Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Theranostics ; 10(10): 4453-4465, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32292507

RESUMEN

Organ ischemia reperfusion injury (IRI), associated with acute hepatocyte death, remains an unresolved problem in clinical orthotopic liver transplantation (OLT). Autophagy, an intracellular self-digesting progress, is responsible for cell reprograming required to regain post-stress homeostasis. Methods: Here, we analyzed the cytoprotective mechanism of pituitary adenylate cyclase-activating polypeptide (PACAP)-promoted hepatocellular autophagy in a clinically relevant mouse model of extended hepatic cold storage (4 °C UW solution for 20 h) followed by syngeneic OLT. Results: In contrast to 41.7% of liver graft failure by day 7 post-transplant in control group, PACAP treatment significantly improved graft survival (91.7% by day 14), and promoted autophagy-associated regeneration programs in OLT. In parallel in vitro studies, PACAP-enhanced autophagy ameliorated cellular damage (LDH/ALT levels), and diminished necrosis in H2O2-stressed primary hepatocytes. Interestingly, PACAP not only induced nuclear cAMP response element-binding protein (CREB), but also triggered reprogramming factor Kruppel-like factor 4 (KLF4) expression in IR-stressed OLT. Indeed, CREB inhibition attenuated hepatic autophagy and recreated hepatocellular injury in otherwise PACAP-protected livers. Furthermore, CREB inhibition suppressed PACAP-induced KLF4 expression, whereas KLF4 blockade abolished PACAP-promoted autophagy and neutralized PACAP-mediated hepatoprotection both in vivo and in vitro. Conclusion: Current study documents the essential neural regulation of PACAP-promoted autophagy in hepatocellular homeostasis in OLT, which provides the emerging therapeutic principle to combat hepatic IRI in OLT.


Asunto(s)
Autofagia/efectos de los fármacos , Hepatocitos/citología , Hígado/patología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/uso terapéutico , Disfunción Primaria del Injerto/tratamiento farmacológico , Daño por Reperfusión , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Hepatocitos/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Hígado/citología , Trasplante de Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Manejo de Especímenes
2.
Am J Transplant ; 15(4): 954-964, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25676534

RESUMEN

Ischemia-reperfusion injury (IRI), an innate immunity-driven local inflammation, remains the major problem in clinical organ transplantation. T cell immunoglobulin and mucin domain (TIM-3)-Galectin-9 (Gal-9) signaling regulates CD4+ Th1 immune responses. Here, we explored TIM-3-Gal-9 function in a clinically relevant murine model of hepatic cold storage and orthotopic liver transplantation (OLT). C57BL/6 livers, preserved for 20 h at 4°C in UW solution, were transplanted to syngeneic mouse recipients. Up-regulation of TIM-3 on OLT-infiltrating activated CD4+ T cells was observed in the early IRI phase (1 h). By 6 h of reperfusion, OLTs in recipients treated with a blocking anti-TIM-3 Ab were characterized by: (1) enhanced hepatocellular damage (sALT levels, liver Suzuki's histological score); (2) polarized cell infiltrate towards Th1/Th17-type phenotype; (3) depressed T cell exhaustion markers (PD-1, LAG3); and (4) elevated neutrophil and macrophage infiltration/activation. In parallel studies, adoptive transfer of CD4+ T cells from naïve WT, but not from TIM-3 Tg donors, readily recreated OLT damage in otherwise IR-resistant RAG(-/-) test recipients. Furthermore, pre-treatment of mice with rGal-9 promoted hepatoprotection against preservation-association liver damage, accompanied by enhanced TIM-3 expression in OLTs. Thus, CD4+ T cell-dependent "negative" TIM-3 costimulation is essential for hepatic homeostasis and resistance against IR stress in OLTs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Frío , Trasplante de Hígado , Receptores Virales/inmunología , Transducción de Señal , Animales , Receptor 2 Celular del Virus de la Hepatitis A , Ratones , Ratones Endogámicos C57BL
3.
J Hepatol ; 62(3): 563-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25450716

RESUMEN

BACKGROUND & AIMS: By binding to T cell immunoglobulin mucin-3 (TIM-3) on activated Th1 cells, galectin-9 (Gal-9) negatively regulates Th1-type alloimmunity. Although T cells contribute to hepatic ischemia-reperfusion injury (IRI), it is unknown whether negative T cell-dependent TIM-3 co-stimulation may rescue IR-stressed orthotopic liver transplants from innate immunity-driven inflammation. METHODS: We used wild type (WT) and TIM-3 transgenic (Tg) mice (C57BL/6) as liver donors and recipients in a clinically-relevant model of hepatic cold storage (20 h at 4°C in UW solution) and syngeneic orthotopic liver transplantation (OLT). RESULTS: Orthotopic liver transplants in WT or TIM-3Tg→TIM-3Tg groups were resistant against IR-stress, evidenced by preserved hepatocellular function (serum ALT levels) and liver architecture (Suzuki's score). In contrast, orthotopic liver transplants in WT or TIM-3Tg→WT groups were susceptible to IRI. TIM-3 induction in circulating CD4+ T cells of the recipient: (1) depressed T-bet/IFN-γ, while amplifying GATA3 and IL-4/IL-10 expression in orthotopic liver transplants; (2) promoted T cell exhaustion (PD-1, LAG-3) phenotype; and (3) depressed neutrophil and macrophage infiltration/function in orthotopic liver transplants. In parallel studies, we documented for the first time that Gal-9, a natural TIM-3 ligand, was produced primarily by and released from IR-stressed hepatocytes, both in vivo and in vitro. Moreover, exogenous recombinant Gal-9 (rGal-9) potentiated liver resistance against IRI by depressing T cell activation and promoting apoptosis of CD4+ T cells. CONCLUSIONS: Harnessing TIM-3/Gal-9 signalling at the T cell-hepatocyte interface facilitates homeostasis in IR-stressed orthotopic liver transplants. Enhancing anti-oxidant hepatocyte Gal-9 potentiates liver IR-resistance. Negative regulation by recipient TIM-3+CD4+ cells provides evidence for cytoprotective functions of a discrete T cell subset, which should be spared when applying T cell-targeted immunosuppression in transplant recipients.


Asunto(s)
Galectinas/inmunología , Hepatocitos/inmunología , Trasplante de Hígado , Receptores Virales/inmunología , Daño por Reperfusión/prevención & control , Linfocitos T/inmunología , Adenosina , Alopurinol , Animales , Apoptosis , Diferenciación Celular , Modelos Animales de Enfermedad , Glutatión , Receptor 2 Celular del Virus de la Hepatitis A , Hepatocitos/patología , Inmunidad Innata , Técnicas In Vitro , Insulina , Trasplante de Hígado/efectos adversos , Activación de Linfocitos , Macrófagos/inmunología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neutrófilos/inmunología , Preservación de Órganos , Soluciones Preservantes de Órganos , Rafinosa , Receptores Virales/genética , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Transducción de Señal/inmunología , Linfocitos T/patología
4.
Transplantation ; 99(1): 48-55, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25340604

RESUMEN

BACKGROUND: Although rapamycin (RPM) have been studied extensively in ischemia models, its functional mechanisms remains to be defined. METHODS: We determined how RPM impacted the pathogenesis of ischemia-reperfusion injury (IRI) in a murine liver partial warm ischemia model, with emphasis on its regulation of hepatocyte death. RESULTS: Rapamycin protected livers from IRI in the presence of fully developed liver inflammatory immune response. Rapamycin enhanced liver autophagy induction at the reperfusion stage. Dual mammalian (mechanistic) target of rapamycin (mTOR)1/2 inhibitor Torin 1, despite its ability to induced autophagy, failed to protect livers from IRI. The treatment with RPM, but not Torin 1, resulted in the enhanced activation of the mTORC2-Akt signaling pathway activation in livers after reperfusion. Inactivation of Akt by Triciribine abolished the liver protective effect of RPM. The differential cytoprotective effect of RPM and Torin 1 was confirmed in vitro in hepatocyte cultures. Rapamycin, but not Trin 1, protected hepatocytes from stress and tumor necrosis factor-α induced cell death; and inhibition of autophagy by chloroquine or Akt by Triciribine abolished RPM-mediated cytoprotection. CONCLUSION: Rapamycin protected livers from IRI by both autophagy and mTORC2-Akt activation mechanisms.


Asunto(s)
Hepatocitos/efectos de los fármacos , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Sustancias Protectoras/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/prevención & control , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/efectos de los fármacos , Línea Celular , Citoprotección , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Activación Enzimática , Hepatocitos/enzimología , Hepatocitos/patología , Hígado/enzimología , Hígado/patología , Hepatopatías/enzimología , Hepatopatías/patología , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones Endogámicos C57BL , Naftiridinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Isquemia Tibia
5.
Transplantation ; 98(7): 721-8, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25171655

RESUMEN

BACKGROUND: Nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of the antioxidant host defense, maintains the cellular redox homeostasis. METHODS: This study was designed to investigate the role and molecular mechanisms by which Nrf2 regulates toll-like receptor (TLR)4-driven inflammation response in a mouse model of hepatic warm ischemia (90 min) and reperfusion (6 hr) injury (IRI). RESULTS: Activation of Nrf2 after preconditioning of wild-type mouse recipients with cobalt protoporphyrin ameliorated liver IRI, evidenced by improved hepatocellular function (serum alanine aminotransferase levels), and preserved tissue architecture (histology Suzuki's score). In marked contrast, ablation of Nrf2 signaling exacerbated IR-induced liver inflammation and damage in Nrf2 knockout hosts irrespective of adjunctive cobalt protoporphyrin treatment. The Nrf2 activation reduced macrophage and neutrophil trafficking, proinflammatory cytokine programs, and hepatocellular necrosis or apoptosis while increasing antiapoptotic functions in IR-stressed livers. At the molecular level, Nrf2 activation augmented heme oxygenase-1 expression and Stat3 phosphorylation and promoted PI3K-Akt while suppressing forkhead box O (Foxo)1 signaling. In contrast, Nrf2 deficiency diminished PI3K-Akt and enhanced Foxo1 expression in the ischemic livers. In parallel in vitro studies, Nrf2 knockdown in lipopolysaccharide-stimulated bone marrow-stimulated bone marrow-derived macrophages (BMMs) decreased heme oxygenase-1 and PI3K-Akt yet increased Foxo1 transcription, leading to enhanced expression of TLR4 proinflammatory mediators. Moreover, pretreatment of bone marrow-derived macrophages with PI3K inhibitor (LY294002) activated Foxo1 signaling, which in turn enhanced TLR4-driven innate responses in vitro. CONCLUSION: Activation of Nrf2 promoted PI3K-Akt, and inhibited Foxo1 activity in IR-triggered local inflammation response. By identifying a novel integrated Nrf2-Akt-Foxo1 signaling network in PI3K-dependent regulation of TLR4-driven innate immune activation, this study provides the rationale for refined therapeutic approaches to manage liver inflammation and IRI in transplant recipients.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis , Cromonas/química , Proteína Forkhead Box O1 , Homeostasis , Inflamación , Lipopolisacáridos/química , Hígado/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Morfolinas/química , Necrosis , Neutrófilos/metabolismo , Oxidación-Reducción , Fosforilación , Protoporfirinas/química , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Isquemia Tibia
6.
Hepatology ; 60(6): 2052-2064, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25066922

RESUMEN

UNLABELLED: Hepatic ischemia-reperfusion injury (IRI), an innate immunity-driven inflammation response, occurs in multiple clinical settings including liver resection, transplantation, trauma, and shock. T-cell immunoglobulin and mucin (TIM)-4, the only TIM protein not expressed on T cells, is found on macrophages and dendritic cells. The regulatory function of macrophage TIM-4 in the engulfment of apoptotic/necrotic bodies in innate immunity-mediated disease states remains unknown. This study focuses on the putative role of TIM-4 signaling in a model of liver warm ischemia (90 minutes) and reperfusion. The ischemia insult triggered TIM-4 expression by stressed hepatocellular phosphatidylserine (PS) presentation, peaking at 6 hours of reperfusion, and coinciding with the maximal hepatocellular damage. TIM-4-deficient or wild-type WT mice treated with antagonistic TIM-4 monoclonal antibody (mAb) were resistant against liver IRI, evidenced by diminished serum alanine aminotransferase (sALT) levels and well-preserved hepatic architecture. Liver hepatoprotection rendered by TIM-4 deficiency was accompanied by diminished macrophage infiltration/chemoattraction, phagocytosis, and activation of Toll-like receptor (TLR)2/4/9-dependent signaling. Correlating with in vivo kinetics, the peak of TIM-4 induction in lipopolysaccharide (LPS)-activated bone marrow derived-macrophages (BMM) was detected in 6-hour cultures. To mimic liver IRI, we employed hydrogen peroxide-necrotic hepatocytes, which readily present PS. Indeed, necrotic hepatocytes were efficiently captured/engulfed by WT (TIM-4+) but not by TIM-4-deficient BMM. Finally, in a newly established model of liver IRI, adoptive transfer of WT but not TIM-4-deficient BMM readily recreated local inflammation response/hepatocellular damage in the CD11b-DTR mouse system. CONCLUSION: These findings document the importance of macrophage-specific TIM-4 activation in the mechanism of hepatic IRI. Macrophage TIM-4 may represent a therapeutic target to minimize innate inflammatory responses in IR-stressed organs.


Asunto(s)
Hepatopatías/inmunología , Macrófagos/fisiología , Proteínas de la Membrana/metabolismo , Fosfatidilserinas/metabolismo , Daño por Reperfusión/inmunología , Animales , Hepatocitos , Inmunidad Innata , Activación de Macrófagos , Masculino , Ratones Endogámicos C57BL , Fagocitosis
7.
Mol Med ; 20: 448-55, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25014792

RESUMEN

Macrophages are instrumental in the pathophysiology of liver ischemia/reperfusion injury (IRI). Although Nrf2 regulates macrophage-specific heme oxygenase-1 (HO-1) antioxidant defense, it remains unknown whether HO-1 induction might rescue macrophage Nrf2-dependent antiinflammatory functions. This study explores the mechanisms by which the Nrf2-HO-1 axis regulates sterile hepatic inflammation responses after adoptive transfer of ex vivo modified HO-1 overexpressing bone marrow-derived macrophages (BMMs). Livers in Nrf2-deficient mice preconditioned with Ad-HO-1 BMMs, but not Ad-ß-Gal-BMMs, ameliorated liver IRI (at 6 h of reperfusion after 90 min of warm ischemia), evidenced by improved hepatocellular function (serum alanine aminotransferase [sALT] levels) and preserved hepatic architecture (Suzuki histological score). Treatment with Ad-HO-1 BMMs decreased neutrophil accumulation, proinflammatory mediators and hepatocellular necrosis/apoptosis in ischemic livers. Moreover, Ad-HO-1 transfection of Nrf2-deficient BMMs suppressed M1 (Nos2(+)) while promoting the M2 (Mrc-1/Arg-1(+)) phenotype. Unlike in controls, Ad-HO-1 BMMs increased the expression of Notch1, Hes1, phosphorylation of Stat3 and Akt in IR-stressed Nrf2-deficient livers as well as in lipopolysaccharide (LPS)-stimulated BMMs. Thus, adoptive transfer of ex vivo generated Ad-HO-1 BMMs rescued Nrf2-dependent antiinflammatory phenotype by promoting Notch1/Hes1/Stat3 signaling and reprogramming macrophages toward the M2 phenotype. These findings provide the rationale for a novel clinically attractive strategy to manage IR liver inflammation/damage.


Asunto(s)
Traslado Adoptivo , Hemo-Oxigenasa 1/metabolismo , Hepatopatías/terapia , Macrófagos , Proteínas de la Membrana/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Daño por Reperfusión/terapia , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular , Hemo-Oxigenasa 1/genética , Proteínas de Homeodominio/metabolismo , Hepatopatías/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Fenotipo , Receptor Notch1/metabolismo , Daño por Reperfusión/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción HES-1
8.
J Hepatol ; 59(6): 1200-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23867319

RESUMEN

BACKGROUND & AIMS: The Keap1-Nrf2 signaling pathway regulates host cell defense responses against oxidative stress and maintains the cellular redox balance. METHODS: We investigated the function/molecular mechanisms by which Keap1-Nrf2 complex may influence liver ischemia/reperfusion injury (IRI) in a mouse model of hepatic cold storage (20h at 4°C) followed by orthotopic liver transplantation (OLT). RESULTS: The Keap1 hepatocyte-specific knockout (HKO) in the donor liver ameliorated post-transplant IRI, evidenced by improved hepatocellular function and OLT outcomes (Keap1 HKO→Keap1 HKO; 100% survival), as compared with controls (WT→WT; 50% survival; p<0.01). By contrast, donor liver Nrf2 deficiency exacerbated IRI in transplant recipients (Nrf2 KO→Nrf2 KO; 40% survival). Ablation of Keap1 signaling reduced macrophage/neutrophil trafficking, pro-inflammatory cytokine programs, and hepatocellular necrosis/apoptosis, while simultaneously promoting anti-apoptotic functions in OLTs. At the molecular level, Keap1 HKO increased Nrf2 levels, stimulated Akt phosphorylation, and enhanced expression of anti-oxidant Trx1, HIF-1α, and HO-1. Pretreatment of liver donors with PI3K inhibitor (LY294002) disrupted Akt/HIF-1A signaling and recreated hepatocellular damage in otherwise IR-resistant Keap1 HKO transplants. In parallel in vitro studies, hydrogen peroxide-stressed Keap1-deficient hepatocytes were characterized by enhanced expression of Nrf2, Trx1, and Akt phosphorylation, in association with decreased release of lactate dehydrogenase (LDH) in cell culture supernatants. CONCLUSIONS: Keap1-Nrf2 complex prevents oxidative injury in IR-stressed OLTs through Keap1 signaling, which negatively regulates Nrf2 pathway. Activation of Nrf2 induces Trx1 and promotes PI3K/Akt, crucial for HIF-1α activity. HIF-1α-mediated overexpression of HO-1/Cyclin D1 facilitates cytoprotection by limiting hepatic inflammatory responses, and hepatocellular necrosis/apoptosis in a PI3K-dependent manner.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Citoesqueleto/fisiología , Trasplante de Hígado , Hígado/irrigación sanguínea , Factor 2 Relacionado con NF-E2/fisiología , Daño por Reperfusión/etiología , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Proteína 1 Asociada A ECH Tipo Kelch , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal
9.
Liver Transpl ; 19(9): 945-56, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23744729

RESUMEN

Hepatic ischemia/reperfusion injury (IRI), an exogenous, antigen-independent, local inflammation response, occurs in multiple clinical settings, including liver transplantation, hepatic resection, trauma, and shock. The nervous system maintains extensive crosstalk with the immune system through neuropeptide and peptide hormone networks. This study examined the function and therapeutic potential of the vasoactive intestinal peptide (VIP) neuropeptide in a murine model of liver warm ischemia (90 minutes) followed by reperfusion. Liver ischemia/reperfusion (IR) triggered an induction of gene expression of intrinsic VIP; this peaked at 24 hours of reperfusion and coincided with a hepatic self-healing phase. Treatment with the VIP neuropeptide protected livers from IRI; this was evidenced by diminished serum alanine aminotransferase levels and well-preserved tissue architecture and was associated with elevated intracellular cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling. The hepatocellular protection rendered by VIP was accompanied by diminished neutrophil/macrophage infiltration and activation, reduced hepatocyte necrosis/apoptosis, and increased hepatic interleukin-10 (IL-10) expression. Strikingly, PKA inhibition restored liver damage in otherwise IR-resistant VIP-treated mice. In vitro, VIP not only diminished macrophage tumor necrosis factor α/IL-6/IL-12 expression in a PKA-dependent manner but also prevented necrosis/apoptosis in primary mouse hepatocyte cultures. In conclusion, our findings document the importance of VIP neuropeptide-mediated cAMP-PKA signaling in hepatic homeostasis and cytoprotection in vivo. Because the enhancement of neural modulation differentially regulates local inflammation and prevents hepatocyte death, these results provide the rationale for novel approaches to managing liver IRI in transplant patients.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Hígado/patología , Daño por Reperfusión/patología , Péptido Intestinal Vasoactivo/química , Animales , Apoptosis , Caspasa 3/metabolismo , Citometría de Flujo/métodos , Hepatocitos/citología , Hepatocitos/metabolismo , Homeostasis , Sistema Inmunológico , Inflamación , Interleucina-10/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Hígado/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Neutrófilos/citología , Peroxidasa/metabolismo , Factores de Tiempo
10.
Hepatology ; 58(1): 351-62, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23408710

RESUMEN

UNLABELLED: Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors, is essential for inducing caspase-1 activation and the consequent secretion of interleukin-1ß (IL-1ß), which is associated with local inflammation during liver ischemia/reperfusion injury (IRI). However, little is known about the mechanisms by which the ASC/caspase-1/IL-1ß axis exerts its function in hepatic IRI. This study was designed to explore the functional roles and molecular mechanisms of ASC/caspase-1/IL-1ß signaling in the regulation of inflammatory responses in vitro and in vivo. With a partial lobar liver warm ischemia (90 minutes) model, ASC-deficient and wild-type mice (C57BL/6) were sacrificed at 6 hours of reperfusion. Separate animal cohorts were treated with an anti-IL-1ß antibody or control immunoglobulin G (10 mg/kg/day intraperitoneally). We found that ASC deficiency inhibited caspase-1/IL-1ß signaling and led to protection against liver ischemia/reperfusion (IR) damage, local enhancement of antiapoptotic functions, and down-regulation of high mobility group box 1 (HMGB1)-mediated, toll-like receptor 4 (TLR4)-driven inflammation. Interestingly, the treatment of ASC-deficient mice with recombinant HMGB1 re-created liver IRI. Moreover, neutralization of IL-1ß ameliorated the hepatocellular damage by inhibiting nuclear factor kappa B (NF-κB)/cyclooxygenase 2 signaling in IR-stressed livers. In parallel in vitro studies, the knockout of ASC in lipopolysaccharide-stimulated bone marrow-derived macrophages depressed HMGB1 activity via the p38 mitogen-activated protein kinase pathway and led to the inhibition of TLR4/NF-κB and ultimately the depression of proinflammatory cytokine programs. CONCLUSION: ASC-mediated caspase-1/IL-1ß signaling promotes HMGB1 to produce a TLR4-dependent inflammatory phenotype and leads to hepatocellular injury. Hence, ASC/caspase-1/IL-1ß signaling mediates the inflammatory response by triggering HMGB1 induction in hepatic IRI. Our findings provide a rationale for a novel therapeutic strategy for managing liver injury due to IR.


Asunto(s)
Caspasa 1/fisiología , Proteínas del Citoesqueleto/fisiología , Proteína HMGB1/biosíntesis , Interleucina-1beta/fisiología , Daño por Reperfusión/inmunología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas del Citoesqueleto/deficiencia , Hígado/lesiones , Masculino , Ratones , Ratones Noqueados , Daño por Reperfusión/prevención & control , Transducción de Señal/fisiología , Receptor Toll-Like 4/fisiología
11.
Hepatology ; 57(3): 1225-37, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22532103

RESUMEN

UNLABELLED: Hepatic ischemia and reperfusion injury (IRI), an exogenous antigen-independent local inflammation response, occurs in multiple clinical settings, including liver transplantation, hepatic resection, trauma, and shock. The immune system and the nervous system maintain extensive communication and mount a variety of integrated responses to danger signals through intricate chemical messengers. This study examined the function and potential therapeutic potential of neuropeptide pituitary adenylate cyclase-activating polypeptides (PACAP) in a murine model of partial liver "warm" ischemia (90 minutes) followed by reperfusion. Liver IRI readily triggered the expression of intrinsic PACAP and its receptors, whereas the hepatocellular damage was exacerbated in PACAP-deficient mice. Conversely, PACAP27, or PACAP38 peptide monotherapy, which elevates intracellular cyclic adenosine monophosphate/protein kinase A (cAMP-PKA) signaling, protected livers from IRI, as evidenced by diminished serum alanine aminotransferase levels and well-preserved tissue architecture. The liver protection rendered by PACAP peptides was accompanied by diminished neutrophil/macrophage infiltration and activation, reduced hepatocyte necrosis/apoptosis, and selectively augmented hepatic interleukin (IL)-10 expression. Strikingly, PKA inhibition readily restored liver damage in otherwise IR-resistant, PACAP-conditioned mice. In vitro, PACAP treatment not only diminished macrophage tumor necrosis factor alpha/IL-6/IL-12 levels in a PKA-dependent manner, but also prevented necrosis and apoptosis in primary mouse hepatocyte cultures. CONCLUSION: Our novel findings document the importance of PACAP-mediated cAMP-PKA signaling in hepatic homeostasis and cytoprotection in vivo. Because the enhancement of neural modulation differentially regulates local inflammation and prevents hepatocyte death, these results provide the rationale for novel approaches to manage liver inflammation and IRI in transplant patients.


Asunto(s)
Factores Inmunológicos/metabolismo , Hepatopatías/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal/inmunología , Animales , Apoptosis/inmunología , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Homeostasis/inmunología , Factores Inmunológicos/inmunología , Hepatopatías/inmunología , Hepatopatías/patología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis/inmunología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
12.
Hepatology ; 57(3): 1203-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23081841

RESUMEN

UNLABELLED: Dendritic cells (DCs) are critical mediators of immune responses that integrate signals from the innate immune system to orchestrate adaptive host immunity. This study was designed to investigate the role and molecular mechanisms of STAT3-induced ß-catenin in the regulation of DC function and inflammatory responses in vitro and in vivo. STAT3 induction in lipopolysaccharide (LPS)-stimulated mouse bone marrow-derived DCs (BMDCs) triggered ß-catenin activation by way of GSK-3ß phosphorylation. The activation of ß-catenin inhibited phosphatase and tensin homolog delete on chromosome 10 (PTEN) and promoted the phosphoinositide 3-kinase (PI3K)/Akt pathway, which in turn down-regulated DC maturation and function. In contrast, knockdown of ß-catenin increased PTEN/TLR4 (Toll-like receptor 4), interferon regulatory factor-3 (IRF3), nuclear factor kappa B (NF-κB) activity, and proinflammatory cytokine programs in response to LPS stimulation. In a mouse model of warm liver ischemia and reperfusion injury (IRI), disruption of ß-catenin signaling increased the hepatocellular damage, enhanced hepatic DC maturation/function, and PTEN/TLR4 local inflammation in vivo. CONCLUSION: These findings underscore the role of ß-catenin to modulate DC maturation and function at the innate-adaptive interface. Activation of ß-catenin triggered PI3K/Akt, which in turn inhibited TLR4-driven inflammatory response in a negative feedback regulatory mechanism. By identifying the molecular pathways by which ß-catenin regulates DC function, our findings provide the rationale for novel therapeutic approaches to manage local inflammation and injury in IR-stressed liver.


Asunto(s)
Inmunidad Adaptativa/fisiología , Células Dendríticas/inmunología , Inmunidad Innata/fisiología , Hepatopatías/inmunología , Daño por Reperfusión/inmunología , beta Catenina/inmunología , Animales , Apoptosis/inmunología , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfohidrolasa PTEN/inmunología , Fosfohidrolasa PTEN/metabolismo , ARN Interferente Pequeño/genética , Daño por Reperfusión/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
13.
Transplantation ; 94(3): 211-7, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22790388

RESUMEN

BACKGROUND: Although endoplasmic reticulum (ER) stress has been implicated in the pathophysiology of organ ischemia-reperfusion injury (IRI), the underlying mechanisms have yet to be fully elucidated. In particular, because tissue proinflammatory immune response is the key mediator of local IRI, how ER stress impacts liver immune cell activation cascade remains to be determined. METHODS: In vitro, ER stress in macrophages and hepatocytes were induced by pharmacological agents. Macrophage Toll-like receptor 4 and hepatocyte tumor necrosis factor α responses were studied. In vivo, the induction of ER stress by ischemia-reperfusion and the impact of ER stress amelioration by a small molecule chaperon 4-phenylbutyric acid on liver immune response were studied in a murine partial liver warm ischemia model. RESULTS: ER-stressed macrophages generated a significantly enhanced proinflammatory immune response against Toll-like receptor 4 stimulation, whereas ER-stressed hepatocytes became more susceptible to tumor necrosis factor α-induced cell death. Ischemia-reperfusion resulted in up-regulations of spliced X-box binding protein 1 and activating transcription factor 6 levels in affected livers. Mice pretreated with 4-phenylbutyric acid were protected from liver IRI, in parallel with diminished local proinflammatory gene induction program. CONCLUSIONS: Our study documents a potential immune regulatory role of ER stress in the mechanism of liver IRI and provides a rationale for targeting stress response as a new therapeutic means to ameliorate tissue inflammation in organ transplant recipients.


Asunto(s)
Retículo Endoplásmico/metabolismo , Hígado/patología , Daño por Reperfusión , Animales , Regulación de la Expresión Génica , Hepatocitos/citología , Sistema Inmunológico , Inflamación , Isquemia/patología , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Fenilbutiratos/farmacología , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
14.
Surgery ; 152(5): 843-50, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22575884

RESUMEN

BACKGROUND: Antithymocyte therapy, specifically antithymocyte globulin (ATG; Thymoglobulin), is increasingly being used in organ transplantation to reduce allograft rejection. The T-lymphocyte has been purported to also play a role in ischemia and reperfusion injury (IRI); however, it has not been well studied. Our aim is to determine if ATG treatment impacts murine intestinal IRI. METHODS: Under anesthesia, male C57BL6 mice underwent 100 minutes of warm intestinal IRI by clamping the superior mesenteric artery. The treatment group received rabbit anti-murine ATG (10 mg/kg) intraperitoneally 6 hours before IRI. Separate survival and analysis groups were performed. Intestinal tissue was procured at 4 and 24 hours after IRI. Tissue analysis included hematoxylin-eosin staining, CD3, CD4, and CD8 immunostaining, myeloperoxidase assay (MPO), quantitative real-time polymerase chain reaction studies, and Western blot. RESULTS: ATG treatment led to marked improvement in 7-day survival and a reduction in tissue injury by histology. MPO was also reduced, and immunostaining confirmed a significant reduction in CD3(+), CD4(+), and CD8(+) infiltrating cells in the treatment group. Quantitative real-time polymerase chain reaction analysis revealed the decreased expression of tumor necrosis factor-α, interferon-inducible protein 10, monocyte chemotactic protein-1, interferon-γ, interleukin-2, and increased production of interleukins -13 and -10 in the treatment group. Western blot analysis revealed decreased caspase-3 and increased signal transducer and activator of transcription 6 levels in the ATG-treated group. CONCLUSION: This study is the first to show that ATG treatment ameliorates intestinal IRI. Treatment with ATG leads to reduced local infiltration by T-lymphocytes, with fewer inflammatory and chemotactic programs and less apoptosis. Treatment also is associated with a T(H)2-type cytokine switch. These novel findings suggest that T-lymphocytes represent important mediators of intestinal IRI and that ATG therapies may be beneficial in the prevention of IRI.


Asunto(s)
Suero Antilinfocítico/uso terapéutico , Enfermedades Intestinales/prevención & control , Intestinos/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Antígenos CD/metabolismo , Western Blotting , Enfermedades Intestinales/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/trasplante , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología
15.
Liver Transpl ; 18(6): 659-70, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22290937

RESUMEN

Hepatic ischemia/reperfusion injury (IRI) occurs in multiple clinical settings, including liver transplantation. The cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) pathway inhibits hepatocellular apoptosis and regulates toll-like receptor 4-triggered inflammation responses in vitro. Here we examined the function and therapeutic potential of cAMP-PKA activation in a murine (C57/BL6) model of liver warm ischemia (90 minutes) followed by reperfusion. Liver IRI triggered cAMP-PKA activation, whereas the administration of its specific inhibitor, H89, exacerbated hepatocellular damage. Conversely, forskolin therapy, which activates PKA by elevating cAMP levels, protected livers from IRI; this was evidenced by diminished serum alanine aminotransferase levels and well-preserved tissue architecture. Liver protection due to cAMP-PKA stimulation was accompanied by diminished neutrophil and macrophage infiltration/activation, reduced hepatocyte necrosis/apoptosis, and increased cAMP response element-binding protein (CREB) expression and augmented interleukin-10 (IL-10) expression. The neutralization of IL-10 restored liver damage in otherwise ischemia/reperfusion-resistant, forskolin-treated mice. In vitro, cAMP-PKA activation diminished macrophage tumor necrosis factor α, IL-6, and IL-12 in an IL-10-dependent manner and prevented necrosis/apoptosis in primary mouse hepatocyte cultures. Our novel findings in a mouse model of liver IRI document the importance of cAMP-PKA signaling in hepatic homeostasis and cytoprotection in vivo. The activation of cAMP-PKA signaling differentially regulates local inflammation and prevents hepatocyte death, and this provides a rationale for novel therapeutic approaches to combating liver IRI in transplant recipients.


Asunto(s)
Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Trasplante de Hígado , Daño por Reperfusión , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/fisiología , Células Cultivadas , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interleucina-10/metabolismo , Isoquinolinas/farmacología , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo , Necrosis/patología , Necrosis/prevención & control , Peroxidasa/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal/fisiología , Sulfonamidas/farmacología , Temperatura
16.
J Hepatol ; 56(2): 359-66, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21756853

RESUMEN

BACKGROUND & AIMS: Signal transducer and activator of transcription 3 (STAT3), a key mediator of anti-inflammatory cytokine signaling, is essential for heme oxygenase-1 (HO-1)-induced cytoprotection. The phosphoinositide 3-kinase (PI3K)/phosphatase and tensin homolog delete on chromosome 10 (PTEN) pathways regulate diverse innate immune responses. This study was designed to investigate the role of STAT3 in the regulation of PI3K/PTEN cascade after HO-1 induction in a mouse model of innate immune-dominated liver ischemia/reperfusion injury (IRI). METHODS: Partial warm ischemia was produced in the left and middle hepatic lobes of C57BL/6 mice for 90 min, followed by 6h of reperfusion. RESULTS: Mice subjected to Ad-HO-1 transfer were resistant to liver IRI, and this cytoprotective effect correlated with increased intrahepatic PI3K/Akt and diminished PTEN expression. In contrast, mice undergoing adjunctive Ad-HO-1 treatment and STAT3 knockdown (siRNA) remained susceptible to IR-mediated local inflammatory response and hepatocellular damage. Consistent with decreased cell apoptosis and inhibited TLR4 expression after PI3K/Akt activation, treatment with specific PI3k inhibitor increased local inflammation and recreated liver IRI despite Ad-HO-1 gene transfer. Parallel in vitro studies with bone marrow derived-macrophages have confirmed that HO-1-STAT3 axis-induced PI3K/Akt negatively regulated PTEN expression in TLR4-dependent fashion. CONCLUSIONS: These findings underscore the role of HO-1 induced STAT3 in modulating PI3K/PTEN in liver IRI cascade. Activating PI3K/Akt provides negative feedback mechanism for TLR4-driven inflammation. Identifying molecular pathways of STAT3 modulation in the innate immune system provides the rationale for novel therapeutic approaches for the management of liver inflammation and IRI in transplant patients.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Inmunidad Innata , Hígado/lesiones , Proteínas de la Membrana/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Factor de Transcripción STAT3/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/genética , Humanos , Hígado/inmunología , Hígado/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Modelos Inmunológicos , FN-kappa B/metabolismo , Fosfohidrolasa PTEN/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Daño por Reperfusión/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Transducción de Señal
17.
Hepatology ; 54(2): 687-96, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21567437

RESUMEN

UNLABELLED: The ubiquitous serine/threonine kinase glycogen synthase kinase 3 beta (Gsk3ß) differentially regulates macrophage Toll-like receptor (TLR)-triggered pro- and anti-inflammatory cytokine programs. This study was designed to determine the in vivo role and therapeutic potential of Gsk3ß modulation in tissue inflammation and injury in a murine model of liver partial warm ischemia/reperfusion injury (IRI). As a constitutively activated liver kinase, Gsk3ß became quickly inactivated (phosphorylated) following IR. The active Gsk3ß, however, was essential for the development of IRI pathology, as administration of its specific inhibitor, SB216763, ameliorated the hepatocellular damage, evidenced by reduced serum alanine aminotransferase (sALT) levels and well-preserved liver architecture compared with controls. The liver protective effect of Gsk3ß inhibition was dependent on an immune regulatory mechanism, rather than direct cytoprotection via mitochondria permeability transition pores (MPTP). Indeed: (1) coadministration of SB216763 and atractyloside (MPTP opener) failed to abrogate a local cytoprotective Gsk3ß inhibition effect; (2) SB216763 selectively inhibited IR-triggered liver pro-inflammatory, but spared interleukin (IL)-10, gene induction programs; and (3) IL-10 neutralization restored liver inflammation and IRI in SB216763-treated mice. Gsk3ß inactivation by IR was a self-regulatory mechanism in liver homeostasis, critically dependent on phosphoinositide 3 (PI3)-kinase activation, as administration of a PI3 kinase inhibitor, wortmannin, reduced Gsk3 phosphorylation and augmented liver damage. In vitro, IL-10 was critical for the suppression of pro-inflammatory gene programs by Gsk3 inhibition in bone marrow-derived macrophages in response to TLR4 stimulation. CONCLUSION: Our novel findings document the key immune regulatory function of Gsk3ß signaling in the pathophysiology of liver IRI, and provide a rationale to target Gsk3ß as a refined therapeutic strategy to ameliorate liver IRI.


Asunto(s)
Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Interleucina-10/inmunología , Hígado/irrigación sanguínea , Daño por Reperfusión/tratamiento farmacológico , Animales , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Daño por Reperfusión/inmunología
18.
Transplantation ; 91(10): 1075-81, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21427633

RESUMEN

BACKGROUND: The sensitized patients can develop an accelerated form of graft rejection mediated by humoral and T-cell-mediated responses, which are resistant to currently used immunosuppression. METHODS AND RESULTS: In our model of fulminant cardiac allograft rejection in sensitized hosts, groups of wild-type (WT) and B-cell-deficient (BKO) mice (B6) were challenged with skin grafts (B/c). Alloreactive CD8 T effector (Teff) activation and T memory (Tmem) differentiation during a 60-day follow-up period were reduced in the absence of B-cell help. The expression of interleukin (IL)-2Rα, IL-7Rα, and IL-15Rα, which support/program CD8 Teff/Tmem expansion, differentiation, and survival, were selectively decreased in BKO hosts. Unlike in WT, in vivo cytotoxic activity analysis of alloreactive Tmem recall response has revealed decreased donor-type (B/c) but not third-party (C3H) cell lysis in sensitized BKO hosts. However, such impaired allo-Ag specific Tmem recall function was insufficient to markedly prolong cardiac allograft survival in sensitized BKO recipients. Indeed, despite quantitative and statistically significant differences between both animal groups, the biological impact of decreased CD8 Teff/Tmem activation and function in the sensitization phase was marginal. Indeed, cardiac allografts underwent fulminant rejection in sensitized BKO, albeit with somewhat delayed kinetics. Interestingly, unlike in naïve counterparts, the rejection cascade remained CD154 blockade-resistant, evidenced by comparable kinetics, and intra-graft cytokine gene profiles in MR1 monoclonal antibody-treated sensitized WT and BKO hosts. CONCLUSION: Although B cells were important for optimal alloreactive CD8 Teff/Tmem function in the sensitization phase, the fulminant rejection of cardiac allografts was B-cell-independent, and CD154 blockade-resistant, as in WT hosts.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Supervivencia de Injerto , Trasplante de Corazón/inmunología , Memoria Inmunológica , Trasplante de Piel/inmunología , Animales , Ligando de CD40/metabolismo , Diferenciación Celular , Citocinas/genética , Citocinas/metabolismo , Citotoxicidad Inmunológica , Inmunoglobulina M/deficiencia , Inmunoglobulina M/genética , Cadenas mu de Inmunoglobulina/genética , Cadenas mu de Inmunoglobulina/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina/metabolismo , Factores de Tiempo , Trasplante Homólogo
19.
Transplantation ; 91(7): 737-43, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21311412

RESUMEN

BACKGROUND: Ischemia-reperfusion (I/R) injury is a major factor leading to intestinal dysfunction or graft loss after intestinal surgery or transplantation. This study investigated the cytoprotective effects and putative mechanisms of interleukin (IL)-13 after intestinal I/R injury in the mouse. METHODS: Mouse warm intestinal I/R injury induced by clamping the superior mesenteric artery for 100 min with tissue analysis at 4 and 24 hr after reperfusion. Treated animals received intravenous recombinant murine IL-13 (rIL-13) and anti-IL-13 antibody, whereas controls received saline. RESULTS: rIL-13 administration markedly prolonged animal survival (100% vs. 50% in saline controls) and resulted in near normal histopathological architecture. rIL-13 treatment also significantly decreased myeloperoxidase activity. Mice conditioned with rIL-13 had a markedly depressed Toll-like receptor-4 expression and increased the expression of Stat6, antioxidant hemeoxygenase-1, and antiapoptotic A20, Bcl-2/Bcl-xl, compared with that of controls. Unlike in controls, the expression of mRNA coding for IL-2/interferon-γ, and interferon-γ-inducible protein (IP)-10/monocyte chemotactic protein-1 remained depressed, whereas that of IL-13/IL-4 reciprocally increased in the mice treated with rIL-13. Administration of anti-IL13 antibody alone or in combination with rIL-13 resulted in outcomes similar to that seen in controls. CONCLUSIONS: This study demonstrates for the first time that IL-13 plays a protective role in intestinal warm I/R injury and a critical role in the regulation of Stat6 and Toll-like receptor-4 signaling. The administration of IL-13 exerts cytoprotective effects in this model by regulating innate and adaptive immunity while the removal of IL-13 using antibody therapy abrogates this effect.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Interleucina-13/farmacología , Intestinos/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Citoprotección , Interleucina-13/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT6/fisiología , Receptor Toll-Like 4/fisiología
20.
Liver Transpl ; 17(2): 201-10, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21280193

RESUMEN

We investigated whether native macrophages overexpressing heme oxygenase 1 (HO-1) could protect rat orthotopic liver transplant (OLT) against cold ischemia/reperfusion injury (IRI). Livers from Sprague-Dawley rats were stored at 4°C in University of Wisconsin solution for 24 hours, and then they were transplanted into syngeneic recipients. Bone marrow-derived macrophages (BMMs) that were transfected ex vivo with heme oxygenase 1 adenovirus (Ad-HO-1), ß-galactosidase adenovirus (Ad-ß-gal), or HO-1 small interfering RNA (siRNA) were infused directly into the OLT before reperfusion. Controls were OLT conditioned with unmodified or scrambled siRNA-transfected cells. The transfer of Ad-HO-1/BMMs increased the survival of OLT to 100% (versus 40%-50% for controls) and decreased serum alanine aminotransferase levels and histological features of hepatocellular damage. In contrast, an infusion of macrophages transfected with HO-1 siRNA/Ad-ß-gal failed to affect IRI. Gene therapy-induced HO-1 suppressed toll-like receptor 4 expression, decreased expression of proinflammatory tumor necrosis factor α, interleukin-1ß, monocyte chemoattractant protein 1, and chemokine (C-X-C motif) ligand 10, and attenuated endothelial intercellular cell adhesion molecule 1 expression with resultant diminished OLT leukocyte sequestration. Although Ad-HO-1/BMMs decreased the frequency of apoptotic cells positive for terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling and ameliorated caspase-3 activity, the expression of interleukin-10 and antiapoptotic B cell lymphoma 2/B cell lymphoma extra large increased in well-functioning OLT. Thus, the transfer of native macrophages transfected ex vivo with HO-1 can rescue rat iso-OLT from IRI. Our study validates a novel and clinically attractive concept: native macrophages transfected ex vivo with the antioxidant HO-1 can be applied at the time of transplantation to mitigate otherwise damaging antigen-independent liver inflammation and injury resulting from the peritransplant harvesting insult. If this new, refined strategy is proven to be effective in allo-OLT recipients, it should be considered in clinical settings to increase the supply of usable donor organs and ultimately improve the overall success of liver transplantation.


Asunto(s)
Isquemia Fría/efectos adversos , Supervivencia de Injerto , Hemo-Oxigenasa 1/metabolismo , Trasplante de Hígado/efectos adversos , Hígado/cirugía , Macrófagos/trasplante , Daño por Reperfusión/prevención & control , Animales , Apoptosis , Caspasa 3/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/metabolismo , Hemo-Oxigenasa 1/genética , Mediadores de Inflamación/metabolismo , Hígado/enzimología , Hígado/inmunología , Hígado/patología , Macrófagos/enzimología , Masculino , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Factores de Tiempo , Receptor Toll-Like 4/metabolismo , Transfección , Trasplante Isogénico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...