Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Cell Mol Med ; 21(12): 3277-3287, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28608983

RESUMO

The use of doxorubicin (DOXO) as a chemotherapeutic drug has been hampered by cardiotoxicity leading to cardiomyopathy and heart failure. Folic acid (FA) is a modulator of endothelial nitric oxide (NO) synthase (eNOS), which in turn is an important player in diseases associated with NO insufficiency or NOS dysregulation, such as pressure overload and myocardial infarction. However, the role of FA in DOXO-induced cardiomyopathy is poorly understood. The aim of this study was to test the hypothesis that FA prevents DOXO-induced cardiomyopathy by modulating eNOS and mitochondrial structure and function. Male C57BL/6 mice were randomized to a single dose of DOXO (20 mg/kg intraperitoneal) or sham. FA supplementation (10 mg/day per oral) was started 7 days before DOXO injection and continued thereafter. DOXO resulted in 70% mortality after 10 days, with the surviving mice demonstrating a 30% reduction in stroke volume compared with sham groups. Pre-treatment with FA reduced mortality to 45% and improved stroke volume (both P < 0.05 versus DOXO). These effects of FA were underlain by blunting of DOXO-induced cardiomyocyte atrophy, apoptosis, interstitial fibrosis and impairment of mitochondrial function. Mechanistically, pre-treatment with FA prevented DOXO-induced increases in superoxide anion production by reducing the eNOS monomer:dimer ratio and eNOS S-glutathionylation, and attenuated DOXO-induced decreases in superoxide dismutase, eNOS phosphorylation and NO production. Enhancing eNOS function by restoring its coupling and subsequently reducing oxidative stress with FA may be a novel therapeutic approach to attenuate DOXO-induced cardiomyopathy.


Assuntos
Antioxidantes/farmacologia , Cardiomiopatias/prevenção & controle , Cardiotônicos/farmacologia , Cardiotoxicidade/prevenção & controle , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Ácido Fólico/farmacologia , Animais , Antibióticos Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/enzimologia , Cardiomiopatias/mortalidade , Cardiotoxicidade/enzimologia , Cardiotoxicidade/mortalidade , Cardiotoxicidade/patologia , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Volume Sistólico/efeitos dos fármacos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo , Análise de Sobrevida
2.
J Cardiovasc Transl Res ; 10(4): 348-358, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28397162

RESUMO

Angiogenic growth factor therapy for ischemic cardiovascular disease carries a risk of stimulating atherosclerotic plaque growth. We evaluated risk benefit ratio of sustained administration of recombinant human placental growth factor (rhPlGF)-2 in mice with advanced atherosclerosis and chronic ischemic cardiomyopathy. We maintained apolipoprotein E-deficient mice on a high cholesterol diet and induced myocardial infarction by transient ligation at 4 weeks. At 8 weeks, we assessed left ventricular (LV) function and randomized mice to receive rhPlGF-2 or vehicle (VEH) subcutaneously for 28 days. Administration of rhPlGF-2 significantly increased PlGF plasma levels without adverse hemodynamic or systemic inflammatory effects. RhPlGF-2 did not increase plaque area, composition, or vulnerability in the aortic arch. RhPlGF-2 significantly improved contractile function and reduced LV end-systolic and end-diastolic volume indices with a concomitant increase in capillary and arteriolar density in ischemic myocardium. RhPlGF-2 may represent a promising therapeutic strategy in chronic ischemic cardiomyopathy.


Assuntos
Indutores da Angiogênese/administração & dosagem , Aorta/efeitos dos fármacos , Doenças da Aorta/tratamento farmacológico , Aterosclerose/tratamento farmacológico , Cardiomiopatias/tratamento farmacológico , Infarto do Miocárdio/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Fator de Crescimento Placentário/administração & dosagem , Função Ventricular Esquerda/efeitos dos fármacos , Indutores da Angiogênese/toxicidade , Animais , Aorta/patologia , Aorta/fisiopatologia , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Cardiomiopatias/diagnóstico por imagem , Cardiomiopatias/fisiopatologia , Colesterol na Dieta , Doença Crônica , Modelos Animais de Doenças , Infusões Subcutâneas , Masculino , Camundongos Knockout para ApoE , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/fisiopatologia , Fator de Crescimento Placentário/toxicidade , Placa Aterosclerótica , Proteínas Recombinantes/administração & dosagem , Recuperação de Função Fisiológica , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Rigidez Vascular/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos
3.
Antioxid Redox Signal ; 26(4): 153-164, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27505125

RESUMO

AIMS: The use of doxorubicin, a potent chemotherapeutic agent, is limited by cardiotoxicity. We tested the hypothesis that decreased soluble guanylate cyclase (sGC) enzyme activity contributes to the development of doxorubicin-induced cardiotoxicity. RESULTS: Doxorubicin administration (20 mg/kg, intraperitoneally [IP]) reduced cardiac sGC activity in wild-type (WT) mice. To investigate whether decreased sGC activity contributes to doxorubicin-induced cardiotoxicity, we studied mice with cardiomyocyte-specific deficiency of the sGC α1-subunit (mice with cardiomyocyte-specific deletion of exon 6 of the sGCα1 allele [sGCα1-/-CM]). After 12 weeks of doxorubicin administration (2 mg/kg/week IP), left ventricular (LV) systolic dysfunction was greater in sGCα1-/-CM than WT mice. To further assess whether reduced sGC activity plays a pathogenic role in doxorubicin-induced cardiotoxicity, we studied a mouse model in which decreased cardiac sGC activity was induced by cardiomyocyte-specific expression of a dominant negative sGCα1 mutant (DNsGCα1) upon doxycycline removal (Tet-off). After 8 weeks of doxorubicin administration, DNsGCα1tg/+, but not WT, mice displayed LV systolic dysfunction and dilatation. The difference in cardiac function and remodeling between DNsGCα1tg/+ and WT mice was even more pronounced after 12 weeks of treatment. Further impairment of cardiac function was attenuated when DNsGCα1 gene expression was inhibited (beginning at 8 weeks of doxorubicin treatment) by administering doxycycline. Furthermore, doxorubicin-associated reactive oxygen species generation was higher in sGCα1-deficient than WT hearts. Innovation and Conclusion: These data demonstrate that a reduction in cardiac sGC activity worsens doxorubicin-induced cardiotoxicity in mice and identify sGC as a potential therapeutic target. Various pharmacological sGC agonists are in clinical development or use and may represent a promising approach to limit doxorubicin-associated cardiotoxicity. Antioxid. Redox Signal. 26, 153-164.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Doxorrubicina/efeitos adversos , Cardiopatias/etiologia , Cardiopatias/metabolismo , Guanilil Ciclase Solúvel/sangue , Animais , Antibióticos Antineoplásicos/administração & dosagem , Cardiotoxicidade , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Cardiopatias/fisiopatologia , Camundongos , Camundongos Knockout , Mutação , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Guanilil Ciclase Solúvel/deficiência , Disfunção Ventricular
4.
J Cardiovasc Pharmacol ; 68(5): 374-382, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27824722

RESUMO

Bmal1-(brain and muscle ARNT-like protein-1) deficient (Bmal1) mice prematurely age because of an increased reactive oxygen species (ROS) production. These mice also show a decline in cardiac function with age. We investigated whether an antioxidant treatment can ameliorate the declining cardiac function in prematurely aged Bmal1 mice. Male Bmal1 and wild-type (Bmal1) mice were exposed for 15 weeks to a high fat and high cholesterol diet with or without the antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL; 5 mmol/L; in drinking water during the last 10 weeks). Echocardiographic analysis revealed that TEMPOL treatment of Bmal1 mice normalized cardiac function, as evidenced by a decrease in left ventricular diastolic and systolic internal diameters, and by an increase in fractional shortening and ejection fraction. The antioxidant did not affect cardiac function in Bmal1 mice. Although TEMPOL did not influence cardiac ROS levels in Bmal1 mice, it significantly protected Bmal1 cardiac telomeres from oxidation, as evidenced by a reduction in the telomere damage score (0.11 ± 0.012% vs. 0.16 ± 0.015%; P = 0.028). Thus, antioxidant treatment normalized cardiac function of Bmal1 mice, probably in part by scavenging ROS.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Senilidade Prematura/tratamento farmacológico , Senilidade Prematura/metabolismo , Antioxidantes/uso terapêutico , Óxidos N-Cíclicos/uso terapêutico , Modelos Animais de Doenças , Animais , Antioxidantes/farmacologia , Colesterol na Dieta/administração & dosagem , Colesterol na Dieta/efeitos adversos , Óxidos N-Cíclicos/farmacologia , Dieta Hiperlipídica/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Marcadores de Spin , Resultado do Tratamento
5.
J Pharmacol Exp Ther ; 356(2): 284-92, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26621143

RESUMO

Enhanced cyclic guanosine monophosphate (cGMP) signaling may attenuate myocardial ischemia-reperfusion injury (I/R) and improve left ventricular (LV) functional recovery after myocardial infarction (MI). We investigated the cardioprotection afforded by inhaled NO (iNO), the phosphodiesterase 5 (PDE5)-specific inhibitor tadalafil (TAD), or their combination (iNO+TAD) in C57Bl6J mice subjected to 6-minute left anterior descending artery ligation followed by reperfusion. We measured plasma and cardiac concentrations of cGMP during early reperfusion, quantified myocardial necrosis and inflammation by serial troponin-I (TnI) and myeloperoxidase-positive cell infiltration at day 3, and evaluated LV function and remodeling after 4 weeks using echocardiography and pressure-conductance catheterization. Administration of iNO, TAD, or both during I/R was safe and hemodynamically well tolerated. Compared with untreated mice (CON), only iNO+TAD increased plasma and cardiac-cGMP levels during early reperfusion (80 ± 12 versus 36 ± 6 pmol/ml and 0.15 ± 0.02 versus 0.05 ± 0.01 pmol/mg protein, P < 0.05 for both). Moreover, iNO+TAD reduced TnI at 4 hours to a greater extent (P < 0.001 versus CON) than either alone (P < 0.05 versus CON) and was associated with significantly less myocardial inflammatory cell infiltration at day 3. After 4 weeks and compared with CON, iNO+TAD was associated with increased fractional shortening (43 ± 1 versus 33 ± 2%, P < 0.01), larger stroke volumes (14.9 ± 1.2 versus 10.2 ± 0.9 µl, P < 0.05), enhanced septal and posterior wall thickening (P < 0.05 and P < 0.001, respectively), and attenuated LV dilatation (P < 0.001), whereas iNO or TAD alone conferred less benefit. Thus, iNO+TAD has superior efficacy to limit early reperfusion injury and attenuate adverse LV remodeling. Combination of inhaled NO with a long-acting PDE5 inhibitor may represent a promising strategy to reduce ischemic damage following reperfusion and better preserve LV function.


Assuntos
Cardiotônicos/administração & dosagem , Traumatismo por Reperfusão Miocárdica/diagnóstico por imagem , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Óxido Nítrico/administração & dosagem , Inibidores da Fosfodiesterase 5/administração & dosagem , Administração por Inalação , Animais , Quimioterapia Combinada , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ultrassonografia
6.
J Clin Invest ; 125(12): 4463-82, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571398

RESUMO

Conditions such as muscular dystrophies (MDs) that affect both cardiac and skeletal muscles would benefit from therapeutic strategies that enable regeneration of both of these striated muscle types. Protocols have been developed to promote induced pluripotent stem cells (iPSCs) to differentiate toward cardiac or skeletal muscle; however, there are currently no strategies to simultaneously target both muscle types. Tissues exhibit specific epigenetic alterations; therefore, source-related lineage biases have the potential to improve iPSC-driven multilineage differentiation. Here, we determined that differential myogenic propensity influences the commitment of isogenic iPSCs and a specifically isolated pool of mesodermal iPSC-derived progenitors (MiPs) toward the striated muscle lineages. Differential myogenic propensity did not influence pluripotency, but did selectively enhance chimerism of MiP-derived tissue in both fetal and adult skeletal muscle. When injected into dystrophic mice, MiPs engrafted and repaired both skeletal and cardiac muscle, reducing functional defects. Similarly, engraftment into dystrophic mice of canine MiPs from dystrophic dogs that had undergone TALEN-mediated correction of the MD-associated mutation also resulted in functional striatal muscle regeneration. Moreover, human MiPs exhibited the same capacity for the dual differentiation observed in murine and canine MiPs. The findings of this study suggest that MiPs should be further explored for combined therapy of cardiac and skeletal muscles.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Mesoderma/metabolismo , Músculo Esquelético/fisiologia , Miocárdio , Regeneração , Animais , Cães , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Mesoderma/citologia , Camundongos , Distrofias Musculares/metabolismo , Distrofias Musculares/terapia , Ratos
7.
Circulation ; 131(9): 815-26, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25561514

RESUMO

BACKGROUND: Microvascular endothelium in different organs is specialized to fulfill the particular needs of parenchymal cells. However, specific information about heart capillary endothelial cells (ECs) is lacking. METHODS AND RESULTS: Using microarray profiling on freshly isolated ECs from heart, brain, and liver, we revealed a genetic signature for microvascular heart ECs and identified Meox2/Tcf15 heterodimers as novel transcriptional determinants. This signature was largely shared with skeletal muscle and adipose tissue endothelium and was enriched in genes encoding fatty acid (FA) transport-related proteins. Using gain- and loss-of-function approaches, we showed that Meox2/Tcf15 mediate FA uptake in heart ECs, in part, by driving endothelial CD36 and lipoprotein lipase expression and facilitate FA transport across heart ECs. Combined Meox2 and Tcf15 haplodeficiency impaired FA uptake in heart ECs and reduced FA transfer to cardiomyocytes. In the long term, this combined haplodeficiency resulted in impaired cardiac contractility. CONCLUSIONS: Our findings highlight a regulatory role for ECs in FA transfer to the heart parenchyma and unveil 2 of its intrinsic regulators. Our insights could be used to develop new strategies based on endothelial Meox2/Tcf15 targeting to modulate FA transfer to the heart and remedy cardiac dysfunction resulting from altered energy substrate usage.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Células Endoteliais/metabolismo , Proteínas de Ligação a Ácido Graxo/biossíntese , Ácidos Graxos/metabolismo , Proteínas de Homeodomínio/fisiologia , Miocárdio/metabolismo , Tecido Adiposo/irrigação sanguínea , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Antígenos CD36/biossíntese , Antígenos CD36/genética , Baixo Débito Cardíaco/etiologia , Baixo Débito Cardíaco/genética , Baixo Débito Cardíaco/metabolismo , Células Cultivadas , Vasos Coronários/citologia , Proteínas de Ligação a Ácido Graxo/genética , Glucose/metabolismo , Heterozigoto , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Humanos , Lipase Lipoproteica/biossíntese , Lipase Lipoproteica/genética , Lipoproteínas VLDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/farmacologia , Análise Serial de Tecidos , Transcriptoma
8.
Circ Res ; 116(3): 425-36, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25520363

RESUMO

RATIONALE: To maintain cardiac mechanical and structural integrity after an ischemic insult, profound alterations occur within the extracellular matrix. Osteoglycin is a small leucine-rich proteoglycan previously described as a marker of cardiac hypertrophy. OBJECTIVE: To establish whether osteoglycin may play a role in cardiac integrity and function after myocardial infarction (MI). METHODS AND RESULTS: Osteoglycin expression is associated with collagen deposition and scar formation in mouse and human MI. Absence of osteoglycin in mice resulted in significantly increased rupture-related mortality with tissue disruption, intramyocardial bleeding, and increased cardiac dysfunction, despite equal infarct sizes. Surviving osteoglycin null mice had greater infarct expansion in comparison with wild-type mice because of impaired collagen fibrillogenesis and maturation in the infarcts as revealed by electron microscopy and collagen polarization. Absence of osteoglycin did not affect cardiomyocyte hypertrophy in the remodeling remote myocardium. In cultured fibroblasts, osteoglycin knockdown or supplementation did not alter transforming growth factor-ß signaling. Adenoviral overexpression of osteoglycin in wild-type mice significantly improved collagen quality, thereby blunting cardiac dilatation and dysfunction after MI. In osteoglycin null mice, adenoviral overexpression of osteoglycin was unable to prevent rupture-related mortality because of insufficiently restoring osteoglycin protein levels in the heart. Finally, circulating osteoglycin levels in patients with heart failure were significantly increased in the patients with a previous history of MI compared with those with nonischemic heart failure and correlated with survival, left ventricular volumes, and other markers of fibrosis. CONCLUSIONS: Increased osteoglycin expression in the infarct scar promotes proper collagen maturation and protects against cardiac disruption and adverse remodeling after MI. In human heart failure, osteoglycin is a promising biomarker for ischemic heart failure.


Assuntos
Cardiomegalia/metabolismo , Colágeno/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Infarto do Miocárdio/metabolismo , Animais , Cardiomegalia/patologia , Cicatriz/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Peptídeos e Proteínas de Sinalização Intercelular/genética , Linfotoxina-alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Ratos , Ratos Endogâmicos Lew , Remodelação Ventricular
9.
J Appl Physiol (1985) ; 117(5): 563-71, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25059236

RESUMO

Transverse aortic constriction (TAC) and angiotensin II (ANG II) subcutaneous osmotic pump infusion are frequently used murine models of pressure overload hypertrophy. The aim of this paper is to investigate time- and stressor-dependent functional and structural changes using echocardiographic B-mode, M-mode, and Doppler characterization. Ten-week-old male C57BL6/J wild-type mice received 4-wk ANG II (1.5 mg·kg(-1)·day(-1), n = 19) or saline (n = 10) infusion followed by echocardiography (Vevo2100, Visual Sonics), or underwent TAC (n = 63) or a sham operation (n = 30). In the TAC protocol, echocardiography was performed after 2 wk (n = 22 TAC, n = 10 sham), after 4 wk (n = 20 TAC, n = 10 sham), and after 10 wk (n = 21 TAC, n = 10 sham). ANG II infusion was associated with a mixed pressure and volume overload, with a variable contribution of volume overload caused by aortic valve insufficiency (grade 0.5-3.5/4). The degree of aortic valve insufficiency correlated with the degree of left ventricular dilation (r(2) = 0.671, P < 0.001). After TAC, all hypertrophic remodeling patterns known in human disease were observed: 1) low-flow, low-gradient with preserved ejection fraction (EF); 2) concentric hypertrophy with normal EF and flow; 3) concentric hypertrophy with moderately decreased EF and/or flow; 4) eccentric hypertrophy with normal EF and flow; 5) eccentric hypertrophy with moderately decreased EF and/or flow; and 6) eccentric hypertrophy with severely depressed EF. Eccentric remodeling was time dependent, with 5% of mice developing this phenotype at 2 wk, 39% at 4 wk, and 59% at 10 wk. Comprehensive echocardiographic analysis allows identification of homogeneous subgroups of mice subjected to hypertrophic stress, reducing variability in experimental results and facilitating clinical translation.


Assuntos
Cardiomegalia/diagnóstico por imagem , Cardiomegalia/fisiopatologia , Angiotensina II , Animais , Aorta Torácica , Cardiomegalia/induzido quimicamente , Constrição Patológica , Interpretação Estatística de Dados , Modelos Animais de Doenças , Ecocardiografia/métodos , Ecocardiografia Doppler/métodos , Camundongos , Camundongos Endogâmicos C57BL , Pressão , Vasoconstritores
10.
Eur J Pharmacol ; 723: 175-80, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24406339

RESUMO

To investigate the chronic effect of sitagliptin (7-[(3R)-3-amino-1-oxo-4-(2,4,5-trifluorophenyl)butyl]-5,6,7,8-tetrahydro-(3-(trifluoromethyl)-1,2,4-triazolo[4,3-a]pyrazine phosphate (1:1) monohydrate, SIT) on metabolism and cardiac function in genetic diabetic Akita mice, 10 weeks old Akita mice were either exposed for 4 months to a high fat and high cholesterol (HF-HC) diet, with or without 10mg/kg/day SIT, or were fed for 3 months with the same diet with or without 50mg/kg/day SIT. SIT treatment of Akita mice at either a low or high dose did not affect body or liver weight. A significant increase in subcutaneous and gonadal fat mass was only observed for the 50mg/kg/day dose of SIT. Furthermore, only the 50mg/kg/day SIT dose resulted in an improvement of glycemic control, as evidenced by a decrease in fasting blood HbA1c levels and an increase in plasma adiponectin levels. Echocardiographic analysis revealed that Akita mice kept on the HF-HC diet with 10mg/kg/day of SIT for 4 months showed an increase in ejection fraction and fractional shortening, whereas the higher dose (50mg/kg/day) had no effect on these parameters, but instead induced left ventricular (LV) hypertrophy as evidenced by an enlarged LV internal diameter, volume and mass. Thus, in the diabetic Akita mouse SIT is cardioprotective at a low dose (10mg/kg/day), whereas improvement of glycemic control requires a higher dose (50mg/kg/day) which, however, induces LV hypertrophy. This mouse model may thus be useful to study the safety of anti-diabetic drugs.


Assuntos
Diabetes Mellitus , Modelos Animais de Doenças , Hipoglicemiantes/farmacologia , Pirazinas/farmacologia , Triazóis/farmacologia , Adiponectina/sangue , Animais , Glicemia/análise , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatologia , Dieta Hiperlipídica , Glucagon/sangue , Peptídeo 1 Semelhante ao Glucagon/sangue , Hemoglobinas Glicadas/análise , Coração/fisiopatologia , Testes de Função Cardíaca , Hipoglicemiantes/sangue , Hipoglicemiantes/farmacocinética , Hipoglicemiantes/uso terapêutico , Insulina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Miocárdio/metabolismo , Pirazinas/sangue , Pirazinas/farmacocinética , Pirazinas/uso terapêutico , Fosfato de Sitagliptina , Triazóis/sangue , Triazóis/farmacocinética , Triazóis/uso terapêutico
11.
J Clin Invest ; 123(8): 3378-82, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23867623

RESUMO

Numerous common genetic variants have been linked to blood pressure, but no underlying mechanism has been elucidated. Population studies have revealed that the variant rs5068 (A/G) in the 3' untranslated region of NPPA, the gene encoding atrial natriuretic peptide (ANP), is associated with blood pressure. We selected individuals on the basis of rs5068 genotype (AG vs. AA) and fed them a low- or high-salt diet for 1 week, after which they were challenged with an intravenous saline infusion. On both diets, before and after saline administration, ANP levels were up to 50% higher in AG individuals than in AA individuals, a difference comparable to the changes induced by high-salt diet or saline infusion. In contrast, B-type natriuretic peptide levels did not differ by rs5068 genotype. We identified a microRNA, miR-425, that is expressed in human atria and ventricles and is predicted to bind the sequence spanning rs5068 for the A, but not the G, allele. miR-425 silenced NPPA mRNA in an allele-specific manner, with the G allele conferring resistance to miR-425. This study identifies miR-425 as a regulator of ANP production, raising the possibility that miR-425 antagonists could be used to treat disorders of salt overload, including hypertension and heart failure.


Assuntos
Fator Natriurético Atrial/sangue , Hipertensão/genética , MicroRNAs/genética , Interferência de RNA , Regiões 3' não Traduzidas , Adulto , Animais , Fator Natriurético Atrial/genética , Células COS , Chlorocebus aethiops , GMP Cíclico/sangue , Feminino , Expressão Gênica/efeitos dos fármacos , Frequência do Gene , Estudos de Associação Genética , Humanos , Hipertensão/sangue , Masculino , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Cloreto de Sódio na Dieta/farmacologia , Adulto Jovem
12.
Eur J Pharmacol ; 700(1-3): 23-31, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23228931

RESUMO

Rosiglitazone ((RS)-5-[4-(2-[methyl(pyridin-2-yl)amino]ethoxy)benzyl]thiazolidine-2,4-dione, RGZ)-induced adverse drug effects in diabetic patients were not adequately predicted by current preclinical rodent models. Therefore, we have used the Akita mouse with genetic predisposition to diabetes to unravel the underlying molecular mechanisms. The effect of RGZ on adipose tissue and on cardiac function was evaluated in diabetic Akita mice kept on a high fat-high cholesterol diet (HF-HCD) for 4 months. When compared to wild-type (WT) mice with the same C57BL/6J genetic background, Akita mice gained significantly less weight (4.4±1.4 g versus 12±0.97 g for WT; P=0.002) and developed less fat (volume of 3.1±1.2 ml versus 16±2.1 ml for WT; P=0.004), associated with adipocyte hypotrophy. Upon treatment with RGZ (10mg/kg/day), Akita mice showed enhanced weight gain (11±0.70 g; P=0.004 versus untreated Akita mice) and fat volume (7.4±0.63 ml; P<0.05 versus untreated Akita mice), without effects on adipocyte or blood vessel size or on macrophage infiltration in adipose tissues. Akita mice kept on HF-HCD for 4 months with administration of RGZ (30 mg/kg/day) showed increased intraventricular septum thickness and cardiac output, without, however, an effect on fractional shortening or ejection fraction. In conclusion, RGZ promotes adiposity and early signs of hypertrophic cardiomyopathy in the diabetic Akita mouse. Thus, this genetically manipulated model may be suitable to test safety of anti-diabetic drugs.


Assuntos
Adiposidade/efeitos dos fármacos , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Tiazolidinedionas/farmacologia , Animais , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Genótipo , Heterozigoto , Hipertrofia/genética , Masculino , Camundongos , Rosiglitazona
13.
PLoS One ; 7(5): e36754, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22586493

RESUMO

The Kruppel-like factor (KLF) family of transcription factors regulates diverse cell biological processes including proliferation, differentiation, survival and growth. Previous studies have shown that KLF15 inhibits cardiac hypertrophy by repressing the activity of pivotal cardiac transcription factors such as GATA4, MEF2 and myocardin. We set out this study to characterize the interaction of KLF15 with putative other transcription factors. We first show that KLF15 interacts with myocardin-related transcription factors (MRTFs) and strongly represses the transcriptional activity of MRTF-A and MRTF-B. Second, we identified a region within the C-terminal zinc fingers of KLF15 that contains the nuclear localization signal. Third, we investigated whether overexpression of KLF15 in the heart would have therapeutic potential. Using recombinant adeno-associated viruses (rAAV) we have overexpressed KLF15 specifically in the mouse heart and provide the first evidence that elevation of cardiac KLF15 levels prevents the development of cardiac hypertrophy in a model of Angiotensin II induced hypertrophy.


Assuntos
Cardiomegalia , Proteínas de Ligação a DNA , Proteínas Nucleares/metabolismo , Proteínas de Fusão Oncogênica , Transativadores/metabolismo , Fatores de Transcrição , Angiotensina II/farmacologia , Animais , Sítios de Ligação , Células COS , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/terapia , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Humanos , Fatores de Transcrição Kruppel-Like , Camundongos , Proteínas Mutantes/metabolismo , Proteínas Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Cardiovasc Res ; 94(1): 115-24, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22308237

RESUMO

AIMS: Thrombospondin-2 (TSP-2) modulates matrix integrity and myocyte survival in the hypertensive or ageing heart. Whether TSP-2 may affect cardiac inflammation and injury, in particular during acute viral myocarditis, is completely unknown. METHODS AND RESULTS: Therefore, mortality, cardiac inflammation, and function were assessed in TSP-2-null (KO) and wild-type (WT) mice in human Coxsackie virus B3 (CVB3)-induced myocarditis. TSP-2 KO had an increased mortality when compared with WT mice during viral myocarditis. The absence of TSP-2 resulted in increased cardiac inflammation and injury at 14 days, which resulted in depressed systolic function [fractional shortening (FS); 34 ± 2.6 in WT vs. 24 ± 1.8 in KO mice, P< 0.05] and increased cardiac dilatation (end-diastolic dimensions, mm; 3.7 ± 0.09 in WT vs. 4.8 ± 0.06 in KO mice, P< 0.05) 35 days post-infection. Lack of TSP-2 resulted in a decreased activation of the anti-inflammatory T-regulatory cells, as indicated by a lower number of CD25-positive T-cells, and significantly decreased gene expression of regulatory T-cell markers, Foxp3 and CTLA-4. Finally, overexpression of TSP-2 in WT hearts using cardiotropic vectors derived from adeno-associated virus serotype 9 (AAV9) inhibited cardiac inflammation and injury at 14 days and improved cardiac function at 35 days post-CVB3 infection when compared with control AAV9. CONCLUSION: TSP-2 has a protective role against cardiac inflammation, injury, and dysfunction in acute viral myocarditis.


Assuntos
Enterovirus Humano B/patogenicidade , Terapia Genética , Ativação Linfocitária , Miocardite/prevenção & controle , Miocárdio/metabolismo , Linfócitos T Reguladores/metabolismo , Trombospondinas/metabolismo , Disfunção Ventricular Esquerda/prevenção & controle , Animais , Antígeno CTLA-4/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Fibrose , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C3H , Camundongos Knockout , Contração Miocárdica , Miocardite/diagnóstico por imagem , Miocardite/genética , Miocardite/imunologia , Miocardite/metabolismo , Miocardite/fisiopatologia , Miocárdio/imunologia , Miocárdio/patologia , Necrose , RNA Mensageiro/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/virologia , Trombospondinas/deficiência , Trombospondinas/genética , Fatores de Tempo , Ultrassonografia , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/imunologia , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda
15.
J Mol Cell Cardiol ; 51(3): 318-28, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21624372

RESUMO

Clinical use of the antineoplastic agent doxorubicin (DOX) is limited by its cardiomyocyte toxicity. Attempts to decrease cardiomyocyte injury showed promising results in vitro, but failed to reduce the adverse effects of DOX in vivo, suggesting that other mechanisms contribute to its cardiotoxicity as well. Evidence that DOX also induces cardiac injury by compromising extracellular matrix integrity is lacking. The matricellular protein thrombospondin-2 (TSP-2) is known for its matrix-preserving function, and for modulating cellular function. Here, we investigated whether TSP-2 modulates the process of doxorubicin-induced cardiomyopathy (DOX-CMP). TSP-2-knockout (TSP-2-KO) and wild-type (WT) mice were treated with DOX (2 mg/kg/week) for 12 weeks to induce DOX-CMP. Mortality was significantly increased in TSP-2-KO compared to WT mice. Surviving DOX-treated TSP-2-KO mice had depressed cardiac function compared to WT animals, accompanied by increased cardiomyocyte apoptosis and matrix damage. Enhanced myocyte damage in the absence of TSP-2 was associated with impaired activation of the Akt signaling pathway in TSP-2-KO compared to WT. The absence of TSP-2, in vivo and in vitro, reduced Akt activation both under non-treated conditions and after DOX. Importantly, inhibition of Akt phosphorylation in cardiomyocytes significantly reduced TSP-2 expression, unveiling a unique feedback loop between Akt and TSP-2. Finally, enhanced matrix disruption in DOX-treated TSP-2-KO hearts went along with increased matrix metalloproteinase-2 levels. Taken together, this study is the first to provide evidence for the implication of the matrix element TSP-2 in protecting against DOX-induced cardiac injury and dysfunction.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Doxorrubicina/toxicidade , Matriz Extracelular/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Trombospondinas/genética , Animais , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Matriz Extracelular/metabolismo , Feminino , Fibrose/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Trombospondinas/metabolismo
16.
Aging Cell ; 10(5): 769-79, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21501375

RESUMO

To understand the process of cardiac aging, it is of crucial importance to gain insight into the age-related changes in gene expression in the senescent failing heart. Age-related cardiac remodeling is known to be accompanied by changes in extracellular matrix (ECM) gene and protein levels. Small noncoding microRNAs regulate gene expression in cardiac development and disease and have been implicated in the aging process and in the regulation of ECM proteins. However, their role in age-related cardiac remodeling and heart failure is unknown. In this study, we investigated the aging-associated microRNA cluster 17-92, which targets the ECM proteins connective tissue growth factor (CTGF) and thrombospondin-1 (TSP-1). We employed aged mice with a failure-resistant (C57Bl6) and failure-prone (C57Bl6 × 129Sv) genetic background and extrapolated our findings to human age-associated heart failure. In aging-associated heart failure, we linked an aging-induced increase in the ECM proteins CTGF and TSP-1 to a decreased expression of their targeting microRNAs 18a, 19a, and 19b, all members of the miR-17-92 cluster. Failure-resistant mice showed an opposite expression pattern for both the ECM proteins and the microRNAs. We showed that these expression changes are specific for cardiomyocytes and are absent in cardiac fibroblasts. In cardiomyocytes, modulation of miR-18/19 changes the levels of ECM proteins CTGF and TSP-1 and collagens type 1 and 3. Together, our data support a role for cardiomyocyte-derived miR-18/19 during cardiac aging, in the fine-tuning of cardiac ECM protein levels. During aging, decreased miR-18/19 and increased CTGF and TSP-1 levels identify the failure-prone heart.


Assuntos
Fator de Crescimento do Tecido Conjuntivo/metabolismo , Insuficiência Cardíaca/patologia , MicroRNAs/metabolismo , Trombospondina 1/metabolismo , Adulto , Idoso , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Biópsia , Colágeno/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibrose/genética , Fibrose/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Coração/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Pessoa de Meia-Idade , Família Multigênica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Ratos Endogâmicos Lew , Trombospondina 1/genética
17.
Hypertension ; 55(2): 249-56, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20048198

RESUMO

Syndecan-1 (Synd1) is a transmembrane heparan sulfate proteoglycan that functions as a coreceptor for various growth factors and modulates signal transduction. The present study investigated whether Synd1, by affecting growth factor signaling, may play a role in hypertension-induced cardiac fibrosis and dysfunction. Expression of Synd1 was increased significantly in mouse hearts with angiotensin II-induced hypertension, which was spatially related to cardiac fibrosis. Angiotensin II significantly impaired fractional shortening and induced cardiac fibrosis in wild-type mice, whereas these effects were blunted in Synd1-null mice. Angiotensin II significantly increased cardiac expression of connective tissue growth factor and collagen type I and III in wild-type mice, which was blunted in Synd1-null mice. These findings were confirmed in vitro, where angiotensin II induced the expression of both connective tissue growth factor and collagen I in fibroblasts. The absence of Synd1 in either Synd1-null fibroblasts, after knockdown of Synd1 by short hairpin RNA, or after inhibition of heparan sulfates by protamine attenuated this increase, which was associated with reduced phosphorylation of Smad2. In conclusion, loss of Synd1 reduces cardiac fibrosis and dysfunction during angiotensin II-induced hypertension.


Assuntos
Angiotensina II/farmacologia , Miocárdio/patologia , Proteína Smad2/metabolismo , Sindecana-1/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Fibrose/patologia , Regulação da Expressão Gênica , Hipertensão/induzido quimicamente , Hipertensão/complicações , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Probabilidade , RNA Mensageiro/análise , Distribuição Aleatória , Proteína Smad2/efeitos dos fármacos , Proteína Smad2/genética , Sindecana-1/genética
18.
Circulation ; 120(16): 1585-97, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19805649

RESUMO

BACKGROUND: The progressive shift from a young to an aged heart is characterized by alterations in the cardiac matrix. The present study investigated whether the matricellular protein thrombospondin-2 (TSP-2) may affect cardiac dimensions and function with physiological aging of the heart. METHODS AND RESULTS: TSP-2 knockout (KO) and wild-type mice were followed up to an age of 60 weeks. Survival rate, cardiac function, and morphology did not differ at a young age in TSP-2 KO compared with wild-type mice. However, >55% of the TSP-2 KO mice died between 24 and 60 weeks of age, whereas <10% of the wild-type mice died. In the absence of TSP-2, older mice displayed a severe dilated cardiomyopathy with impaired systolic function, increased cardiac dilatation, and fibrosis. Ultrastructural analysis revealed progressive myocyte stress and death, accompanied by an inflammatory response and replacement fibrosis, in aging TSP-2 KO animals, whereas capillary or coronary morphology or density was not affected. Importantly, adeno-associated virus-9 gene-mediated transfer of TSP-2 in 7-week-old TSP-2 KO mice normalized their survival and prevented dilated cardiomyopathy. In TSP-2 KO animals, age-related cardiomyopathy was accompanied by increased matrix metalloproteinase-2 and decreased tissue transglutaminase-2 activity, together with impaired collagen cross-linking. At the cardiomyocyte level, TSP-2 deficiency in vivo and its knockdown in vitro decreased the activation of the Akt survival pathway in cardiomyocytes. CONCLUSIONS: TSP-2 expression in the heart protects against age-dependent dilated cardiomyopathy.


Assuntos
Envelhecimento , Cardiomiopatia Dilatada/etiologia , Miocárdio/metabolismo , Trombospondinas/deficiência , Animais , Cardiomiopatia Dilatada/mortalidade , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/prevenção & controle , Morte Celular , Ativação Enzimática , Feminino , Fibrose , Técnicas de Transferência de Genes , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Miocardite/etiologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trombospondinas/genética , Regulação para Cima
19.
J Pathol ; 217(5): 633-41, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19142976

RESUMO

Infection with coxsackievirus B4 (CVB4) may result in an acute severe necrotizing pancreatitis that mostly remains restricted to the acini of the exocrine parenchyma. The mechanisms responsible for this tissue damage, however, remain poorly understood. We here report that COAM, a polyanionic carboxylic acid, provides marked protection against CVB4-induced pancreatitis in a mouse model. Despite the fact that COAM largely reduced disease severity, as detected by serum amylase and lipase levels as well as histologically, titres of replicating CVB4 in the pancreas were virtually unaffected. COAM treatment diminished the infection-associated MMP-9 levels and also resulted in a decreased influx of neutrophils into the infected pancreas. Moreover, we demonstrate that titres of replicating virus in the pancreas did not directly correlate with the severity of disease. In conclusion, our data suggest that immunopathological effects, rather than direct virus-induced destruction, are responsible for the damage to acinar tissue in CVB4-induced pancreatitis.


Assuntos
Infecções por Coxsackievirus/complicações , Enterovirus Humano B/isolamento & purificação , Pancreatite Necrosante Aguda/virologia , Amilose/análogos & derivados , Amilose/uso terapêutico , Animais , Quimiotaxia de Leucócito/efeitos dos fármacos , Infecções por Coxsackievirus/enzimologia , Infecções por Coxsackievirus/patologia , Modelos Animais de Doenças , Enterovirus Humano B/efeitos dos fármacos , Enterovirus Humano B/fisiologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Infiltração de Neutrófilos/efeitos dos fármacos , Pâncreas/enzimologia , Pâncreas/virologia , Pancreatite Necrosante Aguda/enzimologia , Pancreatite Necrosante Aguda/patologia , Pancreatite Necrosante Aguda/prevenção & controle , Replicação Viral/efeitos dos fármacos
20.
J Exp Med ; 206(1): 113-23, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19103879

RESUMO

The matricellular protein SPARC (secreted protein, acidic and rich in cysteine, also known as osteonectin) mediates cell-matrix interactions during wound healing and regulates the production and/or assembly of the extracellular matrix (ECM). This study investigated whether SPARC functions in infarct healing and ECM maturation after myocardial infarction (MI). In comparison with wild-type (WT) mice, animals with a targeted inactivation of SPARC exhibited a fourfold increase in mortality that resulted from an increased incidence of cardiac rupture and failure after MI. SPARC-null infarcts had a disorganized granulation tissue and immature collagenous ECM. In contrast, adenoviral overexpression of SPARC in WT mice improved the collagen maturation and prevented cardiac dilatation and dysfunction after MI. In cardiac fibroblasts in vitro, reduction of SPARC by short hairpin RNA attenuated transforming growth factor beta (TGF)-mediated increase of Smad2 phosphorylation, whereas addition of recombinant SPARC increased Smad2 phosphorylation concordant with increased Smad2 phosphorylation in SPARC-treated mice. Importantly, infusion of TGF-beta rescued cardiac rupture in SPARC-null mice but did not significantly alter infarct healing in WT mice. These findings indicate that local production of SPARC is essential for maintenance of the integrity of cardiac ECM after MI. The protective effects of SPARC emphasize the potential therapeutic applications of this protein to prevent cardiac dilatation and dysfunction after MI.


Assuntos
Ruptura Cardíaca Pós-Infarto/metabolismo , Infarto do Miocárdio/metabolismo , Osteonectina/deficiência , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas da Matriz Extracelular/genética , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Tecido de Granulação/efeitos dos fármacos , Tecido de Granulação/metabolismo , Tecido de Granulação/patologia , Coração/fisiopatologia , Ruptura Cardíaca Pós-Infarto/fisiopatologia , Ruptura Cardíaca Pós-Infarto/prevenção & controle , Hemodinâmica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Osteonectina/genética , Osteonectina/fisiologia , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/genética , Análise de Sobrevida , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA