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.
Proc Natl Acad Sci U S A ; 109(11): 4314-9, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22366318

RESUMO

Although protein S-nitrosylation is increasingly recognized as mediating nitric oxide (NO) signaling, roles for protein denitrosylation in physiology remain unknown. Here, we show that S-nitrosoglutathione reductase (GSNOR), an enzyme that governs levels of S-nitrosylation by promoting protein denitrosylation, regulates both peripheral vascular tone and ß-adrenergic agonist-stimulated cardiac contractility, previously ascribed exclusively to NO/cGMP. GSNOR-deficient mice exhibited reduced peripheral vascular tone and depressed ß-adrenergic inotropic responses that were associated with impaired ß-agonist-induced denitrosylation of cardiac ryanodine receptor 2 (RyR2), resulting in calcium leak. These results indicate that systemic hemodynamic responses (vascular tone and cardiac contractility), both under basal conditions and after adrenergic activation, are regulated through concerted actions of NO synthase/GSNOR and that aberrant denitrosylation impairs cardiovascular function. Our findings support the notion that dynamic S-nitrosylation/denitrosylation reactions are essential in cardiovascular regulation.


Assuntos
Fenômenos Fisiológicos Cardiovasculares , Glutationa Redutase/metabolismo , Álcool Desidrogenase , Animais , Cálcio/metabolismo , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Diástole/efeitos dos fármacos , Feminino , Glutationa Redutase/deficiência , Hemodinâmica/efeitos dos fármacos , Isoproterenol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/efeitos dos fármacos , Miocárdio/citologia , Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Óxido Nítrico Sintase/metabolismo , Nitrosação , Transporte Proteico/efeitos dos fármacos , Receptores Adrenérgicos beta/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Vasodilatação/efeitos dos fármacos
2.
South Med J ; 101(10): 1046-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18791506

RESUMO

Evidence suggests that sepsis is a systemic inflammatory condition complicated by dysequilibrium in coagulation and fibrinolytic homeostasis, with a shift in the balance towards increased coagulation over fibrinolysis. Protein C is a natural anticoagulant consumed and inactivated during sepsis. We present the case of a 33-year-old female, postsplenectomy, with pneumococcal sepsis and purpura fulminans, successfully treated with activated protein C.


Assuntos
Anticoagulantes/uso terapêutico , Vasculite por IgA/tratamento farmacológico , Proteína C/uso terapêutico , Adulto , Feminino , Humanos , Vasculite por IgA/etiologia , Infecções Pneumocócicas/complicações , Sepse/complicações , Sepse/microbiologia
3.
Nitric Oxide ; 16(3): 331-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17307368

RESUMO

Disruption of leptin signaling in the heart may contribute to obesity-related cardiac disease, as leptin deficient (oblob) mice display cardiac hypertrophy, increased cardiac apoptosis and reduced survival. Since leptin maintains a tonic level of neuronal nitric oxide synthase (NOS1) expression in the brain, we hypothesized that leptin deficiency would decrease NOS1 cardiac expression, in turn activating xanthine oxidoreductase (XOR) and creating nitroso-redox imbalance. We studied 2- to 6-month-old oblob (n=26) and C57Bl/6 controls (n=27). Cardiac NOS1 protein abundance (P<0.01) and mRNA expression (P=0.03) were reduced in oblob (n=10 and 6, respectively), while NOS3 protein abundance and mRNA expression were unaltered. Importantly, cardiac NOS1 protein abundance was restored towards normal in oblob mice after leptin treatment (n=3; P<0.05 vs leptin untreated oblob mice). NO metabolite (nitrite and nitrate) production within the myocardium was also reduced in oblob mice (n=5; P=0.02). Furthermore, oxidative stress was increased in oblob mice as GSH/GSSG ratio was decreased (n=4; P=0.02). Whereas XOR activity measured by Amplex Red fluorescence was increased (n=8; P=0.04), XOR and NADPH oxidase subunits protein abundance were not changed in oblob mice (n=6). Leptin deficiency did not disrupt NOS1 subcellular localization, as NOS1 co-localized with ryanodine receptor but not with caveolin-3. In conclusion, leptin deficiency is linked to decreased cardiac expression of NOS1 and NO production, with a concomitant increase in XOR activity and oxidative stress, resulting in nitroso-redox imbalance. These data offer novel insights into potential mechanisms of myocardial dysfunction in obesity.


Assuntos
Leptina/metabolismo , Miocárdio/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Obesidade/metabolismo , Estresse Oxidativo , Animais , Glutationa/metabolismo , Leptina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Miocárdio/citologia , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III , Nitritos/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Xantina Oxidase/genética , Xantina Oxidase/metabolismo
4.
Am J Physiol Heart Circ Physiol ; 292(3): H1328-35, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17071724

RESUMO

Xanthine oxidase (XO) activity contributes to both abnormal excitation-contraction (EC) coupling and cardiac remodeling in heart failure (HF). beta-Adrenergic hyporesponsiveness and abnormalities in Ca(2+) cycling proteins are mechanistically linked features of the HF phenotype. Accordingly, we hypothesized that XO influences beta-adrenergic responsiveness and expression of genes whose products participate in deranged EC coupling. We measured inotropic (dP/dt(max)), lusitropic (tau), and vascular (elastance; E(a)) responses to beta-adrenergic (beta-AR) stimulation with dobutamine in conscious dogs administered allopurinol (100 mg po daily) or placebo during a 4-wk induction of pacing HF. With HF induction, the decreases in both baseline and dobutamine-stimulated inotropic responses were offset by allopurinol. Additionally, allopurinol converted a vasoconstrictor effect to dobutamine to a vasodilator response and enhanced both lusitropic and preload reducing effects. To assess molecular correlates for this phenotype, we measured myocardial sarcoplasmic reticulum Ca(2+)-ATPase 2a (SERCA), phospholamban (PLB), phosphorylated PLB (P-PLB), and Na(+)/Ca(2+) transporter (NCX) gene expression and protein. Although SERCA mRNA and protein concentrations did not change with HF, both PLB and NCX were upregulated (P < 0.05). Additionally, P-PLB and protein kinase A activity were greatly reduced. Allopurinol ameliorated all of these molecular alterations and preserved the PLB-to-SERCA ratio. Preventing maladaptive alterations of Ca(2+) cycling proteins represents a novel mechanism for XO inhibition-mediated preservation of cardiac function in HF, raising the possibility that anti-oxidant therapies for HF may ameliorate transcriptional changes associated with adverse cardiac remodeling and beta-adrenergic hyporesponsiveness.


Assuntos
Alopurinol/uso terapêutico , Proteínas de Ligação ao Cálcio/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Alopurinol/farmacocinética , Animais , Cálcio , Modelos Animais de Doenças , Cães , Coração/efeitos dos fármacos , Coração/fisiopatologia , Contração Miocárdica/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia , Xantina Oxidase/metabolismo
5.
Circ Res ; 98(2): 271-9, 2006 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-16357304

RESUMO

Increased reactive oxygen species (ROS) generation is implicated in cardiac remodeling in heart failure (HF). As xanthine oxidoreductase (XOR) is 1 of the major sources of ROS, we tested whether XOR inhibition could improve cardiac performance and induce reverse remodeling in a model of established HF, the spontaneously hypertensive/HF (SHHF) rat. We randomized Wistar Kyoto (WKY, controls, 18 to 21 months) and SHHF (19 to 21 months) rats to oxypurinol (1 mmol/L; n=4 and n=15, respectively) or placebo (n=3 and n=10, respectively) orally for 4 weeks. At baseline, SHHF rats had decreased fractional shortening (FS) (31+/-3% versus 67+/-3% in WKY, P<0.0001) and increased left-ventricular (LV) end-diastolic dimension (9.7+/-0.2 mm versus 7.0+/-0.4 mm in WKY, P<0.0001). Whereas placebo and oxypurinol did not change cardiac architecture in WKY, oxypurinol attenuated decreased FS and elevated LV end-diastolic dimension, LV end-systolic dimension, and LV mass in SHHF. Increased myocyte width in SHHF was reduced by oxypurinol. Additionally, fetal gene activation, altered calcium cycling proteins, and upregulated phospho-extracellular signal-regulated kinase were restored toward normal by oxypurinol (P<0.05 versus placebo-SHHF). Importantly, SHHF rats exhibited increased XOR mRNA expression and activity, and oxypurinol treatment reduced XOR activity and superoxide production toward normal, but not expression. On the other hand, NADPH oxidase activity remained unchanged, despite elevated subunit protein abundance in treated and untreated SHHF rats. Together these data demonstrate that chronic XOR inhibition restores cardiac structure and function and offsets alterations in fetal gene expression/Ca2+ handling pathways, supporting the idea that inhibiting XOR-derived oxidative stress substantially improves the HF phenotype.


Assuntos
Cardiomiopatia Dilatada/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Remodelação Ventricular/efeitos dos fármacos , Xantina Oxidase/antagonistas & inibidores , Animais , ATPases Transportadoras de Cálcio/metabolismo , Cardiomiopatia Dilatada/enzimologia , Cardiomiopatia Dilatada/fisiopatologia , Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/fisiopatologia , Miocárdio/enzimologia , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Oxipurinol/farmacologia , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Espécies Reativas de Oxigênio , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Xantina Oxidase/fisiologia
6.
Circ Res ; 98(1): 119-24, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16339484

RESUMO

Disruption of leptin signaling is associated with obesity, heart failure, and cardiac hypertrophy, but the role of leptin in cardiac myocyte apoptosis is unknown. We tested the hypothesis that apoptosis increases in leptin-deficient ob/ob and leptin-resistant db/db mice and is associated with aging and left ventricular hypertrophy, increased DNA damage, and decreased survival. We studied young (2- to 3-month-old) and old (12- to 14-month-old) ob/ob and db/db mice and wild-type (WT) controls (n=2 to 4 per group). As expected, ventricular wall thickness and heart weights were similar among young ob/ob, db/db, and WT mice, but higher in old ob/ob and db/db versus old WT. Young ob/ob and db/db showed markedly elevated apoptosis by TUNEL staining and caspase 3 levels compared with WT. Differences in apoptosis were further accentuated with age. Leptin treatment significantly reduced apoptosis in ob/ob mice both in intact hearts and isolated myocytes. Tissue triglycerides were increased in ob/ob hearts, returning to WT levels after leptin repletion. Furthermore, the DNA damage marker, 8oxoG (8-oxo-7,8-dihydroguanidine), was increased, whereas the DNA repair marker, MYH glycosylase, was decreased in old ob/ob and db/db compared with old WT mice. Both ob/ob and db/db mice had decreased survival compared with WT mice. We conclude that leptin-deficient and leptin-resistant mice demonstrate increased apoptosis, DNA damage, and mortality compared with WT mice, suggesting that normal leptin signaling is necessary to prevent excess age-associated DNA damage and premature mortality. These data offer novel insights into potential mechanisms of myocardial dysfunction and early mortality in obesity.


Assuntos
Apoptose , Dano ao DNA , Miócitos Cardíacos/patologia , Obesidade/patologia , Animais , Cardiomegalia/etiologia , Reparo do DNA , Marcação In Situ das Extremidades Cortadas , Leptina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/mortalidade , Estresse Oxidativo , Transdução de Sinais , Triglicerídeos/análise
7.
Circulation ; 112(22): 3415-22, 2005 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-16301341

RESUMO

BACKGROUND: Neuronal nitric oxide synthase (NOS1) plays key cardiac physiological roles, regulating excitation-contraction coupling and exerting an antioxidant effect that maintains tissue NO-redox equilibrium. After myocardial infarction (MI), NOS1 translocates from the sarcoplasmic reticulum to the cell membrane, where it inhibits beta-adrenergic contractility, an effect previously predicted to have adverse consequences. Counter to this idea, we tested the hypothesis that NOS1 has a protective effect after MI. METHODS AND RESULTS: We studied mortality, cardiac remodeling, and upregulation of oxidative stress pathways after MI in NOS1-deficient (NOS1(-/-)) and wild-type C57BL6 (WT) mice. Compared with WT, NOS1(-/-) mice had greater mortality (hazard ratio, 2.06; P=0.036), worse left ventricular (LV) fractional shortening (19.7+/-1.5% versus 27.2+/-1.5%, P<0.05), higher LV diastolic diameter (5.5+/-0.2 versus 4.9+/-0.1 mm, P<0.05), greater residual cellular width (14.9+/-0.5 versus 12.8+/-0.5 microm, P<0.01), and equivalent beta-adrenergic hyporesponsiveness despite similar MI size. Superoxide production increased after MI in both NOS1(-/-) and WT animals, although NO increased only in WT. NADPH oxidase (P<0.05) activity increased transiently in both groups after MI, but NOS1(-/-) mice had persistent basal and post-MI elevations in xanthine oxidoreductase activity. CONCLUSIONS: Together these findings support a protective role for intact NOS1 activity in the heart after MI, despite a potential contribution to LV dysfunction through beta-adrenergic hyporesponsiveness. NOS1 deficiency contributes to an imbalance between oxidative stress and tissue NO signaling, providing a plausible mechanism for adverse consequences of NOS1 deficiency in states of myocardial injury.


Assuntos
Cardiomegalia/enzimologia , Infarto do Miocárdio/complicações , Infarto do Miocárdio/mortalidade , Óxido Nítrico Sintase Tipo I/deficiência , Animais , Cardiomegalia/etiologia , Cardiomegalia/mortalidade , Homeostase , Camundongos , Camundongos Knockout , Mortalidade , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo I/fisiologia , Oxirredução , Estresse Oxidativo , Superóxidos/metabolismo , Disfunção Ventricular Esquerda/etiologia
8.
Int J Cardiol ; 102(2): 333-40, 2005 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-15982506

RESUMO

BACKGROUND: Giant cell myocarditis is a rapidly progressive and often fatal condition without a clear etiology or treatment. A better understanding of giant cell myocarditis pathogenesis is critical to developing treatments to prevent progression and reverse damage. We compared the gene expression of giant cell myocarditis with that of nonfailing hearts. METHODS: Left ventricular samples from two giant cell myocarditis patients harvested during ventricular assist device placement and six unused donor hearts were examined using Affymetrix U133A microarrays. Differential gene expression was defined with a Bonferroni-adjusted p value < or = 0.05 from a Student's t-test and an absolute fold change > or = 2.0. Select gene expression was confirmed with quantitative PCR. RESULTS: Of 115 differentially expressed genes, most were upregulated in giant cell myocarditis and involved in immune response, transcriptional regulation, and metabolism. T-cell activation genes included chemokine receptor 4; chemokine ligands 5, 9, 13, and 18; interleukin-10 receptor alpha; and beta-2 integrin. CONCLUSIONS: Gene expression analysis of giant cell myocarditis offers novel insights into its pathogenesis, namely the role of T-cell activators of the Th1 subset and immune response genes previously implicated in heart failure. This forms the basis for future work aimed at defining novel therapeutic targets for giant cell myocarditis.


Assuntos
Expressão Gênica , Genes MHC da Classe II , Células Gigantes , Miocardite , RNA/genética , Linfócitos T/imunologia , Adulto , Biópsia , Análise por Conglomerados , Feminino , Genes MHC da Classe II/genética , Genes MHC da Classe II/imunologia , Células Gigantes/imunologia , Células Gigantes/patologia , Ventrículos do Coração/imunologia , Ventrículos do Coração/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Miocardite/genética , Miocardite/imunologia , Miocardite/patologia , Reação em Cadeia da Polimerase , RNA Mitocondrial , Reprodutibilidade dos Testes
9.
J Mol Cell Cardiol ; 39(3): 531-6, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15963530

RESUMO

We hypothesized that chronic xanthine oxidase inhibition (XOI) would have favorable effects on both ventricular and vascular performance in evolving heart failure (HF), thereby preserving ventricular-vascular coupling. In HF, XOI reduces oxidative stress and improves both vascular and myocardial function. Dogs were randomized to receive either allopurinol (100 mg/day p.o.) or placebo following surgical instrumentation for chronic measurement of left-ventricular pressure and dimension and during induction of HF by rapid pacing. In the placebo group (n = 8), HF was characterized by increased LV end-diastolic pressure (LVEDP, 10.2 +/- 5.5 and 29.8 +/- 3.9 mmHg, before and after HF, respectively, P < 0.05), end-diastolic dimension (LVEDD, from 29.5 +/- 3.2 to 34.3 +/- 3.2 mm, P < 0.001), and afterload (arterial elastance, Ea, from 17.9 +/- 1.2 to 42.6 +/- 7.9 mmHg/mm, P < 0.05), and reduced contractility (End-systolic ventricular elastance, Ees, from 10.8 +/- 1.3 to 5.6 +/- 2.3 mmHg/mm, P < 0.05). Thus, ventricular-vascular coupling (Ees/Ea ratio) fell 57.6+/-9% (0.61 +/- 0.1 to 0.16 +/- 0.1, P < 0.05). Allopurinol (n = 9) profoundly attenuated both the Ea increase (from 22.3 +/- 3 to 25.6 +/- 4.6 mmHg/mm, P = NS) and the fall in Ees (from 11.8+/-1.1 to 11.7+/-1, P = NS), thereby preserving the Ees/Ea ratio (from 0.58 +/- 0.1 to 0.56 +/- 0.1, P < 0.001 vs. placebo). Allopurinol did not affect the increase in preload (LVEDP and LVEDD). XO cardiac mRNA and protein were similarly upregulated approximately fourfold in both groups. Allopurinol ameliorates increases in afterload and reductions in myocardial contractility during evolving HF, thereby preserving ventricular-vascular coupling. These results demonstrate a unique and potent hemodynamic profile of XOI, thereby providing further rationale for developing XOIs as a novel HF therapy.


Assuntos
Estimulação Cardíaca Artificial/efeitos adversos , Sistema Cardiovascular/fisiopatologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Xantina Oxidase/antagonistas & inibidores , Alopurinol/farmacologia , Animais , Cães , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/etiologia , Hemodinâmica/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , Distribuição Aleatória , Regulação para Cima/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos
10.
Physiol Genomics ; 21(3): 299-307, 2005 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15769906

RESUMO

Cardiomyopathy can be initiated by many factors, but the pathways from unique inciting mechanisms to the common end point of ventricular dilation and reduced cardiac output are unclear. We previously described a microarray-based prediction algorithm differentiating nonischemic (NICM) from ischemic cardiomyopathy (ICM) using nearest shrunken centroids. Accordingly, we tested the hypothesis that NICM and ICM would have both shared and distinct differentially expressed genes relative to normal hearts and compared gene expression of 21 NICM and 10 ICM samples with that of 6 nonfailing (NF) hearts using Affymetrix U133A GeneChips and significance analysis of microarrays. Compared with NF, 257 genes were differentially expressed in NICM and 72 genes in ICM. Only 41 genes were shared between the two comparisons, mainly involved in cell growth and signal transduction. Those uniquely expressed in NICM were frequently involved in metabolism, and those in ICM more often had catalytic activity. Novel genes included angiotensin-converting enzyme-2 (ACE2), which was upregulated in NICM but not ICM, suggesting that ACE2 may offer differential therapeutic efficacy in NICM and ICM. In addition, a tumor necrosis factor receptor was downregulated in both NICM and ICM, demonstrating the different signaling pathways involved in heart failure pathophysiology. These results offer novel insight into unique disease-specific gene expression that exists between end-stage cardiomyopathy of different etiologies. This analysis demonstrates that transcriptome analysis offers insight into pathogenesis-based therapies in heart failure management and complements studies using expression-based profiling to diagnose heart failure of different etiologies.


Assuntos
Cardiomiopatias/genética , Regulação da Expressão Gênica , Insuficiência Cardíaca/genética , Isquemia Miocárdica/genética , Peptidil Dipeptidase A/genética , Enzima de Conversão de Angiotensina 2 , Cardiomiopatias/enzimologia , Cardiomiopatias/terapia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/terapia , Coração Auxiliar , Humanos , Família Multigênica , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/terapia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
11.
J Physiol ; 565(Pt 2): 463-74, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15760936

RESUMO

Impaired leptin signalling in obesity is increasingly implicated in cardiovascular pathophysiology. To explore mechanisms for leptin activity in the heart, we hypothesized that physiological leptin signalling participates in maintaining cardiac beta-adrenergic regulation of excitation-contraction coupling. We studied 10-week-old (before development of cardiac hypertrophy) leptin-deficient (ob/ob, n=12) and C57Bl/6 (wild-type (WT), n=15) mice at baseline and after recombinant leptin infusion (0.3 mg kg-1 day-1 for 28 days, n=6 in each group). Ob/ob-isolated myocytes had attenuated sarcomere shortening and calcium transients ([Ca2+]i) versus WT (P<0.01 for both) following stimulation of the beta-receptor (with isoproterenol (isoprenaline)) or at the post-receptor level (with forskolin and dibutryl-cAMP). In addition, sarcoplasmic reticulum (SR) Ca2+ stores were depressed. Leptin replenishment in ob/ob mice restored each of these abnormalities towards normal without affecting gross (wall thickness) or microscopic (cell size) measures of cardiac architecture. Immunoblots revealed alterations of several proteins involved in excitation-contraction coupling in the ob/ob mice, including decreased abundance of Gsalpha-52 kDa, as well as alterations in the expression of Ca2+ cycling proteins (increased SR Ca2+-ATPase, and depressed phosphorylated phospholamban). In addition, protein kinase A (PKA) activity in ob/ob mice was depressed at baseline and correctable towards the activity found in WT with leptin repletion, a finding that could account for impaired beta-adrenergic responsiveness. Taken together, these data reveal a novel link between the leptin signalling pathway and normal cardiac function and suggest a mechanism by which leptin deficiency or resistance may lead to cardiac depression.


Assuntos
Cardiomegalia/fisiopatologia , Leptina/genética , Leptina/farmacologia , Contração Miocárdica/fisiologia , Obesidade/fisiopatologia , Receptores Adrenérgicos beta/metabolismo , Adenilil Ciclases/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Fatores Etários , Animais , Western Blotting , Cálcio/metabolismo , Cardiomegalia/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Isoproterenol/farmacologia , Leptina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Contração Miocárdica/efeitos dos fármacos , Obesidade/metabolismo , Fenótipo , Receptores para Leptina , Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
Clin Cancer Res ; 10(24): 8683-6, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15623653

RESUMO

The molecular mechanism of lactoferrin-induced cell growth inhibition is incompletely understood. Studying head and neck cancer cells treated with human lactoferrin, we observed growth arrest in three of four cell lines tested. This growth arrest was caused by cell cycle inhibition at the G0-G1 checkpoint. Lactoferrin-induced growth inhibition was associated with a large increase in p27 protein, accompanied by decreased phosphorylation of retinoblastoma protein, and suppression of cyclin E. Decreased levels of phosphorylated Akt were also observed in lactoferrin-sensitive cell lines after treatment. These findings suggest that in head and neck cancer cells the growth inhibitory effects of lactoferrin are mediated through a p27/cyclin E-dependent pathway that may be modulated in part by changes in Akt phosphorylation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fase G1/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/metabolismo , Lactoferrina/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Ciclina E/metabolismo , Inibidor de Quinase Dependente de Ciclina p27 , Relação Dose-Resposta a Droga , Regulação para Baixo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas
13.
Circulation ; 110(22): 3444-51, 2004 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-15557369

RESUMO

BACKGROUND: Gene expression profiling refines diagnostic and prognostic assessment in oncology but has not yet been applied to myocardial diseases. We hypothesized that gene expression differentiates ischemic and nonischemic cardiomyopathy, demonstrating that gene expression profiling by clinical parameters is feasible in cardiology. METHODS AND RESULTS: Affymetrix U133A microarrays of 48 myocardial samples from Johns Hopkins Hospital (JHH) and the University of Minnesota (UM) obtained (1) at transplantation or left ventricular assist device (LVAD) placement (end-stage; n=25), (2) after LVAD support (post-LVAD; n=16), and (3) from newly diagnosed patients (biopsy; n=7) were analyzed with prediction analysis of microarrays. A training set was used to develop the profile and test sets to validate the accuracy of the profile. An etiology prediction profile developed in end-stage JHH samples was tested in independent samples from both JHH and UM with 100% sensitivity and 100% specificity in end-stage samples and 33% sensitivity and 100% specificity in both post-LVAD and biopsy samples. The overall sensitivity was 89% (95% CI 75% to 100%), and specificity was 89% (95% CI 60% to 100%) over 210 random partitions of end-stage samples into training and test sets. Age, gender, and hemodynamic differences did not affect the profile's accuracy in stratified analyses. Select gene expression was confirmed with quantitative polymerase chain reaction. CONCLUSIONS: Gene expression profiling accurately predicts cardiomyopathy etiology, is generalizable to samples from separate institutions, is specific to disease stage, and is unaffected by differences in clinical characteristics. This strongly supports ongoing efforts to incorporate expression profiling-based biomarkers in determining prognosis and response to therapy in heart failure.


Assuntos
Cardiomiopatias/diagnóstico , Cardiomiopatias/genética , Perfilação da Expressão Gênica , Isquemia Miocárdica/complicações , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Cardiomiopatias/classificação , Cardiomiopatias/etiologia , Divisão Celular/genética , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Diagnóstico Diferencial , Estudos de Viabilidade , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Isquemia Miocárdica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Transdução de Sinais/genética
14.
Proc Natl Acad Sci U S A ; 101(45): 15944-8, 2004 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-15486091

RESUMO

Although interactions between superoxide (O(2)(.-)) and nitric oxide underlie many physiologic and pathophysiologic processes, regulation of this crosstalk at the enzymatic level is poorly understood. Here, we demonstrate that xanthine oxidoreductase (XOR), a prototypic superoxide O(2)(.-) -producing enzyme, and neuronal nitric oxide synthase (NOS1) coimmunoprecipitate and colocalize in the sarcoplasmic reticulum of cardiac myocytes. Deficiency of NOS1 (but not endothelial NOS, NOS3) leads to profound increases in XOR-mediated O(2)(.-) production, which in turn depresses myocardial excitation-contraction coupling in a manner reversible by XOR inhibition with allopurinol. These data demonstrate a unique interaction between a nitric oxide and an O(2)(.-) -generating enzyme that accounts for crosstalk between these signaling pathways; these findings demonstrate a direct antioxidant mechanism for NOS1 and have pathophysiologic implications for the growing number of disease states in which increased XOR activity plays a role.


Assuntos
Contração Miocárdica/fisiologia , Miócitos Cardíacos/fisiologia , Óxido Nítrico Sintase/metabolismo , Xantina Oxidase/metabolismo , Animais , Sequência de Bases , Sinalização do Cálcio , DNA Complementar/genética , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Estresse Oxidativo , Retículo Sarcoplasmático/metabolismo , Frações Subcelulares/enzimologia , Superóxidos/metabolismo
15.
Circulation ; 108(16): 2000-6, 2003 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-14517171

RESUMO

BACKGROUND: Although abnormal L-arginine NO signaling contributes to endothelial dysfunction in the aging cardiovascular system, the biochemical mechanisms remain controversial. L-arginine, the NO synthase (NOS) precursor, is also a substrate for arginase. We tested the hypotheses that arginase reciprocally regulates NOS by modulating L-arginine bioavailability and that arginase is upregulated in aging vasculature, contributing to depressed endothelial function. METHODS AND RESULTS: Inhibition of arginase with (S)-(2-boronoethyl)-L-cysteine, HCl (BEC) produced vasodilation in aortic rings from young (Y) adult rats (maximum effect, 46.4+/-9.4% at 10(-5) mol/L, P<0.01). Similar vasorelaxation was elicited with the additional arginase inhibitors N-hydroxy-nor-L-arginine (nor-NOHA) and difluoromethylornithine (DFMO). This effect required intact endothelium and was prevented by 1H-oxadiazole quinoxalin-1-one (P<0.05 and P<0.001, respectively), a soluble guanylyl cyclase inhibitor. DFMO-elicited vasodilation was greater in old (O) compared with Y rat aortic rings (60+/-6% versus 39+/-6%, P<0.05). In addition, BEC restored depressed L-arginine (10(-4) mol/L)-dependent vasorelaxant responses in O rings to those of Y. Arginase activity and expression were increased in O rings, whereas NOS activity and cyclic GMP levels were decreased. BEC and DFMO suppressed arginase activity and restored NOS activity and cyclic GMP levels in O vessels to those of Y. CONCLUSIONS: These findings demonstrate that arginase modulates NOS activity, likely by regulating intracellular L-arginine availability. Arginase upregulation contributes to endothelial dysfunction of aging and may therefore be a therapeutic target.


Assuntos
Envelhecimento/metabolismo , Arginase/metabolismo , Vasos Sanguíneos/fisiopatologia , Endotélio Vascular/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Envelhecimento/patologia , Animais , Arginase/antagonistas & inibidores , Arginina/farmacologia , Vasos Sanguíneos/enzimologia , Vasos Sanguíneos/patologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase , Técnicas In Vitro , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Guanilil Ciclase Solúvel , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
16.
Int J Cancer ; 107(1): 46-52, 2003 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-12925955

RESUMO

Alterations in chromosomal number and structure are found in most solid malignancies including head and neck squamous cell carcinoma (HNSC), however, the presence of ongoing, chromosomal instability in HNSC and its relation to spindle assembly checkpoint defects has not been formally demonstrated. We investigated the status of chromosomal instability (CIN) in HNSC primary tumors and cell lines as well as spindle assembly checkpoint integrity in HNSC cell lines. Centromeric fluorescence in situ hybridization (FISH) was carried out on expanded single cell-derived colonies from HNSC cell lines and primary HNSC touch preparations. The deviation of chromosomes from the modal number in single cell derived colonies was 18.4-27% in 6 HNSC cell lines, and 2-3% in a control cell line, HCT116. Twelve primary tumors and 4 normal controls were also studied; all primary tumors demonstrated significant deviation from the modal chromosomal number (average 33.7%, range = 29.9-43.9%), compared to normal controls (average 4.6%, range = 3.6-5.6%). Additional characterization of the rate of chromosomal breakage was carried out by dual color FISH simultaneously using centromeric and telomeric probes for individual chromosomes on expanded singe cell-derived colonies and primary HNSC. Control HCT 116 colonies demonstrated a mean discordance between number of centromeric and telomeric hybridization signals in 21% (range = 19-23%) of cells, whereas HNSC cell line colonies demonstrated a mean discordance of 50% (range = 38-55%), with the majority of instances of discordant signal indicating telomeric loss. Similarly, touch preparations from primary HNSC demonstrated discordance in hybridization signal of centromeric vs. telomeric signal of 26.3% (range = 18.5-42%), with normal controls showing a rate of discordance of 6.4% (range = 4-8%). Finally, all 6 HNSC cell lines demonstrated partial impairment of mitotic arrest in response to nocodazole, indicating that impairment of the spindle assembly checkpoint may contribute to chromosomal instability in HNSC. Ongoing instability in chromosomal number and structure are consistent features of primary HNSC and cell lines. Spindle assembly checkpoint impairment occurs in HNSC cell lines and may contribute to chromosomal instability in HNSC.


Assuntos
Carcinoma de Células Escamosas/genética , Aberrações Cromossômicas , Neoplasias de Cabeça e Pescoço/genética , Fuso Acromático/genética , Centrossomo , Cromossomos/genética , DNA de Neoplasias/análise , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Mitose , Células Tumorais Cultivadas
17.
Circ Res ; 92(12): 1322-9, 2003 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-12764022

RESUMO

The mechanisms by which nitric oxide (NO) influences myocardial Ca2+ cycling remain controversial. Because NO synthases (NOS) have specific spatial localization in cardiac myocytes, we hypothesized that neuronal NOS (NOS1) found in cardiac sarcoplasmic reticulum (SR) preferentially regulates SR Ca2+ release and reuptake resulting in potentiation of the cardiac force-frequency response (FFR). Transesophageal pacing (660 to 840 bpm) in intact C57Bl/6 mice (WT) stimulated both contractility (dP/dtmax normalized to end-diastolic volume; dP/dt-EDV) by 51+/-5% (P<0.001) and lusitropy (tau; tau) by 20.3+/-2.0% (P<0.05). These responses were markedly attenuated in mice lacking NOS1 (NOS1-/-) (15+/-2% increase in dP/dt-EDV; P<0.001 versus WT; and no change in tau; P<0.01 versus WT). Isolated myocytes from NOS1-/- (approximately 2 months of age) also exhibited suppressed frequency-dependent sarcomere shortening and Ca2+ transients ([Ca2+]i) compared with WT. SR Ca2+ stores, a primary determinant of the FFR, increased at higher frequencies in WT (caffeine-induced [Ca2+]i at 4 Hz increased 107+/-23% above 1 Hz response) but not in NOS1-/- (13+/-26%; P<0.01 versus WT). In contrast, mice lacking NOS3 (NOS3-/-) had preserved FFR in vivo, as well as in isolated myocytes with parallel increases in sarcomere shortening, [Ca2+]i, and SR Ca2+ stores. NOS1-/- had increased SR Ca2+ ATPase and decreased phospholamban protein abundance, suggesting compensatory increases in SR reuptake mechanisms. Together these data demonstrate that NOS1 selectively regulates the cardiac FFR via influences over SR Ca2+ cycling. Thus, there is NOS isoform-specific regulation of different facets of rate-dependent excitation-contraction coupling; inactivation of NOS1 has the potential to contribute to the pathophysiology of states characterized by diminished frequency-dependent inotropic responses.


Assuntos
Cálcio/metabolismo , Contração Miocárdica/fisiologia , Óxido Nítrico/fisiologia , Animais , Pressão Sanguínea/fisiologia , Western Blotting , Tamanho Celular/fisiologia , Estimulação Elétrica , Feminino , Genótipo , Hemodinâmica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Musculares/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Sarcômeros/fisiologia , Retículo Sarcoplasmático/metabolismo , Função Ventricular Esquerda/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...