Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Med ; 27(1): 18, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33632134

RESUMO

BACKGROUND: Hepatic ischemia/reperfusion (I/R) injury can be a major complication following liver surgery contributing to post-operative liver dysfunction. Maresin 1 (MaR1), a pro-resolving lipid mediator, has been shown to suppress I/R injury. However, the mechanisms that account for the protective effects of MaR1 in I/R injury remain unknown. METHODS: WT (C57BL/6J) mice were subjected to partial hepatic warm ischemia for 60mins followed by reperfusion. Mice were treated with MaR1 (5-20 ng/mouse), Boc2 (Lipoxin A4 receptor antagonist), LY294002 (Akt inhibitor) or corresponding controls just prior to liver I/R or at the beginning of reperfusion. Blood and liver samples were collected at 6 h post-reperfusion. Serum aminotransferase, histopathologic changes, inflammatory cytokines, and oxidative stress were analyzed to evaluate liver injury. Signaling pathways were also investigated in vitro using primary mouse hepatocyte (HC) cultures to identify underlying mechanisms for MaR1 in liver I/R injury. RESULTS: MaR1 treatment significantly reduced ALT and AST levels, diminished necrotic areas, suppressed inflammatory responses, attenuated oxidative stress and decreased hepatocyte apoptosis in liver after I/R. Akt signaling was significantly increased in the MaR1-treated liver I/R group compared with controls. The protective effect of MaR1 was abrogated by pretreatment with Boc2, which together with MaR1-induced Akt activation. MaR1-mediated liver protection was reversed by inhibition of Akt. CONCLUSIONS: MaR1 protects the liver against hepatic I/R injury via an ALXR/Akt signaling pathway. MaR1 may represent a novel therapeutic agent to mitigate the detrimental effects of I/R-induced liver injury.


Assuntos
Ácidos Docosa-Hexaenoicos/uso terapêutico , Hepatopatias/tratamento farmacológico , Substâncias Protetoras/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Formil Peptídeo/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Sobrevivência Celular/efeitos dos fármacos , Citocinas/sangue , Ácidos Docosa-Hexaenoicos/farmacologia , Glutationa Peroxidase/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Hepatopatias/sangue , Hepatopatias/metabolismo , Hepatopatias/patologia , Masculino , Malondialdeído/metabolismo , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Receptores de Formil Peptídeo/genética , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos
2.
Hepatology ; 72(4): 1394-1411, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31997373

RESUMO

BACKGROUND AND AIMS: Itaconate, a metabolite of the tricarboxylic acid cycle, plays anti-inflammatory roles in macrophages during endotoxemia. The mechanisms underlying its anti-inflammatory roles have been shown to be mediated by the modulation of oxidative stress, an important mechanism of hepatic ischemia-reperfusion (I/R) injury. However, the role of itaconate in liver I/R injury is unknown. APPROACH AND RESULTS: We found that deletion of immune-responsive gene 1 (IRG1), encoding for the enzyme producing itaconate, exacerbated liver injury and systemic inflammation. Furthermore, bone marrow adoptive transfer experiments indicated that deletion of IRG1 in both hematopoietic and nonhematopoietic compartments contributes to the protection mediated by IRG1 after I/R. Interestingly, the expression of IRG1 was up-regulated in hepatocytes after I/R and hypoxia/reoxygenation-induced oxidative stress. Modulation of the IRG1 expression levels in hepatocytes regulated hepatocyte cell death. Importantly, addition of 4-octyl itaconate significantly improved liver injury and hepatocyte cell death after I/R. Furthermore, our data indicated that nuclear factor erythroid 2-related factor 2 (Nrf2) is required for the protective effect of IRG1 on mouse and human hepatocytes against oxidative stress-induced injury. Our studies document the important role of IRG1 in the acute setting of sterile injury induced by I/R. Specifically, we provide evidence that the IRG1/itaconate pathway activates Nrf2-mediated antioxidative response in hepatocytes to protect liver from I/R injury. CONCLUSIONS: Our data expand on the importance of IRG1/itaconate in nonimmune cells and identify itaconate as a potential therapeutic strategy for this unfavorable postsurgical complication.


Assuntos
Anti-Inflamatórios/farmacologia , Carboxiliases/fisiologia , Hepatócitos/metabolismo , Fígado/irrigação sanguínea , Fator 2 Relacionado a NF-E2/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Succinatos/farmacologia , Animais , Humanos , Hidroliases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Transdução de Sinais/fisiologia , Succinatos/uso terapêutico
3.
Am J Physiol Gastrointest Liver Physiol ; 314(6): G655-G667, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29446653

RESUMO

Liver ischemia-reperfusion (I/R) injury occurs through induction of oxidative stress and release of damage-associated molecular patterns (DAMPs), including cytosolic DNA released from dysfunctional mitochondria or from the nucleus. Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) is a cytosolic DNA sensor known to trigger stimulator of interferon genes (STING) and downstream type 1 interferon (IFN-I) pathways, which are pivotal innate immune system responses to pathogen. However, little is known about the role of cGAS/STING in liver I/R injury. We subjected C57BL/6 (WT), cGAS knockout (cGAS-/-), and STING-deficient (STINGgt/gt) mice to warm liver I/R injury and that found cGAS-/- mice had significantly increased liver injury compared with WT or STINGgt/gt mice, suggesting a protective effect of cGAS independent of STING. Liver I/R upregulated cGAS in vivo and also in vitro in hepatocytes subjected to anoxia/reoxygenation (A/R). We confirmed a previously published finding that hepatocytes do not express STING under normoxic conditions or after A/R. Hepatocytes and liver from cGAS-/- mice had increased cell death and reduced induction of autophagy under hypoxic conditions as well as increased apoptosis. Protection could be restored in cGAS-/- hepatocytes by overexpression of cGAS or by pretreatment of mice with autophagy inducer rapamycin. Our findings indicate a novel protective role for cGAS in the regulation of autophagy during liver I/R injury that occurs independently of STING. NEW & NOTEWORTHY Our studies are the first to document the important role of cGAS in the acute setting of sterile injury induced by I/R. Specifically, we provide evidence that cGAS protects liver from I/R injury in a STING-independent manner.


Assuntos
Autofagia/fisiologia , Interferon Tipo I , Fígado , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/metabolismo , Traumatismo por Reperfusão , Animais , Apoptose/fisiologia , DNA Nucleotidiltransferases/fisiologia , Indutores de Interferon/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Fígado/irrigação sanguínea , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Substâncias Protetoras/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais
4.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226571

RESUMO

The sensing of double-stranded RNA (dsRNA) in the liver is important for antiviral defenses but can also contribute to sterile inflammation during liver injury. Hepatocytes are often the target of viral infection and are easily injured by inflammatory insults. Here we sought to establish the pathways involved in the production of type I interferons (IFN-I) in response to extracellular poly(I:C), a dsRNA mimetic, in hepatocytes. This was of interest because hepatocytes are long-lived and, unlike most immune cells that readily die after activation with dsRNA, are not viewed as cells with robust antimicrobial capacity. We found that poly(I:C) leads to rapid up-regulation of inducible nitric oxide synthase (iNOS), double-stranded RNA-dependent protein kinase (PKR), and Src. The production of IFN-ß was dependent on iNOS, PKR, and Src and partially dependent on TLR3/Trif. iNOS and Src up-regulation was partially dependent on TLR3/Trif but entirely dependent on PKR. The phosphorylation of TLR3 on tyrosine 759 was shown to increase in parallel to IFN-ß production in an iNOS- and Src-dependent manner, and Src was found to directly interact with TLR3 in the endosomal compartment of poly(I:C)-treated cells. Furthermore, we identified a robust NO/cGMP/PKG-dependent feedforward pathway for the amplification of iNOS expression. These data identify iNOS/NO as an integral component of IFN-ß production in response to dsRNA in hepatocytes in a pathway that involves the coordinated activities of TLR3/Trif and PKR.


Assuntos
Hepatócitos/imunologia , Hepatócitos/metabolismo , Interferon beta/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/farmacologia , Receptor 3 Toll-Like/metabolismo , eIF-2 Quinase/metabolismo , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/deficiência , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Células Cultivadas , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Hepatócitos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/genética , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética , Tirosina/química , Regulação para Cima/efeitos dos fármacos , eIF-2 Quinase/deficiência , eIF-2 Quinase/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
5.
J Immunol ; 193(5): 2405-15, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25070845

RESUMO

Autophagy, an evolutionarily conserved homeostasis process regulating biomass quantity and quality, plays a critical role in the host response to sepsis. Recent studies show its calcium dependence, but the calcium-sensitive regulatory cascades have not been defined. In this study, we describe a novel mechanism in which calcium/calmodulin-dependent protein kinase IV (CaMKIV), through inhibitory serine phosphorylation of GSK-3ß and inhibition of FBXW7 recruitment, prevents ubiquitin proteosomal degradation of mammalian target of rapamycin (mTOR) and thereby augments autophagy in both the macrophage and the kidney. Under the conditions of sepsis studied, mTOR expression and activity were requisite for autophagy, a paradigm countering the current perspective that prototypically, mTOR inhibition induces autophagy. CaMKIV-mTOR-dependent autophagy was fundamentally important for IL-6 production in vitro and in vivo. Similar mechanisms were operant in the kidney during endotoxemia and served a cytoprotective role in mitigating acute kidney injury. Thus, CaMKIV-mTOR-dependent autophagy is conserved in both immune and nonimmune/parenchymal cells and is fundamental for the respective functional and adaptive responses to septic insult.


Assuntos
Injúria Renal Aguda/imunologia , Autofagia/efeitos dos fármacos , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Macrófagos/imunologia , Serina-Treonina Quinases TOR/imunologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Autofagia/genética , Autofagia/imunologia , Proteína Quinase Tipo 4 Dependente de Cálcio-Calmodulina/genética , Linhagem Celular , Proteínas F-Box/genética , Proteínas F-Box/imunologia , Proteína 7 com Repetições F-Box-WD , Regulação da Expressão Gênica/imunologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/imunologia , Glicogênio Sintase Quinase 3 beta , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Serina-Treonina Quinases TOR/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
6.
J Biol Chem ; 287(43): 35887-98, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22898814

RESUMO

We and others have previously shown that the inducible nitric-oxide synthase (iNOS) and nitric oxide (NO) are hepatoprotective in a number of circumstances, including endotoxemia. In vitro, hepatocytes are protected from tumor necrosis factor (TNF) α-induced apoptosis via cGMP-dependent and cGMP-independent mechanisms. We have shown that the cGMP-dependent protective mechanisms involve the inhibition of death-inducing signaling complex formation. We show here that LPS-induced iNOS expression leads to rapid TNF receptor shedding from the surface of hepatocytes via NO/cGMP/protein kinase G-dependent activation and surface translocation of TNFα-converting enzyme (TACE/ADAM17). The activation of TACE is associated with the up-regulation of iRhom2 as well as the interaction and phosphorylation of TACE and iRhom2, which are also NO/cGMP/protein kinase G-dependent. These findings suggest that one mechanism of iNOS/NO-mediated protection of hepatocytes involves the rapid shedding of TNF receptor 1 to limit TNFα signaling.


Assuntos
Proteínas ADAM/metabolismo , GMP Cíclico/metabolismo , Hepatócitos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Proteínas ADAM/genética , Proteína ADAM17 , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Células Cultivadas , GMP Cíclico/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Hepatócitos/citologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
7.
Nitric Oxide ; 31: 9-19, 2013 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-23474170

RESUMO

iNOS localizes to both the cytosol and peroxisomes in hepatocytes in vitro and in vivo. The structural determinants for iNOS localization are not known. One plausible mechanism for iNOS localization to the peroxisome is through the interaction with peroxisomal import proteins PEX5 or PEX7. siRNA knockdown of PEX7 reduced iNOS colocalization with the peroxisomal protein PMP70. Proteomic studies using MALDI-MS identified iNOS association with the 50-kD ezrin binding PDZ protein (EBP50). Confocal microscopy studies and immunoelectron microscopy confirmed iNOS association with EBP50, with greatest colocalization occurring at 8h of cytokine exposure. EBP50 associated with peroxisomes in a PEX5 and PEX7-dependent manner. iNOS localization to peroxisomes was contingent on EBP50 expression in LPS-treated mice. Thus, iNOS targeting to peroxisomes in hepatocytes involves interaction with PEX7 and EBP50. The targeting of iNOS protein to the peroxisome may shift the balance of metabolic processes that rely on heme proteins susceptible to modification by radical oxygen and nitrogen radicals.


Assuntos
Hepatócitos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Peroxissomos/metabolismo , Fosfoproteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Hepatócitos/química , Hepatócitos/enzimologia , Fígado/química , Fígado/enzimologia , Fígado/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia de Fluorescência , Receptor 2 de Sinal de Orientação para Peroxissomos , Fosfoproteínas/genética , RNA Interferente Pequeno/genética , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais , Trocadores de Sódio-Hidrogênio/genética
8.
Am J Pathol ; 179(3): 1221-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21741934

RESUMO

Tumor necrosis factor receptor 1 (TNFR1) activation in hepatocytes can trigger apoptotic or inflammatory signaling. The factors that determine which signaling pathway dominates are not clear and are thought to relate to the efficiency of death-inducing signaling complex (DISC) formation. However, the steps involved in DISC formation in hepatocytes are poorly understood. In characterizing DISC formation within cultured hepatocytes, we demonstrated that TNF-α exposure leads to the rapid formation of a DISC involving TNF-α, the TNFR-associated death domain adaptor molecule (TRADD), the Fas-associated death domain adaptor molecule (FADD), caspase-8, TNFR-associated factor 2 (TRAF2), and receptor-interacting protein (RIP). The inclusion of the sensitizing agent actinomycin D both accelerated and amplified the appearance of the DISC. Notably, TNFR1 along with some DISC components also appeared within mitochondria within 30 minutes. Whereas TNFR1 consistently co-localized with the TRADD, FADD, the caspase-8, and TRAF2 in the cytosolic fraction, TNFR1 in the mitochondria was associated only with caspase-8 after TNF-α exposure. Similar observations were made in vivo using TNF-α with D-galactosamine. Actinomycin D alone also enhanced the appearance of DISC components in both cytosol and the mitochondria. Thus the DISC that includes TNFR1 forms in the cytosol of hepatocytes under both survival and pro-apoptotic conditions. The observations also suggest that TNF-α-mediated signaling includes the translocation of TNFR1 to mitochondria.


Assuntos
Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/biossíntese , Hepatócitos/efeitos dos fármacos , Mitocôndrias/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Células Cultivadas , Dactinomicina/farmacologia , Proteína de Domínio de Morte Associada a Fas/metabolismo , Galactose/farmacologia , Hepatócitos/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Fator 2 Associado a Receptor de TNF/metabolismo
9.
Hepatology ; 53(6): 2053-62, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21437926

RESUMO

UNLABELLED: Adaptive responses to sepsis are necessary to prevent organ failure and death. Cellular signaling responses that limit cell death and structural damage allow a cell to withstand insult from sepsis to prevent irreversible organ dysfunction. One such protective pathway to reduce hepatocellular injury is the up-regulation of heme oxygenase-1 (HO-1) signaling. HO-1 is up-regulated in the liver in response to multiple stressors, including sepsis and lipopolysaccharide (LPS), and has been shown to limit cell death. Another recently recognized rudimentary cellular response to injury is autophagy. The aim of these investigations was to test the hypothesis that HO-1 protects against hepatocyte cell death in experimental sepsis in vivo or LPS in vitro via induction of autophagy. These data demonstrate that both HO-1 and autophagy are up-regulated in the liver after cecal ligation and puncture (CLP) in C57BL/6 mice or in primary mouse hepatocytes after treatment with LPS (100 ng/mL). CLP or LPS results in minimal hepatocyte cell death. Pharmacological inhibition of HO-1 activity using tin protoporphyrin or knockdown of HO-1 prevents the induction of autophagic signaling in these models and results in increased hepatocellular injury, apoptosis, and death. Furthermore, inhibition of autophagy using 3-methyladenine or small interfering RNA specific to VPS34, a class III phosphoinositide 3-kinase that is an upstream regulator of autophagy, resulted in hepatocyte apoptosis in vivo or in vitro. LPS induced phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK), in part, via HO-dependent signaling. Moreover, inhibition of p38 MAPK prevented CLP- or LPS-induced autophagy. CONCLUSION: Sepsis or LPS-induced autophagy protects against hepatocellular death, in part via an HO-1 p38 MAPK-dependent signaling. Further investigations are needed to elucidate how autophagic signaling prevents apoptosis and cell death.


Assuntos
Apoptose/fisiologia , Autofagia/fisiologia , Heme Oxigenase-1/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatopatias/complicações , Hepatopatias/microbiologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Ceco/lesões , Células Cultivadas , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/genética , Hepatócitos/efeitos dos fármacos , Ligadura/efeitos adversos , Lipopolissacarídeos/farmacologia , Hepatopatias/etiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Punções/efeitos adversos , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Curr Protoc ; 2(1): e342, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35038380

RESUMO

The protocols presented here describe steps for cryosectioning tissue samples to be used in light microscopy methodologies including histochemistry, enzyme immunohistochemistry, and immunofluorescence. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Cryosectioning.


Assuntos
Crioultramicrotomia , Imunofluorescência , Imuno-Histoquímica , Fixação de Tecidos
11.
Curr Protoc ; 2(1): e343, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35030299

RESUMO

Gravity flow whole-body perfusion maintains effective and reproducible preservation of tissue architecture critical to investigate pathobiology of multiple organs from the same specimen. The purpose of the protocols described within this article is to help the researcher optimize tissue harvest for multisystem pathobiology comparison. The protocols presented here describe tissue harvest for processing and cryopreservation to generate optimal samples for microscopy and parallel biochemical and molecular biology analysis. First, this paper outlines a protocol for tissue perfusion and organ harvest that allows the researcher multiple analysis options from the same research subject simultaneously. Second, this paper outlines a model to optimize ex-vivo tissue fixation for precious human sample preparation. Finally, this paper outlines a methodology for freezing tissue samples to optimize their capacity for biochemical and immunohistochemical analysis. Benefits and alternative approaches to retain cellular morphology in tissue harvest and processing are discussed. Also described within each section are common technical issues to assist problem-solving and troubleshooting. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Whole body in vivo tissue perfusion by gravity flow: preparation and surgical procedures Alternate Protocol: Human ex vivo tissue fixation Basic Protocol 2: Freezing of tissue samples.


Assuntos
Manejo de Espécimes , Coleta de Tecidos e Órgãos , Humanos , Perfusão , Fixação de Tecidos
12.
Curr Protoc ; 2(9): e549, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36102926

RESUMO

Immunohistochemistry is an essential technique for the localization and measurement of proteins in cells and tissues. This article describes methods for labeling proteins in adherent and suspension cell cultures and in tissue sections. Choices of antibodies and detection methods are discussed, and detailed troubleshooting guidelines are provided. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Immunofluorescent labeling of cells grown as adherent monolayers Alternate Protocol 1: Immunofluorescent labeling of cells in suspension Basic Protocol 2: Immunofluorescent labeling of tissue sections Alternate Protocol 2: Immunofluorescent labeling using streptavidin-biotin conjugates Alternate Protocol 3: Immunofluorescent double-labeling of tissue sections Alternate Protocol 4: Immunofluorescent double-labeling of tissue sections with two primary antibodies from the same host species.


Assuntos
Anticorpos , Biotina , Antígenos , Imuno-Histoquímica , Proteínas , Estreptavidina
13.
Blood Coagul Fibrinolysis ; 33(1): 14-24, 2022 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-34889809

RESUMO

Acute kidney injury (AKI) is common after trauma, but contributory factors are incompletely understood. Increases in plasma von Willebrand Factor (vWF) with concurrent decreases in ADAMTS13 are associated with renal microvascular thrombosis in other disease states, but similar findings have not been shown in trauma. We hypothesized that molecular changes in circulating vWF and ADAMTS13 promote AKI following traumatic injury. VWF antigen, vWF multimer composition and ADAMTS13 levels were compared in plasma samples from 16 trauma patients with and without trauma-induced AKI, obtained from the Prehospital Air Medical Plasma (PAMPer) biorepository. Renal histopathology and function, vWF and ADAMTS13 levels were assessed in parallel in a murine model of polytrauma and haemorrhage. VWF antigen was higher in trauma patients when compared with healthy controls [314% (253-349) vs. 100% (87-117)] [median (IQR)], while ADAMTS13 activity was lower [36.0% (30.1-44.7) vs. 100.0% (83.1-121.0)]. Patients who developed AKI showed significantly higher levels of high molecular weight multimeric vWF at 72-h when compared with non-AKI counterparts [32.9% (30.4-35.3) vs. 27.8% (24.6-30.8)]. Murine plasma cystatin C and vWF were elevated postpolytrauma model in mice, with associated decreases in ADAMTS13, and immunohistologic analysis demonstrated renal injury with small vessel plugs positive for fibrinogen and vWF. Following traumatic injury, the vWF-ADAMTS13 axis shifted towards a prothrombotic state in both trauma patients and a murine model. We further demonstrated that vWF-containing, microangiopathic deposits were concurrently produced as the prothrombotic changes were sustained during the days following trauma, potentially contributing to AKI development.


Assuntos
Injúria Renal Aguda , Fator de von Willebrand , Proteína ADAMTS13 , Animais , Humanos , Rim , Camundongos , Peso Molecular , Plasma
14.
J Cereb Blood Flow Metab ; 42(12): 2255-2269, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35854408

RESUMO

Epinephrine is the principal resuscitation therapy for pediatric cardiac arrest (CA). Clinical data suggest that although epinephrine increases the rate of resuscitation, it fails to improve neurological outcome, possibly secondary to reductions in microvascular flow. We characterized the effect of epinephrine vs. placebo administered at resuscitation from pediatric asphyxial CA on microvascular and macrovascular cortical perfusion assessed using in vivo multiphoton microscopy and laser speckle flowmetry, respectively, and on brain tissue oxygenation (PbO2), behavioral outcomes, and neuropathology in 16-18-day-old rats. Epinephrine-treated rats had a more rapid return of spontaneous circulation and brisk immediate cortical reperfusion during 1-3 min post-CA vs. placebo. However, at the microvascular level, epinephrine-treated rats had penetrating arteriole constriction and increases in both capillary stalling (no-reflow) and cortical capillary transit time 30-60 min post-CA vs. placebo. Placebo-treated rats had increased capillary diameters post-CA. The cortex was hypoxic post-CA in both groups. Epinephrine treatment worsened reference memory performance vs. shams. Hippocampal neuron counts did not differ between groups. Resuscitation with epinephrine enhanced immediate reperfusion but produced microvascular alterations during the first hour post-resuscitation, characterized by vasoconstriction, capillary stasis, prolonged cortical transit time, and absence of compensatory cortical vasodilation. Targeted therapies mitigating the deleterious microvascular effects of epinephrine are needed.


Assuntos
Reanimação Cardiopulmonar , Parada Cardíaca , Animais , Ratos , Microscopia , Circulação Cerebrovascular/fisiologia , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/complicações , Epinefrina/farmacologia , Epinefrina/uso terapêutico , Ressuscitação
15.
Mol Med ; 17(9-10): 1031-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21666957

RESUMO

Adaptive immune responses are induced in liver after major stresses such as hemorrhagic shock (HS) and trauma. There is emerging evidence that the inflammasome, the multiprotein platform that induces caspase-1 activation and promotes interleukin (IL)-1ß and IL-18 processing, is activated in response to cellular oxidative stress, such as after hypoxia, ischemia and HS. Additionally, damage-associated molecular patterns, such as those released after injury, have been shown to activate the inflammasome and caspase-1 through the NOD-like receptor (NLR) NLRP3. However, the role of the inflammasome in organ injury after HS and trauma is unknown. We therefore investigated inflammatory responses and end-organ injury in wild-type (WT) and caspase-1(-/-)mice in our model of HS with bilateral femur fracture (HS/BFF). We found that caspase-1(-/-) mice had higher levels of systemic inflammatory cytokines than WT mice. This result corresponded to higher levels of liver damage, cell death and neutrophil influx in caspase-1(-/-) liver compared with WT, although there was no difference in lung damage between experimental groups. To determine if hepatoprotection also depended on NLRP3, we subjected NLRP3(-/-) mice to HS/BFF, but found inflammatory responses and liver damage in these mice was similar to WT. Hepatoprotection was also not due to caspase-1-dependent cytokines, IL-1ß and IL-18. Altogether, these data suggest that caspase-1 is hepatoprotective, in part through regulation of cell death pathways in the liver after major trauma, and that caspase-1 activation after HS/BFF does not depend on NLRP3. These findings may have implications for the treatment of trauma patients and may lead to progress in prevention or treatment of multiple organ failure (MOF).


Assuntos
Caspase 1/metabolismo , Fraturas do Fêmur/fisiopatologia , Inflamação/enzimologia , Fígado/enzimologia , Choque Hemorrágico/fisiopatologia , Animais , Apoptose , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 1/genética , Ativação Enzimática , Inflamassomos/metabolismo , Inflamação/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Proteína 3 que Contém Domínio de Pirina da Família NLR , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Receptores de Interleucina-18/genética , Receptores de Interleucina-18/metabolismo
16.
Am J Physiol Regul Integr Comp Physiol ; 299(5): R1175-82, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702808

RESUMO

Although complement activation is known to occur in the setting of severe hemorrhagic shock and tissue trauma (HS/T), the extent to which complement drives the initial inflammatory response and end-organ damage is uncertain. In this study, complement factor 3-deficient (C3(-/-)) mice and wild-type control mice were subjected to 1.5-h hemorrhagic shock, bilateral femur fracture, and soft tissue injury, followed by 4.5-h resuscitation (HS/T). C57BL/6 mice were also given 15 U of cobra venom factor (CVF) or phosphate-buffered saline injected intraperitoneally, followed by HS/T 24 h later. The results showed that HS/T resulted in C3 consumption in wild-type mice and C3 deposition in injured livers. C3(-/-) mice had significantly lower serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and circulating DNA levels, together with much lower circulating interleukin (IL)-6, IL-10, and high-mobility group box 1 (HMGB1) levels. Temporary C3 depletion by CVF preconditioning also led to reduced transaminases and a blunted cytokine release. C3(-/-) mice displayed well-preserved hepatic structure. C3(-/-) mice subjected to HS/T had higher levels of heme oxygenase-1, which has been associated with tissue protection in HS models. Our data indicate that complement activation contributes to inflammatory pathways and liver damage in HS/T. This suggests that targeting complement activation in the setting of severe injury could be useful.


Assuntos
Ativação do Complemento , Complemento C3/deficiência , Hepatopatias/prevenção & controle , Fígado/imunologia , Choque Hemorrágico/imunologia , Síndrome de Resposta Inflamatória Sistêmica/prevenção & controle , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Biomarcadores/sangue , Complemento C3/genética , DNA de Cadeia Simples/sangue , Modelos Animais de Doenças , Venenos Elapídicos/administração & dosagem , Fraturas do Fêmur/complicações , Fraturas do Fêmur/imunologia , Proteína HMGB1/sangue , Heme Oxigenase (Desciclizante)/sangue , Injeções Intraperitoneais , Interleucina-10/sangue , Interleucina-6/sangue , Fígado/metabolismo , Hepatopatias/sangue , Hepatopatias/genética , Hepatopatias/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Choque Hemorrágico/sangue , Choque Hemorrágico/genética , Lesões dos Tecidos Moles/complicações , Lesões dos Tecidos Moles/imunologia , Síndrome de Resposta Inflamatória Sistêmica/sangue , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Fatores de Tempo
17.
Front Immunol ; 11: 229, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32328059

RESUMO

High-mobility group box-1 (HMGB1), a ubiquitous nuclear protein, acts as a late mediator of lethality when released extracellularly during sepsis. The major source of circulating HMGB1 in sepsis is hepatocytes. However, the mechanism of HMGB1 release of hepatocytes during sepsis is not very clear. We have previously shown that bacterial endotoxin [lipopolysaccharide (LPS)] sensing pathways, including Toll-like receptor (TLR)4 and caspase-11, regulate hepatocyte HMGB1 release in response to LPS. Here, we report the novel function of caspase-11 and gasdermin D (GsdmD) in LPS-induced active HMGB1 released from hepatocytes. HMGB1 release during endotoxemia was caspase-11/GsdmD dependent via an active way in vivo and in vitro. Caspase-11/GsdmD was responsible for HMGB1 translocation from nucleus to the cytoplasm via calcium changing-induced phosphorylation of calcium-calmodulin kinase kinase (camkk)ß during endotoxemia. Cleaved GsdmD accumulated on the endoplasmic reticulum, suggesting this may lead to calcium leak and intracellular calcium increase. Furthermore, we investigated that exosome was an important pathway for HMGB1 release from hepatocytes; this process was dependent on TLR4, independent of caspase-11 and GsdmD in vivo and in vitro. These findings provide a novel mechanism that TLR4 signaling results in an increase in caspase-11 expression, as well as increased exosome release, while caspase-11/GsdmD activation/cleavage leads to accumulation of HMGB1 in the cytoplasm through a process associated with the release of calcium from the endoplasmic reticulum and camkkß activation.


Assuntos
Caspases Iniciadoras/metabolismo , Exossomos/metabolismo , Proteína HMGB1/metabolismo , Hepatócitos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Sepse/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Cálcio/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Caspases Iniciadoras/genética , Retículo Endoplasmático/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Ligação a Fosfato/genética , Transdução de Sinais , Receptor 4 Toll-Like/genética
18.
J Cereb Blood Flow Metab ; 39(5): 913-925, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29192562

RESUMO

Decreased cerebral blood flow (CBF) after cardiac arrest (CA) contributes to secondary ischemic injury in infants and children. We previously reported cortical hypoperfusion with tissue hypoxia early in a pediatric rat model of asphyxial CA. In order to identify specific alterations as potential therapeutic targets to improve cortical hypoperfusion post-CA, we characterize the CBF alterations at the cortical microvascular level in vivo using multiphoton microscopy. We hypothesize that microvascular constriction and disturbances of capillary red blood cell (RBC) flow contribute to cortical hypoperfusion post-CA. After resuscitation from 9 min asphyxial CA, transient dilation of capillaries and venules at 5 min was followed by pial arteriolar constriction at 30 and 60 min (19.6 ± 1.3, 19.3 ± 1.2 µm at 30, 60 min vs. 22.0 ± 1.2 µm at baseline, p < 0.05). At the capillary level, microcirculatory disturbances were highly heterogeneous, with RBC stasis observed in 25.4% of capillaries at 30 min post-CA. Overall, the capillary plasma mean transit time was increased post-CA by 139.7 ± 51.5%, p < 0.05. In conclusion, pial arteriolar constriction, the no-reflow phenomenon and increased plasma transit time were observed post-CA. Our results detail the microvascular disturbances in a pediatric asphyxial CA model and provide a powerful platform for assessing specific vascular-targeted therapies.


Assuntos
Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Parada Cardíaca/complicações , Microcirculação , Fenômeno de não Refluxo/etiologia , Animais , Encéfalo/fisiopatologia , Parada Cardíaca/fisiopatologia , Masculino , Fenômeno de não Refluxo/fisiopatologia , Ratos Sprague-Dawley , Vasodilatação
19.
Apoptosis ; 13(8): 983-92, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18543108

RESUMO

FADD/MORT1 (The adaptor protein of Fas Associate Death Domain/Mediator of Receptor Induced Toxicity) is essential for signal transduction of death receptor signaling. We have previously shown that FADD is significantly up-regulated in TNFalpha/ActD induced apoptosis. Over-expression of FADD also induces death of lung cancer cells and primary hepatocytes. We hypothesize that the increase in detectable FADD levels require the proximal steps in apoptotic signaling and speculated that FADD would be redistributed in cells destined to undergo apoptosis. We show that monomeric non-phosphorylated FADD is up-regulated in hepatocytes treated with TNFalpha/ActD and that it accumulates in the cytoplasm. Nuclear phosphorylated FADD decreases with TNFalpha/ActD treatment. Dimeric FADD in the cytoplasm remains constant with TNFalpha/ActD. The change in FADD levels and distribution was dependent on caspase-3, caspase-8 activity and the presence of BID. Thus, changes in FADD levels and distribution are downstream of caspase activation and mitochondria changes that are initiated by the formation of the DISC complex. Changes in FADD levels and distribution may represent a novel feed-forward mechanism to propagate apoptosis signaling in hepatocytes.


Assuntos
Apoptose/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Caspases/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Hepatócitos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Apoptose/efeitos dos fármacos , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/efeitos dos fármacos , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 8/efeitos dos fármacos , Caspase 8/metabolismo , Caspases/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Dactinomicina/farmacologia , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Proteína de Domínio de Morte Associada a Fas/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
20.
Nitric Oxide ; 18(4): 256-65, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18313411

RESUMO

There is evidence that NO can regulate CO production, however less is known about CO regulation of NO synthesis. Our studies were undertaken to define how CO regulates iNOS in cultured hepatocytes. CO (250ppm) exposure resulted in a significant decrease in iNOS protein, nitrite production, level of active iNOS dimer and cytosolic iNOS activity in cells stimulated with cytokines (IL-1beta) or transfected with the human iNOS gene. However, IL-1beta-stimulated iNOS mRNA expression was unaffected by CO. These effects of CO on iNOS protein levels were inhibited when CO was scavenged using hemoglobin. HO-1 induction with an adenoviral vector carrying HO-1 showed a decrease in total iNOS protein, nitrite production, and iNOS dimer level from cells stimulated by IL-1beta. iNOS protein level was significantly higher in lung endothelial cells isolated from HO-1 knockout mice compared to wild type cultures stimulated with cytokines mixture. CO was found to increase p38 phosphorylation and p38 inhibition using SB203580 increased iNOS protein levels in response to IL-1beta. Interestingly, proteasome inhibitors (MG132 and Lactacystin) and an autophagy inhibitor (3-methyladenine) reversed CO influence iNOS levels. Our results imply that CO exposure decreases NO production by suppressing dimer formation and increasing iNOS degradation through a process involving p38 activation.


Assuntos
Monóxido de Carbono/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Interleucina-1beta/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Adenoviridae/genética , Animais , Células Cultivadas , Cobalto/química , Dimerização , Ativação Enzimática , Regulação da Expressão Gênica , Vetores Genéticos/genética , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Nitritos/metabolismo , Porfirinas/química , Porfirinas/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Ratos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA