Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-37047281

RESUMO

Mass spectrometry is a powerful technique for investigating renal pathologies and identifying biomarkers, and efficient protein extraction from kidney tissue is essential for bottom-up proteomic analyses. Detergent-based strategies aid cell lysis and protein solubilization but are poorly compatible with downstream protein digestion and liquid chromatography-coupled mass spectrometry, requiring additional purification and buffer-exchange steps. This study compares two well-established detergent-based methods for protein extraction (in-solution sodium deoxycholate (SDC); suspension trapping (S-Trap)) with the recently developed sample preparation by easy extraction and digestion (SPEED) method, which uses strong acid for denaturation. We compared the quantitative performance of each method using label-free mass spectrometry in both sheep kidney cortical tissue and plasma. In kidney tissue, SPEED quantified the most unique proteins (SPEED 1250; S-Trap 1202; SDC 1197). In plasma, S-Trap produced the most unique protein quantifications (S-Trap 150; SDC 148; SPEED 137). Protein quantifications were reproducible across biological replicates in both tissue (R2 = 0.85-0.90) and plasma (SPEED R2 = 0.84; SDC R2 = 0.76, S-Trap R2 = 0.65). Our data suggest SPEED as the optimal method for proteomic preparation in kidney tissue and S-Trap or SPEED as the optimal method for plasma, depending on whether a higher number of protein quantifications or greater reproducibility is desired.


Assuntos
Detergentes , Espectrometria de Massas em Tandem , Animais , Ovinos , Detergentes/química , Espectrometria de Massas em Tandem/métodos , Proteômica/métodos , Reprodutibilidade dos Testes , Proteínas
2.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-35055195

RESUMO

One-quarter of patients with acute decompensated heart failure (ADHF) experience acute kidney injury (AKI)-an abrupt reduction or loss of kidney function associated with increased long-term mortality. There is a critical need to identify early and real-time markers of AKI in ADHF; however, to date, no protein biomarkers have exhibited sufficient diagnostic or prognostic performance for widespread clinical uptake. We aimed to identify novel protein biomarkers of AKI associated with ADHF by quantifying changes in protein abundance in the kidneys that occur during ADHF development and recovery in an ovine model. Relative quantitative protein profiling was performed using sequential window acquisition of all theoretical fragment ion spectra-mass spectrometry (SWATH-MS) in kidney cortices from control sheep (n = 5), sheep with established rapid-pacing-induced ADHF (n = 8), and sheep after ~4 weeks recovery from ADHF (n = 7). Of the 790 proteins quantified, we identified 17 candidate kidney injury markers in ADHF, 1 potential kidney marker of ADHF recovery, and 2 potential markers of long-term renal impairment (differential abundance between groups of 1.2-2.6-fold, adjusted p < 0.05). Among these 20 candidate protein markers of kidney injury were 6 candidates supported by existing evidence and 14 novel candidates not previously implicated in AKI. Proteins of differential abundance were enriched in pro-inflammatory signalling pathways: glycoprotein VI (activated during ADHF development; adjusted p < 0.01) and acute phase response (repressed during recovery from ADHF; adjusted p < 0.01). New biomarkers for the early detection of AKI in ADHF may help us to evaluate effective treatment strategies to prevent mortality and improve outcomes for patients.


Assuntos
Injúria Renal Aguda/diagnóstico , Biomarcadores/metabolismo , Insuficiência Cardíaca/metabolismo , Proteômica/métodos , Injúria Renal Aguda/sangue , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/urina , Animais , Biomarcadores/sangue , Biomarcadores/urina , Modelos Animais de Doenças , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/urina , Humanos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/urina , Prognóstico , Ovinos
3.
Int J Mol Sci ; 21(12)2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560137

RESUMO

Hydrogen sulfide (H2S) is recognized as an endogenous gaseous signaling molecule generated by cystathionine γ-lyase (CSE) in cardiovascular tissues. H2S up-regulation has been shown to reduce ischemic injury, and H2S donors are cardioprotective in rodent models when administered concurrent with myocardial ischemia. We evaluated the potential utility of H2S therapy in ameliorating cardiac remodeling with administration delayed until 2 h post-infarction in mice with or without cystathionine γ-lyase gene deletion (CSE-/-). The slow-release H2S donor, GYY4137, was administered from 2 h after surgery and daily for 28 days following myocardial infarction (MI) induced by coronary artery ligation, comparing responses in CSE-/- with wild-type (WT) mice (n = 5-10/group/genotype). Measures of cardiac function and expression of key genes associated with cardiac hypertrophy, fibrosis, and apoptosis were documented in atria, ventricle, and kidney tissues. Post-MI GYY4137 administration reduced infarct area and restored cardiac function, accompanied by reduction of the elevated ventricular expression of genes mediating cardiac remodeling to near-normal levels. Few differences between WT and CSE-/- mice were observed, except CSE-/- mice had higher blood pressures, and higher atrial Mir21a expression across all treatment groups. These findings suggest endogenous CSE gene deletion does not substantially exacerbate the long-term response to MI. Moreover, the H2S donor GYY4137 administered after onset of MI preserves cardiac function and protects against adverse cardiac remodeling in both WT and CSE-deficient mice.


Assuntos
Cistationina gama-Liase/genética , Sulfeto de Hidrogênio/metabolismo , Morfolinas/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Compostos Organotiofosforados/administração & dosagem , Animais , Modelos Animais de Doenças , Testes de Função Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , Morfolinas/farmacologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Compostos Organotiofosforados/farmacologia , Recuperação de Função Fisiológica , Regulação para Cima
4.
J Card Fail ; 22(1): 64-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26362519

RESUMO

BACKGROUND: The (pro)renin receptor [(P)RR] is implicated in the pathogenesis of cardiovascular disease. We investigated the effects of (P)RR blockade after myocardial infarction (MI) in a mouse coronary-ligation model. METHODS AND RESULTS: Mice underwent sham control surgeries (n = 8) or induction of MI followed by 28 days' treatment with a vehicle control (n = 8) or (P)RR antagonist (n = 8). Compared with sham control subjects, MI + vehicle mice demonstrated reduced left ventricular (LV) ejection fraction (LVEF: P < .001) and fractional shortening (P < .001), and increased LV end-systolic and -diastolic volumes (LVESV: P < .001; LVEDV: P < .001) 28 days after MI. In addition, MI decreased LV posterior wall and septal diameters (both P < .001), increased heart weight-body weight ratios (P < .05), LV collagen deposition, and cardiomyocyte diameter (both P < .001), and up-regulated collagen 1 (P < .01) and ß-myosin heavy chain (ß-MHC: P < .05) mRNA. Compared with MI + vehicle mice, (P)RR antagonism after MI reduced infarct size (P < .01), improved LVEF (P < .001), fractional shortening (P < .001), and stroke volume (P < .05), and decreased LVESV (P < .001) and LVEDV (P < .001). (P)RR antagonism also reversed MI-induced transmural thinning (P < .001) and reduced LV fibrosis (P < .01), cardiomyocyte size (P < .001), and ventricular collagen 1 (P < .01), ß-MHC (P = .06), transforming growth factor ß1 (P < .01), and angiotensin-converting enzyme (P < .05) expression. CONCLUSIONS: The present study found that (P)RR blockade after MI in mice ameliorates infarct size, cardiac fibrosis/hypertrophy, and cardiac dysfunction and identifies the receptor as a potential therapeutic target in this setting.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Miocárdio/patologia , Oligopeptídeos/farmacologia , ATPases Translocadoras de Prótons/antagonistas & inibidores , Receptores de Superfície Celular/antagonistas & inibidores , Remodelação Ventricular/efeitos dos fármacos , Animais , Cardiomegalia/prevenção & controle , Fibrose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/uso terapêutico , Função Ventricular Esquerda/efeitos dos fármacos
5.
J Cardiovasc Pharmacol ; 65(3): 269-75, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25658462

RESUMO

The impact of chronic urocortin 2 (Ucn2) treatment after myocardial infarction (MI) has not previously been investigated. In this study, we examined the effects of 30-day Ucn2 administration (415 µg·kg·d SC per day) in mice post-MI. Compared with surgical sham + vehicle controls (n = 10), MI + vehicle animals (n = 10) after 30 days demonstrated decreased ejection fraction (75.6 ± 1.2 vs. 43.6% ± 0.8%, P < 0.001) and fractional shortening (38.20 ± 0.83 vs. 18.4% ± 0.54%, P < 0.001) in association with increased heart weight-to-body weight ratio (4.57 ± 0.25 vs. 5.29 ± 0.18, P < 0.01), left ventricular (LV) mass (91 ± 7 vs. 126 ± 8 mg, P < 0.01), LV internal diameters at both systole (1.91 ± 0.14 vs. 3.45 ± 0.09 mm, P < 0.001) and diastole (3.14 ± 0.15 vs. 4.25 ± 0.10 mm, P < 0.001), LV end systolic volumes (0.02 ± 0.01 vs. 0.11 ± 0.01 mL, P < 0.001), and ventricular collagen 1 and ß-myosin heavy chain gene expression. Compared with MI + vehicle mice, MI + Ucn2 animals (n = 10) exhibited significantly reduced infarct size (4.00 ± 0.39 vs. 1.83 ± 0.44 mm, P < 0.01), heart weight-to-body weight ratio (4.75 ± 0.19, P = 0.06), LV mass (101 ± 6 mg, P < 0.01), LV internal diameters (systole 2.61 ± 0.09 mm, P < 0.001; diastole 3.78 ± 0.09 mm, P < 0.001), and end systolic volumes (0.14 ± 0.02 mL, P < 0.01) in conjunction with improved ejection fraction (65.2% ± 0.9%, P < 0.001) and fractional shortening (18.4 ± 0.5 vs. 30.5% ± 0.5%, P < 0.001). Ucn2 treatment also decreased collagen 1 and ß-myosin heavy chain expression. In conclusion, chronic Ucn2 treatment significantly improves cardiovascular function and attenuates cardiac injury and remodeling in experimental MI.


Assuntos
Cardiotônicos/farmacologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Infarto do Miocárdio/tratamento farmacológico , Urocortinas/farmacologia , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Fibrose , Regulação da Expressão Gênica , Hemodinâmica/efeitos dos fármacos , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
6.
Am J Physiol Endocrinol Metab ; 302(5): E576-84, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22185842

RESUMO

The aim of this study was to create a comprehensive mouse model of the metabolic syndrome by crossing aromatase-deficient (ArKO) mice with apolipoprotein E-deficient (ApoE(-/-)) mice. Successive crossbreeding of ArKO with ApoE(-/-)-deficient mice generated double knockout, MetS-Tg mice. The phenotypic characteristics of the MetS-Tg mice were assessed at 3, 6, and 12 mo of age and compared with age- and sex-matched wild-type (WT) controls. Blood pressure and heart rate were recorded by a noninvasive, computerized tail-cuff system. Oral glucose and intraperitoneal insulin tolerance tests were performed. Serum cholesterol levels were measured by a combined quantitative colorimetric assay. Plasma adiponectin, C-reactive protein (CRP), insulin, interleukin-6 (IL-6), leptin, resistin, and tumor necrosis factor-α (TNF-α) were measured by multiplexed ELISA. MetS-Tg mice displayed significantly increased body weight, central obesity, and elevated blood pressure at all three ages compared with WT mice. Elevated serum cholesterol was associated with higher triglycerides and LDL/VLDL cholesterol particles and was accompanied by a decrease in HDL and histological evidence of fatty liver. MetS-Tg mice of all ages showed impaired glucose tolerance. At 12 mo, MetS-Tg mice had elevated plasma levels of CRP, IL-6, leptin, and TNF-α, but resistin levels were largely unchanged. We now report that this combination of gene knockouts produces a novel strain of mice that display the diverse clinical features of the metabolic syndrome, including central obesity, progressive hypertension, an adverse serum lipid profile, fatty liver, glucose intolerance, insulin resistance, and evidence of an inflammatory state.


Assuntos
Apolipoproteínas E/fisiologia , Aromatase/fisiologia , Modelos Animais de Doenças , Síndrome Metabólica/fisiopatologia , Tecido Adiposo Branco/imunologia , Tecido Adiposo Branco/patologia , Animais , Aorta/imunologia , Aorta/patologia , Apolipoproteínas E/genética , Aromatase/genética , Aterosclerose/etiologia , Proteína C-Reativa/análise , Cruzamentos Genéticos , Citocinas/sangue , Fígado Gorduroso/etiologia , Feminino , Intolerância à Glucose/etiologia , Hiperlipidemias/etiologia , Hipertensão/etiologia , Resistência à Insulina , Fígado/imunologia , Fígado/patologia , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/imunologia , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Obesidade Abdominal/etiologia
7.
J Am Heart Assoc ; 10(18): e021312, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34533033

RESUMO

BACKGROUND Acute decompensated heart failure (ADHF) is associated with deterioration in renal function-an important risk factor for poor outcomes. Whether ADHF results in permanent kidney damage/dysfunction is unknown. METHODS AND RESULTS We investigated for the first time the renal responses to the development of, and recovery from, ADHF using an ovine model. ADHF development induced pronounced hemodynamic changes, neurohormonal activation, and decline in renal function, including decreased urine, sodium and urea excretion, and creatinine clearance. Following ADHF recovery (25 days), creatinine clearance reductions persisted. Kidney biopsies taken during ADHF and following recovery showed widespread mesangial cell prominence, early mild acute tubular injury, and medullary/interstitial fibrosis. Renal transcriptomes identified altered expression of 270 genes following ADHF development and 631 genes following recovery. A total of 47 genes remained altered post-recovery. Pathway analysis suggested gene expression changes, driven by a network of inflammatory cytokines centered on IL-1ß (interleukin 1ß), lead to repression of reno-protective eNOS (endothelial nitric oxide synthase) signaling during ADHF development, and following recovery, activation of glomerulosclerosis and reno-protective pathways and repression of proinflammatory/fibrotic pathways. A total of 31 dysregulated genes encoding proteins detectable in urine, serum, and plasma identified potential candidate markers for kidney repair (including CNGA3 [cyclic nucleotide gated channel subunit alpha 3] and OIT3 [oncoprotein induced transcript 3]) or long-term renal impairment in ADHF (including ACTG2 [actin gamma 2, smooth muscle] and ANGPTL4 [angiopoietin like 4]). CONCLUSIONS In an ovine model, we provide the first direct evidence that an episode of ADHF leads to an immediate decline in kidney function that failed to fully resolve after ≈4 weeks and is associated with persistent functional/structural kidney injury. We identified molecular pathways underlying kidney injury and repair in ADHF and highlighted 31 novel candidate biomarkers for acute kidney injury in this setting.


Assuntos
Injúria Renal Aguda , Insuficiência Cardíaca , Injúria Renal Aguda/genética , Animais , Biomarcadores , Creatinina , Insuficiência Cardíaca/genética , Rim/fisiologia , Ovinos , Carneiro Doméstico , Transcriptoma
8.
Biochim Biophys Acta ; 1792(12): 1175-84, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19782130

RESUMO

The heart adapts to an increased workload through the activation of a hypertrophic response within the cardiac ventricles. This response is characterized by both an increase in the size of the individual cardiomyocytes and an induction of a panel of genes normally expressed in the embryonic and neonatal ventricle, such as atrial natriuretic peptide (ANP). ANP and brain natriuretic peptide (BNP) exert their biological actions through activation of the natriuretic peptide receptor-1 (Npr1). The current study examined mice lacking Npr1 (Npr1(-/-)) activity and investigated the effects of the absence of Npr1 signaling during cardiac development on embryo viability, cardiac structure and gene and protein expression. Npr1(-/-)embryos were collected at embryonic day (ED) 12.5, 15.5 and neonatal day 1 (ND 1). Npr1(-/-)embryos occurred at the expected Mendelian frequency at ED 12.5, but knockout numbers were significantly decreased at ED 15.5 and ND 1. There was no indication of cardiac structural abnormalities in surviving embryos. However, Npr1(-/-)embryos exhibited cardiac enlargement (without fibrosis) from ED 15.5 as well as significantly increased ANP mRNA and protein expression compared to wild-type (WT) mice, but no concomitant increase in expression of the hypertrophy-related transcription factors, Mef2A, Mef2C, GATA-4, GATA-6 or serum response factor (SRF). However, there was a significant decrease in Connexin-43 (Cx43) gene and protein expression at mid-gestation in Npr1(-/-)embryos. Our findings suggest that the mechanism by which natriuretic peptide signaling influences cardiac development in Npr1(-/-) mice is distinct from that seen during the development of pathological cardiac hypertrophy and fibrosis. The decreased viability of Npr1(-/-)embryos may result from a combination of cardiomegaly and dysregulated Cx43 protein affecting cardiac contractility.


Assuntos
Cardiomegalia/mortalidade , Cardiomegalia/patologia , Embrião de Mamíferos/citologia , Coração/embriologia , Receptores do Fator Natriurético Atrial/fisiologia , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Pressão Sanguínea , Western Blotting , Cardiomegalia/metabolismo , Conexina 43/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Coração/fisiologia , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Knockout , Peptídeo Natriurético Encefálico/genética , Peptídeo Natriurético Encefálico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
9.
Endocrinology ; 149(11): 5828-34, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18653707

RESUMO

After myocardial infarction (MI), the heart may undergo progressive ventricular remodeling, resulting in a deterioration of cardiac function. TGF-beta is a key cytokine that both initiates and terminates tissue repair, and its sustained production underlies the development of tissue fibrosis, particularly after MI. We investigated the effects of a novel orally active specific inhibitor of the TGF-beta receptor 1 (SD-208) in an experimental model of MI. Mice underwent ligation of the left coronary artery to induce MI and were subsequently treated for 30 d after infarction with either SD-208 or a vehicle control. Blockade of TGF-beta signaling reduced mean arterial pressure in all groups. SD-208 treatment after MI resulted in a trend for reduced ventricular and renal gene expression of TGF-beta-activated kinase-1 (a downstream modulator of TGF-beta signaling) and a significant decrease in collagen 1, in association with a marked decrease in cardiac mass. Post-MI SD-208 treatment significantly reduced circulating levels of plasma renin activity as well as down-regulating the components of the cardiac and renal renin-angiotensin system (angiotensinogen, angiotensin converting enzyme, and angiotensin II type I receptor). Our findings indicate that blockade of the TGF-beta signaling pathway results in significant amelioration of deleterious cardiac remodeling after infarction.


Assuntos
Infarto do Miocárdio/fisiopatologia , Pteridinas/farmacologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Remodelação Ventricular/efeitos dos fármacos , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Pteridinas/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Renina/sangue , Renina/metabolismo , Sistema Renina-Angiotensina/fisiologia , Fator de Crescimento Transformador beta/fisiologia
10.
J Mol Endocrinol ; 38(1-2): 245-57, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17293444

RESUMO

The natriuretic peptides, atrial (ANP) and brain natriuretic peptide (BNP) are known to suppress cardiac hypertrophy and fibrosis. Both ANP and BNP exert their bioactivities through the Npr1 receptor, and Npr1 knockout mice (Npr1-/-) exhibit marked cardiac hypertrophy and fibrosis. In this study, we investigated which genes within the hypertrophic and fibrotic pathways are influenced by the lack of Npr1 signalling. cDNA microarray and quantitative real-time PCR (RT-PCR) analyses were performed on cardiac ventricles from Npr1-/-mice. Gene expression at early and late stages during development of hypertrophy was investigated in male and female Npr1-/-mice at 8 weeks and 6 months of age. Heart weight to body weight ratios (HW:BW) were maximally increased in 8-week males (P<0 x 01), whilst HW:BW in females continued to increase progressively up to 6 months (P<0 x 01). This was despite blood pressure being similarly elevated at both the ages in male and female knockout when compared with wild-type (WT) mice (P<0 x 001). Microarray analysis identified altered gene expression at the earliest steps in the hypertrophy-signalling cascade in Npr1-/- mice, particularly calcium-calmodulin signalling and ion channels, with subsequent changes in the expression of intracellular messengers including protein kinases and transcription factors. Real-time PCR analysis confirmed significant differences in gene expression of ANP, BNP, calmodulin 1, histone deacetylase 7a (HDAC7a), protein kinase C (PKC)iota, (GATA) 4, collagen 1, phospholamban and transforming growth factor-beta1 in Npr1-/- mice when compared with WT (P<0 x 05). The present study implicates the calmodulin-CaMK-Hdac-Mef2 and PKC-MAPK-GATA4 pathways in Npr1 mediation of cardiac hypertrophy.


Assuntos
Cardiomegalia/genética , Fibrose/genética , Regulação da Expressão Gênica/fisiologia , Guanilato Ciclase/fisiologia , Miocárdio/patologia , Receptores do Fator Natriurético Atrial/fisiologia , Animais , Cardiomegalia/metabolismo , Fibrose/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/metabolismo
11.
Nutrients ; 9(7)2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28686204

RESUMO

There is extensive evidence that walnut consumption is protective against cardiovascular disease and diabetes in the healthy population, but the beneficial effects of walnut consumption in individuals with the metabolic syndrome (MetS) remain uncertain. We compared a range of cardio-metabolic traits and related tissue gene expression associated with 21 weeks of dietary walnut supplementation in a mouse model of MetS (MetS-Tg) and wild-type (WT) mice (n = 10 per genotype per diet, equal males and females). Compared to standard diet, walnuts did not significantly alter food consumption or body weight trajectory of either MetS-Tg or WT mice. In MetS-Tg mice, walnuts were associated with reductions in oral glucose area under the curve (gAUC, standard diet 1455 ± 54, walnut 1146 ± 91, p = 0.006) and mean arterial blood pressure (MAP, standard diet 100.6 ± 1.9, walnut 73.2 ± 1.8 mmHg, p < 0.001), with neutral effects on gAUC and MAP in WT mice. However, in MetS-Tg mice, walnuts were also associated with trends for higher plasma cholesterol (standard diet 4.73 ± 0.18, walnut 7.03 ± 1.99 mmol/L, p = 0.140) and triglyceride levels (standard diet 2.4 ± 0.5, walnut 5.4 ± 1.6 mmol/L, p = 0.061), despite lowering cholesterol and having no effect on triglycerides in WT mice. Moreover, in both MetS-Tg and WT mice, walnuts were associated with significantly increased liver expression of genes associated with metabolism (Fabp1, Insr), cell stress (Atf6, Ddit3, Eif2ak3), fibrosis (Hgf, Sp1, Timp1) and inflammation (Tnf, Ptpn22, Pparg). In conclusion, dietary walnuts were associated with modest favourable effects in WT mice, but a combination of beneficial and adverse effects in MetS-Tg mice, and up-regulation of hepatic pro-fibrotic and pro-inflammatory genes in both mouse strains.


Assuntos
Juglans/química , Síndrome Metabólica/fisiopatologia , Nozes/química , Extratos Vegetais/administração & dosagem , Extratos Vegetais/efeitos adversos , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aromatase/deficiência , Aromatase/genética , Pressão Sanguínea/efeitos dos fármacos , Cruzamentos Genéticos , Suplementos Nutricionais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos/genética
12.
Sci Rep ; 7(1): 8250, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811555

RESUMO

Study of microRNA (miRNAs) using sheep models is limited due to lack of miRNA information. We therefore investigated oar-miRNAs and their regulation in an ovine model of heart failure (HF). Left ventricular (LV) tissue was collected from normal (Cont), HF (LV pacing @ ~220bpm for 13-days) and HF-recovery sheep (HF-R, 26-days after pacing cessation). MiRNA expression was profiled using next-generation sequencing (NGS) and miRNA array, and validated by stem-loop qPCR. Detected sequences were mapped against the ovine genome (Oar v4.0) and aligned with known miRNAs (miRBase v21). A total of 36,438,340 raw reads were obtained with a peak distribution of 18-23 nt. Of these, 637 miRNAs were detected by NGS and mapped to the ovine genome. With cut-off at 10 counts, 275 novel miRNAs were identified (with 186 showing 100% alignment and 89 showing 70-99% alignment with human/mouse and/or rat miRNAs, respectively), and 78 known oar-miRNAs. Cardiac-enriched miRNA-1, -133a, -208a/b and -499 were highly expressed in the LV. With HF induction, miRNA-133b-3p, -208b-3p, -125a-5p, -125b-5p, -126-3p, -21-5p, -210-3p, -29a-3p, -320a and -494-3p were significantly up-regulated relative to Cont and tended to return to normal levels following HF-recovery. This study has expanded the sheep miRNA database, and demonstrated HF-induced regulation of miRNAs.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Miocárdio/metabolismo , Ovinos/genética , Transcriptoma , Animais , Biomarcadores , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica , Sequenciamento de Nucleotídeos em Larga Escala , Especificidade de Órgãos/genética , Reprodutibilidade dos Testes
13.
Int J Cardiol ; 197: 56-65, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26113475

RESUMO

BACKGROUND: Renal dysfunction is a frequent complication and crucial determinant of outcome in acute decompensated heart failure (ADHF). The aim of the study was to assess urocortin 2 (Ucn2) as a short-term therapy in ADHF with a focus on its renal effects. Comparison was made with dobutamine to distinguish effects beyond pure inotropism. METHODS: Sheep with ADHF received a 2-day infusion of a vehicle-control, Ucn2 or dobutamine (n=6/grp). RESULTS: Compared to controls, Ucn2 and dobutamine produced matched improvements in cardiac contractility and output and arterial pressure, whereas reductions in central venous and left atrial pressures were greater with Ucn2. Both agents comparably repressed ADHF-activated hormone systems with the exception of the natriuretic peptides, which fell significantly during dobutamine but not Ucn2. While Ucn2 and dobutamine increased creatinine clearance and urine volume similarly, only Ucn2 induced a significant natriuresis. Ucn2 also decreased collagen deposition in the heart and kidney and suppressed gene expression of collagen-1, transforming growth factor-ß1, components of the angiotensin system (angiotensinogen, angiotensin-converting enzyme, type-1 receptor) and markers of hypertrophy (GATA binding protein-4, ß-myosin heavy chain), apoptosis (caspase3) and inflammation/injury (interleukin-18, cystatin C, neutrophil gelatinase-associated lipocalin, kidney injury molecule-1) in these tissues, while increasing cardiac natriuretic peptide mRNA. In contrast, dobutamine produced reduced or opposite effects on expression of these factors. CONCLUSIONS: Ucn2 administration in ADHF has favorable effects on hemodynamics, the natriuretic peptides and tissue mediators of inflammation, fibrosis, apoptosis and hypertrophy that stand in contrast to dobutamine. These data support Ucn2's potential as a renoprotective therapeutic in this setting.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/uso terapêutico , Modelos Animais de Doenças , Dobutamina/uso terapêutico , Insuficiência Cardíaca/prevenção & controle , Nefropatias/prevenção & controle , Urocortinas/uso terapêutico , Animais , Biomarcadores/metabolismo , Colágeno/metabolismo , Feminino , Expressão Gênica , Coração/efeitos dos fármacos , Insuficiência Cardíaca/metabolismo , Rim/efeitos dos fármacos , Nefropatias/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Ovinos , Fator de Crescimento Transformador beta1/metabolismo
14.
Endocrinology ; 144(6): 2191-4, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12746273

RESUMO

Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) are cardiac hormones, secreted by the atria and ventricles, respectively, in the normal adult heart. They participate in the regulation of blood pressure and body fluid homeostasis and modify growth and development of cardiovascular tissues and bone. Levels of ANP are higher in the fetal circulation than in adults, and fetal ventricles express higher levels of ANP and BNP than adult ventricles. The reappearance of ventricular ANP expression in adults is recognized as a marker of the induction of the embryonic gene program in ventricular hypertrophy. The natriuretic peptide system appears to be functional by midgestation, to respond to volume stimuli, and to regulate blood pressure and salt and water balance in the developing embryo. In addition, the natriuretic peptides may help regulate the blood supply to the fetus, acting as vasodilators in the placental vasculature. Peaks of ANP and BNP expression during gestation coincide with significant events in cardiac organogenesis, suggesting a role for ANP/BNP in the formation of the heart. In knockout mice lacking the natriuretic peptide receptor (NPR)-A gene (Npr1(-/-)), survival is reduced, with hearts enlarged at birth and possible cardiac developmental abnormalities. Surviving adult Npr1(-/-) mice have elevated blood pressure and marked cardiac hypertrophy and fibrosis, indicating that the ANP/BNP system is an important regulator of myocyte growth during development.


Assuntos
Fator Natriurético Atrial/fisiologia , Sistema Cardiovascular/embriologia , Coração/embriologia , Circulação Placentária/fisiologia , Animais , Feminino , Coração/fisiologia , Humanos , Gravidez
15.
Eur J Heart Fail ; 15(10): 1138-47, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23696613

RESUMO

AIMS: Since their identification in the circulation, microRNAs have received considerable interest as putative biomarkers of cardiovascular disease. We have investigated the diagnostic utility of microRNAs in differentiating between patients with heart failure (HF) and non-HF-related breathlessness, and between HF with reduced (HF-REF) and preserved (HF-PEF) EF. METHODS AND RESULTS: MicroRNA profiling was performed on plasma from 32 HF and 15 COPD patients, as well as 14 healthy controls. Seventeen microRNAs were selected for validation in 44 HF, 32 COPD, 59 other breathless, and 15 controls. Cases of HF were split evenly between HF-REF and HF-PEF. Diagnostic utility was compared with NT-proBNP and high sensitivity troponin T (hs-troponin T). MiR-103 [area under the curve (AUC) = 0.642, P = 0.007], miR-142-3p (AUC = 0.668, P = 0.002), miR-199a-3p (AUC = 0.668, P = 0.002), miR-23a (AUC = 0.637, P = 0.010), miR-27b (AUC = 0.642, P = 0.008), miR-324-5p (AUC = 0.621, P = 0.023), and miR-342-3p (AUC = 0.644, P = 0.007) were associated with HF diagnosis in regression and receiver operating characteristic (ROC) analyses. Individually, NT-proBNP (AUC = 0.896, P = 9.68 × 10(-14)) and hs-troponin T (AUC = 0.750, P = 2.50 × 10(-6)) exhibited greater sensitivity and specificity. However, combining significantly associated microRNAs with NT-proBNP improved the AUC of NT-proBNP by 4.6% (P = 0.013). Four microRNAs, miR-103, miR-142-3p, miR-30b, and miR-342-3p, were differentially expressed between HF and controls, COPD, and other breathless patients (P = 0.002-0.030). Eight microRNAs that distinguished between HF-REF and HF-PEF in screening (P = 0.017-0.049) were not replicated in the validation. CONCLUSIONS: Four microRNAs distinguished between HF and exacerbation of COPD, other causes of dyspnoea, and controls. Seven were associated with HF diagnosis in regression and ROC analysis. Although individually NT-proBNP was far superior in predicting HF, combining microRNA levels with NT-proBNP may add diagnostic value.


Assuntos
Dispneia/sangue , Insuficiência Cardíaca/sangue , MicroRNAs/sangue , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Estudos de Casos e Controles , Dispneia/etiologia , Feminino , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/genética , Curva ROC , Análise de Regressão , Volume Sistólico , Troponina T/sangue
16.
Circ Heart Fail ; 5(5): 645-52, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22798523

RESUMO

BACKGROUND: The (pro)renin receptor (P)RR is implicated in blood pressure regulation and the pathophysiology of heart failure (HF). The effects of (P)RR blockade in HF have not been previously investigated. METHODS AND RESULTS: Eight sheep received on 2 separate days a vehicle control and incremental intravenous boluses of a (P)RR antagonist, ovine handle region peptide (HRP) (1, 5, and 25 mg at 90-minute intervals), both before (normal) and after induction of HF by rapid left ventricular pacing. In normal sheep, HRP reduced heart rate (P<0.001) and hematocrit (P=0.019) compared with time-matched control data, without significantly affecting any other hemodynamic, hormonal, or renal variables. In sheep with HF, HRP treatment induced progressive falls in mean arterial pressure (P<0.001) in association with decreases in left atrial pressure (P<0.001), peripheral resistance (P=0.014), and hematocrit (P<0.001). Cardiac contractility tended to decline (P=0.096), whereas cardiac output was unaltered. HRP administration produced a dose-dependent decrease in plasma renin activity (P=0.004), with similar trends observed for plasma angiotensin II and aldosterone (P=0.093 and P=0.088, respectively). Circulating natriuretic peptides, endothelin-1, and catecholamine levels were unchanged. HRP also induced a reduction in plasma sodium concentrations relative to control (P=0.024), a natriuresis (P=0.046), and a tendency for creatinine excretion and clearance to improve. CONCLUSIONS: (P)RR antagonism in experimental HF resulted in cardiovascular and renal benefits in association with inhibition of the renin-angiotensin-aldosterone system. These findings suggest that (P)RR contributes to pressure/volume regulation in HF and identifies the receptor as a potential therapeutic target in this disease.


Assuntos
Fármacos Cardiovasculares/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Hemodinâmica/efeitos dos fármacos , Hormônios/sangue , Rim/efeitos dos fármacos , Oligopeptídeos/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Pressão Arterial/efeitos dos fármacos , Função do Átrio Esquerdo/efeitos dos fármacos , Pressão Atrial/efeitos dos fármacos , Biomarcadores/sangue , Débito Cardíaco/efeitos dos fármacos , Estimulação Cardíaca Artificial , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Rim/metabolismo , Rim/fisiopatologia , Contração Miocárdica/efeitos dos fármacos , Natriurese/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Ovinos , Fatores de Tempo , Resistência Vascular/efeitos dos fármacos , Receptor de Pró-Renina
17.
Hypertension ; 57(6): 1136-44, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21536988

RESUMO

Although acute administration of urocortin 2 has beneficial actions in heart failure, the integrated hemodynamic, hormonal, and renal effects of sustained urocortin 2 treatment in this disease have not been investigated. In the current study, we administered a 4-day infusion of a vehicle control (0.9% saline; n=6) or urocortin 2 (0.75 µg/kg per hour; n=6) to sheep with pacing-induced heart failure. Compared with time-matched controls, infusion of urocortin 2 produced rapid (30-minute) and persistent (4-day) improvements in cardiac contractility (day 4: control 905±73 versus urocortin 2 1424±158 mm Hg/s; P<0.001) and output (2.6±0.1 versus 3.8±0.3 L/min; P<0.001), together with reductions in left atrial pressure (28±1 versus 12±1 mm Hg; P<0.001) and peripheral resistance (30±2 versus 20±2 mm Hg/L per min; P<0.001). In contrast, urocortin 2-induced falls in mean arterial pressure were not established until the second day (day 4: 74±2 versus 72±2 mm Hg; P<0.05). Prolonged urocortin 2 administration was associated with sustained (days 0 to 4) declines in plasma renin activity (day 4: 1.33±0.27 versus 0.73±0.20 nmol/L per hour; P<0.001), aldosterone (970±383 versus 396±96 pmol/L; P<0.05), vasopressin (2.4±0.8 versus 1.3±0.1 pmol/L; P<0.05), endothelin 1 (7.2±0.7 versus 4.5±0.4 pmol/L; P<0.01), and atrial (269±27 versus 150±19 pmol/L; P<0.001) and B-type (65±9 versus 29±6 pmol/L; P<0.001) natriuretic peptides, as well as an acute transient rise in plasma cortisol (day 1: P<0.001). Chronic urocortin 2 also persistently augmented urinary sodium (day 4: 4-fold increase; P<0.001) and creatinine (1.4-fold; P<0.001) excretion and creatinine clearance (1.5-fold; P<0.01) compared with control. Food consumption was temporarily suppressed (P<0.05). In conclusion, 4-day urocortin 2 administration induces sustained improvements in hemodynamics and renal function, in association with inhibition of multiple vasoconstrictor/volume-retaining systems. These findings support the therapeutic potential for urocortin 2 in heart failure.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Urocortinas/farmacologia , Resistência Vascular/efeitos dos fármacos , Aldosterona/sangue , Animais , Pressão Sanguínea/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Creatinina/urina , Sistema Endócrino/efeitos dos fármacos , Sistema Endócrino/fisiopatologia , Endotelina-1/sangue , Feminino , Coração/efeitos dos fármacos , Coração/fisiopatologia , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/urina , Infusões Intravenosas , Rim/efeitos dos fármacos , Rim/fisiopatologia , Camundongos , Oligopeptídeos/sangue , Renina/sangue , Ovinos , Sódio/urina , Fatores de Tempo , Urocortinas/administração & dosagem , Vasopressinas/sangue
18.
BMC Med Genomics ; 1: 64, 2008 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-19114010

RESUMO

BACKGROUND: Reliability of real-time PCR (RT-qPCR) data is dependent on the use of appropriate reference gene(s) for normalization. To date, no validated reference genes have been reported for normalizing gene expression in human myocardium. This study aimed to identify validated reference genes for use in gene expression studies of failed and non-failed human myocardium. METHODS: Bioinformatic analysis of published human heart gene expression arrays (195 failed hearts, 16 donor hearts) was used to identify 10 stable and abundant genes for further testing. The expression stability of these genes was investigated in 28 failed and 28 non-failed human myocardium samples by RT-qPCR using geNorm software. RESULTS: Signal recognition particle 14 kDa (SRP14), tumor protein, translationally-controlled 1 (TPT1) and eukaryotic elongation factor 1A1 (EEF1A1) were ranked the most stable genes. The commonly used reference gene, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was ranked the least stable of the genes tested. The normalization strategy was tested by comparing RT-qPCR data of both normalized and raw expression levels of brain natriuretic peptide precursor (NPPB), a gene known to be up-regulated in heart failure. Non-normalized levels of NPPB exhibited a marginally significant difference between failed and non-failed samples (p = 0.058). In contrast, normalized NPPB expression levels were significantly higher in heart-failed patients compared with controls (p = 0.023). CONCLUSION: This study used publicly available gene array data to identify a strategy for normalization involving two reference genes in combination that may have broad application for accurate and reliable normalization of RT-qPCR data in failed and non-failed human myocardium.

19.
Am J Physiol Heart Circ Physiol ; 291(4): H1952-8, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973826

RESUMO

This study investigated whether gene expression profiles of myofibroblasts derived from infarcted myocardium differ from normal cardiac fibroblasts. We compared the expression of cytoskeletal proteins in cultured ovine cardiac fibroblasts derived from infarcted (ID) and noninfarcted ovine myocardium (NID) and the levels of expression of the natriuretic peptide receptors (NPR)-A and NPR-B in response to treatment with transforming growth factor (TGF)-beta1 and/or platelet-derived growth factor (PDGF). Transformation of cultured cardiac fibroblasts to myofibroblasts, as indicated by alpha-smooth muscle actin and vimentin expression, was independent of the presence of TGF-beta1, PDGF, or cell origin. ID fibroblasts had higher basal levels than NID fibroblasts of NPR-A (ID: 58.0 +/- 32.2 arbitrary density units, NID: undetectable), NPR-B (ID: 780 +/- 155, NID: 330 +/- 38 arbitrary density units) and collagen I (ID: 17.2 +/- 0.5, NID: 10.5 +/- 1.7 pg mRNA/mug total RNA, P < 0.05) but lower levels of alpha-SMa expression (ID: 50.2 +/- 7.9, NID: 76.9 +/- 3.2 fluorescence units, P < 0.05). NPR-A mRNA in ID fibroblasts showed a rapid fourfold increase in response to TGF-beta1 and/or PDGF at 4 and 2 h, respectively, followed by a profound decline; in NID cells, NPR-A mRNA was undetectable. In ID fibroblasts, cytokines reduced NPR-B mRNA below control levels; in NID fibroblasts, TGF-beta1 and PDGF elicited prompt increments in expression: a fourfold increase with TGF-beta1 at 8 h and a twofold increase with PDGF at 4 h (P < 0.05). In summary, transformation of cardiac fibroblasts to myofibroblasts in culture is independent of cytokine treatment. Moreover, whether the cultured cardiac fibroblasts are from infarct tissue is a major determinant of NPR expression levels and cytokine responses, even after four to five passages.


Assuntos
Citocinas/farmacologia , Fibroblastos/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/citologia , Miocárdio/patologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores do Fator Natriurético Atrial/genética , Ovinos , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1
20.
Am J Physiol Heart Circ Physiol ; 283(2): H707-14, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12124219

RESUMO

Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) are cardiac hormones that regulate blood pressure and volume, and exert their biological actions via the natriuretic peptide receptor-A gene (Npr1). Mice lacking Npr1 (Npr(-/-)) have marked cardiac hypertrophy and fibrosis disproportionate to their increased blood pressure. This study examined the relationships between ANP and BNP gene expression, immunoreactivity and fibrosis in cardiac tissue, circulating ANP levels, and ANP and BNP mRNA during embryogenesis in Npr1(-/-) mice. Disruption of the Npr1 signaling pathway resulted in augmented ANP and BNP gene and ANP protein expression in the cardiac ventricles, most pronounced for ANP mRNA in females [414 +/- 57 in Npr1(-/-) ng/mg and 124 +/- 25 ng/mg in wild-type (WT) by Taqman assay, P < 0.001]. This increased expression was highly correlated to the degree of cardiac hypertrophy and was localized to the left ventricle (LV) inner free wall and to areas of ventricular fibrosis. In contrast, plasma ANP was significantly greater than WT in male but not female Npr1(-/-) mice. Increased ANP and BNP gene expression was observed in Npr1(-/-) embryos from 16 days of gestation. Our study suggests that cardiac ventricular expression of ANP and BNP is more closely associated with local hypertrophy and fibrosis than either systemic blood pressure or circulating ANP levels.


Assuntos
Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Cardiomiopatias/metabolismo , Guanilato Ciclase/deficiência , Miocárdio/metabolismo , Peptídeo Natriurético Encefálico/metabolismo , Receptores do Fator Natriurético Atrial/deficiência , Animais , Fator Natriurético Atrial/sangue , Fator Natriurético Atrial/genética , Cardiomegalia/etiologia , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Embrião de Mamíferos/metabolismo , Feminino , Fibrose , Guanilato Ciclase/genética , Ventrículos do Coração , Hipertensão/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout/genética , Peptídeo Natriurético Encefálico/sangue , Peptídeo Natriurético Encefálico/genética , RNA Mensageiro/metabolismo , Receptores do Fator Natriurético Atrial/genética , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA