors, Angiotensin/metabolism , Renin/genetics , Renin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
11.
Circ Res ; 87(12): 1123-32, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11110769

ABSTRACT

The renin-angiotensin system is upregulated with diabetes, and this may contribute to the development of a dilated myopathy. Angiotensin II (Ang II) locally may lead to oxidative damage, activating cardiac cell death. Moreover, diabetes and hypertension could synergistically impair myocardial structure and function. Therefore, apoptosis and necrosis were measured in ventricular myocardial biopsies obtained from diabetic and diabetic-hypertensive patients. Accumulation of a marker of oxidative stress, nitrotyrosine, and Ang II labeling were evaluated quantitatively. The diabetic heart showed cardiac hypertrophy, cavitary dilation, and depressed ventricular performance. These alterations were more severe with diabetes and hypertension. Diabetes was characterized by an 85-fold, 61-fold, and 26-fold increase in apoptosis of myocytes, endothelial cells, and fibroblasts, respectively. Apoptosis in cardiac cells did not increase additionally with diabetes and hypertension. Diabetes increased necrosis by 4-fold in myocytes, 9-fold in endothelial cells, and 6-fold in fibroblasts. However, diabetes and hypertension increased necrosis by 7-fold in myocytes and 18-fold in endothelial cells. Similarly, Ang II labeling in myocytes and endothelial cells increased more with diabetes and hypertension than with diabetes alone. Nitrotyrosine localization in cardiac cells followed a comparable pattern. In spite of the difference in the number of nitrotyrosine-positive cells with diabetes and with diabetes and hypertension, apoptosis and necrosis of myocytes, endothelial cells, and fibroblasts were detected only in cells containing this modified amino acid. In conclusion, local increases in Ang II with diabetes and with diabetes and hypertension may enhance oxidative damage, activating cardiac cell apoptosis and necrosis.


Subject(s)
Apoptosis , Cardiomegaly/etiology , Diabetes Mellitus, Type 2/complications , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Angiotensin II/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Diabetes Mellitus, Type 2/metabolism , Female , Heart Failure/etiology , Humans , Hypertension/etiology , Male , Middle Aged , Oxidative Stress , Reactive Oxygen Species/metabolism , Renin-Angiotensin System
12.
Eur J Cell Biol ; 77(2): 117-23, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9840461

ABSTRACT

We investigated the expression of the retinoblastoma protein (pRB) in adipocytes and its possible interaction with the adipogenic transcription factor CCAAT/enhancer-binding protein alpha (C/EBPalpha) in controlling the acquisition of the terminally differentiated adipocyte phenotype. The pRB was expressed (as measured by immunoblotting and/or immunofluorescence) in mice brown and white adipose tissue and in cultured adipocytes that showed lipid accumulation and expressed specific differentiation markers such as aP2 (measured using a specific cDNA probe) and in the case of brown adipocytes UCP-1 (measured using specific antibodies), but was undetectable in proliferative undifferentiated preadipocytes. Transient transfection experiments revealed a functional interaction between pRB and C/EBPalpha affecting transcription from the ucp-1 gene promoter. Thus, in immortalized brown adipocytes, co-transfection of both a C/EBPalpha and a pRB expression vectors maximally enhanced the expression of reporter chloramphenicol acetyltransferase driven by the ucp-1 promoter. Interestingly, C/EBPalpha inhibited reporter gene expression in CHO cells in an effect that was also potentiated in the presence of pRB. A positive effect of pRB on transcription from the ucp-1 promoter could be detected in C/EBPalpha-/-fibroblasts only after forced to overexpress C/EBPalpha, suggesting that the effect of pRB is dependent on its interaction with C/EBPalpha. We also found evidence that pRB and C/EBPalpha can directly bind to each other in vitro. Our results show that the expression of pRB is restricted to differentiated adipocytes, and provide evidence of a physical and functional interaction between pRB and C/EBPalpha that affects the transcriptional activity of the later on a brown adipocyte-specific gene.


Subject(s)
Adipocytes/cytology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , CHO Cells , Cell Differentiation , Cricetinae , DNA-Binding Proteins/genetics , Humans , Mice , Nuclear Proteins/genetics , Rats , Retinoblastoma Protein/genetics , Transcription Factors/genetics
13.
J Cell Sci ; 111 ( Pt 8): 1081-93, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9512504

ABSTRACT

Terminally differentiated skeletal muscle myotubes are arrested in the G0 phase of the cell cycle, and this arrest is not reversed by stimulation with serum or growth factors. The myotubes have been shown to be refractory to apoptosis even under low serum conditions. When the SV40 large T antigen is induced in the C2SVTts11 myotubes, which stably harbor the T antigen gene linked to an inducible promoter, the terminally differentiated cells reenter the cell cycle to resume nuclear DNA replication representing S phase. We show here that the large T-expressing myotubes further proceeded to M phase represented by the appearance of mitotic figures with centrosomes, condensed chromosomes, and mitotic spindles. The myotubes eventually cleaved and midbodies were formed at the cleavage sites of the cytoplasm. In some cases actin filaments, reminiscent of the contractile rings, accumulated at the cleavage furrows. Thus, terminally differentiated myotubes remain able to resume at least one round of the cell cycle and consequently are considered to be capable of dedifferentiation. A subset of myotubes expressing large T did not undergo mitosis. Some of them were degenerative and contained deformed giant nuclei and pulverized nuclei. The others suffered apoptotic cell death, which was identified by morphological changes of the nuclei and the labeling with dUTP at the ends of chromatin DNA fragments. The induction of apoptosis was unlikely to be confined to a particular phase of the cell cycle. These results imply that terminally differentiated myotubes also retain a complete set of machinery for apoptosis.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Apoptosis/physiology , Mitosis/physiology , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Animals , Biotin , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , DNA Fragmentation , Deoxyuracil Nucleotides , Fluorescent Antibody Technique , Gene Expression Regulation, Viral/physiology , Mice , Muscle Development , Muscle, Skeletal/growth & development , Staining and Labeling , Transfection
14.
J Cell Biol ; 139(3): 579-87, 1997 Nov 03.
Article in English | MEDLINE | ID: mdl-9348276

ABSTRACT

The mammalian nucleus is highly organized, and nuclear processes such as DNA replication occur in discrete nuclear foci, a phenomenon often termed "functional organization" of the nucleus. We describe the identification and characterization of a bipartite targeting sequence (amino acids 1-28 and 111-179) that is necessary and sufficient to direct DNA ligase I to nuclear replication foci during S phase. This targeting sequence is located within the regulatory, NH2-terminal domain of the protein and is dispensable for enzyme activity in vitro but is required in vivo. The targeting domain functions position independently at either the NH2 or the COOH termini of heterologous proteins. We used the targeting sequence of DNA ligase I to visualize replication foci in vivo. Chimeric proteins with DNA ligase I and the green fluorescent protein localized at replication foci in living mammalian cells and thus show that these subnuclear functional domains, previously observed in fixed cells, exist in vivo. The characteristic redistribution of these chimeric proteins makes them unique markers for cell cycle studies to directly monitor entry into S phase in living cells.


Subject(s)
DNA Ligases/genetics , DNA Ligases/metabolism , DNA Replication , Peptide Mapping , Amino Acid Sequence , Animals , COS Cells , Cell Cycle , Cell Nucleus/metabolism , DNA Ligase ATP , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Deletion
15.
J Clin Invest ; 100(5): 1294-304, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9276748

ABSTRACT

To identify the cis-acting regulatory element(s) which control the induction of the atrial natriuretic factor (ANF) gene in acute pressure overload, DNA constructs consisting of promoter elements linked to a reporter gene were injected into the myocardium of dogs, which underwent aortic banding or were sham-operated. Expression of a reporter gene construct harboring the ANF promoter (-3400ANF) was induced 6-12-fold after 7 d of pressure overload. An internal deletion of 556 bp (nucleotide sequence -693 to -137) completely abrogated the inducibility of the ANF reporter gene construct. An activator protein-1 (AP1)-like site (-496 to -489) and a cAMP regulatory element (CRE) (-602 to -596) are located within the deleted sequence. Site-directed mutagenesis of the AP1-like site but not the CRE completely prevented the induction of this construct to acute pressure overload. Further, the AP1-like site was able to confer inducibility of a heterologous promoter (beta-myosin heavy chain) to higher values than controls. Gel mobility shift assay (GMSA) supershift analysis was performed using a radiolabeled probe of the ANF promoter (-506/-483) that included the AP1-like site (ATGAATCA) sequence, as well as a probe converted to contain an AP1 consensus sequence (ATGACTCA). GMSA analysis demonstrated that the ANF AP1-like element could bind both a constitutively expressed factor and the AP1 proteins, and conversion to a true AP1 site increased its affinity for AP1. However, 7 d after the onset of pressure overload, the AP1 proteins were present only at low levels, and the major complex formed by the ANF AP1-like probe was not supershifted by a jun antibody. Using a large animal model of pressure overload, we have demonstrated that a unique cis-acting element was primarily responsible for the overload induction of the ANF gene.


Subject(s)
Atrial Natriuretic Factor/genetics , Cardiomegaly/genetics , Gene Expression Regulation , Genes, Regulator , Animals , DNA-Binding Proteins/metabolism , Dogs , Promoter Regions, Genetic , Receptors, Atrial Natriuretic Factor/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation
16.
Mol Cell Biol ; 17(5): 2745-55, 1997 May.
Article in English | MEDLINE | ID: mdl-9111345

ABSTRACT

The muscle-specific regulatory region of the alpha-cardiac myosin heavy-chain (MHC) gene contains the thyroid hormone response element (TRE) and two A/T-rich DNA sequences, designated A/T1 and A/T2, the putative myocyte-specific enhancer factor 2 (MEF2) binding sites. We investigated the roles of the TRE and MEF2 binding sites and the potential interaction between thyroid hormone receptor (TR) and MEF2 proteins regulating the alpha-MHC promoter. Deletion mutation analysis indicated that both the A/T2 motif and TRE were required for muscle-specific expression of the alpha-MHC gene. The alpha-MHC enhancer containing both the A/T2 motif and TRE was synergistically activated by coexpression of MEF2 and TR in nonmuscle cells, whereas neither factor by itself activated the alpha-MHC reporters. The reporter construct containing the A/T2 sequence and the TRE linked to a heterologous promoter also showed synergistic activation by coexpression of MEF2 and TR in nonmuscle cells. Moreover, protein binding assays demonstrated that MEF2 and TR specifically bound to one another in vitro and in vivo. The MADS domain of MEF2 and the DNA-binding domain of TR were necessary and sufficient to mediate their physical interaction. Our results suggest that the members of the MADS family (MEF2) and steroid receptor superfamily (TR) interact with one another to synergistically activate the alpha-cardiac MHC gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Myosin Heavy Chains/genetics , Receptors, Thyroid Hormone/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , DNA/metabolism , Drug Synergism , Homeodomain Proteins/metabolism , MEF2 Transcription Factors , Myogenic Regulatory Factors , Regulatory Sequences, Nucleic Acid
17.
J Cell Biochem ; 62(3): 405-10, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8872611

ABSTRACT

Cellular transcription factor E2F1 is thought to regulate the expression of genes important for cell cycle progression and cell proliferation. Deregulated E2F1 expression induces S-phase entry in quiescent cells and inhibits myogenic differentiation. We show here that E2F1 inhibits the activation of gene transcription by myogenic basic helix-loop-helix proteins myoD and myogenin. Transfection assay using different deletion constructs indicates that both the DNA binding and the transactivation domains of E2F1 are required for its inhibition of myoD transcription activation. However, the retinoblastoma protein (RB) binding domain is not required. Furthermore, co-transfection with the RB, which inhibits the transcription activity of E2F1, can also repress E2F1 inhibition of myoD transactivation. These results suggest an essential role of E2F1-mediated transcription in its inhibition of myogenesis.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , MyoD Protein/genetics , Myogenin/genetics , Transcription Factors/genetics , Transcription Factors/pharmacology , Transcription, Genetic , Animals , Binding Sites , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Creatine Kinase/drug effects , Creatine Kinase/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Helix-Loop-Helix Motifs/drug effects , Helix-Loop-Helix Motifs/genetics , Mice , Muscles/cytology , MyoD Protein/drug effects , Myogenin/drug effects , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/pharmacology , Retinoblastoma-Binding Protein 1 , Sequence Deletion , Transcription Factor DP1 , Transcription Factors/metabolism
18.
J Mol Cell Cardiol ; 28(7): 1453-61, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8841933

ABSTRACT

Regulation of ionic currents in the heart is partly achieved by signaling cascades which alter intracellular levels of cyclic nucleotides. Changes in cyclic nucleotide levels can regulate channels either directly, like the direct binding of cAMP to the i(f) channel in pacemaker tissues, or indirectly through phosphorylation of channels by cAMP-dependent, or cGMP-dependent protein kinases. These types of regulation generally alter the voltage sensitivities of channels. A class of voltage-insensitive channels, first discovered in retinal rods and olfactory neurons, were recently identified in the heart. These channels are opened by the direct binding of cyclic nucleotides, providing a means of regulating ionic currents outside the influence of membrane voltage. Since different isoforms have different affinities for cAMP and cGMP, it is important to determine which isoforms are expressed in heart in order to predict their roles in heart function. We have cloned the olfactory channel from mouse heart, and find that although the message is very rare, Western blot analysis indicates the olfactory channel protein is stable in heart sarcolemma. Our data also suggest the olfactory channel protein forms homomeric channels in the heart since other isoforms or splice variants were not detected either by PCR amplification or by RNase protection. In addition, we have isolated and sequenced the mouse olfactory cyclic nucleotide-gated channel gene, and show the genomic organization is remarkably similar to that found in the human retinal channel gene. Part of this work was presented in abstract form.


Subject(s)
Ion Channels/metabolism , Myocardium/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Epithelium/metabolism , Heart/physiology , Immunoblotting , Ion Channels/genetics , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Rabbits , Sequence Homology, Amino Acid
19.
Mech Dev ; 57(1): 103-12, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8817457

ABSTRACT

MEF2 proteins are a family of transcription factors that have muscle-specific DNA binding activity and bind to conserved A/T rich elements in the regulatory regions of numerous muscle-specific genes. They are thought have an important role in the development and differentiation of skeletal muscle. Recent in situ hybridization studies using mouse MEF2 probes have shown that MEF2 gene transcripts are detected very early in development at high levels in the myogenic cells of the myotome and embryonic heart. However, MEF2A and MEF2D transcripts have been detected in many adult tissues where they are not translated or the corresponding proteins are rapidly degraded. Therefore, it is important to establish the temporal and spatial correlation between MEF2 RNA and protein expression. In the present study we have performed in situ immunohistochemistry of whole mount mouse embryos at different stages of development using polyclonal antibodies specific for the MEF2A, MEF2C and MEF2D isoforms. At day 8.5 of development, all three MEF2 isoforms are expressed in the heart. MEF2A and C are detected in a few cells in the rostral-most somite by day 9 of development. Their expression then proceeds in a rostro-caudal direction concurrent with somite maturation. The pattern of expression of the MEF2D isoform is similar to that of MEF2C but the amount detected is much lower. Interestingly, MEF2A is also detected as early as day 8.5 p.c. in cells of the embryonic vasculature and non-myogenic cells. These results demonstrate that MEF2 proteins are detected early in development in the somites and heart, thus supporting their importance in the early stages of the hierarchical cascade of myogenesis. Their presence in non-muscle cells further suggests they could also play a role in the determination of other mesodermal derivatives, including cells of the vasculature.


Subject(s)
DNA-Binding Proteins/biosynthesis , Heart/embryology , Muscles/embryology , Transcription Factors/biosynthesis , Age Factors , Animals , Cell Lineage , DNA-Binding Proteins/immunology , Heart/physiology , Immunohistochemistry , MEF2 Transcription Factors , Mice , Muscles/cytology , Muscles/physiology , Myogenic Regulatory Factors , Somites/metabolism , Tissue Distribution , Transcription Factors/immunology
20.
J Biol Chem ; 271(24): 14371-5, 1996 Jun 14.
Article in English | MEDLINE | ID: mdl-8662987

ABSTRACT

The mammalian achaete-scute homolog 1 (MASH1) protein is required for the early development of the nervous system. However, the molecular and biochemical mechanism by which MASH1 acts to determine neurogenesis are still unknown. The myocyte enhancer factor 2A (MEF2A) is a MADS transcription factor that is essential for the specification and differentiation of the muscle lineage. Here we show that MEF2A and MASH1 are coordinately induced during the differentiation of the teratocarcinoma cell line P19 along a neuronal lineage and that in transient transfection assays, MEF2A and MASH1 cooperatively activate gene expression. This cooperativity appears to be due to a specific physical interaction between MEF2A and MASH1. Taken together, these findings suggest that MASH1 via a cooperative interaction with MEF2A may regulate the expression of specific genes that are critical for neuronal differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Binding Sites , Cell Differentiation/drug effects , Cell Line , Chloramphenicol O-Acetyltransferase/biosynthesis , Chlorocebus aethiops , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/isolation & purification , MEF2 Transcription Factors , Mammals , Mice , Mutagenesis , Myogenic Regulatory Factors , Neurons/cytology , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Deletion , Teratocarcinoma , Transcription Factors/biosynthesis , Transcription Factors/isolation & purification , Transcriptional Activation , Transfection , Tretinoin/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...