Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Adv Physiol Educ ; 48(3): 488-497, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38602008

RESUMO

The traditional case study has been used as a learning tool for the past 100 years, and in our program, graduate physiology students are presented with a real-world scenario and must determine the diagnosis and treatment of the patient. We found that students defaulted to memorization of disease with treatment and bypassed gaining an understanding of the mechanistic physiology behind disease and treatment. To adjust our student's approach, we developed a novel way to enhance student learning. To accomplish this shift from memorization to physiological mastery, we created the Inverted Case Study. This approach diverges from the traditional model in that students are given the diagnosis and treatment beforehand and are tasked with explaining the actual physiology of the case. In this way, students can no longer rely on the memorization of symptoms-disease-treatment but rather gain a solid understanding of the physiological mechanisms of the disease since that is the focus of the Inverted Case Study Technique. The Inverted Case Study approach is an effective approach to apply and hone critical thinking skills.NEW & NOTEWORTHY This article presents a novel approach to century-old learning techniques that enhances students' self-reported learning and also their attitudes toward learning mechanistic physiology and increases their perception of preparedness for professional school.


Assuntos
Fisiologia , Humanos , Fisiologia/educação , Aprendizagem Baseada em Problemas/métodos , Avaliação Educacional/métodos , Aprendizagem
2.
J Clin Invest ; 128(11): 4843-4855, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30153110

RESUMO

Cardiac two-pore domain potassium channels (K2P) exist in organisms from Drosophila to humans; however, their role in cardiac function is not known. We identified a K2P gene, CG8713 (sandman), in a Drosophila genetic screen and show that sandman is critical to cardiac function. Mice lacking an ortholog of sandman, TWIK-related potassium channel (TREK-1, also known Kcnk2), exhibit exaggerated pressure overload-induced concentric hypertrophy and alterations in fetal gene expression, yet retain preserved systolic and diastolic cardiac function. While cardiomyocyte-specific deletion of TREK-1 in response to in vivo pressure overload resulted in cardiac dysfunction, TREK-1 deletion in fibroblasts prevented deterioration in cardiac function. The absence of pressure overload-induced dysfunction in TREK-1-KO mice was associated with diminished cardiac fibrosis and reduced activation of JNK in cardiomyocytes and fibroblasts. These findings indicate a central role for cardiac fibroblast TREK-1 in the pathogenesis of pressure overload-induced cardiac dysfunction and serve as a conceptual basis for its inhibition as a potential therapy.


Assuntos
Cardiomegalia/metabolismo , Fibroblastos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Drosophila melanogaster , Fibroblastos/patologia , Fibrose , Humanos , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miócitos Cardíacos/patologia , Canais de Potássio de Domínios Poros em Tandem/genética , Domínios Proteicos
3.
Basic Res Cardiol ; 112(3): 23, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28299467

RESUMO

The myocardial response to pressure overload involves coordination of multiple transcriptional, posttranscriptional, and metabolic cues. The previous studies show that one such metabolic cue, O-GlcNAc, is elevated in the pressure-overloaded heart, and the increase in O-GlcNAcylation is required for cardiomyocyte hypertrophy in vitro. Yet, it is not clear whether and how O-GlcNAcylation participates in the hypertrophic response in vivo. Here, we addressed this question using patient samples and a preclinical model of heart failure. Protein O-GlcNAcylation levels were increased in myocardial tissue from heart failure patients compared with normal patients. To test the role of OGT in the heart, we subjected cardiomyocyte-specific, inducibly deficient Ogt (i-cmOgt -/-) mice and Ogt competent littermate wild-type (WT) mice to transverse aortic constriction. Deletion of cardiomyocyte Ogt significantly decreased O-GlcNAcylation and exacerbated ventricular dysfunction, without producing widespread changes in metabolic transcripts. Although some changes in hypertrophic and fibrotic signaling were noted, there were no histological differences in hypertrophy or fibrosis. We next determined whether significant differences were present in i-cmOgt -/- cardiomyocytes from surgically naïve mice. Interestingly, markers of cardiomyocyte dedifferentiation were elevated in Ogt-deficient cardiomyocytes. Although no significant differences in cardiac dysfunction were apparent after recombination, it is possible that such changes in dedifferentiation markers could reflect a larger phenotypic shift within the Ogt-deficient cardiomyocytes. We conclude that cardiomyocyte Ogt is not required for cardiomyocyte hypertrophy in vivo; however, loss of Ogt may exert subtle phenotypic differences in cardiomyocytes that sensitize the heart to pressure overload-induced ventricular dysfunction.


Assuntos
Cardiomegalia/metabolismo , Insuficiência Cardíaca/metabolismo , Miócitos Cardíacos/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Humanos , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase
4.
Cell Signal ; 28(10): 1580-92, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27169346

RESUMO

ß2-Adrenergic receptors (ß2AR) transactivate epidermal growth factor receptors (EGFR) through formation of a ß2AR-EGFR complex that requires activation of Src to mediate signaling. Here, we show that both lipid and protein kinase activities of the bifunctional phosphoinositide 3-kinase (PI3K) enzyme are required for ß2AR-stimulated EGFR transactivation. Mechanistically, the generation of phosphatidylinositol (3,4,5)-tris-phosphate (PIP3) by the lipid kinase function stabilizes ß2AR-EGFR complexes while the protein kinase activity of PI3K regulates Src activation by direct phosphorylation. The protein kinase activity of PI3K phosphorylates serine residue 70 on Src to enhance its activity and induce EGFR transactivation following ßAR stimulation. This newly identified function for PI3K, whereby Src is a substrate for the protein kinase activity of PI3K, is of importance since Src plays a key role in pathological and physiological signaling.


Assuntos
Receptores ErbB/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ativação Transcricional/genética , Quinases da Família src/metabolismo , Sequência de Aminoácidos , Técnicas Biossensoriais , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , Isoproterenol/farmacologia , Espectrometria de Massas , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinases da Família src/química
5.
Circulation ; 131(24): 2120-30, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-25995315

RESUMO

BACKGROUND: Whether biomechanical force on the heart can induce exosome secretion to modulate cardiovascular function is not known. We investigated the secretion and activity of exosomes containing a key receptor in cardiovascular function, the angiotensin II type I receptor (AT1R). METHODS AND RESULTS: Exosomes containing AT1Rs were isolated from the media overlying AT1R-overexpressing cells exposed to osmotic stretch and from sera of mice undergoing cardiac pressure overload. The presence of AT1Rs in exosomes was confirmed by both electron microscopy and radioligand receptor binding assays and shown to require ß-arrestin2, a multifunctional adaptor protein essential for receptor trafficking. We show that functional AT1Rs are transferred via exosomes in an in vitro model of cellular stretch. Using mice with global and cardiomyocyte conditional deletion of ß-arrestin2, we show that under conditions of in vivo pressure overload the cellular source of the exocytosis of exosomes containing AT1R is the cardiomyocyte. Exogenously administered AT1R-enriched exosomes target cardiomyocytes, skeletal myocytes, and mesenteric resistance vessels and are sufficient to confer blood pressure responsiveness to angiotensin II infusion in AT1R knockout mice. CONCLUSIONS: AT1R-enriched exosomes are released from the heart under conditions of in vivo cellular stress to likely modulate vascular responses to neurohormonal stimulation. In the context of the whole organism, the concept of G protein-coupled receptor trafficking should consider circulating exosomes as part of the reservoir of functional AT1Rs.


Assuntos
Exossomos/química , Miócitos Cardíacos/química , Receptor Tipo 1 de Angiotensina/sangue , Estresse Mecânico , Animais , Arrestinas/deficiência , Arrestinas/genética , Arrestinas/fisiologia , Pressão Sanguínea , Constrição , Exossomos/fisiologia , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Células Musculares/metabolismo , Miócitos Cardíacos/ultraestrutura , Pressão Osmótica , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ensaio Radioligante , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/genética , Resistência Vascular , beta-Arrestinas
6.
J Biol Chem ; 289(43): 29665-76, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25183011

RESUMO

Derangements in metabolism and related signaling pathways characterize the failing heart. One such signal, O-linked ß-N-acetylglucosamine (O-GlcNAc), is an essential post-translational modification regulated by two enzymes, O-GlcNAc transferase and O-GlcNAcase (OGA), which modulate the function of many nuclear and cytoplasmic proteins. We recently reported reduced OGA expression in the failing heart, which is consistent with the pro-adaptive role of increased O-GlcNAcylation during heart failure; however, molecular mechanisms regulating these enzymes during heart failure remain unknown. Using miRNA microarray analysis, we observed acute and chronic changes in expression of several miRNAs. Here, we focused on miR-539 because it was predicted to target OGA mRNA. Indeed, co-transfection of the OGA-3'UTR containing reporter plasmid and miR-539 overexpression plasmid significantly reduced reporter activity. Overexpression of miR-539 in neonatal rat cardiomyocytes significantly suppressed OGA expression and consequently increased O-GlcNAcylation; conversely, the miR-539 inhibitor rescued OGA protein expression and restored O-GlcNAcylation. In conclusion, this work identifies the first target of miR-539 in the heart and the first miRNA that regulates OGA. Manipulation of miR-539 may represent a novel therapeutic target in the treatment of heart failure and other metabolic diseases.


Assuntos
Insuficiência Cardíaca/genética , MicroRNAs/metabolismo , Regulação para Cima/genética , beta-N-Acetil-Hexosaminidases/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Sequência de Bases , Hipóxia Celular/genética , Regulação para Baixo/genética , Glicosilação , Células HEK293 , Testes de Função Cardíaca , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Dados de Sequência Molecular , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Ratos Sprague-Dawley
7.
Circ Heart Fail ; 7(4): 634-42, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24762972

RESUMO

BACKGROUND: Cardiac hypertrophy and heart failure are associated with metabolic dysregulation and a state of chronic energy deficiency. Although several disparate changes in individual metabolic pathways have been described, there has been no global assessment of metabolomic changes in hypertrophic and failing hearts in vivo. Hence, we investigated the impact of pressure overload and infarction on myocardial metabolism. METHODS AND RESULTS: Male C57BL/6J mice were subjected to transverse aortic constriction or permanent coronary occlusion (myocardial infarction [MI]). A combination of LC/MS/MS and GC/MS techniques was used to measure 288 metabolites in these hearts. Both transverse aortic constriction and MI were associated with profound changes in myocardial metabolism affecting up to 40% of all metabolites measured. Prominent changes in branched-chain amino acids were observed after 1 week of transverse aortic constriction and 5 days after MI. Changes in branched-chain amino acids after MI were associated with myocardial insulin resistance. Longer duration of transverse aortic constriction and MI led to a decrease in purines, acylcarnitines, fatty acids, and several lysolipid and sphingolipid species but a marked increase in pyrimidines as well as ascorbate, heme, and other indices of oxidative stress. Cardiac remodeling and contractile dysfunction in hypertrophied hearts were associated with large increases in myocardial, but not plasma, levels of the polyamines putrescine and spermidine as well as the collagen breakdown product prolylhydroxyproline. CONCLUSIONS: These findings reveal extensive metabolic remodeling common to both hypertrophic and failing hearts that are indicative of extracellular matrix remodeling, insulin resistance and perturbations in amino acid, and lipid and nucleotide metabolism.


Assuntos
Cardiomegalia/metabolismo , Metabolismo Energético/fisiologia , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Estresse Oxidativo , Animais , Cardiomegalia/diagnóstico , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Ecocardiografia , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Volume Sistólico , Espectrometria de Massas em Tandem
8.
Am J Physiol Heart Circ Physiol ; 306(1): H142-53, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24186210

RESUMO

The singly coded gene O-linked-ß-N-acetylglucosamine (O-GlcNAc) transferase (Ogt) resides on the X chromosome and is necessary for embryonic stem cell viability during embryogenesis. In mature cells, this enzyme catalyzes the posttranslational modification known as O-GlcNAc to various cellular proteins. Several groups, including our own, have shown that acute increases in protein O-GlcNAcylation are cardioprotective both in vitro and in vivo. Yet, little is known about how OGT affects cardiac function because total body knockout (KO) animals are not viable. Presently, we sought to establish the potential involvement of cardiomyocyte Ogt in cardiac maturation. Initially, we characterized a constitutive cardiomyocyte-specific (cm)OGT KO (c-cmOGT KO) mouse and found that only 12% of the c-cmOGT KO mice survived to weaning age (4 wk old); the surviving animals were smaller than their wild-type littermates, had dilated hearts, and showed overt signs of heart failure. Dysfunctional c-cmOGT KO hearts were more fibrotic, apoptotic, and hypertrophic. Several glycolytic genes were also upregulated; however, there were no gross changes in mitochondrial O2 consumption. Histopathology of the KO hearts indicated the potential involvement of endoplasmic reticulum stress, directing us to evaluate expression of 78-kDa glucose-regulated protein and protein disulfide isomerase, which were elevated. Additional groups of mice were subjected to inducible deletion of cmOGT, which did not produce overt dysfunction within the first couple of weeks of deletion. Yet, long-term loss (via inducible deletion) of cmOGT produced gradual and progressive cardiomyopathy. Thus, cardiomyocyte Ogt is necessary for maturation of the mammalian heart, and inducible deletion of cmOGT in the adult mouse produces progressive ventricular dysfunction.


Assuntos
Miócitos Cardíacos/metabolismo , N-Acetilglucosaminiltransferases/genética , Animais , Apoptose , Cardiomiopatia Dilatada/congênito , Cardiomiopatia Dilatada/patologia , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático , Fibrose/congênito , Fibrose/patologia , Deleção de Genes , Glicólise , Insuficiência Cardíaca/congênito , Insuficiência Cardíaca/patologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , N-Acetilglucosaminiltransferases/metabolismo , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo
9.
PLoS One ; 8(12): e83174, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24367585

RESUMO

Preclinical studies of animals with risk factors, and how those risk factors contribute to the development of cardiovascular disease and cardiac dysfunction, are clearly needed. One such approach is to feed mice a diet rich in fat (i.e. 60%). Here, we determined whether a high fat diet was sufficient to induce cardiac dysfunction in mice. We subjected mice to two different high fat diets (lard or milk as fat source) and followed them for over six months and found no significant decrement in cardiac function (via echocardiography), despite robust adiposity and impaired glucose disposal. We next determined whether antecedent and concomitant exposure to high fat diet (lard) altered the murine heart's response to infarct-induced heart failure; high fat feeding during, or before and during, heart failure did not significantly exacerbate cardiac dysfunction. Given the lack of a robust effect on cardiac dysfunction with high fat feeding, we then examined a commonly used mouse model of overt diabetes, hyperglycemia, and obesity (db/db mice). db/db mice (or STZ treated wild-type mice) subjected to pressure overload exhibited no significant exacerbation of cardiac dysfunction; however, ischemia-reperfusion injury significantly depressed cardiac function in db/db mice compared to their non-diabetic littermates. Thus, we were able to document a negative influence of a risk factor in a relevant cardiovascular disease model; however, this did not involve exposure to a high fat diet. High fat diet, obesity, or hyperglycemia does not necessarily induce cardiac dysfunction in mice. Although many investigators use such diabetes/obesity models to understand cardiac defects related to risk factors, this study, along with those from several other groups, serves as a cautionary note regarding the use of murine models of diabetes and obesity in the context of heart failure.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Animais , Respiração Celular/efeitos dos fármacos , Complicações do Diabetes/etiologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Complicações do Diabetes/fisiopatologia , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/análise , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Hiperglicemia/complicações , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Dilatação Mitocondrial/efeitos dos fármacos
10.
PLoS One ; 8(1): e53951, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308291

RESUMO

This study was designed to test whether reduced levels of cardiac fructose-2,6-bisphosphate (F-2,6-P(2)) exacerbates cardiac damage in response to pressure overload. F-2,6-P(2) is a positive regulator of the glycolytic enzyme phosphofructokinase. Normal and Mb transgenic mice were subject to transverse aortic constriction (TAC) or sham surgery. Mb transgenic mice have reduced F-2,6-P(2) levels, due to cardiac expression of a transgene for a mutant, kinase deficient form of the enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2) which controls the level of F-2,6-P(2). Thirteen weeks following TAC surgery, glycolysis was elevated in FVB, but not in Mb, hearts. Mb hearts were markedly more sensitive to TAC induced damage. Echocardiography revealed lower fractional shortening in Mb-TAC mice as well as larger left ventricular end diastolic and end systolic diameters. Cardiac hypertrophy and pulmonary congestion were more severe in Mb-TAC mice as indicated by the ratios of heart and lung weight to tibia length. Expression of α-MHC RNA was reduced more in Mb-TAC hearts than in FVB-TAC hearts. TAC produced a much greater increase in fibrosis of Mb hearts and this was accompanied by 5-fold more collagen 1 RNA expression in Mb-TAC versus FVB-TAC hearts. Mb-TAC hearts had the lowest phosphocreatine to ATP ratio and the most oxidative stress as indicated by higher cardiac content of 4-hydroxynonenal protein adducts. These results indicate that the heart's capacity to increase F-2,6-P(2) during pressure overload elevates glycolysis which is beneficial for reducing pressure overload induced cardiac hypertrophy, dysfunction and fibrosis.


Assuntos
Aorta/patologia , Cardiomegalia/metabolismo , Frutosedifosfatos/metabolismo , Glicólise , Miocárdio/metabolismo , Fosfofrutoquinase-2/deficiência , Trifosfato de Adenosina/metabolismo , Animais , Aorta/cirurgia , Cardiomegalia/fisiopatologia , Constrição Patológica , Fibrose/metabolismo , Fibrose/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Estresse Oxidativo , Fosfocreatina/metabolismo , Fosfofrutoquinase-2/genética , Remodelação Ventricular
11.
Am J Physiol Heart Circ Physiol ; 302(10): H2122-30, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22408028

RESUMO

The regulation of cardiomyocyte hypertrophy is a complex interplay among many known and unknown processes. One specific pathway involves the phosphatase calcineurin, which regulates nuclear translocation of the essential cardiac hypertrophy transcription factor, nuclear factor of activated T-cells (NFAT). Although metabolic dysregulation is frequently described during cardiac hypertrophy, limited insights exist regarding various accessory pathways. One metabolically derived signal, beta-O-linked N-acetylglucosamine (O-GlcNAc), has emerged as a highly dynamic posttranslational modification of serine and threonine residues regulating physiological and stress processes. Given the metabolic dysregulation during hypertrophy, we hypothesized that NFAT activation is dependent on O-GlcNAc signaling. Pressure overload-induced hypertrophy (via transverse aortic constriction) in mice or treatment of neonatal rat cardiac myocytes with phenylephrine significantly enhanced global O-GlcNAc signaling. NFAT-luciferase reporter activity revealed O-GlcNAc-dependent NFAT activation during hypertrophy. Reversal of enhanced O-GlcNAc signaling blunted cardiomyocyte NFAT-induced changes during hypertrophy. Taken together, these results demonstrate a critical role of O-GlcNAc signaling in NFAT activation during hypertrophy and provide evidence that O-GlcNAc signaling is coordinated with the onset and progression of cardiac hypertrophy. This represents a potentially significant and novel mechanism of cardiac hypertrophy, which may be of particular interest in future in vivo studies of hypertrophy.


Assuntos
Acetilglucosamina/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais/fisiologia , Transcrição Gênica/fisiologia , Animais , Cardiomegalia/patologia , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenilefrina/farmacologia , Processamento de Proteína Pós-Traducional/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol Heart Circ Physiol ; 301(5): H2073-80, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21873502

RESUMO

Cardiac failure is associated with increased levels of oxidized DNA, especially mitochondrial (mtDNA). It is not known if oxidized mtDNA contributes to cardiac dysfunction. To test if protection of mtDNA can reduce cardiac injury, we produced transgenic mice with cardiomyocyte-specific overexpression of the DNA repair enzyme 8-oxoguanine DNA glycosylase 1 (OGG1) isoform 2a. In one line of mice, the transgene increased OGG1 activity by 115% in mitochondria and by 28% in nuclei. OGG1 transgenic mice demonstrated significantly lower cardiac mitochondrial levels of the DNA guanine oxidation product 7,8-dihydro-8-oxoguanine (8-oxo-dG) under basal conditions, after doxorubicin administration, or after transaortic constriction (TAC), but the transgene produced no detectable reduction in nuclear 8-oxo-dG content. OGG1 mice were tested for protection from the cardiac effects of TAC 13 wk after surgery. Compared with FVB-TAC mice, hearts from OGG1-TAC mice had lower levels of ß-myosin heavy chain mRNA but they did not display significant differences in the ratio of heart weight to tibia length or protection of cardiac function measured by echocardiography. The principle benefit of OGG1 overexpression was a significant decrease in TAC-induced cardiac fibrosis. This protection was indicated by reduced Sirius red staining on OGG1 cardiac sections and by significantly decreased induction of collagen 1 and 3 mRNA expression in OGG1 hearts after TAC surgery. These results provide a new model to assess the damaging cardiac effects of 8-oxo-dG formation and suggest that increased repair of 8-oxo-dG in mtDNA decreases cardiac pathology.


Assuntos
DNA Glicosilases/metabolismo , DNA Mitocondrial/metabolismo , Cardiopatias/enzimologia , Mitocôndrias Cardíacas/enzimologia , Miocárdio/enzimologia , Animais , Antibióticos Antineoplásicos/farmacologia , Aorta/cirurgia , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Constrição , Dano ao DNA , DNA Glicosilases/genética , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Fibrose , Regulação da Expressão Gênica , Genótipo , Guanina/análogos & derivados , Guanina/metabolismo , Cardiopatias/diagnóstico por imagem , Cardiopatias/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias Cardíacas/efeitos dos fármacos , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Estresse Oxidativo , Fenótipo , RNA Mensageiro/metabolismo , Ultrassonografia , Regulação para Cima
13.
Amino Acids ; 40(3): 895-911, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20798965

RESUMO

O-linked ß-N-acetylglucosamine (O-GlcNAc) is an inducible, dynamically cycling and reversible post-translational modification of Ser/Thr residues of nucleocytoplasmic and mitochondrial proteins. We recently discovered that O-GlcNAcylation confers cytoprotection in the heart via attenuating the formation of mitochondrial permeability transition pore (mPTP) and the subsequent loss of mitochondrial membrane potential. Because Ca(2+) overload and reactive oxygen species (ROS) generation are prominent features of post-ischemic injury and favor mPTP formation, we ascertained whether O-GlcNAcylation mitigates mPTP formation via its effects on Ca(2+) overload and ROS generation. Subjecting neonatal rat cardiac myocytes (NRCMs, n ≥ 6 per group) to hypoxia, or mice (n ≥ 4 per group) to myocardial ischemia reduced O-GlcNAcylation, which later increased during reoxygenation/reperfusion. NRCMs (n ≥ 4 per group) infected with an adenovirus carrying nothing (control), adenoviral O-GlcNAc transferase (adds O-GlcNAc to proteins, AdOGT), adenoviral O-GlcNAcase (removes O-GlcNAc to proteins, AdOGA), vehicle or PUGNAc (blocks OGA; increases O-GlcNAc levels) were subjected to hypoxia-reoxygenation or H(2)O(2), and changes in Ca(2+) levels (via Fluo-4AM and Rhod-2AM), ROS (via DCF) and mPTP formation (via calcein-MitoTracker Red colocalization) were assessed using time-lapse fluorescence microscopy. Both OGT and OGA overexpression did not significantly (P > 0.05) alter baseline Ca(2+) or ROS levels. However, AdOGT significantly (P < 0.05) attenuated both hypoxia and oxidative stress-induced Ca(2+) overload and ROS generation. Additionally, OGA inhibition mitigated both H(2)O(2)-induced Ca(2+) overload and ROS generation. Although AdOGA exacerbated both hypoxia and H(2)O(2)-induced ROS generation, it had no effect on H(2)O(2)-induced Ca(2+) overload. We conclude that inhibition of Ca(2+) overload and ROS generation (inducers of mPTP) might be one mechanism through which O-GlcNAcylation reduces ischemia/hypoxia-mediated mPTP formation.


Assuntos
Acetilglucosamina/metabolismo , Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Transdução de Sinais , Animais , Células Cultivadas , Glicosilação , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Ratos , Ratos Sprague-Dawley
14.
Proc Natl Acad Sci U S A ; 107(41): 17797-802, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20876116

RESUMO

The failing heart is subject to elevated metabolic demands, adverse remodeling, chronic apoptosis, and ventricular dysfunction. The interplay among such pathologic changes is largely unknown. Several laboratories have identified a unique posttranslational modification that may have significant effects on cardiovascular function. The O-linked ß-N-acetylglucosamine (O-GlcNAc) posttranslational modification (O-GlcNAcylation) integrates glucose metabolism with intracellular protein activity and localization. Because O-GlcNAc is derived from glucose, we hypothesized that altered O-GlcNAcylation would occur during heart failure and figure prominently in its pathophysiology. After 5 d of coronary ligation in WT mice, cardiac O-GlcNAc transferase (OGT; which adds O-GlcNAc to proteins) and levels of O-GlcNAcylation were significantly (P < 0.05) elevated in the surviving remote myocardium. We used inducible, cardiac myocyte-specific Cre recombinase transgenic mice crossed with loxP-flanked OGT mice to genetically delete cardiomyocyte OGT (cmOGT KO) and ascertain its role in the failing heart. After tamoxifen induction, cardiac O-GlcNAcylation of proteins and OGT levels were significantly reduced compared with WT, but not in other tissues. WT and cardiomyocyte OGT KO mice underwent nonreperfused coronary ligation and were followed for 4 wk. Although OGT deletion caused no functional change in sham-operated mice, OGT deletion in infarcted mice significantly exacerbated cardiac dysfunction compared with WT. These data provide keen insights into the pathophysiology of the failing heart and illuminate a previously unrecognized point of integration between metabolism and cardiac function in the failing heart.


Assuntos
Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/fisiopatologia , Miocárdio/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Remodelação Ventricular/fisiologia , Acilação , Animais , Ecocardiografia , Imunofluorescência , Hemodinâmica , Técnicas Histológicas , Immunoblotting , Ligadura , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Tamoxifeno
15.
J Mol Cell Cardiol ; 45(2): 313-25, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18539296

RESUMO

O-linked beta-N-acetylglucosamine (O-GlcNAc) is a dynamic, inducible, and reversible post-translational modification of nuclear and cytoplasmic proteins on Ser/Thr amino acid residues. In addition to its putative role as a nutrient sensor, we have recently shown pharmacologic elevation of O-GlcNAc levels positively affected myocyte survival during oxidant stress. However, no rigorous assessment of the contribution of O-GlcNAc transferase has been performed, particularly in the post-hypoxic setting. Therefore, we hypothesized that pharmacological or genetic manipulation of O-GlcNAc transferase (OGT), the enzyme that adds O-GlcNAc to proteins, would affect cardiac myocyte survival following hypoxia/reoxygenation (H/R). Adenoviral overexpression of OGT (AdOGT) in cardiac myocytes augmented O-GlcNAc levels and reduced post-hypoxic damage. Conversely, pharmacologic inhibition of OGT significantly attenuated O-GlcNAc levels, exacerbated post-hypoxic cardiac myocyte death, and sensitized myocytes to mitochondrial membrane potential collapse. Both genetic deletion of OGT using a cre-lox approach and translational silencing via RNAi also resulted in significant reductions in OGT protein and O-GlcNAc levels, and, exacerbated post-hypoxic cardiac myocyte death. Inhibition of OGT reduced O-GlcNAc levels on voltage dependent anion channel (VDAC) in isolated mitochondria and sensitized to calcium-induced mitochondrial permeability transition pore (mPTP) formation, indicating that mPTP may be an important target of O-GlcNAc signaling and confirming the aforementioned mitochondrial membrane potential results. These data demonstrate that OGT exerts pro-survival actions during hypoxia-reoxygenation in cardiac myocytes, particularly at the level of mitochondria.


Assuntos
Acetilglucosaminidase/fisiologia , Hipóxia/metabolismo , Membranas Intracelulares/fisiologia , Mitocôndrias/metabolismo , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , N-Acetilglucosaminiltransferases/fisiologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/fisiologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/patologia , N-Acetilglucosaminiltransferases/genética , Necrose , Permeabilidade , Ratos , Ratos Sprague-Dawley
16.
Am J Physiol Heart Circ Physiol ; 294(6): H2889-97, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18456722

RESUMO

During ischemia and heart failure, there is an increase in cardiac glycolysis. To understand if this is beneficial or detrimental to the heart, we chronically elevated glycolysis by cardiac-specific overexpression of phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2) in transgenic mice. PFK-2 controls the level of fructose-2,6-bisphosphate (Fru-2,6-P2), an important regulator of phosphofructokinase and glycolysis. Transgenic mice had over a threefold elevation in levels of Fru-2,6-P2. Cardiac metabolites upstream of phosphofructokinase were significantly reduced, as would be expected by the activation of phosphofructokinase. In perfused hearts, the transgene caused a significant increase in glycolysis that was less sensitive to inhibition by palmitate. Conversely, oxidation of palmitate was reduced by close to 50%. The elevation in glycolysis made isolated cardiomyocytes highly resistant to contractile inhibition by hypoxia, but in vivo the transgene had no effect on ischemia-reperfusion injury. Transgenic hearts exhibited pathology: the heart weight-to-body weight ratio was increased 17%, cardiomyocyte length was greater, and cardiac fibrosis was increased. However, the transgene did not change insulin sensitivity. These results show that the elevation in glycolysis provides acute benefits against hypoxia, but the chronic increase in glycolysis or reduction in fatty acid oxidation interferes with normal cardiac metabolism, which may be detrimental to the heart.


Assuntos
Glicólise , Mutação , Traumatismo por Reperfusão Miocárdica/enzimologia , Miocárdio/enzimologia , Fosfofrutoquinase-2/metabolismo , Animais , Hipóxia Celular , Modelos Animais de Doenças , Hipertrofia , Insulina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Contração Muscular , Infarto do Miocárdio/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Miócitos Cardíacos/enzimologia , Oxirredução , Ácido Palmítico/metabolismo , Fosfofrutoquinase-2/genética , Fatores de Tempo , Regulação para Cima
17.
Am J Physiol Heart Circ Physiol ; 291(6): H2743-51, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16844920

RESUMO

3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors increase endothelial nitric oxide synthase (eNOS) activity by multiple mechanisms. We previously reported that genetic overexpression of eNOS improves survival and cardiac function in congestive heart failure (CHF). In the present study, we tested the hypothesis that low-dose treatment with an 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor exerts beneficial effects on survival and/or cardiac function in a murine model of CHF. Mice were subjected to permanent ligation of the left coronary artery and randomized to receive either saline vehicle or simvastatin (0.25 mg/kg) 2 h after myocardial infarction and daily (0.25 mg/kg) for 7 days, followed by 21 days of administration every other day for a total duration of 28 days. Myocardial infarct size was not reduced by simvastatin therapy (P = not significant between groups). Simvastatin treatment did significantly (P < 0.05) improve survival (45%) compared with vehicle treatment (25%). In addition, simvastatin treatment significantly improved (P < 0.01) left ventricular function and significantly (P < 0.01) abrogated cardiac hypertrophy and pulmonary edema compared with vehicle treatment. The protective effects of simvastatin were abrogated by delayed initiation of treatment or genetic ablation of eNOS. In conclusion, low-dose simvastatin therapy significantly improves survival and cardiac function and reduces both cardiac hypertrophy and pulmonary edema via an eNOS-dependent mechanism in a murine model of CHF.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/enzimologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Sinvastatina/farmacologia , Disfunção Ventricular Esquerda/tratamento farmacológico , Disfunção Ventricular Esquerda/enzimologia , Animais , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Insuficiência Cardíaca/fisiopatologia , Inibidores de Hidroximetilglutaril-CoA Redutases/sangue , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Hipertrofia Ventricular Esquerda/enzimologia , Lipídeos/sangue , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/enzimologia , Óxido Nítrico Sintase Tipo III/genética , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/enzimologia , Índice de Gravidade de Doença , Sinvastatina/sangue , Taxa de Sobrevida , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...