Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
J Intern Med ; 286(1): 2-15, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30623510

RESUMO

Progress in the testing of therapies targeting the immune response following trauma, a leading cause of morbidity and mortality worldwide, has been slow. We propose that the design of interventional trials in trauma would benefit from a scheme or platform that could support the identification and implementation of prognostic strategies for patient stratification. Here, we propose a stratification scheme based on defined time periods or windows following the traumatic event. This 'time-window' model allows for the incorporation of prognostic variables ranging from circulating biomarkers and clinical data to patient-specific information such as gene variants to predict adverse short- or long-term outcomes. A number of circulating biomarkers, including cell injury markers and damage-associated molecular patterns (DAMPs), and inflammatory mediators have been shown to correlate with adverse outcomes after trauma. Likewise, several single nucleotide polymorphisms (SNPs) associate with complications or death in trauma patients. This review summarizes the status of our understanding of the prognostic value of these classes of variables in predicting outcomes in trauma patients. Strategies for the incorporation of these prognostic variables into schemes designed to stratify trauma patients, such as our time-window model, are also discussed.


Assuntos
Modelos Teóricos , Ferimentos e Lesões/imunologia , Biomarcadores/análise , Ensaios Clínicos como Assunto/métodos , Humanos , Prognóstico , Fatores de Tempo , Ferimentos e Lesões/classificação , Ferimentos e Lesões/terapia
2.
Cell Death Differ ; 23(9): 1428-47, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-26943325

RESUMO

Infection is a common clinical complication following tissue damage resulting from surgery and severe trauma. Studies have suggested that cell pre-activation by antecedent trauma/tissue damage profoundly impacts the response of innate immune cells to a secondary infectious stimulus. Cell necroptosis, a form of regulated inflammatory cell death, is one of the mechanisms that control cell release of inflammatory mediators from important innate immune executive cells such as macrophages (Mφ), which critically regulate the progress of inflammation. In this study, we investigated the mechanism and role of trauma/tissue damage in the regulation of LPS-induced Mφ necroptosis using a mouse model simulating long-bone fracture. We demonstrate that LPS acting through Toll-like receptor (TLR) 4 promotes Mφ necroptosis. However, necroptosis is ameliorated by high-mobility group box 1 (HMGB1) release from damaged tissue. We show that HMGB1 acting through cell surface receptor for advanced glycation end products (RAGE) upregulates caveolin-1 expression, which in turn induces caveolae-mediated TLR4 internalization and desensitization to decrease Mφ necroptosis. We further show that RAGE-MyD88 activation of Cdc42 and subsequent activation of transcription factor Sp1 serves as a mechanism underlying caveolin-1 transcriptional upregulation. These results reveal a previous unidentified protective role of damage-associated molecular pattern (DAMP) molecules in restricting inflammation in response to exogenous pathogen-associated molecular pattern molecules.


Assuntos
Apoptose/efeitos dos fármacos , Fraturas Ósseas/patologia , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Necrose/fisiopatologia , Alarminas/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Células Cultivadas , Fraturas Ósseas/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fator 88 de Diferenciação Mieloide/metabolismo , Fármacos Neuroprotetores/farmacologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Regulação para Cima
4.
J Intern Med ; 276(5): 425-43, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24935761

RESUMO

High-mobility group box 1 (HMGB1) was originally defined as a ubiquitous nuclear protein, but it was later determined that the protein has different roles both inside and outside of cells. Nuclear HMGB1 regulates chromatin structure and gene transcription, whereas cytosolic HMGB1 is involved in inflammasome activation and autophagy. Extracellular HMGB1 has drawn attention because it can bind to related cell signalling transduction receptors, such as the receptor for advanced glycation end products, Toll-like receptor (TLR)2, TLR4 and TLR9. It also participates in the development and progression of a variety of diseases. HMGB1 is actively secreted by stimulation of the innate immune system, and it is passively released by ischaemia or cell injury. This review focuses on the important role of HMGB1 in the pathogenesis of acute and chronic sterile inflammatory conditions. Strategies that target HMGB1 have been shown to significantly decrease inflammation in several disease models of sterile inflammation, and this may represent a promising clinical approach for treatment of certain conditions associated with sterile inflammation.


Assuntos
Proteína HMGB1/fisiologia , Inflamação/fisiopatologia , Animais , Compartimento Celular , Quimiocina CXCL12/metabolismo , Proteína HMGB1/imunologia , Proteína HMGB1/metabolismo , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Oxirredução , Receptor para Produtos Finais de Glicação Avançada , Receptores CXCR4/metabolismo , Receptores Imunológicos/metabolismo , Receptores Toll-Like/metabolismo
5.
Cell Death Differ ; 21(8): 1229-39, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24769733

RESUMO

Macrophages can be activated and regulated by high-mobility group box 1 (HMGB1), a highly conserved nuclear protein. Inflammatory functions of HMGB1 are mediated by binding to cell surface receptors, including the receptor for advanced glycation end products (RAGE), Toll-like receptor (TLR)2, TLR4, and TLR9. Pyroptosis is a caspase-1-dependent programmed cell death, which features rapid plasma membrane rupture, DNA fragmentation, and release of proinflammatory intracellular contents. Pyroptosis can be triggered by various stimuli, however, the mechanism underlying pyroptosis remains unclear. In this study, we identify a novel pathway of HMGB1-induced macrophage pyroptosis. We demonstrate that HMGB1, acting through RAGE and dynamin-dependent signaling, initiates HMGB1endocytosis, which in turn induces cell pyroptosis. The endocytosis of HMGB1 triggers a cascade of molecular events, including cathepsin B release from ruptured lysosomes followed by pyroptosome formation and caspase-1 activation. We further confirm that HMGB1-induced macrophage pyroptosis also occurs in vivo during endotoxemia, suggesting a pathophysiological significance for this form of pyroptosis in the development of inflammation. These findings shed light on the regulatory role of ligand-receptor internalization in directing cell fate, which may have an important role in the progress of inflammation following infection and injury.


Assuntos
Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Animais , Apoptose/fisiologia , Endocitose/fisiologia , Proteína HMGB1/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transfecção
6.
Oncogene ; 33(5): 567-77, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23318458

RESUMO

Tumor cells require increased adenosine triphosphate (ATP) to support anabolism and proliferation. The precise mechanisms regulating this process in tumor cells are unknown. Here, we show that the receptor for advanced glycation endproducts (RAGE) and one of its primary ligands, high-mobility group box 1 (HMGB1), are required for optimal mitochondrial function within tumors. We found that RAGE is present in the mitochondria of cultured tumor cells as well as primary tumors. RAGE and HMGB1 coordinately enhanced tumor cell mitochondrial complex I activity, ATP production, tumor cell proliferation and migration. Lack of RAGE or inhibition of HMGB1 release diminished ATP production and slowed tumor growth in vitro and in vivo. These findings link, for the first time, the HMGB1-RAGE pathway with changes in bioenergetics. Moreover, our observations provide a novel mechanism within the tumor microenvironment by which necrosis and inflammation promote tumor progression.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Proteína HMGB1/metabolismo , Neoplasias Pancreáticas/patologia , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Trifosfato de Adenosina/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Butadienos/farmacologia , Antígeno CD24/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cicloeximida/farmacologia , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Metabolismo Energético , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína HMGB1/efeitos dos fármacos , Humanos , Inflamação/metabolismo , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Nitrilas/farmacologia , Neoplasias Pancreáticas/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor para Produtos Finais de Glicação Avançada/genética , Rotenona/farmacologia , Transdução de Sinais , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Microambiente Tumoral , Desacopladores
7.
Am J Transplant ; 12(11): 2938-48, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23016570

RESUMO

Ischemia/reperfusion injury (IRI) is the most common cause of early mortality following lung transplantation (LTx). We hypothesized that nitrite, an endogenous source of nitric oxide (NO), may protect lung grafts from IRI. Rat lung grafts were stored in preservation solution at 4°C for 6 hours. Both grafts and recipients were treated with nitrite. Nitrite treatment was associated with significantly higher levels of tissue oxygenation, lower levels of cytokines and neutrophil/macrophage infiltration, lower myeloperoxidase activity, reduced oxidative injury and increased cGMP levels in grafts than in the controls. Treatment with either a nitric oxide scavenger or a soluble guanylyl cyclase (sGC) inhibitor diminished the beneficial effects of nitrite and decreased cGMP concentrations. These results suggest that nitric oxide, generated from nitrite, is the molecule responsible for the effects of nitrite via the nitric oxide/sGC/cGMP pathway. Allopurinol, a xanthine oxidoreductase (XOR) inhibitor, abrogated the protective effects of nitrite, suggesting that XOR is a key enzyme in the conversion of nitrite to nitric oxide. In vitro experiments demonstrated that nitrite prevented apoptosis in pulmonary endothelial cells. Nitrite also exhibits longer survival rate in recipients than control. In conclusion, nitrite inhibits lung IRI following cold preservation and had higher survival rate in LTx model.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Transplante de Pulmão/efeitos adversos , Nitritos/farmacologia , Estresse Oxidativo/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Lesão Pulmonar Aguda/etiologia , Animais , Modelos Animais de Doenças , Rejeição de Enxerto , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Pulmão/métodos , Masculino , Óxido Nítrico/metabolismo , Peroxidase/metabolismo , Distribuição Aleatória , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Valores de Referência
8.
Neurogastroenterol Motil ; 24(7): 658-69, e294, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22489868

RESUMO

BACKGROUND: Early murine endotoxin-induced ileus at 6 h is exclusively mediated by non-hemopoietic TLR4/MyD88 signaling despite molecular activation of hemopoietic cells which included a significant IL-6 mRNA induction. Our objective was to define the role of hemopoietic cells in LPS/TLR4-triggered ileus and inflammation over time, and identify mechanisms of ileus. METHODS: CSF-1(-/-) , TLR4 non-chimera and TLR4 chimera mice were single-shot intraperitoneal injected with ultrapure lipopolysaccharide (UP-LPS) and studied up to 4 days. Subgroups of TLR4(WT) mice were additionally intravenously injected with exogenous recombinant IL-6 (rmIL-6) or murine soluble IL-6 receptor blocking antibody (anti-sIL-6R mAB). KEY RESULTS: Hemopoietic TLR4 signaling independently mediated UP-LPS-induced ileus at 24 h, but chemotactic muscularis neutrophil extravasation was not causatively involved and mice lacking CSF-1-dependent macrophages died prematurely. Synergy of hemopoietic and non-hemopoietic cells determined ileus severity and mortality which correlated with synergistic cell lineage specific transcription of inflammatory mediators like IL-6 within the intestinal muscularis. Circulating IL-6 levels were LPS dose dependent, but exogenous rmIL-6 did not spark off a self-perpetuating inflammatory response triggering ileus. Sustained therapeutic inhibition of functional IL-6 signaling efficiently ameliorated late ileus while preemptive antibody-mediated IL-6R blockade was marginally effective in mitigating ileus. However, IL-6R blockade did not prevent endotoxin-associated mortality nor did it alter circulating IL-6 levels. CONCLUSIONS & INFERENCES: A time-delayed bone marrow-driven mechanism of murine endotoxin-induced ileus exists, and hemopoietic cells synergize with non-hemopoietic cells thereby prolonging ileus and fueling intestinal inflammation. Importantly, IL-6 signaling via IL-6R/gp130 drives late ileus, yet it did not regulate mortality in endotoxic shock.


Assuntos
Íleus/metabolismo , Interleucina-6/metabolismo , Choque Séptico/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem da Célula , Íleus/patologia , Imuno-Histoquímica , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Contração Muscular/fisiologia , Músculo Liso/fisiologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Choque Séptico/patologia
9.
Mucosal Immunol ; 3(6): 578-93, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20664577

RESUMO

In humans, environmental exposure to a high dose of lipopolysaccharide (LPS) protects from allergic asthma, the immunological underpinnings of which are not well understood. In mice, exposure to a high LPS dose blunted house dust mite-induced airway eosinophilia and T-helper 2 (Th2) cytokine production. Although adoptively transferred Th2 cells induced allergic airway inflammation in control mice, they were unable to do so in LPS-exposed mice. LPS promoted the development of a CD11b(+)Gr1(int)F4/80(+) lung-resident cell resembling myeloid-derived suppressor cells in a Toll-like receptor 4 and myeloid differentiation factor 88 (MyD88)-dependent manner that suppressed lung dendritic cell (DC)-mediated reactivation of primed Th2 cells. LPS effects switched from suppressive to stimulatory in MyD88(-/-) mice. Suppression of Th2 effector function was reversed by anti-interleukin-10 (IL-10) or inhibition of arginase 1. Lineage(neg) bone marrow progenitor cells could be induced by LPS to develop into CD11b(+)Gr1(int)F4/80(+)cells both in vivo and in vitro that when adoptively transferred suppressed allergen-induced airway inflammation in recipient mice. These data suggest that CD11b(+)Gr1(int)F4/80(+) cells contribute to the protective effects of LPS in allergic asthma by tempering Th2 effector function in the tissue.


Assuntos
Hipersensibilidade/imunologia , Lipopolissacarídeos/administração & dosagem , Pulmão/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Transferência Adotiva , Animais , Anticorpos Bloqueadores/administração & dosagem , Antígenos de Diferenciação/biossíntese , Antígeno CD11b/biossíntese , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Eosinofilia , Humanos , Hipersensibilidade/fisiopatologia , Hipersensibilidade/terapia , Terapia de Imunossupressão , Interleucina-10/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células Mieloides/patologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Pyroglyphidae/imunologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/patologia
10.
Libyan J Med ; 4(3): 97-103, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21483522

RESUMO

Traumatic injury/hemorrhagic shock (T/HS) elicits an acute inflammatory response that may result in death. Inflammation describes a coordinated series of molecular, cellular, tissue, organ, and systemic responses that drive the pathology of various diseases including T/HS and traumatic brain injury (TBI). Inflammation is a finely tuned, dynamic, highly-regulated process that is not inherently detrimental, but rather required for immune surveillance, optimal post-injury tissue repair, and regeneration. The inflammatory response is driven by cytokines and chemokines and is partially propagated by damaged tissue-derived products (Damage-associated Molecular Patterns; DAMP's). DAMPs perpetuate inflammation through the release of pro-inflammatory cytokines, but may also inhibit anti-inflammatory cytokines. Various animal models of T/HS in mice, rats, pigs, dogs, and non-human primates have been utilized in an attempt to move from bench to bedside. Novel approaches, including those from the field of systems biology, may yield therapeutic breakthroughs in T/HS and TBI in the near future.

11.
Am J Transplant ; 8(10): 2015-24, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18727697

RESUMO

Ischemia/reperfusion (I/R) injury during small intestinal transplantation (SITx) frequently causes complications including dysmotility, inflammation and organ failure. Recent evidence indicates hydrogen inhalation eliminates toxic hydroxyl radicals. Syngeneic, orthotopic SITx was performed in Lewis rats with 3 h of cold ischemic time. Both donor and recipient received perioperative air or 2% hydrogen inhalation. SITx caused a delay in gastrointestinal transit and decreased jejunal circular muscle contractile activity 24 h after surgery. Hydrogen treatment resulted in significantly improved gastrointestinal transit, as well as jejunal smooth muscle contractility in response to bethanechol. The transplant induced upregulation in the inflammatory mediators CCL2, IL-1 beta, IL-6 and TNF-alpha were mitigated by hydrogen. Hydrogen significantly diminished lipid peroxidation compared to elevated tissue malondialdehyde levels in air-treated grafts demonstrating an antioxidant effect. Histopathological mucosal erosion and increased gut permeability indicated a breakdown in posttransplant mucosal barrier function which was significantly attenuated by hydrogen treatment. In recipient lung, hydrogen treatment also resulted in a significant abatement in inflammatory mRNA induction and reduced neutrophil recruitment. Hydrogen inhalation significantly ameliorates intestinal transplant injury and prevents remote organ inflammation via its antioxidant effects. Administration of perioperative hydrogen gas may be a potent and clinically applicable therapeutic strategy for intestinal I/R injury.


Assuntos
Hidrogênio/uso terapêutico , Intestinos/patologia , Estresse Oxidativo , Traumatismo por Reperfusão/terapia , Transplante/métodos , Administração por Inalação , Animais , Antioxidantes/metabolismo , Gases , Hidrogênio/administração & dosagem , Inflamação , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Transplantes/efeitos adversos
12.
J Leukoc Biol ; 83(3): 558-63, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17938274

RESUMO

High mobility group box 1 (HMGB1) is a nuclear factor released extracellularly as a late mediator of lethality in sepsis and as an early mediator of inflammation following injury. In contrast to the proinflammatory role of HMGB1, recent evidence suggests beneficial applications of HMGB1 in injury states. One such application is the use of HMGB1 as a preconditioning stimulus. Preconditioning is a phenomenon whereby a low level of stressful stimuli confers protection against subsequent injury. Preconditioning has been demonstrated in multiple species, can be induced by various stimuli, and is applicable in different organ systems. Only with the recent introduction of the concept of endogenous molecules, such as HMGB1, as signals and mediators for inflammation during injury states has the use of endogenous molecules been investigated for this use. This review will focus on the use of endogenous molecules, specifically HMGB1, as a preconditioning stimulus and its mechanism of protection, as well as other protective applications for HMGB1.


Assuntos
Proteína HMGB1/fisiologia , Inflamação/fisiopatologia , Sepse/fisiopatologia , Animais , Humanos , Precondicionamento Isquêmico , Transdução de Sinais , Receptor 4 Toll-Like
13.
Am J Transplant ; 7(10): 2279-90, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17711551

RESUMO

Carbon monoxide (CO) provides protection against oxidative stress via anti-inflammatory and cytoprotective actions. In this study, we tested the hypothesis that a low concentration of exogenous (inhaled) CO would protect transplanted lung grafts from cold ischemia-reperfusion injury via a mechanism involving the mitogen-activated protein kinase (MAPK) signaling pathway. Lewis rats underwent orthotopic syngeneic or allogeneic left lung transplantation with 6 h of cold static preservation. Exposure of donors and recipients (1 h before and then continuously post-transplant) to 250 ppm CO resulted in significant improvement in gas exchange, reduced leukocyte sequestration, preservation of parenchymal and endothelial cell ultrastructure and reduced inflammation compared to animals exposed to air. The beneficial effects of CO were associated with p38 MAPK phosphorylation and were significantly prevented by treatment with a p38 MAPK inhibitor, suggesting that CO's efficacy is at least partially mediated by activation of p38 MAPK. Furthermore, CO markedly suppressed inflammatory events in the contralateral naïve lung. This study demonstrates that perioperative exposure of donors and recipients to CO at a low concentration can impart potent anti-inflammatory and cytoprotective effects in a clinically relevant model of lung transplantation and support further evaluation for potential clinical use.


Assuntos
Monóxido de Carbono/uso terapêutico , Transplante de Pulmão/fisiologia , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Anti-Inflamatórios/uso terapêutico , Ciclo-Oxigenase 2/genética , Interleucinas/genética , Pulmão/ultraestrutura , Masculino , Neutrófilos/fisiologia , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Homólogo , Transplante Isogênico
14.
Biochem Soc Trans ; 34(Pt 5): 957-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17052236

RESUMO

Hepatic I/R (ischaemia/reperfusion) injury occurs in a variety of clinical settings including transplantation, elective liver resections and trauma. One of the challenges in studying the pathophysiology of I/R injury is the fact that the liver plays a central role in a variety of metabolic pathways in addition to governing aspects of immune surveillance and tolerance. The pathways activated in response to insults as varied as toxins, microbial and endogenous ligands and I/R may share common elements. The multiple intracellular signalling cascades involved in this process and the initiating events are still under investigation. Recent work on the role of TLRs (Toll-like receptors) in I/R injury has elucidated some of the more proximal signalling events in the pathway. In addition to the well-established role of signalling molecules such as NO (nitric oxide) in mediating damage or protection following hepatic I/R, more recent studies have focused on the participation of endogenous danger signals or DAMPs (damage-associated molecular patterns) such as HMGB1 (high-mobility group box 1). The complex interplay between HMGB1, TLRs and the many intracellular signalling molecules and pathways is illustrative of how our understanding of hepatic I/R injury is continually evolving.


Assuntos
Fígado , Traumatismo por Reperfusão/fisiopatologia , Animais , Humanos , Camundongos , Camundongos Knockout , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/fisiologia
15.
Apoptosis ; 11(3): 441-51, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16538385

RESUMO

Cyclic AMP (cAMP) and cyclic GMP (cGMP) suppress apoptosis in many cell types, including hepatocytes. We have previously shown that membrane-permeable cAMP and cGMP analogs attenuate tumor necrosis factor alpha plus actinomycin D (TNFalpha/ActD)-induced apoptosis in hepatocytes at a step upstream of caspase activation and cytochrome c release. Recently we have also shown that FADD levels increase 10 folds in response to TNFalpha/ActD. Therefore we hypothesized that cAMP and cGMP would inhibit FADD upregulation. We show here that cyclic nucleotide analogs dibutyryl cAMP (db-cAMP) and 8-bromo-cGMP (Br-cGMP) inhibit cell death and the cleavages of multiple caspases including caspase-10, -9, -8, -3, and -2, as well as suppress FADD protein up-regulation in TNFalpha/ActD-induced apoptosis. The inhibitory effects of cAMP were seen at lower concentrations than cGMP. Both cAMP and cGMP prevented FADD overexpression and cell death in hepatocytes transfected with the FADD gene. A protein kinase A (PKA) inhibitor, KT 5720, reversed the inhibition of FADD protein levels induced by cAMP or cGMP. In conclusion, our findings indicate that cAMP and cGMP prevent TNFalpha/ActD-induced apoptosis in hepatocytes and that this occurs in association with a near complete inhibition of the upregulation of FADD via a PKA-dependent mechanism.


Assuntos
Apoptose/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Hepatócitos/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Bucladesina/metabolismo , Sobrevivência Celular , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , GMP Cíclico/análogos & derivados , Dactinomicina/metabolismo , Proteína de Domínio de Morte Associada a Fas/genética , Marcação In Situ das Extremidades Cortadas , Masculino , Inibidores da Síntese de Ácido Nucleico/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Regulação para Cima
16.
Cell Death Differ ; 13(3): 512-23, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16167066

RESUMO

Dexamethasone (DEX) pretreatment protected hepatocytes from TNF-alpha plus actinomycin D (ActD)-induced apoptosis by suppressing caspase-8 activation and the mitochondria-dependent apoptosis pathway. DEX treatment upregulated cellular FLICE inhibitory protein (cFLIP) expression, but did not alter the protein levels of Bcl-2, Bcl-xL, Mcl-1, and cIAP as well as Akt activation. The increased cFLIP mRNA level by DEX was inhibited by ActD, indicating that DEX upregulates cFLIP expression at the transcriptional step. DEX also inhibited Jo2-mediated hepatocyte apoptosis by blocking the formation of the death-inducing signaling complex and caspase-8 activation. Specific downregulation of cFLIP expression using siRNA reversed the antiapoptotic effect of DEX by increasing caspase-8 activation. Moreover, DEX administration into mice increased cFLIP expression in the liver and prevented Jo2-induced hepatic injury by inhibiting caspase-8 and -3 activities. Our results indicate that DEX exerts a protective role in death receptor-induced in vitro and in vivo hepatocyte apoptosis by upregulating cFLIP expression.


Assuntos
Apoptose/efeitos dos fármacos , Dexametasona/farmacologia , Hepatócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Animais , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Caspase 3 , Caspase 8 , Caspase 9 , Inibidores de Caspase , Caspases/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Dactinomicina/antagonistas & inibidores , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Galactosamina/antagonistas & inibidores , Hepatócitos/citologia , Hepatócitos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Inibidores da Síntese de Ácido Nucleico/farmacologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Ativação Transcricional , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regulação para Cima , Receptor fas/metabolismo
17.
Gene Ther ; 13(4): 330-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16195699

RESUMO

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF family, is a type II transmembrane cytokine molecule. Soluble TRAIL has been shown to induce apoptosis in a wide variety of cancer cells in vitro and to suppress tumor growth specifically without damaging normal cells and tissues in vivo. In our previous report, we have demonstrated that an artificial gene encoding the polypeptide composed of the three functional elements (a secretion signal, a trimerization domain and an apoptosis-inducing moiety of TRAIL gene sequence) expresses and secretes highly apoptotic trimeric TRAIL into the culture supernatant. Here, as an approach to TRAIL-based cancer gene therapy, we developed an adenoviral vector delivering the gene that encodes our secretable trimeric TRAIL (stTRAIL). This adenovirus (Ad-stTRAIL) potently induced apoptosis in vitro in cancer cell lines such as HeLa, MDA-MB-231, A549, HCT116 and U-87MG. In an animal xenograft tumor model bearing a human glioma cell line U-87MG, intratumoral delivery of Ad-stTRAIL dramatically suppressed tumor growth without showing detectable adverse side effects. Histological analysis revealed that Ad-stTRAIL suppresses tumor growth by inducing apoptotic cell death. Contrary to the known rapid clearance of systemically delivered TRAIL protein from the blood circulation, stTRAIL expressed by Ad-stTRAIL in tumor tissues persisted for more than 4 days. In a comparison of tumor suppressor activity between Ad-stTRAIL and Ad-flTRAIL (delivering the full-length TRAIL gene) after mixing infected cells with uninfected cells and implanting these mixed cells in nude mice, Ad-stTRAIL showed higher tumor suppressor activity than that of Ad-flTRAIL. Our data reveal that a gene therapy using Ad-stTRAIL has a promising potential to treat human cancers including gliomas.


Assuntos
Adenoviridae/genética , Proteínas Reguladoras de Apoptose/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Glioma/terapia , Glicoproteínas de Membrana/metabolismo , Neoplasias de Tecido Nervoso/terapia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/uso terapêutico , Linhagem Celular Tumoral , Células Cultivadas , Expressão Gênica , Vetores Genéticos/genética , Glioma/patologia , Histocitoquímica , Humanos , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/uso terapêutico , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias de Tecido Nervoso/patologia , Ligante Indutor de Apoptose Relacionado a TNF , Transplante Heterólogo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/uso terapêutico
18.
Biophys J ; 90(5): 1546-59, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16339882

RESUMO

We propose a mathematical model for mitochondria-dependent apoptosis, in which kinetic cooperativity in formation of the apoptosome is a key element ensuring bistability. We examine the role of Bax and Bcl-2 synthesis and degradation rates, as well as the number of mitochondrial permeability transition pores (MPTPs), on the cell response to apoptotic stimuli. Our analysis suggests that cooperative apoptosome formation is a mechanism for inducing bistability, much more robust than that induced by other mechanisms, such as inhibition of caspase-3 by the inhibitor of apoptosis (IAP). Simulations predict a pathological state in which cells will exhibit a monostable cell survival if Bax degradation rate is above a threshold value, or if Bax expression rate is below a threshold value. Otherwise, cell death or survival occur depending on initial caspase-3 levels. We show that high expression rates of Bcl-2 can counteract the effects of Bax. Our simulations also demonstrate a monostable (pathological) apoptotic response if the number of MPTPs exceeds a threshold value. This study supports our contention, based on mathematical modeling, that cooperativity in apoptosome formation is critically important for determining the healthy responses to apoptotic stimuli, and helps define the roles of Bax, Bcl-2, and MPTP vis-à-vis apoptosome formation.


Assuntos
Apoptose/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Canais Iônicos/metabolismo , Mitocôndrias/fisiologia , Modelos Biológicos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Sobrevivência Celular , Simulação por Computador , Humanos , Proteínas de Transporte da Membrana Mitocondrial , Poro de Transição de Permeabilidade Mitocondrial
19.
Cell Mol Biol (Noisy-le-grand) ; 51(3): 329-35, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16191401

RESUMO

Under normoxic conditions, nitric oxide (NO) suppresses hepatocyte apoptosis. In contrast, NO contributes to hepatocellular injury in conditions associated with ischemia and reperfusion. To understand this paradoxical effect further, we compared the effects of various doses of NO, delivered from the chemical NO donor S-nitroso-N-acetylpenicillamine (SNAP), under both normoxic and hypoxic tissue culture conditions. We found that the cell death induced by NO under hypoxic conditions, which increased the production of reactive oxygen species, was accompanied by a necrotic morphology with a concomitant early decrease in ATP levels. The NO-induced death of hypoxic hepatocytes was reversed by co-incubation with the anti-oxidant N-acetylcysteine. We conclude that hypoxia-induced oxidative stress subsequent to ATP depletion can switch NO from an anti-apoptotic to a hepatotoxic agent. These findings may have implications for NO-induced liver damage in settings of tissue hypoxia.


Assuntos
Apoptose , Hipóxia Celular , Hepatócitos/fisiologia , Óxido Nítrico/fisiologia , Acetilcisteína/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Hepatócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doadores de Óxido Nítrico/farmacologia , Oxirredução , Estresse Oxidativo/fisiologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/fisiopatologia
20.
Proc Natl Acad Sci U S A ; 102(39): 13837-42, 2005 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-16172396

RESUMO

Hepatocytes are capable of repeated inducible NO synthase (iNOS) expression, which occurs under inflammatory and stress conditions. This iNOS expression regulates a number of cellular functions as well as cell viability. To better understand the posttranslational mechanisms that regulate the fate of iNOS in these cells, we characterized the iNOS distributed within peroxisomes. The selective permeabilization of membranes (plasma vs. peroxisomal) confirmed that there are cytosolic and peroxisomal pools of iNOS in cytokine-stimulated hepatocytes and that the iNOS protein associates with peroxisome. Detergent solubilization of the membrane fraction released iNOS to the soluble fraction. iNOS localized to membrane fraction is predominantly monomeric, but dimerization is partially reconstituted rapidly upon incubation with tetrahydrobiopterin. The reconstituted iNOS exhibits a lower specific activity than iNOS isolated from the soluble pool. Depletion of intracellular tetrahydrobiopterin with an inhibitor of de novo pterin synthesis resulted in a predominance of monomeric iNOS without a greater relative distribution of iNOS to the peroxisomal pool. Thus, iNOS exists in a least two pools in hepatocytes: a soluble pool composed of both active dimer and monomer and a peroxisomal pool of monomeric iNOS. iNOS might localize to peroxisomes in long-lived cells such as hepatocytes as a protective mechanism to remove incompetent enzyme.


Assuntos
Hepatócitos/enzimologia , Peroxissomos/enzimologia , Animais , Membrana Celular/enzimologia , Citocinas/fisiologia , Digitonina/farmacologia , Dimerização , Hepatócitos/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...