Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 42(11): 1333-1350, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36288290

RESUMO

BACKGROUND: The harmful vascular effects of smoking are well established, but the effects of chronic use of electronic cigarettes (e-cigarettes) on endothelial function are less understood. We hypothesized that e-cigarette use causes changes in blood milieu that impair endothelial function. METHODS: Endothelial function was measured in chronic e-cigarette users, chronic cigarette smokers, and nonusers. We measured effects of participants' sera, or e-cigarette aerosol condensate, on NO and H2O2 release and cell permeability in cultured endothelial cells (ECs). RESULTS: E-cigarette users and smokers had lower flow-mediated dilation (FMD) than nonusers. Sera from e-cigarette users and smokers reduced VEGF (vascular endothelial growth factor)-induced NO secretion by ECs relative to nonuser sera, without significant reduction in endothelial NO synthase mRNA or protein levels. E-cigarette user sera caused increased endothelial release of H2O2, and more permeability than nonuser sera. E-cigarette users and smokers exhibited changes in circulating biomarkers of inflammation, thrombosis, and cell adhesion relative to nonusers, but with distinct profiles. E-cigarette user sera had higher concentrations of the receptor for advanced glycation end products (RAGE) ligands S100A8 and HMGB1 (high mobility group box 1) than smoker and nonuser sera, and receptor for advanced glycation end product inhibition reduced permeability induced by e-cigarette user sera but did not affect NO production. CONCLUSIONS: Chronic vaping and smoking both impair FMD and cause changes in the blood that inhibit endothelial NO release. Vaping, but not smoking, causes changes in the blood that increase microvascular endothelial permeability and may have a vaping-specific effect on intracellular oxidative state. Our results suggest a role for RAGE in e-cigarette-induced changes in endothelial function.


Assuntos
Sistemas Eletrônicos de Liberação de Nicotina , Proteína HMGB1 , Vaping , Humanos , Vaping/efeitos adversos , Fator A de Crescimento do Endotélio Vascular , Receptor para Produtos Finais de Glicação Avançada , Fumar/efeitos adversos , Células Endoteliais , Peróxido de Hidrogênio , Aerossóis , Biomarcadores , RNA Mensageiro , Óxido Nítrico Sintase
2.
J Neuroinflammation ; 19(1): 118, 2022 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-35610647

RESUMO

BACKGROUND: The transient receptor potential vanilloid 1 (TRPV1) participates in thermosensation and inflammatory pain, but its immunomodulatory mechanisms remain enigmatic. N-Oleoyl dopamine (OLDA), an endovanilloid and endocannabinoid, is a TRPV1 agonist that is produced in the central nervous system and the peripheral nervous system. We studied the anti-inflammatory effects and TRPV1-dependent mechanisms of OLDA in models of inflammation and sepsis. METHODS: Mice were challenged intratracheally or intravenously with LPS, or intratracheally with S. aureus to induce pneumonia and sepsis, and then were treated intravenously with OLDA. Endpoints included plasma cytokines, leukocyte activation marker expression, mouse sepsis scores, lung histopathology, and bacterial counts. The role of TRPV1 in the effects of OLDA was determined using Trpv1-/- mice, and mice with TRPV1 knockdown pan-neuronally, in peripheral nervous system neurons, or in myeloid cells. Circulating monocytes/macrophages were depleted using clodronate to determine their role in the anti-inflammatory effects of OLDA in endotoxemic mice. Levels of exogenous OLDA, and of endovanilloids and endocannabinoids, at baseline and in endotoxemic mice, were determined by LC-MS/MS. RESULTS: OLDA administration caused an early anti-inflammatory response in endotoxemic and septic mice with high serum levels of IL-10 and decreased levels of pro-inflammatory cytokines. OLDA also reduced lung injury and improved mouse sepsis scores. Blood and lung bacterial counts were comparable between OLDA- and carrier-treated mice with S. aureus pneumonia. OLDA's effects were reversed in mice with pan-neuronal TRPV1 knockdown, but not with TRPV1 knockdown in peripheral nervous system neurons or myeloid cells. Depletion of monocytes/macrophages reversed the IL-10 upregulation by OLDA in endotoxemic mice. Brain and blood levels of endovanilloids and endocannabinoids were increased in endotoxemic mice. CONCLUSIONS: OLDA has strong anti-inflammatory actions in mice with endotoxemia or S. aureus pneumonia. Prior studies focused on the role of peripheral nervous system TRPV1 in modulating inflammation and pneumonia. Our results suggest that TRPV1-expressing central nervous system neurons also regulate inflammatory responses to endotoxemia and infection. Our study reveals a neuro-immune reflex that during acute inflammation is engaged proximally by OLDA acting on neuronal TRPV1, and through a multicellular network that requires circulating monocytes/macrophages, leads to the systemic production of IL-10.


Assuntos
Endotoxemia , Sepse , Animais , Sistema Nervoso Central/metabolismo , Cromatografia Líquida , Citocinas/metabolismo , Dopamina/metabolismo , Endocanabinoides , Endotoxemia/induzido quimicamente , Endotoxemia/tratamento farmacológico , Inflamação/metabolismo , Interleucina-10/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Sepse/tratamento farmacológico , Staphylococcus aureus , Canais de Cátion TRPV/metabolismo , Espectrometria de Massas em Tandem
3.
Int Immunopharmacol ; 32: 76-86, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26802603

RESUMO

The activation of hepatic stellate cells (HSCs) plays a critical role in the development of liver fibrosis. The induction of apoptosis in activated HSCs during the recovery phase of hepatic fibrosis represents a potential anti-fibrotic therapy. We have previously shown that Smad2 protects against hepatic fibrogenesis; however, the role of Smad2 in the regulation of activated HSC apoptosis remains unknown. We hypothesized that Smad2 regulates the apoptosis of activated HSCs, leading to the resolution of liver fibrosis. To test this hypothesis, the livers of rats were harvested at 0 and 4 weeks after hepatic fibrosis was established by CCl4 injection. Furthermore, TGF-ß1-activated HSCs were treated with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) following the silencing or overexpression of Smad2. Both the phosphorylation of Smad2 and TRAIL were detected in fibrotic liver tissues. The results of TUNEL and α-SMA double-staining showed an increase in the apoptosis of activated HSCs during the spontaneous recovery phase. The knockdown of Smad2 reduced TRAIL-induced apoptosis in TGF-ß1-activated human LX-2 cells and resulted in an increased expression of α-SMA and collagen I (Col. I). In contrast, the overexpression of Smad2 increased TRAIL-induced HSC apoptosis and reduced the expression of α-SMA and Col. I. The mechanisms underlying these findings were associated with the Smad2-mediated down-regulation of X-linked inhibitor of apoptosis protein (XIAP), resulting in enhanced caspase-3 activity and apoptosis. In conclusion, Smad2 enhances TRAIL-induced apoptosis in activated HSCs, which facilitates the resolution of hepatic fibrosis.


Assuntos
Apoptose/fisiologia , Células Estreladas do Fígado/metabolismo , Proteína Smad2/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Tetracloreto de Carbono , Caspase 3/metabolismo , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Masculino , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Proteína Smad2/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
4.
Toxicol Appl Pharmacol ; 289(2): 163-76, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26435214

RESUMO

SIRT1 (silent information regulator 1), a conserved NAD+-dependent histone deacetylase, is closely related with various biological processes. Moreover, the important role of SIRT1 in alcoholic liver disease, nonalcoholic fatty liver and HCC had been widely reported. Recently, a novel role of SIRT1 was uncovered in organ fibrosis diseases. Here, we investigated the inhibitory effect of SIRT1 in liver fibrogenesis. SIRT1 protein was dramatically decreased in CCl4-treated mice livers. Stimulation of LX-2 cells with TGF-ß1 also resulted in a significant suppression of SIRT1 protein. Nevertheless, TGF-ß1-induced LX-2 cell activation was inhibited by SIRT1 plasmid, and this was accompanied by up-regulation of cell apoptosis-related proteins. Overexpression of SIRT1 also attenuated TGF-ß1-induced expression of myofibroblast markers α-SMA and COL1a. However, the important characteristic of the recovery of liver fibrosis is not only the apoptosis of activated stellate cells but also the reversal of the myofibroblast-like phenotype to a quiescent-like phenotype. Restoration of SIRT1 protein was observed in the in vivo spontaneously liver fibrosis reversion model and in vitro MDI (isobutylmethylxanthine, dexamethasone, and insulin)-induced reversed stellate cells, and forced expression of SIRT1 also promoted the reversal of activated stellate cells. Furthermore, lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) was increased in liver fibrosis. RNAi-mediated suppression of MALAT1 resulted in a decrease of myofibroblast markers and restoration of SIRT1 protein. These observations suggested that SIRT1 contributed to apoptosis and reversion of activated LX-2 cells and SIRT1 might be regulated by MALAT1 in liver fibrosis. Therefore, SIRT1 could be considered as a valuable therapeutic target for translational studies of liver fibrosis.


Assuntos
Apoptose , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Células Estreladas do Fígado/enzimologia , Cirrose Hepática Experimental/prevenção & controle , Fígado/enzimologia , Sirtuína 1/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Tetracloreto de Carbono , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Humanos , Insulina/farmacologia , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/enzimologia , Cirrose Hepática Experimental/genética , Cirrose Hepática Experimental/patologia , Masculino , Camundongos Endogâmicos C57BL , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Fenótipo , Interferência de RNA , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Sirtuína 1/genética , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/farmacologia , Xantinas/farmacologia
5.
Sci Signal ; 8(391): ra86, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26307013

RESUMO

Inflammatory critical illness is a syndrome that is characterized by acute inflammation and organ injury, and it is triggered by infections and noninfectious tissue injury, both of which activate innate immune receptors and pathways. Although reports suggest an anti-inflammatory role for the mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5), we previously found that ERK5 mediates proinflammatory responses in primary human cells in response to stimulation of Toll-like receptor 2 (TLR2). We inhibited the kinase activities and reduced the abundances of ERK5 and MEK5, a MAPK kinase directly upstream of ERK5, in primary human vascular endothelial cells and monocytes, and found that ERK5 promoted inflammation induced by a broad range of microbial TLR agonists and by the proinflammatory cytokines interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α). Furthermore, we found that inhibitors of MEK5 or ERK5 reduced the plasma concentrations of proinflammatory cytokines in mice challenged with TLR ligands or heat-killed Staphylococcus aureus, as well as in mice that underwent sterile lung ischemia-reperfusion injury. Finally, we found that inhibition of ERK5 protected endotoxemic mice from death. Together, our studies support a proinflammatory role for ERK5 in primary human endothelial cells and monocytes, and suggest that ERK5 is a potential therapeutic target in diverse disorders that cause inflammatory critical illness.


Assuntos
Células Endoteliais da Veia Umbilical Humana/imunologia , Proteína Quinase 7 Ativada por Mitógeno/imunologia , Monócitos/imunologia , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Animais , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Interleucina-1beta/imunologia , Masculino , Camundongos , Monócitos/patologia , Síndrome de Resposta Inflamatória Sistêmica/patologia , Síndrome de Resposta Inflamatória Sistêmica/terapia , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/imunologia
6.
Stem Cells Transl Med ; 4(7): 832-40, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25999518

RESUMO

UNLABELLED: : Mesenchymal stem cells (MSCs) can be derived from multiple tissue sources. However, the optimal source of MSCs for cell-based therapy for acute lung injury (ALI) is unclear. In the present experiments, we studied bone marrow (BM)-derived and embryonic stem cell-derived human MSC (ES-MSCs) as a therapeutic agent in Escherichia coli endotoxin-induced ALI in mice. We hypothesized that ES-MSCs would be more potent than BM-MSCs owing to its more primitive source of origin. ALI was induced by the intratracheal instillation of endotoxin at 4 mg/kg into 10-12-week-old C57BL/6 mice with or without BM-MSCs, ES-MSCs, or normal human lung fibroblasts as a cellular control. Compared with the endotoxin-injured mice at 48 hours, the administration of ES-MSCs provided results similar to those of BM-MSCs, significantly reducing the influx of white blood cells and neutrophils and decreasing the secretion of the inflammatory cytokines, macrophage inflammatory protein-2 and tumor necrosis factor-α, in the injured alveolus. BM-MSCs also reduced extravascular lung water, a measure of pulmonary edema, by 60% and the total protein levels, a measure of lung permeability, by 66%. However, surprisingly, ES-MSCs did not have these protective effects, which was partially explained by the increased secretion of matrix metallopeptidase 9 by ES-MSCs, an enzyme known to increase lung protein permeability. In conclusion, both BM-MSCs and ES-MSCs markedly decreased endotoxin-induced inflammation. However, ES-MSCs did not show any beneficial effect on reducing pulmonary edema and lung protein permeability compared with BM-MSCs, suggesting that not all MSCs behave in a similar fashion. Our results highlight the need perhaps for a disease-specific potency assay for MSCs. SIGNIFICANCE: To determine the optimal source of mesenchymal stem cells (MSCs) for cell-based therapy for acute lung injury, bone marrow (BM)- and embryonic stem cell-derived human MSC (ES-MSCs) were compared as therapeutic agents for Escherichia coli endotoxin-induced lung injury in mice. ES-MSCs behaved similarly to BM-MSCs by markedly decreasing the inflammatory response induced by endotoxin. However, unlike BM-MSCs, ES-MSCs provided no protective effects against increasing lung water and protein permeability, in part because of an increase in expression of matrix metallopeptidase 9 by ES-MSCs. In patients with acute respiratory distress syndrome, impaired alveolar fluid clearance (i.e., no resolution of pulmonary edema fluid) has been associated with higher mortality rates. Although ES-MSCs might ultimately be found to have properties superior to those of BM-MSCs, such as for immunomodulation, these results highlight the need for a disease-specific potency assay for stem cell-based therapy.

7.
Biochim Biophys Acta ; 1842(11): 2204-15, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25201080

RESUMO

Long noncoding RNAs (lncRNAs) are being increasingly recognized as major players in governing fundamental biological processes through diverse mechanisms. Maternally expressed gene 3 (MEG3) is an imprinted gene located at 14q32 that encodes a lncRNA correlated with several human cancers. Recently, the methylation-dependent downregulation of MEG3 has been described in liver cancers. However, its biological functional role in liver fibrosis remains unknown. In our study, MEG3 levels were remarkably decreased in CCl4-induced mouse liver fibrosis models and human fibrotic livers as demonstrated by real-time quantitative PCR. Moreover, the expression of MEG3 was downregulated in human hepatic stellate cell lines LX-2 cells in response to transforming growth factor-ß1 (TGF-ß1) stimulation in dose and time-dependent manner. Enforced expression of MEG3 in LX-2 cells inhibited TGF-ß1-induced cell proliferation, while promoting cell apoptosis. In addition, hypermethylation of MEG3 promoter was identified by methylation-specific PCR and MEG3 expression was robustly increased by the inhibition of methylation with either 5-aza-2-deoxycytidine (5-azadC), or siRNA to DNA methyltransferase 1 (DNMT1) in TGF-ß1-induced LX-2 cells. More importantly, overexpression of MEG3 could activate p53 and mediate cytochrome c release, subsequently leading to caspase-3-dependent apoptosis in TGF-ß1-treated LX-2 cells. These findings suggested that MEG3 may play an important role in stellate cell activation and liver fibrosis progression and act as a novel potential therapeutic target for liver fibrosis.

8.
Innate Immun ; 18(4): 602-16, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22186927

RESUMO

The vascular endothelium is integrally involved in the host response to infection and in organ failure during acute inflammatory disorders such as sepsis. Gram-negative and Gram-positive bacterial lipoproteins circulate in sepsis and can directly activate the endothelium by binding to endothelial cell (EC) TLR2. In this report, we perform the most comprehensive analysis to date of the immune-related genes regulated after activation of endothelial TLR2 by bacterial di- and triacylated lipopeptides. We found that TLR2 activation specifically induces the expression of the genes IL-6, IL-8, CSF2, CSF3, ICAM1 and SELE by human umbilical vein ECs and human lung microvascular ECs. These proteins participate in neutrophil recruitment, adherence and activation at sites of inflammation. Significantly, our studies demonstrate that TLR2-mediated EC responses are specifically geared towards recruitment, activation, and survival of neutrophils and not mononuclear leukocytes, that ECs do not require priming by other inflammatory stimuli to respond to bacterial lipopeptides and, unlike mononuclear leukocytes, TLR2 agonists do not induce ECs to secrete TNF-α. This study suggests that endothelial TLR2 may be an important regulator of neutrophil trafficking to sites of infection in general, and that direct activation of lung endothelial TLR2 may contribute to acute lung injury during sepsis.


Assuntos
Endotélio Vascular/imunologia , Ativação de Neutrófilo , Neutrófilos/imunologia , Sepse/imunologia , Receptor 2 Toll-Like/metabolismo , Proteínas de Bactérias/farmacologia , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Selectina E/genética , Selectina E/metabolismo , Endotélio Vascular/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Lipoproteínas/farmacologia , Ativação de Neutrófilo/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Regulação para Cima
9.
Nephrol Dial Transplant ; 26(6): 1813-20, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21068143

RESUMO

BACKGROUND: Immunosuppressive agents have been investigated in renal ischaemia-reperfusion injury (IRI) and have frequently demonstrated a beneficial effect. Most studies focused on treatment of the recipient at the time of transplantation. Pre-treatment of these organs before injury (pharmacological pre-conditioning) may particularly protect these organs. This study aimed to investigate the possible protective effects of donor pre-treatment with cyclosporine (CsA) or the mTOR inhibitor everolimus or their combination against IRI during renal transplantation in a rat model. METHODS: Donors received vehicle, CsA (5 mg/kg), everolimus (0.5 mg/kg) or CsA + everolimus. Two oral doses were administered to the donors at 24 h and again at 6 h prior to donor kidney removal. Syngeneic rat kidneys were preserved in UW solution for 24 h prior to transplantation. After 24 h of reperfusion, blood and tissue samples were collected from recipients for further analysis. RESULTS: Renal functions as determined by creatinine and necrosis scores were not different between the experimental groups. Cleaved caspase-3, heat shock protein 70 (HSP70), tumor-necrosis factor-alpha (TNF-α) and nitrotyrosine protein levels were not statistically different between the four treatment groups at 24 h post-transplantation. Blood NMR analysis on metabolic markers for IRI reveals no beneficial effects of donor pre-treatment on the 24-h outcome in transplantation. CONCLUSIONS: When given alone or as a combination to donors before organ recovery, cyclosporine or everolimus does not appear to ameliorate IRI.


Assuntos
Ciclosporina/uso terapêutico , Imunossupressores/uso terapêutico , Transplante de Rim/efeitos adversos , Traumatismo por Reperfusão/etiologia , Sirolimo/análogos & derivados , Animais , Everolimo , Rejeição de Enxerto/etiologia , Testes de Função Renal , Masculino , Ratos , Ratos Endogâmicos Lew , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/fisiopatologia , Sirolimo/uso terapêutico , Doadores de Tecidos , Condicionamento Pré-Transplante , Resultado do Tratamento
10.
J Surg Res ; 158(1): 43-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19394963

RESUMO

BACKGROUND: Prolonged hepatic warm ischemia during surgery remains a significant problem, particularly in the setting of liver resection and reduced remaining liver mass. The goal of the present study is to evaluate the effect of passive cooling caused by exposure to ambient conditions on hepatic injury in rats during warm ischemia followed by hepatectomy. METHODS: The left and median lobes of male rats were exposed to 75 min of ischemia under either normothermic (37 degrees C) or mildly hypothermic (34 degrees C) conditions. After 75 min of ischemia, the right lobe was resected, leaving the animal with only the remaining ischemic lobes. Animals were allowed to survive indefinitely or sacrificed at 4 h after reperfusion for determination of injury and inflammatory gene expression. RESULTS: Survival was already markedly higher in mildly hypothermic rats than normothermic rats at 24 h. Short passive cooling for the time course of the ischemic event significantly increased the hepatic induction of heat shock proteins 70 and 32 (both 3-fold versus normothermia, P<0.05) in response to ischemia/reperfusion whereas it significantly decreased the induction of tumor necrosis factor-alpha (TNF-alpha) and macrophage inflammatory protein-2 (MIP-2) in the liver. Biochemical markers of hepatic injury were significantly lower in the passive cooling group than in normothermic animals: aspartate aminotransferase (AST) serum concentrations were 9277+/-3461IU/L versus 15106+/-4104IU/L (P<0.01), and alanine aminotransferase (ALT) levels 5986+/-2246IU/L versus 9429+/-3643IU/L (P<0.01). CONCLUSION: We demonstrated in a clinically relevant model of hepatic ischemia/reperfusion that mild hypothermia significantly reduces hepatic injury and improves survival.


Assuntos
Citocinas/biossíntese , Proteínas de Choque Térmico/biossíntese , Hepatectomia , Hipotermia Induzida , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Caspase 3/análise , Hepatectomia/métodos , Hepatectomia/mortalidade , Fígado/patologia , Ratos , Ratos Zucker , Traumatismo por Reperfusão/prevenção & controle
11.
Surgery ; 143(2): 243-51, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18242341

RESUMO

BACKGROUND: Arterial inflow occlusion is a well-known mechanism of renal injury during major vascular surgery. In contrast, renal injury from venous outflow obstruction is poorly understood. The goal of this study was to examine the injury pattern of renal venous outflow obstruction, compare this with the traditional model of arterial occlusion, and examine possible mechanisms. METHODS: Male Fisher rats were used for the renal warm ischemia model. Twenty-five minutes of renal ischemia was induced by selectively occluding either the renal artery or vein. After 24 h of reperfusion, whole blood and kidney tissue were collected for further analysis. RESULTS: Serum creatinine (SCr) concentrations taken 24 h after reperfusion were significantly greater in the venous occlusion group (V) when compared to the arterial group (A). While histology did not demonstrate significant differences in extent of necrosis between both groups, a stronger inflammatory response resulted from venous occlusion. Specifically, significantly greater MCP-1 mRNA and significantly greater MCP-1, TNF-alpha, and HO-1 protein levels were found in the venous group, while no differences in MIP-2, ICAM-1, and VCAM-1 mRNA expression existed between A and V. Further analysis demonstrated presence of increased cleaved caspase-3 protein in the artery group than in the venous group. CONCLUSIONS: Venous renal outflow obstruction results in more severe functional renal injury when compared to arterial inflow occlusion. Macrophage activation and neutrophilic infiltration appear to be exaggerated during venous occlusion.


Assuntos
Artéria Renal/cirurgia , Circulação Renal , Veias Renais/cirurgia , Traumatismo por Reperfusão/fisiopatologia , Animais , Constrição , Modelos Animais , Ratos , Procedimentos Cirúrgicos Urológicos
12.
Anesthesiology ; 108(3): 402-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18292678

RESUMO

BACKGROUND: Chronic hyperglycemia is known to increase renal injury, particularly during ischemia-reperfusion episodes. The goal of this study was to examine whether transient hyperglycemia during or after renal ischemia-reperfusion increased renal dysfunction. METHODS: Male Lewis rats underwent sham operations or unilateral nephrectomies followed by contralateral renal ischemia-reperfusion. Hyperglycemic rats were given 25% dextrose to induce transient hyperglycemia lasting throughout the duration of ischemia (PI rats) or beginning 2 h after initiation of reperfusion (PR rats). Additional vehicle control rats received saline and underwent ischemia-reperfusion surgery as with PI and PR rats. Twenty-five minutes of mild renal ischemia followed by 24 h of reperfusion was induced by occluding the renal artery and vein. RESULTS: Terminal serum creatinine concentrations were significantly higher in the PI rats when compared with the PR or vehicle control rats. Histology demonstrated significantly increased necrosis in the PI rats relative to PR and control animals. Tissue analyses demonstrated significantly higher heat shock protein 70, heat shock protein 32, and cleaved caspase-3 protein levels in the PI rats. Oxidative stress generated through the xanthine pathway in the PI group was significantly increased compared with the oxidative stress in the PR and vehicle control rats. In contrast, vascular endothelial growth factor and erythropoietin were significantly decreased in the PI rats compared with the PR rats and controls. CONCLUSIONS: Hyperglycemia that occurred during renal ischemia-reperfusion resulted in severe functional injury compared with normoglycemia or with hyperglycemia that occurred after reperfusion. Investigated molecular pathways are more profoundly affected by hyperglycemia that occurs before renal ischemia-reperfusion.


Assuntos
Hiperglicemia/patologia , Rim/irrigação sanguínea , Rim/patologia , Traumatismo por Reperfusão/patologia , Animais , Hiperglicemia/sangue , Hiperglicemia/complicações , Masculino , Ratos , Ratos Endogâmicos Lew , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/etiologia
13.
Mol Pharmacol ; 71(1): 193-200, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17018578

RESUMO

Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Under stress conditions, some porphyrin derivatives can inhibit HO1 and trigger cell death. Motexafin gadolinium (MGd) is an expanded porphyrin that selectively targets cancer cells through a process of futile redox cycling that decreases intracellular reducing metabolites and protein thiols. Here, we report that hematopoietic-derived cell lines that constitutively express HO1 are more susceptible to MGd-induced apoptosis than those that do not. MGd used in combination with tin protoporphyrin IX, an inhibitor of HO1, resulted in synergistic cell killing. Consistent with these cell culture observations, we found that MGd is an inhibitor of heme oxygenase-1 activity in vitro. We demonstrate that inhibition of HO1 reflects an interaction of MGd with NADPH-cytochrome P450 reductase, the electron donor for HO1, that results in diversion of reducing equivalents from heme oxidation to oxygen reduction. In accord with this mechanism, MGd is also an in vitro inhibitor of CYP2C9, CYP3A4, and CYP4A1. Inhibition of HO1 by MGd may contribute to its anticancer activity, whereas its in vitro inhibition of a broad spectrum of P450 enzymes indicates that a potential exists for drug-drug interactions.


Assuntos
Divisão Celular/efeitos dos fármacos , Heme Oxigenase-1/genética , Metaloporfirinas/farmacologia , NADPH-Ferri-Hemoproteína Redutase/antagonistas & inibidores , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Espectrofotometria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA