Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(5): e22282, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35344224

RESUMEN

Inflammatory bowel disease (IBD) represents a set of idiopathic and chronic inflammatory diseases of the gastrointestinal tract. Central to the pathogenesis of IBD is a dysregulation of normal intestinal epithelial homeostasis. cGAS is a DNA-sensing receptor demonstrated to promote autophagy, a mechanism that removes dysfunctional cellular components. Beclin-1 is a crucial protein involved in the initiation of autophagy. We hypothesized that cGAS plays a key role in intestinal homeostasis by upregulating Beclin-1-mediated autophagy. We evaluated intestinal cGAS levels in humans with IBD and in murine colonic tissue after performing a 2% dextran sulfate sodium (DSS) colitis model. Autophagy and cell death mechanisms were studied in cGAS KO and WT mice via qPCR, WB analysis, H&E, IF, and TUNEL staining. Autophagy was measured in stimulated intestinal epithelial cells (IECs) via WB analysis. Our data demonstrates cGAS to be upregulated during human and murine colitis. Furthermore, cGAS deficiency leads to worsened colitis and decreased levels of autophagy proteins including Beclin-1 and LC3-II. Co-IP demonstrates a direct binding between cGAS and Beclin-1 in IECs. Transfection of cGAS in stimulated HCT-116 cells leads to increased autophagy. IECs isolated from cGAS KO have diminished autophagic flux. cGAS KO mice subjected to DSS have increased cell death and cleaved caspase-3. Lastly, treatment of cGAS KO mice with rapamycin decreased the severity of colitis. Our data suggest that cGAS maintains intestinal epithelial homeostasis during human IBD and murine colitis by upregulating Beclin-1-mediated autophagy and preventing IEC death. Rescue of autophagy can attenuate the severity of colitis associated with cGAS deficiency.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Autofagia/fisiología , Beclina-1/genética , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Homeostasis , Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Nucleótidos Cíclicos , Nucleotidiltransferasas/genética
2.
Hepatology ; 73(6): 2494-2509, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32924145

RESUMEN

BACKGROUND AND AIMS: Liver ischemia/reperfusion injury (IRI) induces local and systemic inflammation in which neutrophil extracellular traps (NETs) are major drivers. IRI markedly augments metastatic growth, which is consistent with the notion that the liver IRI can serve as a premetastatic niche. Exercise training (ExT) confers a sustainable protection, reducing IRI in some animal models, and has been associated with improved survival in patients with cancer; however, the impact of ExT on liver IRI or development of hepatic metastases is unknown. APPROACH AND RESULTS: Mice were randomized into exercise (ExT) and sedentary groups before liver IRI and tumor injection. Computerized dynamic network analysis of 20 inflammatory mediators was used to dissect the sequence of mediator interactions after ischemia/reperfusion (I/R) that induce injury. ExT mice showed a significant decrease in hepatic IRI and tissue necrosis. This coincided with disassembly of complex networks among inflammatory mediators seen in sedentary mice. Neutrophil infiltration and NET formation were decreased in the ExT group, which suppressed the expression of liver endothelial cell adhesion molecules. Concurrently, ExT mice revealed a distinct population of infiltrating macrophages expressing M2 phenotypic genes. In a metastatic model, fewer metastases were present 3 weeks after I/R in the ExT mice, a finding that correlated with a marked increase in tumor-suppressing T cells within the tumor microenvironment. CONCLUSIONS: ExT preconditioning mitigates the inflammatory response to liver IRI, protecting the liver from injury and metastases. In light of these findings, potential may exist for the reduction of liver premetastatic niches induced by liver IRI through the use of ExT as a nonpharmacologic therapy before curative surgical approaches.


Asunto(s)
Trampas Extracelulares/inmunología , Inflamación , Hepatopatías , Metástasis de la Neoplasia , Infiltración Neutrófila/inmunología , Condicionamiento Físico Animal/métodos , Daño por Reperfusión , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Inmunidad , Inflamación/etiología , Inflamación/inmunología , Inflamación/terapia , Hepatopatías/inmunología , Hepatopatías/patología , Hepatopatías/terapia , Ratones , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/terapia , Factores Protectores , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Resultado del Tratamiento
3.
Mol Med ; 27(1): 18, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33632134

RESUMEN

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.


Asunto(s)
Ácidos Docosahexaenoicos/uso terapéutico , Hepatopatías/tratamiento farmacológico , Sustancias Protectoras/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Formil Péptido/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Supervivencia Celular/efectos de los fármacos , Citocinas/sangre , Ácidos Docosahexaenoicos/farmacología , Glutatión Peroxidasa/metabolismo , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hepatopatías/sangre , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Malondialdehído/metabolismo , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Receptores de Formil Péptido/genética , Daño por Reperfusión/sangre , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos
4.
Hepatology ; 72(4): 1394-1411, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31997373

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Carboxiliasas/fisiología , Hepatocitos/metabolismo , Hígado/irrigación sanguínea , Factor 2 Relacionado con NF-E2/fisiología , Daño por Reperfusión/prevención & control , Succinatos/farmacología , Animales , Humanos , Hidroliasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Transducción de Señal/fisiología , Succinatos/uso terapéutico
5.
J Immunol ; 202(1): 268-277, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30504418

RESUMEN

Hepatic ischemia reperfusion (I/R) is a clinically relevant model of acute sterile inflammation leading to a reverberating, self-sustaining inflammatory response with resultant necrosis. We hypothesized that computerized dynamic network analysis (DyNA) of 20 inflammatory mediators could help dissect the sequence of post-I/R mediator interactions that induce injury. Although the majority of measured inflammatory mediators become elevated in the first 24 h, we predicted that only a few would be secreted early in the process and serve as organizational centers of downstream intermediator complexity. In support of this hypothesis, DyNA inferred a central organizing role for IL-17A during the first 3 h of reperfusion. After that, DyNA revealed connections among almost all the inflammatory mediators, representing an ongoing cytokine storm. Blocking IL-17A immediately after reperfusion disassembled the inflammatory networks and protected the liver from injury. Disassembly of the networks was not achieved if IL-17A blockage was delayed two or more hours postreperfusion. Network disassembly was accompanied by decrease in neutrophil infiltration and neutrophil extracellular trap (NET) formation. By contrast, administration of recombinant IL-17A increased neutrophil infiltration, NET formation, and liver necrosis. The administration of DNase, a NET inhibitor, significantly reduced hepatic damage despite prior administration of IL-17A, and DNase also disassembled the inflammatory networks. In vitro, IL-17A was a potent promoter of NET formation. Therefore, computational analysis identified IL-17A's early, central organizing role in the rapid evolution of a network of inflammatory mediators that induce neutrophil infiltration and NET formation responsible for hepatic damage after liver I/R.


Asunto(s)
Biología Computacional/métodos , Simulación por Computador , Trampas Extracelulares/inmunología , Interleucina-17/metabolismo , Hígado/patología , Neutrófilos/inmunología , Daño por Reperfusión/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Células Cultivadas , Desoxirribonucleasas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-17/inmunología , Hígado/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Infiltración Neutrófila , Mapas de Interacción de Proteínas
6.
Mol Med ; 26(1): 115, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33238880

RESUMEN

BACKGROUND: Circulating high-mobility group box 1 (HMGB1) plays important roles in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Intracellular HMGB1 is critical for the biology of hepatocytes. However, the intracellular role of HMGB1 in hepatocellular steatosis is unknown. Therefore, we aimed to investigate the role of hepatocyte-specific HMGB1 (HC-HMGB1) in development of hepatic steatosis. METHODS: Wild type (WT) C57BL/6 and HC-HMGB1-/- mice were fed high-fat diet (HFD) or low-fat diet (LFD) for up to 16 weeks. RESULTS: As expected, HMGB1 translocated from nuclear into cytoplasm and released into circulation after HFD treatment. HC-HMGB1 deficiency significantly reduced circulating HMGB1, suggesting that hepatocyte is a major source of circulating HMGB1 during NAFLD. Unexpectedly, HC-HMGB1 deficiency promoted rapid weight gain with enhanced hepatic fat deposition compared with WT at as early as 4 weeks after HFD treatment. Furthermore, there was no difference between WT and HC-HMGB1-/- mice in glucose tolerance, energy expenditure, liver damage or systemic inflammation. Interestingly, hepatic gene expression related to free fatty acid (FFA) ß-oxidation was significantly down-regulated in HC-HMGB1-/- mice compared with WT, and endoplasmic reticulum (ER) stress markers were significantly higher in livers of HC-HMGB1-/- mice. In vitro experiments using primary mouse hepatocytes showed absence of HMGB1 increased FFA-induced intracellular lipid accumulation, accompanied by increased ER-stress, significant downregulation of FFA ß-oxidation, and reduced oxidative phosphorylation. CONCLUSIONS: Our findings suggest that hepatocyte HMGB1 protects against dysregulated lipid metabolism via maintenance of ß-oxidation and prevention of ER stress. This represents a novel mechanism for HMGB1-regulation of hepatocellular steatosis, and suggests that stabilizing HMGB1 in hepatocytes may be effective strategies for prevention and treatment of NAFLD.


Asunto(s)
Dieta Alta en Grasa , Hígado Graso/etiología , Hígado Graso/metabolismo , Proteína HMGB1/genética , Hepatocitos/metabolismo , Estrés Fisiológico , Animales , Biopsia , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/patología , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo , Oxidación-Reducción
7.
Mol Med ; 26(1): 69, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641037

RESUMEN

BACKGROUND: We previously showed that the autophagy inhibitor chloroquine (CQ) increases inflammatory cleaved caspase-1 activity in myocytes, and that caspase-1/11 is protective in sterile liver injury. However, the role of caspase-1/11 in the recovery of muscle from ischemia caused by peripheral arterial disease is unknown. We hypothesized that caspase-1/11 mediates recovery in muscle via effects on autophagy and this is modulated by CQ. METHODS: C57Bl/6 J (WT) and caspase-1/11 double-knockout (KO) mice underwent femoral artery ligation (a model of hind-limb ischemia) with or without CQ (50 mg/kg IP every 2nd day). CQ effects on autophagosome formation, microtubule associated protein 1A/1B-light chain 3 (LC3), and caspase-1 expression was measured using electron microscopy and immunofluorescence. Laser Doppler perfusion imaging documented perfusion every 7 days. After 21 days, in situ physiologic testing in tibialis anterior muscle assessed peak force contraction, and myocyte size and fibrosis was also measured. Muscle satellite cell (MuSC) oxygen consumption rate (OCR) and extracellular acidification rate was measured. Caspase-1 and glycolytic enzyme expression was detected by Western blot. RESULTS: CQ increased autophagosomes, LC3 consolidation, total caspase-1 expression and cleaved caspase-1 in muscle. Perfusion, fibrosis, myofiber regeneration, muscle contraction, MuSC fusion, OCR, ECAR and glycolytic enzyme expression was variably affected by CQ depending on presence of caspase-1/11. CQ decreased perfusion recovery, fibrosis and myofiber size in WT but not caspase-1/11KO mice. CQ diminished peak force in whole muscle, and myocyte fusion in MuSC and these effects were exacerbated in caspase-1/11KO mice. CQ reductions in maximal respiration and ATP production were reduced in caspase-1/11KO mice. Caspase-1/11KO MuSC had significant increases in protein kinase isoforms and aldolase with decreased ECAR. CONCLUSION: Caspase-1/11 signaling affects the response to ischemia in muscle and effects are variably modulated by CQ. This may be critically important for disease treated with CQ and its derivatives, including novel viral diseases (e.g. COVID-19) that are expected to affect patients with comorbidities like cardiovascular disease.


Asunto(s)
Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Cloroquina/farmacología , Infecciones por Coronavirus/patología , Isquemia/patología , Músculo Esquelético/patología , Neumonía Viral/patología , Animales , Autofagosomas/metabolismo , Autofagia/efectos de los fármacos , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Glucólisis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Células Musculares/metabolismo , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Fosforilación Oxidativa , Pandemias , Enfermedad Arterial Periférica/patología , Neumonía Viral/tratamiento farmacológico , Regeneración , SARS-CoV-2 , Transducción de Señal , Tratamiento Farmacológico de COVID-19
8.
Hepatology ; 68(4): 1347-1360, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29631332

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a progressive, inflammatory form of fatty liver disease. It is the most rapidly rising risk factor for the development of hepatocellular carcinoma (HCC), which can arise in NASH with or without cirrhosis. The inflammatory signals promoting the progression of NASH to HCC remain largely unknown. The propensity of neutrophils to expel decondensed chromatin embedded with inflammatory proteins, known as neutrophil extracellular traps (NETs), has been shown to be important in chronic inflammatory conditions and in cancer progression. In this study, we asked whether NET formation occurs in NASH and contributes to the progression of HCC. We found elevated levels of a NET marker in serum of patients with NASH. In livers from STAM mice (NASH induced by neonatal streptozotocin and high-fat diet), early neutrophil infiltration and NET formation were seen, followed by an influx of monocyte-derived macrophages, production of inflammatory cytokines, and progression of HCC. Inhibiting NET formation, through treatment with deoxyribonuclease (DNase) or using mice knocked out for peptidyl arginine deaminase type IV (PAD4-/- ), did not affect the development of a fatty liver but altered the consequent pattern of liver inflammation, which ultimately resulted in decreased tumor growth. Mechanistically, we found that commonly elevated free fatty acids stimulate NET formation in vitro. CONCLUSION: Our findings implicate NETs in the protumorigenic inflammatory environment in NASH, suggesting that their elimination may reduce the progression of liver cancer in NASH. (Hepatology 2018).


Asunto(s)
Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Biomarcadores/metabolismo , Biopsia con Aguja , Carcinoma Hepatocelular/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Pronóstico , Distribución Aleatoria , Medición de Riesgo
9.
Ann Hematol ; 98(7): 1603-1610, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31020347

RESUMEN

Platelets are activated in solid cancers, including pancreatic ductal adenocarcinoma (PDA), a highly aggressive malignancy with a devastating prognosis and limited therapeutic options. The mechanisms by which activated platelets regulate tumor progression are poorly understood. The nucleotide-binding domain leucine-rich repeat containing protein 3 (NLRP3) inflammasome is a key inflammatory mechanism recently identified in platelets, which controls platelet activation and aggregation. In an orthotopic PDA mouse model involving surgical implantation of Panc02 murine cancer cells into the tail of the pancreas, we show that the NLRP3 inflammasome in circulating platelets is upregulated in pancreatic cancer. Pharmacological inhibition or genetic ablation of NLRP3 in platelets resulted in decreased platelet activation, platelet aggregation, and tumor progression. Moreover, interfering with platelet NLRP3 signaling significantly improved survival of tumor-bearing mice. Hence, the platelet NLRP3 inflammasome plays a critical role in PDA and might represent a novel therapeutic target.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Pancreáticas/metabolismo , Agregación Plaquetaria , Regulación hacia Arriba , Animales , Plaquetas , Línea Celular Tumoral , Inflamasomas/genética , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología
10.
Am J Physiol Gastrointest Liver Physiol ; 314(6): G655-G667, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29446653

RESUMEN

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.


Asunto(s)
Autofagia/fisiología , Interferón Tipo I , Hígado , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , Daño por Reperfusión , Animales , Apoptosis/fisiología , ADN Nucleotidiltransferasas/fisiología , Inductores de Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Hígado/irrigación sanguínea , Hígado/metabolismo , Hígado/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Sustancias Protectoras/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Transducción de Señal
11.
Immunol Cell Biol ; 96(9): 935-947, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29672927

RESUMEN

Sepsis is defined as infection with organ dysfunction due to a dysregulated immune response. The lung is one of the most vulnerable organs at the onset of sepsis. Interleukin (IL)-33 can be released by injured epithelial and endothelial cells in the lung and regulate immune activation and infiltration. Therefore, we postulated that IL-33 would contribute to the immune response in the lung during sepsis. Using the cecal ligation and puncture (CLP) sepsis model, we show here that IL-33 contributes significantly to both sepsis-induced inflammation in the lung and systemic inflammatory response in the early phase of sepsis. Despite the higher intra-peritoneal bacterial burden, the absence of IL-33 resulted in less infiltration of neutrophils and monocytes into the lungs in association with lower circulating, lung and liver cytokine levels as well as reduced lung injury at 6 h after sepsis. IL-33 was required for the upregulation of IL-5 in type 2 Innate Lymphoid Cells (ILC2), while IL-5 neutralization suppressed neutrophil and monocyte infiltration in the lungs during CLP sepsis. This reduction in leukocyte infiltration in IL-33-deficient mice was reversed by administration of recombinant IL-5. These results indicate that IL-33 plays a major role in the local inflammatory changes in the lung, in part, by regulating IL-5 and this axis contributes to lung injury early after the onset of sepsis.


Asunto(s)
Interleucina-33/inmunología , Interleucina-5/inmunología , Linfocitos/inmunología , Neumonía/inmunología , Sepsis/inmunología , Animales , Modelos Animales de Enfermedad , Inmunidad Innata , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila
12.
Hepatology ; 65(1): 253-268, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27774630

RESUMEN

Sterile liver inflammation, such as liver ischemia-reperfusion, hemorrhagic shock after trauma, and drug-induced liver injury, is initiated and regulated by endogenous mediators including DNA and reactive oxygen species. Here, we identify a mechanism for redox-mediated regulation of absent in melanoma 2 (AIM2) inflammasome activation in hepatocytes after redox stress in mice, which occurs through interaction with cytosolic high mobility group box 1 (HMGB1). We show that in liver during hemorrhagic shock in mice and in hepatocytes after hypoxia with reoxygenation, cytosolic HMGB1 associates with AIM2 and is required for activation of caspase-1 in response to cytosolic DNA. Activation of caspase-1 through AIM2 leads to subsequent hepatoprotective responses such as autophagy. HMGB1 binds to AIM2 at a non-DNA-binding site on the hematopoietic interferon-inducible nuclear antigen domain of AIM2 to facilitate inflammasome and caspase-1 activation in hepatocytes. Furthermore, binding of HMGB1 to AIM2 is stronger with fully reduced all-thiol HMGB1 than with partially oxidized disulfide-HMGB1, and binding strength corresponds to caspase-1 activation. These data suggest that HMGB1 redox status regulates AIM2 inflammasome activation. CONCLUSION: These findings suggest a novel and important mechanism for regulation of AIM2 inflammasome activation in hepatocytes during redox stress and may suggest broader implications for how this and other inflammasomes are activated and how their activation is regulated during cell stress, as well as the mechanisms of inflammasome regulation in nonimmune cell types. (Hepatology 2017;65:253-268).


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hepatocitos/metabolismo , Inflamasomas/metabolismo , Hepatopatías/etiología , Animales , Caspasa 1/metabolismo , Proteína HMGB1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción
13.
Hepatology ; 66(1): 182-197, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28370295

RESUMEN

The ability of cancer cells to survive and grow under hypoxic conditions has been known for decades, but the mechanisms remain poorly understood. Under certain conditions, cancer cells undergo changes in their bioenergetic profile to favor mitochondrial respiration by activating the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) and up-regulating mitochondrial biogenesis. In this study, we hypothesized that augmented mitochondrial biogenesis plays a critical role for cancer cells to survive hypoxia. Consistent with this hypothesis, both hypoxic human hepatocellular carcinoma (HCC) tumors and HCC cell lines subjected to hypoxia increase mitochondrial biogenesis. Silencing of PGC-1α in hypoxic HCC cell lines halts their proliferation. Mechanistic investigations in vitro indicated that intracellular high mobility group box 1 (HMGB1) protein, a nuclear protein overexpressed in HCC, is essential for the process. Silencing of HMGB1 in hypoxic HCC cell lines resulted in a significant decrease in PGC-1α activation and mitochondrial biogenesis. Without HMGB1, hypoxic HCC cells had significantly reduced adenosine triphosphate production, decreased cellular proliferation, and increased apoptosis. In a diethylnitrosamine-induced murine model of HCC, genetic blocking of HMGB1 in hypoxic tumors resulted in a significant decrease in tumor growth. Tumors lacking HMGB1 had a significant reduction in mitochondrial biogenesis and a significant increase in mitochondrial dysfunction. Further in vitro mechanistic experiments indicated that during hypoxia HMGB1 translocates from the nucleus to the cytoplasm and binds to cytoplasmic Toll-like receptor-9. This binding leads to activation of p38 and subsequent phosphorylation of PGC-1α, with resultant up-regulation of mitochondrial biogenesis. CONCLUSION: Taken together, our findings suggest that during hypoxia HMGB1 up-regulates mitochondrial biogenesis in HCC cancer cells, promoting tumor survival and proliferation. (Hepatology 2017;66:182-197).


Asunto(s)
Carcinoma Hepatocelular/genética , Proteína HMGB1/genética , Neoplasias Hepáticas/genética , Biogénesis de Organelos , Receptor Toll-Like 9/metabolismo , Animales , Carcinoma Hepatocelular/patología , Hipoxia de la Célula , Supervivencia Celular , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distribución Aleatoria , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas
14.
J Biol Chem ; 291(29): 15093-107, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226571

RESUMEN

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.


Asunto(s)
Hepatocitos/inmunología , Hepatocitos/metabolismo , Interferón beta/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Bicatenario/inmunología , ARN Bicatenario/farmacología , Receptor Toll-Like 3/metabolismo , eIF-2 Quinasa/metabolismo , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Hepatocitos/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Tirosina/química , Regulación hacia Arriba/efectos de los fármacos , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
15.
J Hepatol ; 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28943296

RESUMEN

BACKGROUND & AIMS: Neutrophils and liver sinusoidal endothelial cells (LSECs) both contribute to sterile inflammatory injury during ischemia/reperfusion (I/R), a well-known liver surgical stress. Interleukin-33 (IL-33) has been shown to drive neutrophil infiltration during inflammatory responses through its receptor ST2. We recently reported that infiltrating neutrophils form neutrophil extracellular traps (NETs), which exacerbate sterile inflammatory injury in liver I/R. Here, we sought to determine the role of IL-33 in NET formation during liver sterile inflammation. METHODS: Evaluation of IL-33 forming NETs was investigated using a partial liver I/R model to generate sterile injury in healthy WT, IL-33 and ST2 knockouts. Serum levels of IL-33 and myeloperoxidase (MPO)-DNA complex were measured in both humans and mice after the first surgery. Liver damage was assessed. Mouse neutrophil depletion was performed by intraperitoneal injection of anti-Ly6G antibody before I/R. RESULTS: Patients undergoing liver resection showed a significant increase in serum IL-33 compared to healthy volunteers. This coincided with higher serum MPO-DNA complexes. NET formation was decreased in IL-33 and ST2 knockout mice compared with control mice, after liver I/R. IL-33 or ST2 deficiency protected livers from I/R injury, whereas rIL-33 administration during I/R exacerbated hepatotoxicity and systemic inflammation. In vitro, IL-33 is released from LSECs to promote NET formation. IL-33 deficient LSECs failed to induce NETs. ST2 deficient neutrophils limited their capacity to form NETs in vitro and adoptive transfer of ST2 knockout neutrophils to neutrophil-depleted WT mice significantly decreased NET formation. CONCLUSIONS: Data establish that IL-33, mainly released from LSECs, causes excessive sterile inflammation after hepatic I/R by inducing NET formation. Therapeutic targeting of IL-33/ST2 might extend novel strategies to minimize organ damage in various clinical settings associated with sterile inflammation. LAY SUMMARY: Liver ischemia and reperfusion injury results in the formation of neutrophil extracellular traps, which contribute to organ damage in liver surgeries. Herein, we show that IL-33 is released from liver sinusoidal endothelial cells to promote NET formation during liver I/R, which exacerbates inflammatory cascades and sterile inflammation.

16.
Biochem Biophys Res Commun ; 478(1): 143-148, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27449608

RESUMEN

Platelets are circulating cellular sensors that express and release the damage-associated molecular pattern molecule (DAMP) high-mobility group box 1 (HMGB1) at sites of disrupted vascular and tissue integrity. We have recently identified platelet-derived HMGB1 as a critical mediator of thrombosis. The role of platelet-derived HMGB1 in mediating interactions with monocytes remains unknown. In transgenic mice with platelet-specific ablation of HMGB1 and neutralization studies, we show that HMGB1 derived from platelets promotes recruitment of monocytes and prevents monocytes from undergoing apoptosis. During experimental trauma and hemorrhagic shock, infiltrated monocytes in the lung and liver were significantly attenuated in mice lacking HMGB1 in platelets. Platelet-derived HMGB1 mediated monocyte migration via the receptor for advanced glycation end products (RAGE) and suppressed apoptosis via toll-like receptor 4 (TLR4)-dependent activation of MAPK/ERK (extracellular signal-regulated kinase) in monocytes. In conclusion, we identify platelet-derived HMGB1 as a critical regulator of monocyte recruitment and apoptosis, with potential implications in disease states associated with thrombosis and inflammation.


Asunto(s)
Apoptosis/inmunología , Proteína HMGB1/inmunología , Inflamación/inmunología , Monocitos/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Receptor Toll-Like 4/inmunología , Animales , Movimiento Celular/inmunología , Células Cultivadas , Inflamación/patología , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Transgénicos , Monocitos/citología , Monocitos/patología
17.
Hepatology ; 62(2): 600-14, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25855125

RESUMEN

UNLABELLED: Innate immunity plays a crucial role in the response to sterile inflammation such as liver ischemia/reperfusion (I/R) injury. The initiation of liver I/R injury results in the release of damage-associated molecular patterns, which trigger an innate immune and inflammatory cascade through pattern recognition receptors. Neutrophils are recruited to the liver after I/R and contribute to organ damage and innate immune and inflammatory responses. Formation of neutrophil extracellular traps (NETs) has been recently found in response to various stimuli. However, the role of NETs during liver I/R injury remains unknown. We show that NETs form in the sinusoids of ischemic liver lobes in vivo. This was associated with increased NET markers, serum level of myeloperoxidase-DNA complexes, and tissue level of citrullinated-histone H3 compared to control mice. Treatment with peptidyl-arginine-deiminase 4 inhibitor or DNase I significantly protected hepatocytes and reduced inflammation after liver I/R as evidenced by inhibition of NET formation, indicating the pathophysiological role of NETs in liver I/R injury. In vitro, NETs increase hepatocyte death and induce Kupffer cells to release proinflammatory cytokines. Damage-associated molecular patterns, such as High Mobility Group Box 1 and histones, released by injured hepatocytes stimulate NET formation through Toll-like receptor (TLR4)- and TLR9-MyD88 signaling pathways. After neutrophil depletion in mice, the adoptive transfer of TLR4 knockout or TLR9 knockout neutrophils confers significant protection from liver I/R injury with a significant decrease in NET formation. In addition, we found inhibition of NET formation by the peptidyl-arginine-deiminase 4 inhibitor and that DNase I reduces High Mobility Group Box 1 and histone-mediated liver I/R injury. CONCLUSION: Damage-associated molecular patterns released during liver I/R promote NET formation through the TLR signaling pathway. Development of NETs subsequently exacerbates organ damage and initiates inflammatory responses during liver I/R.


Asunto(s)
Trampas Extracelulares/metabolismo , Proteína HMGB1/metabolismo , Hígado/lesiones , Neutrófilos/inmunología , Transducción de Señal/genética , Animales , Biomarcadores/sangre , Células Cultivadas , Modelos Animales de Enfermedad , Trampas Extracelulares/inmunología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Hepatocitos/inmunología , Hepatocitos/metabolismo , Histonas/metabolismo , Inmunidad Innata/inmunología , Immunoblotting , Inflamación/metabolismo , Inflamación/fisiopatología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/inmunología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Biología Molecular , Neutrófilos/metabolismo , Distribución Aleatoria , Sensibilidad y Especificidad
18.
J Immunol ; 193(5): 2405-15, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070845

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/inmunología , Autofagia/efectos de los fármacos , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Serina-Treonina Quinasas TOR/inmunología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Autofagia/genética , Autofagia/inmunología , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina/genética , Línea Celular , Proteínas F-Box/genética , Proteínas F-Box/inmunología , Proteína 7 que Contiene Repeticiones F-Box-WD , Regulación de la Expresión Génica/inmunología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/inmunología , Glucógeno Sintasa Quinasa 3 beta , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Serina-Treonina Quinasas TOR/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
19.
Mol Med ; 20: 639-48, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25375408

RESUMEN

Toll-like receptor 4 (TLR4) is ubiquitously expressed on parenchymal and immune cells of the liver and is the most studied TLR responsible for the activation of proinflammatory signaling cascades in liver ischemia and reperfusion (I/R). Since pharmacological inhibition of TLR4 during the sterile inflammatory response of I/R has not been studied, we sought to determine whether eritoran, a TLR4 antagonist trialed in sepsis, could block hepatic TLR4-mediated inflammation and end organ damage. When C57BL/6 mice were pretreated with eritoran and subjected to warm liver I/R, there was significantly less hepatocellular injury compared to control counterparts. Additionally, we found that eritoran is protective in liver I/R through inhibition of high-mobility group box protein B1 (HMGB1)-mediated inflammatory signaling. When eritoran was administered in conjunction with recombinant HMGB1 during liver I/R, there was significantly less injury, suggesting that eritoran blocks the HMGB1-TLR4 interaction. Not only does eritoran attenuate TLR4-dependent HMGB1 release in vivo, but this TLR4 antagonist also dampened HMGB1's release from hypoxic hepatocytes in vitro and thereby weakened HMGB1's activation of innate immune cells. HMGB1 signaling through TLR4 makes an important contribution to the inflammatory response seen after liver I/R. This study demonstrates that novel blockade of HMGB1 by the TLR4 antagonist eritoran leads to the amelioration of liver injury.


Asunto(s)
Disacáridos/farmacología , Proteína HMGB1/antagonistas & inhibidores , Hepatopatías/metabolismo , Daño por Reperfusión/metabolismo , Fosfatos de Azúcar/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Línea Celular , Células Cultivadas , Proteína HMGB1/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hepatopatías/tratamiento farmacológico , Hepatopatías/patología , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Hepatology ; 59(5): 1984-1997, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24375466

RESUMEN

UNLABELLED: High-mobility group box 1 (HMGB1) is an abundant chromatin-associated nuclear protein and released into the extracellular milieu during liver ischemia-reperfusion (I/R), signaling activation of proinflammatory cascades. Because the intracellular function of HMGB1 during sterile inflammation of I/R is currently unknown, we sought to determine the role of intracellular HMGB1 in hepatocytes after liver I/R. When hepatocyte-specific HMGB1 knockout (HMGB1-HC-KO) and control mice were subjected to a nonlethal warm liver I/R, it was found that HMGB1-HC-KO mice had significantly greater hepatocellular injury after I/R, compared to control mice. Additionally, there was significantly greater DNA damage and decreased chromatin accessibility to repair with lack of HMGB1. Furthermore, lack of hepatocyte HMGB1 led to excessive poly(ADP-ribose)polymerase 1 activation, exhausting nicotinamide adenine dinucleotide and adenosine triphosphate stores, exacerbating mitochondrial instability and damage, and, consequently, leading to increased cell death. We found that this was also associated with significantly more oxidative stress (OS) in HMGB1-HC-KO mice, compared to control. Increased nuclear instability led to a resultant increase in the release of histones with subsequently more inflammatory cytokine production and organ damage through activation of Toll-like receptor 9. CONCLUSION: The lack of HMGB1 within hepatocytes leads to increased susceptibility to cellular death after OS conditions.


Asunto(s)
Citoprotección , Proteína HMGB1/fisiología , Hepatocitos/metabolismo , Hígado/irrigación sanguínea , Daño por Reperfusión/etiología , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Daño del ADN , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NAD/metabolismo , Estrés Oxidativo , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Receptor Toll-Like 9/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA