Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 9(1): 3699, 2018 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-30194428

RESUMO

This article contains errors in Figs. 5 and 6, for which we apologize. In Fig. 5f, the image 'E12.5 tail' was inadvertently replaced with a duplicate of the image 'E12.5 trunk' from the same panel. In Figure 6d, the image 'E9.5/OH-TAM E8.5, embryo' was inadvertently replaced with a duplicate of the image 'E10.5/ OH-TAM E8.5, embryo' from Fig. 6b. The corrected versions of these figures appear in the Author Correction associated with this Article.

2.
Nat Commun ; 9(1): 75, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311541

RESUMO

Tissue macrophages in many adult organs originate from yolk sac (YS) progenitors, which invade the developing embryo and persist by means of local self-renewal. However, the route and characteristics of YS macrophage trafficking during embryogenesis are incompletely understood. Here we show the early migration dynamics of YS-derived macrophage progenitors in vivo using fate mapping and intravital microscopy. From embryonic day 8.5 (E8.5) CX3CR1+ pre-macrophages are present in the mouse YS where they rapidly proliferate and gain access to the bloodstream to migrate towards the embryo. Trafficking of pre-macrophages and their progenitors from the YS to tissues peaks around E10.5, dramatically decreases towards E12.5 and is no longer evident from E14.5 onwards. Thus, YS progenitors use the vascular system during a restricted time window of embryogenesis to invade the growing fetus. These findings close an important gap in our understanding of the development of the innate immune system.


Assuntos
Movimento Celular , Células-Tronco Embrionárias/citologia , Macrófagos/citologia , Saco Vitelino/citologia , Animais , Circulação Sanguínea , Linhagem da Célula , Proliferação de Células , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Células-Tronco Hematopoéticas/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Fatores de Tempo , Saco Vitelino/embriologia
3.
Cell Death Differ ; 22(9): 1402-12, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26088163

RESUMO

Muscular dystrophy (MD) refers to a clinically and genetically heterogeneous group of degenerative muscle disorders characterized by progressive muscle wasting and often premature death. Although the primary defect underlying most forms of MD typically results from a loss of sarcolemmal integrity, the secondary molecular mechanisms leading to muscle degeneration and myofiber necrosis is debated. One hypothesis suggests that elevated or dysregulated cytosolic calcium is the common transducing event, resulting in myofiber necrosis in MD. Previous measurements of resting calcium levels in myofibers from dystrophic animal models or humans produced equivocal results. However, recent studies in genetically altered mouse models have largely solidified the calcium hypothesis of MD, such that models with artificially elevated calcium in skeletal muscle manifest fulminant dystrophic-like disease, whereas models with enhanced calcium clearance or inhibited calcium influx are resistant to myofiber death and MD. Here, we will review the field and the recent cadre of data from genetically altered mouse models, which we propose have collectively mostly proven the hypothesis that calcium is the primary effector of myofiber necrosis in MD. This new consensus on calcium should guide future selection of drugs to be evaluated in clinical trials as well as gene therapy-based approaches.


Assuntos
Cálcio/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Distrofias Musculares/patologia
4.
Arch Biochem Biophys ; 579: 40-6, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26032335

RESUMO

The mitochondrial peptidyl prolyl isomerase cyclophilin D (CypD) activates permeability transition (PT). To study the role of CypD in this process we compared the functions of brain mitochondria isolated from wild type (BMWT) and CypD knockout (Ppif(-/-)) mice (BMKO) with and without CypD inhibitor Cyclosporin A (CsA) under normal and Ca(2+) stress conditions. Our data demonstrate that BMKO are characterized by higher rates of glutamate/malate-dependent oxidative phosphorylation, higher membrane potential and higher resistance to detrimental Ca(2+) effects than BMWT. Under the elevated Ca(2+) and correspondingly decreased membrane potential the dose response in BMKO shifts to higher Ca(2+) concentrations as compared to BMWT. However, significantly high Ca(2+) levels result in complete loss of membrane potential in BMKO, too. CsA diminishes the loss of membrane potential in BMWT but has no protecting effect in BMKO. The results are in line with the assumption that PT is regulated by CypD under the control of matrix Ca(2+). Due to missing of CypD the BMKO can favor PT only at high Ca(2+) concentrations. It is concluded that CypD sensitizes the brain mitochondria to PT, and its inhibition by CsA or CypD absence improves the complex I-related mitochondrial function and increases mitochondria stability against Ca(2+) stress.


Assuntos
Encéfalo/fisiologia , Cálcio/metabolismo , Ciclofilinas/metabolismo , Mitocôndrias/fisiologia , Estresse Oxidativo/fisiologia , Oxigênio/metabolismo , Animais , Respiração Celular/fisiologia , Células Cultivadas , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Complexo I de Transporte de Elétrons/metabolismo , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
5.
J Thromb Haemost ; 12(12): 2089-101, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25330904

RESUMO

BACKGROUND: Calcium-dependent signaling mechanisms play a critical role in platelet activation. Unlike calcium-activated protease and kinase, the contribution of calcium-activated protein serine/threonine phosphatase in platelet activation is poorly understood. OBJECTIVE: To assess the role of catalytic subunit of protein phosphatase 2B (PP2B) or calcineurin in platelet function. RESULTS: Here, we showed that an increase in PP2B activity was associated with agonist-induced activation of human and murine platelets. Pharmacological inhibitors of the catalytic subunit of protein phosphatase 2B (PP2B-A) such as cyclosporine A or tacrolimus (FK506) potentiated aggregation of human platelets. Murine platelets lacking the ß isoform of PP2B-A (PP2B-Aß(-/-) ) displayed increased aggregation with low doses of agonist concentrations. Loss of PP2B-Aß did not affect agonist-induced integrin αII b ß3 inside-out signaling, but increased basal Src activation and outside-in αII b ß3 signaling to p38 mitogen-activated protein kinase (MAPK), with a concomitant enhancement in platelet spreading on immobilized fibrinogen and greater fibrin clot retraction. Fibrinogen-induced increased p38 activation in PP2B-Aß(-/-) platelets were blocked by Src inhibitor. Both PP2B-Aß(-/-) platelets and PP2B-Aß-depleted human embryonal kidney 293 αII b ß3 cells displayed increased adhesion to immobilized fibrinogen. Filamin A, an actin crosslinking phosphoprotein that is known to associate with ß3 , was dephosphorylated on Ser(2152) in fibrinogen-adhered wild-type but not in PP2B-Aß(-/-) platelets. In a FeCl3 injury thrombosis model, PP2B-Aß(-/-) mice showed decreased time to occlusion in the carotid artery. CONCLUSION: These observations indicate that PP2B-Aß by suppressing outside-in αII b ß3 integrin signaling limits platelet response to vascular injury.


Assuntos
Plaquetas/citologia , Calcineurina/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Animais , Artérias Carótidas/metabolismo , Domínio Catalítico , Adesão Celular , Cloretos/química , Feminino , Compostos Férricos/química , Fibrinogênio/metabolismo , Citometria de Fluxo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Fosforilação , Agregação Plaquetária , Isoformas de Proteínas , Transdução de Sinais , Trombose/fisiopatologia
6.
Cell Death Differ ; 21(8): 1209-17, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24658400

RESUMO

The mitochondrial phosphate carrier (PiC) is critical for ATP synthesis by serving as the primary means for mitochondrial phosphate import across the inner membrane. In addition to its role in energy production, PiC is hypothesized to have a role in cell death as either a component or a regulator of the mitochondrial permeability transition pore (MPTP) complex. Here, we have generated a mouse model with inducible and cardiac-specific deletion of the Slc25a3 gene (PiC protein). Loss of PiC protein did not prevent MPTP opening, suggesting it is not a direct pore-forming component of this complex. However, Slc25a3 deletion in the heart blunted MPTP opening in response to Ca(2+) challenge and led to a greater Ca(2+) uptake capacity. This desensitization of MPTP opening due to loss or reduction in PiC protein attenuated cardiac ischemic-reperfusion injury, as well as partially protected cells in culture from Ca(2+) overload induced death. Intriguingly, deletion of the Slc25a3 gene from the heart long-term resulted in profound hypertrophy with ventricular dilation and depressed cardiac function, all features that reflect the cardiomyopathy observed in humans with mutations in SLC25A3. Together, these results demonstrate that although the PiC is not a direct component of the MPTP, it can regulate its activity, suggesting a novel therapeutic target for reducing necrotic cell death. In addition, mice lacking Slc25a3 in the heart serve as a novel model of metabolic, mitochondrial-driven cardiomyopathy.


Assuntos
Cardiomiopatias/genética , Mitocôndrias Cardíacas/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Simportadores de Próton-Fosfato/genética , Animais , Cálcio/metabolismo , Cardiomiopatias/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Poro de Transição de Permeabilidade Mitocondrial , Estresse Oxidativo/genética , Simportadores de Próton-Fosfato/deficiência
7.
Mol Metab ; 3(2): 124-34, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24634818

RESUMO

Insulin resistance is associated with mitochondrial dysfunction, but the mechanism by which mitochondria inhibit insulin-stimulated glucose uptake into the cytoplasm is unclear. The mitochondrial permeability transition pore (mPTP) is a protein complex that facilitates the exchange of molecules between the mitochondrial matrix and cytoplasm, and opening of the mPTP occurs in response to physiological stressors that are associated with insulin resistance. In this study, we investigated whether mPTP opening provides a link between mitochondrial dysfunction and insulin resistance by inhibiting the mPTP gatekeeper protein cyclophilin D (CypD) in vivo and in vitro. Mice lacking CypD were protected from high fat diet-induced glucose intolerance due to increased glucose uptake in skeletal muscle. The mitochondria in CypD knockout muscle were resistant to diet-induced swelling and had improved calcium retention capacity compared to controls; however, no changes were observed in muscle oxidative damage, insulin signaling, lipotoxic lipid accumulation or mitochondrial bioenergetics. In vitro, we tested 4 models of insulin resistance that are linked to mitochondrial dysfunction in cultured skeletal muscle cells including antimycin A, C2-ceramide, ferutinin, and palmitate. In all models, we observed that pharmacological inhibition of mPTP opening with the CypD inhibitor cyclosporin A was sufficient to prevent insulin resistance at the level of insulin-stimulated GLUT4 translocation to the plasma membrane. The protective effects of mPTP inhibition on insulin sensitivity were associated with improved mitochondrial calcium retention capacity but did not involve changes in insulin signaling both in vitro and in vivo. In sum, these data place the mPTP at a critical intersection between alterations in mitochondrial function and insulin resistance in skeletal muscle.

8.
Clin Exp Immunol ; 158(3): 317-24, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19747209

RESUMO

Transforming growth factor (TGF) beta1) is an immunoregulatory cytokine involved in self-tolerance and lymphocyte homeostasis. Tgfb1 knock-out (KO) mice develop severe multi-focal autoimmune inflammatory lesions due to [Ca(2+)]i deregulation in T cells, and die within 3 weeks after birth. Because the calcineurin inhibitor FK506 inhibits the hyperresponsiveness of Tgfb1(-/-) thymocytes, and because calcineurin Abeta (CNAbeta)-deficient mice do not reject allogenic tumours, we have generated Tgfb1(-/-) Cnab(-/-) mice to address whether CNAbeta deficiency prevents T cell activation and inflammation in Tgfb1(-/-) mice. Here we show that in Tgfb1(-/-) Cnab(-/-) mice inflammation is reduced significantly relative to that in Tgfb1(-/-) mice. However, both CD4(+) and CD8(+) T cells in double knock-out (DKO) mice are activated, as revealed by up-regulation of CD11a lymphocyte function-associated antigen-1 (LFA-1), CD44 and CD69 and down-regulation of CD62L. These data suggest that deficiency of CNAbeta decreases inflammatory lesions but does not prevent activation of autoreactive T cells. Also Tgfb1(-/-) T cells can undergo activation in the absence of CNAbeta, probably by using the other isoform of calcineurin (CNAalpha) in a compensatory manner. CNAbeta-deficient T cells undergo spontaneous activation in vivo and are activated upon anti-T cell receptor stimulation in vitro. Understanding the role of calcineurin in T cell regulation should open up new therapeutic opportunities for inflammation and cancer.


Assuntos
Doenças Autoimunes/imunologia , Calcineurina/deficiência , Inflamação/imunologia , Fator de Crescimento Transformador beta1/imunologia , Animais , Doenças Autoimunes/patologia , Doenças Autoimunes/prevenção & controle , Calcineurina/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Inflamação/patologia , Inflamação/prevenção & controle , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Camundongos , Camundongos Knockout , Baço/imunologia , Análise de Sobrevida , Subpopulações de Linfócitos T/imunologia
9.
Mucosal Immunol ; 1(4): 289-96, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19079190

RESUMO

Periostin is an extracellular matrix protein that has been primarily studied in the context of the heart, where it has been shown to promote cardiac repair and remodeling. In this study, we focused on the role of periostin in an allergic eosinophilic inflammatory disease (eosinophilic esophagitis (EE)) known to involve extensive tissue remodeling. Periostin was indeed markedly overexpressed (35-fold) in the esophagus of EE patients, particularly in the papillae, compared with control individuals. Periostin expression was downstream from transforming growth factor-beta and interleukin-13, as these cytokines were elevated in EE esophageal samples and markedly induced periostin production by primary esophageal fibroblasts (107- and 295-fold, respectively, at 10 ng ml(-1)). A functional role for periostin in eliciting esophageal eosinophilia was demonstrated, as periostin-null mice had a specific defect in allergen-induced eosinophil recruitment to the lungs and esophagus (66 and 72% decrease, respectively). Mechanistic analyses revealed that periostin increased (5.8-fold) eosinophil adhesion to fibronectin. As such, these findings extend the involvement of periostin to esophagitis and uncover a novel role for periostin in directly regulating leukocyte (eosinophil) accumulation in T helper type 2-associated mucosal inflammation in both mice and humans.


Assuntos
Moléculas de Adesão Celular/fisiologia , Eosinófilos/fisiologia , Esofagite/imunologia , Hipersensibilidade/imunologia , Eosinofilia Pulmonar/imunologia , Animais , Asma/imunologia , Asma/patologia , Adesão Celular/fisiologia , Moléculas de Adesão Celular/genética , Dermatite Atópica/imunologia , Dermatite Atópica/patologia , Eosinófilos/imunologia , Esofagite/patologia , Esôfago/metabolismo , Esôfago/patologia , Fibroblastos/fisiologia , Humanos , Hipersensibilidade/patologia , Interleucina-13/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Eosinofilia Pulmonar/patologia , Rinite/imunologia , Rinite/patologia , Fator de Crescimento Transformador beta/imunologia
10.
Am J Physiol Heart Circ Physiol ; 284(2): H425-30, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12388248

RESUMO

Echocardiograms have been assessed only at 56 days in mice overexpressing calcineurin (CN mice). Age-dependent echocardiographic changes were evaluated because the development of sudden death is time dependent. Because cyclosporin A (CsA) reverses hypertrophy in CN mice, its effects on the time course of the development of sudden death and cardiac dysfunction were assessed. In wild-type (WT) mice, the left ventricular (LV) internal end-diastolic dimension (LVIDd) increased and the LV mass index (LVMI) decreased with age. In CN mice, two distinct phases of pathophysiology were found. After 14 days, in CN mice, the LVIDd and LVMI were significantly increased, but sudden death had not occurred. After 28 days, in CN mice, relative dilation of the left ventricle occurred, whereas the LVMI decreased. Sudden death developed during progressive dilation associated with systolic and diastolic dysfunction. CsA treatment reversed hypertrophy in CN mice but did not reverse systolic and diastolic dysfunction and exaggerated sudden death. Sudden cardiac death was associated with systolic and diastolic dysfunction but was not related to isolated cardiac hypertrophy in CN mice.


Assuntos
Calcineurina/metabolismo , Coração/fisiopatologia , Animais , Cardiomegalia/complicações , Cardiomegalia/diagnóstico por imagem , Ciclosporina/farmacologia , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/prevenção & controle , Diástole , Progressão da Doença , Ecocardiografia , Coração/efeitos dos fármacos , Ventrículos do Coração , Camundongos , Camundongos Transgênicos , Análise de Sobrevida , Sístole , Fatores de Tempo
11.
Mol Cell Biol ; 21(21): 7460-9, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11585926

RESUMO

The zinc finger-containing transcription factor GATA4 has been implicated as a critical regulator of multiple cardiac-expressed genes as well as a regulator of inducible gene expression in response to hypertrophic stimulation. Here we demonstrate that GATA4 is itself regulated by the mitogen-activated protein kinase signaling cascade through direct phosphorylation. Site-directed mutagenesis and phospho-specific GATA4 antiserum revealed serine 105 as the primary site involved in agonist-induced phosphorylation of GATA4. Infection of cultured cardiomyocytes with an activated MEK1-expressing adenovirus induced robust phosphorylation of serine 105 in GATA4, while a dominant-negative MEK1-expressing adenovirus blocked agonist-induced phosphorylation of serine 105, implicating extracellular signal-regulated kinase (ERK) as a GATA4 kinase. Indeed, bacterially purified ERK2 protein directly phosphorylated purified GATA4 at serine 105 in vitro. Phosphorylation of serine 105 enhanced the transcriptional potency of GATA4, which was sensitive to U0126 (MEK1 inhibitor) but not SB202190 (p38 inhibitor). Phosphorylation of serine 105 also modestly enhanced the DNA binding activity of bacterially purified GATA4. Finally, induction of cardiomyocyte hypertrophy with an activated MEK1-expressing adenovirus was blocked with a dominant-negative GATA4-engrailed-expressing adenovirus. These results suggest a molecular pathway whereby MEK1-ERK1/2 signaling regulates cardiomyocyte hypertrophic growth through the transcription factor GATA4 by direct phosphorylation of serine 105, which enhances DNA binding and transcriptional activation.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/citologia , Serina/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Western Blotting , Butadienos/farmacologia , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Fator de Transcrição GATA4 , Genes Dominantes , Glutationa Transferase/metabolismo , Imidazóis/farmacologia , Imuno-Histoquímica , Leucina/metabolismo , Luciferases/metabolismo , MAP Quinase Quinase 1 , Sistema de Sinalização das MAP Quinases , Proteína Quinase 3 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Nitrilas/farmacologia , Fosforilação , Plasmídeos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Ativação Transcricional , Dedos de Zinco
12.
Genes Dev ; 15(20): 2702-19, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11641276

RESUMO

Rho-like GTPases play a pivotal role in the orchestration of changes in the actin cytoskeleton in response to receptor stimulation, and have been implicated in transcriptional activation, cell growth regulation, and oncogenic transformation. Recently, a role for RhoA in the regulation of cardiac contractility and hypertrophic cardiomyocyte growth has been suggested but the mechanisms underlying RhoA function in the heart remain undefined. We now report that transcription factor GATA-4, a key regulator of cardiac genes, is a nuclear mediator of RhoA signaling and is involved in the control of sarcomere assembly in cardiomyocytes. Both RhoA and GATA-4 are essential for sarcomeric reorganization in response to hypertrophic growth stimuli and overexpression of either protein is sufficient to induce sarcomeric reorganization. Consistent with convergence of RhoA and GATA signaling, RhoA potentiates the transcriptional activity of GATA-4 via a p38 MAPK-dependent pathway that phosphorylates GATA-4 activation domains and GATA binding sites mediate RhoA activation of target cardiac promoters. Moreover, a dominant-negative GATA-4 protein abolishes RhoA-induced sarcomere reorganization. The identification of transcription factor GATA-4 as a RhoA mediator in sarcomere reorganization and cardiac gene regulation provides a link between RhoA effects on transcription and cell remodeling.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Miocárdio/metabolismo , Fatores de Transcrição/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Adenoviridae/genética , Northern Blotting , Western Blotting , Células Cultivadas , DNA/genética , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Endotelina-1/farmacologia , Elementos Facilitadores Genéticos , Imunofluorescência , Fator de Transcrição GATA4 , Regulação da Expressão Gênica , Humanos , Hibridização In Situ , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfoproteínas Fosfatases/farmacologia , Regiões Promotoras Genéticas , RNA/metabolismo , Sarcômeros/fisiologia , Fatores de Transcrição/genética , Proteínas Quinases p38 Ativadas por Mitógeno , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Circ Res ; 89(1): 20-5, 2001 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-11440973

RESUMO

The advent of conditional and tissue-specific recombination systems in gene-targeted or transgenic mice has permitted an assessment of single gene function in a temporally regulated and cell-specific manner. Here we generated transgenic mice expressing a tamoxifen-inducible Cre recombinase protein fused to two mutant estrogen-receptor ligand-binding domains (MerCreMer) under the control of the alpha-myosin heavy chain promoter. These transgenic mice were crossed with the ROSA26 lacZ-flox-targeted mice to examine Cre recombinase activity and the fidelity of the system. The data demonstrate essentially no Cre-mediated recombination in the embryonic, neonatal, or adult heart in the absence of inducing agent but >80% recombination after only four tamoxifen injections. Expression of the MerCreMer fusion protein within the adult heart did not affect cardiac performance, cellular architecture, or expression of hypertrophic marker genes, demonstrating that the transgene-encoded protein is relatively innocuous. In summary, MerCreMer transgenic mice represent a tool for temporally regulated inactivation of any loxP-targeted gene within the developing and adult heart or for specifically directing recombination and expression of a loxP-inactivated cardiac transgene in the heart.


Assuntos
Coração/embriologia , Integrases/genética , Miocárdio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Proteínas Virais/genética , Animais , Regulação da Expressão Gênica , Integrases/metabolismo , Cinética , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , RNA Mensageiro/biossíntese , Receptores de Estrogênio/genética , Proteínas Recombinantes de Fusão , Recombinação Genética , Transgenes , Proteínas Virais/metabolismo
14.
Physiol Genomics ; 6(1): 19-28, 2001 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-11395543

RESUMO

To define molecular mechanisms of cardiac hypertrophy, genes whose expression was perturbed by any of four different transgenic mouse hypertrophy models [protein kinase C-epsilon activation peptide (PsiepsilonRACK), calsequestrin (CSQ), calcineurin (CN), and Galpha(q)] were compared by DNA microarray analyses using the approximately 8,800 genes present on the Incyte mouse GEM1. The total numbers of regulated genes (tens to hundreds) correlated with phenotypic severity of the model (Galpha(q) > CN > CSQ > PsiepsilonRACK), but demonstrated that no single gene was consistently upregulated. Of the three models exhibiting pathological hypertrophy, only atrial natriuretic peptide was consistently upregulated, suggesting that transcriptional alterations are highly specific to individual genetic causes of hypertrophy. However, hierarchical-tree and K-means clustering analyses revealed that subsets of the upregulated genes did exhibit coordinate regulatory patterns that were unique or overlapping across the different hypertrophy models. One striking set consisted of apoptotic genes uniquely regulated in the apoptosis-prone Galpha(q) model. Thus, rather than identifying a single common hypertrophic cardiomyopathy gene program, these data suggest that extensive groups of genes may be useful for the prediction of specific underlying genetic determinants and condition-specific therapeutic approaches.


Assuntos
Cardiomegalia/genética , Animais , Apoptose/genética , Calcineurina/genética , Calsequestrina/genética , Cardiomegalia/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Perfilação da Expressão Gênica , Proteínas Heterotriméricas de Ligação ao GTP/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , RNA Mensageiro/biossíntese , Transcrição Gênica , Regulação para Cima
16.
Dev Biol ; 232(2): 351-61, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11401397

RESUMO

Retinoic acid (RA), a potent teratogen, produces a characteristic set of embryonic cardiovascular malformations similar to those observed in neural crest ablated avians. While the effects of RA on neural crest are well described, the molecular mechanism(s) of RA action on these cells is less clear. The present study examines the relationship between RA and mitogen-activated protein kinase signaling in neural crest cells and demonstrates that c-Jun N-terminal kinase (JNK) activation is severely repressed by RA. RA suppressed migration and proliferation of primary cultures of mouse neural crest cells treated in vitro as well as from animals treated in vivo. On Western blots, JNK activation/phosphorylation in neural crest cultures was reduced, while neither extracellular signal-regulated kinase (ERK) nor p38 pathways were affected. Both the dose-dependent stimulation of neural crest outgrowth and JNK phosphorylation by platelet-derived growth factor AA, which promotes outgrowth but not proliferation of neural crest cultures, were completely abrogated by RA. To establish the relevance of the JNK signaling pathway to cardiac neural crest migration, dominant negative adenoviral constructs were used to inhibit upstream activation of JNK or c-Jun downstream responses. Both adenoviral constructs markedly reduced neural crest cell outgrowth, while a dominant negative inhibitor of the p38 pathway had no effect. These data demonstrate that the JNK signaling pathway and c-Jun activation are critical for cardiac neural crest outgrowth and are potential targets for the action of RA.


Assuntos
Coração Fetal/efeitos dos fármacos , Coração Fetal/inervação , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Crista Neural/efeitos dos fármacos , Tretinoína/toxicidade , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Coração Fetal/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Crista Neural/citologia , Crista Neural/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais , Teratogênicos/toxicidade , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
17.
J Biol Chem ; 276(32): 30245-53, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11356841

RESUMO

The zinc finger-containing transcription factors GATA4 and GATA6 are important regulators of basal and inducible gene expression in cardiac and smooth muscle cell types. Here we demonstrate a direct functional role for GATA4 and GATA6 as regulators of cardiomyocyte hypertrophic growth and gene expression. To model the increase in endogenous GATA4 and GATA6 transcriptional activity that occurs in response to hypertrophic stimulation, each factor was overexpressed in cardiomyocytes using recombinant adenovirus. Overexpression of either GATA4 or GATA6 was sufficient to induce cardiomyocyte hypertrophy characterized by enhanced sarcomeric organization, a greater than 2-fold increase in cell surface area, and a significant increase in total protein accumulation. In vivo, transgenic mice with 2.5-fold overexpression of GATA4 within the adult heart demonstrated a slowly progressing increase in heart to body weight ratio, histological features of cardiomyopathy, and activation of hypertrophy-associated genes, suggesting that GATA factors are sufficient regulators of cardiomyocyte hypertrophy in vitro and in vivo. To evaluate the requirement of GATA factors as downstream transcriptional mediators of hypertrophy, a dominant negative GATA4-engrailed repressor fusion-encoding adenovirus was generated. Expression of GATA4-engrailed blocked GATA4- and GATA6-directed transcriptional responses and agonist-induced cardiomyocyte hypertrophy, demonstrating that cardiac-expressed GATA factors are necessary mediators of this process.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Miocárdio/metabolismo , Fatores de Transcrição/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , DNA/metabolismo , DNA Complementar/metabolismo , Embrião de Mamíferos/metabolismo , Fator de Transcrição GATA4 , Fator de Transcrição GATA6 , Técnicas de Transferência de Genes , Proteínas de Homeodomínio/metabolismo , Hipertrofia , Luciferases/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção
18.
Genes Dev ; 15(7): 839-44, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11297508

RESUMO

GATA-family transcription factors are critical to the development of diverse tissues. In particular, GATA-4 has been implicated in formation of the vertebrate heart. As the mouse Gata-4 knock-out is early embryonic lethal because of a defect in ventral morphogenesis, the in vivo function of this factor in heart development remains unresolved. To search for a requirement for Gata4 in heart development, we created mice harboring a single amino acid replacement in GATA-4 that impairs its physical interaction with its presumptive cardiac cofactor FOG-2. Gata4(ki/ki) mice die just after embryonic day (E) 12.5 exhibiting features in common with Fog2(-/-) embryos as well as additional semilunar cardiac valve defects and a double-outlet right ventricle. These findings establish an intrinsic requirement for GATA-4 in heart development. We also infer that GATA-4 function is dependent on interaction with FOG-2 and, very likely, an additional FOG protein for distinct aspects of heart formation.


Assuntos
Anomalias dos Vasos Coronários/genética , Vasos Coronários/embriologia , Proteínas de Ligação a DNA/fisiologia , Coração Fetal/crescimento & desenvolvimento , Cardiopatias Congênitas/genética , Fatores de Transcrição/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Anomalias dos Vasos Coronários/embriologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Fatores de Ligação de DNA Eritroide Específicos , Coração Fetal/patologia , Fator de Transcrição GATA4 , Genes Letais , Idade Gestacional , Cardiopatias Congênitas/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Morfogênese/genética , Mutagênese Sítio-Dirigida , Conformação Proteica , Fatores de Transcrição/biossíntese , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica , Valina/química , Proteínas de Peixe-Zebra
19.
J Biol Chem ; 276(19): 15913-9, 2001 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-11279073

RESUMO

Multiple intracellular signaling pathways have been shown to regulate the hypertrophic growth of cardiac myocytes including mitogen-activated protein kinase (MAPK) and calcineurin-nuclear factor of activated T-cells. However, it is uncertain if individual regulatory pathways operate in isolation or if interconnectivity between unrelated pathways is required for the orchestration of the entire hypertrophic response. To this end, we investigated the interconnectivity between calcineurin-mediated cardiac myocyte hypertrophy and p38 MAPK signaling in vitro and in vivo. We show that calcineurin promotes down-regulation of p38 MAPK activity and enhances expression of the dual specificity phosphatase MAPK phosphatase-1 (MKP-1). Transgenic mice expressing activated calcineurin in the heart were characterized by inactivation of p38 and increased MKP-1 expression during early postnatal development, before the onset of cardiac hypertrophy. In vitro, cultured neonatal cardiomyocytes infected with a calcineurin-expressing adenovirus and stimulated with phenylephrine demonstrated reduced p38 phosphorylation and increased MKP-1 protein levels. Activation of endogenous calcineurin with the calcium ionophore decreased p38 phosphorylation and increased MKP-1 protein levels. Inhibition of endogenous calcineurin with cyclosporin A decreased MKP-1 protein levels and increased p38 activation in response to agonist stimulation. To further investigate potential cross-talk between calcineurin and p38 through alteration in MKP-1 expression, the MKP-1 promoter was characterized and determined to be calcineurin-responsive. These data suggest that calcineurin enhances MKP-1 expression in cardiac myocytes, which is associated with p38 inactivation.


Assuntos
Calcineurina/metabolismo , Proteínas de Ciclo Celular , Proteínas Imediatamente Precoces/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/metabolismo , Fosfoproteínas Fosfatases , Proteínas Tirosina Fosfatases/genética , Animais , Animais Recém-Nascidos , Células COS , Calcimicina/farmacologia , Calcineurina/genética , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Ciclosporina/farmacologia , Fosfatase 1 de Especificidade Dupla , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica , Coração/efeitos dos fármacos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Miocárdio/citologia , Fenilefrina/farmacologia , Regiões Promotoras Genéticas , Proteína Fosfatase 1 , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno
20.
Proc Natl Acad Sci U S A ; 98(6): 3322-7, 2001 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-11248077

RESUMO

The Ca(2+)-calmodulin-activated Ser/Thr protein phosphatase calcineurin and the downstream transcriptional effectors of calcineurin, nuclear factor of activated T cells, have been implicated in the hypertrophic response of the myocardium. Recently, the calcineurin inhibitory agents cyclosporine A and FK506 have been extensively used to evaluate the importance of this signaling pathway in rodent models of cardiac hypertrophy. However, pharmacologic approaches have rendered equivocal results necessitating more specific or genetic-based inhibitory strategies. In this regard, we have generated Tg mice expressing the calcineurin inhibitory domains of Cain/Cabin-1 and A-kinase anchoring protein 79 specifically in the heart. DeltaCain and DeltaA-kinase-anchoring protein Tg mice demonstrated reduced cardiac calcineurin activity and reduced hypertrophy in response to catecholamine infusion or pressure overload. In a second approach, adenoviral-mediated gene transfer of DeltaCain was performed in the adult rat myocardium to evaluate the effectiveness of an acute intervention and any potential species dependency. DeltaCain adenoviral gene transfer inhibited cardiac calcineurin activity and reduced hypertrophy in response to pressure overload without reducing aortic pressure. These results provide genetic evidence implicating calcineurin as an important mediator of the cardiac hypertrophic response in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Inibidores de Calcineurina , Calcineurina/fisiologia , Cardiomegalia/prevenção & controle , Fosfoproteínas/fisiologia , Proteínas de Ancoragem à Quinase A , Adenoviridae , Animais , Pressão Sanguínea , Calcineurina/genética , Cardiomegalia/induzido quimicamente , Cardiomegalia/patologia , Cardiotônicos/efeitos adversos , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Expressão Gênica , Vetores Genéticos , Coração/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Isoproterenol/efeitos adversos , Camundongos , Camundongos Transgênicos , Fenótipo , Fosfoproteínas/genética , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...