Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Cell Cardiol ; 186: 111-124, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38039845

RESUMO

The mechanistic target of rapamycin (mTOR) is evolutionarily conserved from yeast to humans and is one of the most fundamental pathways of living organisms. Since its discovery three decades ago, mTOR has been recognized as the center of nutrient sensing and growth, homeostasis, metabolism, life span, and aging. The role of dysregulated mTOR in common diseases, especially cancer, has been extensively studied and reported. Emerging evidence supports that mTOR critically regulates innate immune responses that govern the pathogenesis of various cardiovascular diseases. This review discusses the regulatory role of mTOR in macrophage functions in acute inflammation triggered by ischemia and in atherosclerotic cardiovascular disease (ASCVD) and heart failure with preserved ejection fraction (HFpEF), in which chronic inflammation plays critical roles. Specifically, we discuss the role of mTOR in trained immunity, immune senescence, and clonal hematopoiesis. In addition, this review includes a discussion on the architecture of mTOR, the function of its regulatory complexes, and the dual-arm signals required for mTOR activation to reflect the current knowledge state. We emphasize future research directions necessary to understand better the powerful pathway to take advantage of the mTOR inhibitors for innovative applications in patients with cardiovascular diseases associated with aging and inflammation.


Assuntos
Doenças Cardiovasculares , Insuficiência Cardíaca , Humanos , Sirolimo/farmacologia , Volume Sistólico , Serina-Treonina Quinases TOR/metabolismo , Inflamação , Macrófagos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina
2.
J Mol Cell Cardiol ; 159: 62-79, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34139235

RESUMO

Monocytes and macrophages are cellular forces that drive and resolve inflammation triggered by acute myocardial ischemia. One of the most important but least understood regulatory mechanisms is how these cells sense cues from the micro-milieu and integrate environmental signals with their response that eventually determines the outcome of myocardial repair. In the current study, we investigated if the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) plays this role. We present evidence that support a robustly activated mTORC1 pathway in monocytes and macrophages in the infarcting myocardium.. Specific mTORC1 inhibition transformed the landscape of cardiac monocytes and macrophages into reparative cells that promoted myocardial healing. As the result, mTORC1 inhibition diminished remodeling and reduced mortality from acute ischemia by 80%. In conclusion, our data suggest a critical role of mTORC1 in regulating the functions of cardiac monocytes and macrophages, and specific mTORC1 inhibition protects the heart from inflammatory injury in acute ischemia. As mTOR/mTORC1 is a master regulator that integrates external signals with cellular responses, the study sheds light on how the cardiac monocytes and macrophages sense and respond to the ischemic environment..


Assuntos
Coração/fisiopatologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Isquemia Miocárdica/metabolismo , Miocárdio/metabolismo , Fagócitos/metabolismo , Remodelação Ventricular/fisiologia , Animais , Macrófagos/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Transdução de Sinais/fisiologia
3.
Acta Neurochir (Wien) ; 163(5): 1383-1389, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33404872

RESUMO

BACKGROUND: Impaired cerebrovascular reactivity appears to be linked to worse global outcome in adult traumatic brain injury (TBI). Literature suggests that current treatments administered in TBI care, in the intensive care unit (ICU), fail to greatly impact recorded cerebrovascular reactivity measures. In particular, the impact of sedation on cerebrovascular reactivity in traumatic brain injury (TBI) remains unclear in vivo. The goal of this study was to preliminarily assess the relationship between objectively measured depth of sedation and cerebrovascular reactivity in TBI. METHODS: Within, we describe a case series of 5 adult TBI patients with TBI, during which objective high-frequency physiology for sedation depth, using bispectral index (BIS), and both intracranial pressure (ICP) and arterial blood pressure (ABP) were recorded. Pressure reactivity index (PRx) and RAP (a metric of cerebral compensatory reserve) were derived. Relationships between cerebrovascular reactivity and compensatory reserve monitoring with BIS metrics were explored using descriptive plots. RESULTS: A total of 5 cases in our prospectively maintained database with high-frequency physiology for ICP, ABP, and BIS. Through error bar plotting, it can be seen that each patient displays a parabolic relationship between BIS and PRx. This suggests a potential "optimal" depth of sedation where cerebrovascular reactivity is the most intact. CONCLUSIONS: This small series highlights the potential impact of depth of sedation on cerebrovascular reactivity in TBI. It suggests that there may be an individual optimal depth of sedation, so as to optimize cerebrovascular reactivity. Further study of objective depth of sedation and its impact on cerebrovascular physiology in TBI is required.


Assuntos
Anestesia , Lesões Encefálicas Traumáticas/fisiopatologia , Circulação Cerebrovascular/fisiologia , Cuidados Críticos , Medicina de Precisão , Adulto , Idoso , Monitores de Consciência , Feminino , Humanos , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Adulto Jovem
4.
J Clin Monit Comput ; 35(2): 307-315, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31989415

RESUMO

Continuous cerebrovascular reactivity assessment in traumatic brain injury (TBI) has been limited by the need for invasive monitoring of either cerebral physiology or arterial blood pressure (ABP). This restricts the application of continuous measures to the acute phase of care, typically in the intensive care unit. It remains unknown if ongoing impairment of cerebrovascular reactivity occurs in the subacute and long-term phase, and if it drives ongoing morbidity in TBI. We describe an entirely non-invasive method for continuous assessment of cerebrovascular reactivity. We describe the technique for entirely non-invasive continuous assessment of cerebrovascular reactivity utilizing near-infrared spectroscopy (NIRS) and robotic transcranial Doppler (rTCD) technology, with details provided for NIRS. Recent advances in continuous high-frequency non-invasive ABP measurement, combined with NIRS or rTCD, can be employed to derive continuous and entirely non-invasive cerebrovascular reactivity metrics. Such non-invasive measures can be obtained during any aspect of patient care post-TBI, and even during outpatient follow-up, avoiding classical intermittent techniques and costly neuroimaging based metrics obtained only at specialized centers. This combination of technology and signal analytic techniques creates avenues for future investigation of the long-term consequences of cerebrovascular reactivity, integrating high-frequency non-invasive cerebral physiology, neuroimaging, proteomics and clinical phenotype at various stages post-injury.


Assuntos
Lesões Encefálicas Traumáticas , Circulação Cerebrovascular , Lesões Encefálicas Traumáticas/diagnóstico por imagem , Homeostase , Humanos , Espectroscopia de Luz Próxima ao Infravermelho , Ultrassonografia Doppler Transcraniana
6.
Am J Physiol Gastrointest Liver Physiol ; 315(1): G66-G79, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29597356

RESUMO

Complement plays a crucial role in microbial defense and clearance of apoptotic cells. Emerging evidence suggests complement is an important contributor to alcoholic liver disease. While complement component 1, Q subcomponent (C1q)-dependent complement activation contributes to ethanol-induced liver injury, the role of the alternative pathway in ethanol-induced injury is unknown. Activation of complement via the classical and alternative pathways was detected in alcoholic hepatitis patients. Female C57BL/6J [wild type (WT)], C1q-deficient ( C1qa-/-, lacking classical pathway activation), complement protein 4-deficient ( C4-/-, lacking classical and lectin pathway activation), complement factor D-deficient ( FD-/-, lacking alternative pathway activation), and C1qa/FD-/- (lacking classical and alternative pathway activation) mice were fed an ethanol-containing liquid diet or pair-fed control diet for 4 or 25 days. Following chronic ethanol exposure, liver injury, steatosis, and proinflammatory cytokine expression were increased in WT but not C1qa-/-, C4-/-, or C1qa/FD-/- mice. In contrast, liver injury, steatosis, and proinflammatory mediators were robustly increased in ethanol-fed FD-/- mice compared with WT mice. Complement activation, assessed by hepatic accumulation of C1q and complement protein 3 (C3) cleavage products (C3b/iC3b/C3c), was evident in livers of WT mice in response to both short-term and chronic ethanol. While C1q accumulated in ethanol-fed FD-/- mice (short term and chronic), C3 cleavage products were detected after short-term but not chronic ethanol. Consistent with impaired complement activation, chronic ethanol induced the accumulation of apoptotic cells and fibrogenic responses in the liver of FD-/- mice. These data highlight the protective role of complement factor D (FD) and suggest that FD-dependent amplification of complement is an adaptive response that promotes hepatic healing and recovery in response to chronic ethanol. NEW & NOTEWORTHY Complement, a component of the innate immune system, is an important pathophysiological contributor to ethanol-induced liver injury. We have identified a novel role for factor D, a component of the alternative pathway, in protecting the liver from ethanol-induced inflammation, accumulation of apoptotic hepatocytes, and profibrotic responses. These data indicate a dual role of complement with regard to inflammatory and protective responses and suggest that accumulation of apoptotic cells impairs hepatic healing/recovery during alcoholic liver disease.


Assuntos
Etanol , Inflamação , Hepatopatias Alcoólicas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Depressores do Sistema Nervoso Central/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Fator D do Complemento/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/fisiologia , Citocinas/imunologia , Etanol/metabolismo , Etanol/farmacologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/metabolismo
7.
Circulation ; 137(24): 2613-2634, 2018 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-29437120

RESUMO

BACKGROUND: Myocardium irreversibly injured by ischemic stress must be efficiently repaired to maintain tissue integrity and contractile performance. Macrophages play critical roles in this process. These cells transform across a spectrum of phenotypes to accomplish diverse functions ranging from mediating the initial inflammatory responses that clear damaged tissue to subsequent reparative functions that help rebuild replacement tissue. Although macrophage transformation is crucial to myocardial repair, events governing this transformation are poorly understood. METHODS: Here, we set out to determine whether innate immune responses triggered by cytoplasmic DNA play a role. RESULTS: We report that ischemic myocardial injury, along with the resulting release of nucleic acids, activates the recently described cyclic GMP-AMP synthase-stimulator of interferon genes pathway. Animals lacking cyclic GMP-AMP synthase display significantly improved early survival after myocardial infarction and diminished pathological remodeling, including ventricular rupture, enhanced angiogenesis, and preserved ventricular contractile function. Furthermore, cyclic GMP-AMP synthase loss of function abolishes the induction of key inflammatory programs such as inducible nitric oxide synthase and promotes the transformation of macrophages to a reparative phenotype, which results in enhanced repair and improved hemodynamic performance. CONCLUSIONS: These results reveal, for the first time, that the cytosolic DNA receptor cyclic GMP-AMP synthase functions during cardiac ischemia as a pattern recognition receptor in the sterile immune response. Furthermore, we report that this pathway governs macrophage transformation, thereby regulating postinjury cardiac repair. Because modulators of this pathway are currently in clinical use, our findings raise the prospect of new treatment options to combat ischemic heart disease and its progression to heart failure.


Assuntos
Citosol/enzimologia , DNA/metabolismo , Macrófagos/enzimologia , Infarto do Miocárdio/enzimologia , Miocárdio/metabolismo , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Animais , Macrófagos/patologia , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia , Nucleotidiltransferases/genética , Remodelação Ventricular
9.
Mol Immunol ; 72: 9-18, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26922040

RESUMO

BACKGROUND: Ethanol feeding in mice activates complement via C1q binding to apoptotic cells in the liver; complement contributes to ethanol-induced inflammation and injury. Despite the critical role of C1q in ethanol-induced injury, the mechanism by which ethanol activates C1q remains poorly understood. Secretory IgM (sIgM), traditionally considered to act as an anti-microbial, also has critical housekeeping functions, facilitating clearance of apoptotic cells, at least in part through activation of C1q. Therefore, we hypothesized that (1) ethanol-induced apoptosis in the liver recruits sIgM, facilitating the activation of C1q and complement and (2) C1INH (C1 esterase inhibitor), which inhibits C1 functional activity, prevents complement activation and decreases ethanol-induced liver injury. METHODS: Female C57BL/6 wild-type, C1qa(-/-), BID(-/-) and sIgM(-/-) mice were fed ethanol containing liquid diets or pair-fed control diets. C1INH or vehicle was given via tail vein injection to ethanol- or pair-fed wild-type mice at 24 and 48h prior to euthanasia. RESULTS: Ethanol exposure increased apoptosis in the liver, as well as the accumulation of IgM in the liver. In the early stages of ethanol feeding, C1q co-localized with IgM in the peri-sinusoidal space of the liver and accumulation of IgM and C3b was dependent on ethanol-induced BID-dependent apoptosis. sIgM(-/-) mice were protected from both ethanol-induced activation of complement and early ethanol-induced liver injury when compared to wild-type mice. Treatment with C1INH also decreased hepatic C3b deposition and ethanol-induced injury. CONCLUSION: These data indicate that sIgM contributes to activation of complement and ethanol-induced increases in inflammatory cytokine expression and hepatocyte injury in the early stages of ethanol-induced liver injury.


Assuntos
Apoptose , Ativação do Complemento , Complemento C1q/imunologia , Imunoglobulina M/imunologia , Hepatopatias Alcoólicas/imunologia , Animais , Proteínas Inativadoras do Complemento 1/farmacologia , Proteína Inibidora do Complemento C1 , Complemento C1q/antagonistas & inibidores , Citocinas/biossíntese , Feminino , Hepatócitos/imunologia , Hepatopatias Alcoólicas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Solubilidade
11.
Cereb Cortex ; 25(3): 578-97, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24046077

RESUMO

Electrophysiological oscillations are thought to create temporal windows of communication between brain regions. We show here that human cortical slices maintained in vitro can generate oscillations similar to those observed in vivo. We have characterized these oscillations using local field potential and whole-cell recordings obtained from neocortical slices acquired during epilepsy surgery. We confirmed that such neocortical slices maintain the necessary cellular and circuitry components, and in particular inhibitory mechanisms, to manifest oscillatory activity when exposed to glutamatergic and cholinergic agonists. The generation of oscillations was dependent on intact synaptic activity and muscarinic receptors. Such oscillations differed in electrographic and pharmacological properties from epileptiform activity. Two types of activity, theta oscillations and high gamma activity, uniquely characterized this model-activity not typically observed in animal cortical slices. We observed theta oscillations to be synchronous across cortical laminae suggesting a novel role of theta as a substrate for interlaminar communication. As well, we observed cross-frequency coupling (CFC) between theta phase and high gamma amplitude similar to that observed in vivo. The high gamma "bursts" generated by such CFC varied in their frequency content, suggesting that this variability may underlie the broadband nature of high gamma activity.


Assuntos
Ritmo Gama , Interneurônios/fisiologia , Neocórtex/fisiologia , Células Piramidais/fisiologia , Lobo Temporal/fisiologia , Ritmo Teta , Adulto , Idoso , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ritmo Gama/efeitos dos fármacos , Humanos , Técnicas In Vitro , Interneurônios/efeitos dos fármacos , Ácido Caínico/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Pessoa de Meia-Idade , Neocórtex/efeitos dos fármacos , Células Piramidais/efeitos dos fármacos , Receptores Muscarínicos/fisiologia , Lobo Temporal/efeitos dos fármacos , Ritmo Teta/efeitos dos fármacos , Adulto Jovem
12.
Pharmacol Res Perspect ; 2(5): e00061, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25089199

RESUMO

The hypoxia-sensing transcriptional factor HIF1α is implicated in a variety of hepato-pathological conditions; however, the contribution of hepatocyte-derived HIF1α during progression of alcoholic liver injury is still controversial. HIF1α induces a variety of genes including those involved in apoptosis via p53 activation. Increased hepatocyte apoptosis is critical for progression of liver inflammation, stellate cell activation and fibrosis. Using hepatocyte-specific HIF1α-deficient mice (ΔHepHIF1α-/-), here we investigated the contribution of HIF1α to ethanol-induced hepatocyte apoptosis and its role in amplification of fibrosis after carbon tetrachloride (CCl4) exposure. Moderate ethanol feeding (11% of Kcal) induced accumulation of hypoxia-sensitive pimonidazole adducts and HIF1α expression in the liver within 4 days of ethanol feeding. Chronic CCl4 treatment increased M30-positive cells, a marker of hepatocyte apoptosis in pair-fed control mice. Concomitant ethanol feeding (11% of Kcal) amplified CCl4-induced hepatocyte apoptosis in livers of wild-type mice, associated with elevated p53K386acetylation, PUMA expression and Ly6c+ cell infiltration. Subsequent to increased apoptosis, ethanol enhanced induction of pro-fibrotic markers including stellate cell activation, collagen 1 expression and extracellular matrix deposition, following CCl4 exposure. Ethanol-induced exacerbation of hepatocyte apoptosis, p53K386 acetylation and PUMA expression following CCl4 exposure was attenuated in livers of ΔHepHIF1α-/- mice. This protection was also associated with a reduction in Ly6c+ cell infiltration and decreased fibrosis in livers of ΔHepHIF1α-/- mice. In summary, these results indicate that moderate ethanol exposure leads to hypoxia/HIF1α-mediated signaling in hepatocytes and induction of p53-dependent apoptosis of hepatocytes, resulting in increased hepatic fibrosis during chronic CCl4 exposure.

13.
Expert Rev Cardiovasc Ther ; 12(9): 1087-98, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25047512

RESUMO

Heart failure has become a huge public health problem. The treatment options for heart failure, however, are considerably limited. The significant disparity between the scope of a prominent health problem and the restricted means of therapy propagates heart failure epidemics. Delineating novel mechanisms of heart failure is imperative. Emerging evidence suggests that epigenetic regulation may take part in the pathogenesis of heart failure. Epigenetic regulation involves DNA and histone modifications that lead to changes in DNA-based transcriptional programs without altering the DNA sequence. Although more and more mechanisms are being discovered, the best understood epigenetic modifications are achieved through covalent biochemical reactions including histone acetylation, histone methylation and DNA methylation. Connecting environmental stimuli with genomic programs, epigenetic regulation remains important in maintaining homeostases and the pathogeneses of diseases. This review summarizes the most recent developments regarding individual epigenetic modifications and their implications in the pathogenesis of heart failure. Understanding this strategically important mechanism is potentially the key for developing powerful interventions in the future.


Assuntos
Metilação de DNA , Epigênese Genética , Insuficiência Cardíaca/fisiopatologia , Acetilação , Animais , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/terapia , Histonas/metabolismo , Humanos
15.
Clin Liver Dis ; 18(1): 157-63, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24274871

RESUMO

Alcoholic liver disease (ALD) remains a major cause of chronic liver diseases and liver failure. Population-based prospective studies and patient cohort studies have demonstrated that obesity and the metabolic syndrome exacerbate progression of ALD and increase hepatocellular carcinoma (HCC) incidence and mortality. Emerging evidence also suggests a synergism between alcohol and obesity in mortality and HCC incidence. Recognition of these increased risks and detection of early-stage liver disease may offer the opportunity to address these modifiable risk factors and prevent disease progression in these patients.


Assuntos
Hepatopatias Alcoólicas/complicações , Síndrome Metabólica/complicações , Obesidade/complicações , Carcinoma Hepatocelular/etiologia , Complicações do Diabetes/etiologia , Progressão da Doença , Humanos , Hepatopatias Alcoólicas/mortalidade , Hepatopatias Alcoólicas/terapia , Neoplasias Hepáticas/etiologia , Fatores de Risco
16.
PLoS One ; 8(7): e69114, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874883

RESUMO

UNLABELLED: The effect of moderate alcohol consumption on liver fibrosis is not well understood, but evidence suggests that adenosine may play a role in mediating the effects of moderate ethanol on tissue injury. Ethanol increases the concentration of adenosine in the liver. Adenosine 2A receptor (A2AR) activation is known to enhance hepatic stellate cell (HSC) activation and A2AR deficient mice are protected from fibrosis in mice. Making use of a novel mouse model of moderate ethanol consumption in which female C57BL/6J mice were allowed continued access to 2% (vol/vol) ethanol (11% calories) or pair-fed control diets for 2 days, 2 weeks or 5 weeks and superimposed with exposure to CCl4, we tested the hypothesis that moderate ethanol consumption increases fibrosis in response to carbon tetrachloride (CCl4) and that treatment of mice with an A2AR antagonist prevents and/or reverses this ethanol-induced increase in liver fibrosis. Neither the expression or activity of CYP2E1, required for bio-activation of CCl4, nor AST and ALT activity in the plasma were affected by ethanol, indicating that moderate ethanol did not increase the direct hepatotoxicity of CCl4. However, ethanol feeding enhanced HSC activation and exacerbated liver fibrosis upon exposure to CCl4. This was associated with an increased sinusoidal angiogenic response in the liver. Treatment with A2AR antagonist both prevented and reversed the ability of ethanol to exacerbate liver fibrosis. CONCLUSION: Moderate ethanol consumption exacerbates hepatic fibrosis upon exposure to CCl4. A2AR antagonism may be a potential pharmaceutical intervention to decrease hepatic fibrosis in response to ethanol.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Bebidas Alcoólicas/efeitos adversos , Tetracloreto de Carbono/toxicidade , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/prevenção & controle , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Feminino , Cirrose Hepática/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Azeite de Oliva , Óleos de Plantas , Purinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Am Heart Assoc ; 2(2): e000016, 2013 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-23568341

RESUMO

BACKGROUND: Mechanical assist device therapy has emerged recently as an important and rapidly expanding therapy in advanced heart failure, triggering in some patients a beneficial reverse remodeling response. However, mechanisms underlying this benefit are unclear. METHODS AND RESULTS: In a model of mechanical unloading of the left ventricle, we observed progressive myocyte atrophy, autophagy, and robust activation of the transcription factor FoxO3, an established regulator of catabolic processes in other cell types. Evidence for FoxO3 activation was similarly detected in unloaded failing human myocardium. To determine the role of FoxO3 activation in cardiac muscle in vivo, we engineered transgenic mice harboring a cardiomyocyte-specific constitutively active FoxO3 mutant (caFoxO3(flox);αMHC-Mer-Cre-Mer). Expression of caFoxO3 triggered dramatic and progressive loss of cardiac mass, robust increases in cardiomyocyte autophagy, declines in mitochondrial biomass and function, and early mortality. Whereas increases in cardiomyocyte apoptosis were not apparent, we detected robust increases in Bnip3 (Bcl2/adenovirus E1B 19-kDa interacting protein 3), an established downstream target of FoxO3. To test the role of Bnip3, we crossed the caFoxO3(flox);αMHC-Mer-Cre-Mer mice with Bnip3-null animals. Remarkably, the atrophy and autophagy phenotypes were significantly blunted, yet the early mortality triggered by FoxO3 activation persisted. Rather, declines in cardiac performance were attenuated by proteasome inhibitors. Consistent with involvement of FoxO3-driven activation of the ubiquitin-proteasome system, we detected time-dependent activation of the atrogenes program and sarcomere protein breakdown. CONCLUSIONS: In aggregate, these data point to FoxO3, a protein activated by mechanical unloading, as a master regulator that governs both the autophagy-lysosomal and ubiquitin-proteasomal pathways to orchestrate cardiac muscle atrophy.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Insuficiência Cardíaca/terapia , Coração Auxiliar , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Remodelação Ventricular/fisiologia , Animais , Atrofia , Autofagia , Modelos Animais de Doenças , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Transplante de Coração , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Transdução de Sinais , Complexos Ubiquitina-Proteína Ligase/fisiologia
18.
Nature ; 491(7426): 692-7, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23192146

RESUMO

A new data set of middle- and upper-stratospheric temperatures based on reprocessing of satellite radiances provides a view of stratospheric climate change during the period 1979-2005 that is strikingly different from that provided by earlier data sets. The new data call into question our understanding of observed stratospheric temperature trends and our ability to test simulations of the stratospheric response to emissions of greenhouse gases and ozone-depleting substances. Here we highlight the important issues raised by the new data and suggest how the climate science community can resolve them.


Assuntos
Altitude , Atmosfera , Temperatura , Incerteza , Atmosfera/análise , Clima , Aquecimento Global/história , Aquecimento Global/estatística & dados numéricos , História do Século XX , História do Século XXI , Atividades Humanas , Modelos Teóricos , Astronave , Fatores de Tempo
19.
J Hum Reprod Sci ; 5(1): 26-31, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22870011

RESUMO

OBJECTIVE: The aim of the present study was to evaluate the prevalence of metabolic syndrome in women with polycystic ovary syndrome (PCOS). SETTING: Infertility clinic in a tertiary care hospital. STUDY DESIGN: A prospective cross-sectional study. MATERIALS AND METHODS: All the women attending the infertility clinic categorized as polycystic ovary syndrome according to Rotterdam criteria (2003) during the study period were included in the study. The women with PCOS underwent screening for metabolic syndrome as defined by the modified American Heart Association/National Heart Lung Blood Institute (AHA/NHLBI) modified ATP 111 (2005) definition. A multivariate logistic regression analysis was applied and significant predictors identified for the prediction of metabolic syndrome. RESULTS: The overall prevalence of metabolic syndrome according to the modified AHA/NHLBI ATP III (2005) criteria was 37.5%. A total of 5.8 % cases were detected to have diabetes mellitus, 8.3% had impaired fasting glucose, and 11.7 % had an impaired glucose test. Dyslipidemia was present in 93.3% cases of PCOS. Among all the risk factors, age and waist hip ratio ≥0.85 were strongly associated with the presence of metabolic syndrome. CONCLUSION: Infertile women with PCOS, particularly those with age ≥25 years or with central obesity (a waist hip ratio of ≥0.85), are at a higher risk of developing metabolic syndrome and should be offered screening tests.

20.
Alcohol Clin Exp Res ; 36(7): 1139-47, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22273278

RESUMO

BACKGROUND: Correlative evidence indicates that apoptosis is associated with the progression of alcoholic liver disease. If apoptosis contributes to ethanol (EtOH)-induced steatohepatitis and/or fibrosis, then mice deficient in Bid, a key pro-apoptotic Bcl-2 family member, or mice treated with a pan-caspase inhibitor (VX166) should be resistant to EtOH-induced liver injury. METHODS: This hypothesis was tested in mice using a model of chronic, heavy EtOH-induced liver injury, as well as in a model in which moderate EtOH feeding accelerated the appearance of early markers of hepatic fibrosis in response to acute carbon tetrachloride (CCl(4) ) exposure. RESULTS: Chronic EtOH feeding to mice increased TUNEL- and cytokeratin-18-positive cells in the liver, as well as the expression of receptor-interacting protein kinase 3 (RIP3), a marker of necroptosis. In this model, Bid-/- mice or wild-type mice treated with VX166 were protected from EtOH-induced apoptosis, but not EtOH-induced RIP3 expression. Bid deficiency or inhibition of caspase activity did not protect mice from EtOH-induced increases in plasma alanine and aspartate amino transferase activity, steatosis, or mRNA expression of some inflammatory cytokines. Moderate EtOH feeding to mice enhanced the response of mice to acute CCl(4) exposure, resulting in increased expression of α-smooth muscle actin and accumulation of extracellular matrix protein. VX166-treatment attenuated EtOH-mediated acceleration of these early indicators of CCl(4) -induced hepatic fibrosis, decreasing the expression of α-smooth muscle actin, and the accumulation of extracellular matrix protein. CONCLUSIONS: EtOH-induced apoptosis of hepatocytes was mediated by Bid. Apoptosis played a critical role in the accelerating the appearance of early markers of CCl(4) -induced fibrosis by moderate EtOH but did not contribute to EtOH-induced hepatocyte injury, steatosis, or expression of mRNA for some inflammatory cytokines.


Assuntos
Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/deficiência , Tetracloreto de Carbono/toxicidade , Etanol/toxicidade , Fígado Gorduroso Alcoólico/patologia , Mediadores da Inflamação/toxicidade , Hepatopatias Alcoólicas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Fígado Gorduroso Alcoólico/metabolismo , Fígado Gorduroso Alcoólico/prevenção & controle , Feminino , Hepatite Alcoólica/metabolismo , Hepatite Alcoólica/patologia , Hepatite Alcoólica/prevenção & controle , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Cirrose Hepática Alcoólica/metabolismo , Cirrose Hepática Alcoólica/patologia , Cirrose Hepática Alcoólica/prevenção & controle , Hepatopatias Alcoólicas/patologia , Hepatopatias Alcoólicas/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA