Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 15(11): 1064-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25240383

RESUMEN

It remains largely unclear how antigen-presenting cells (APCs) encounter effector or memory T cells efficiently in the periphery. Here we used a mouse contact hypersensitivity (CHS) model to show that upon epicutaneous antigen challenge, dendritic cells (DCs) formed clusters with effector T cells in dermal perivascular areas to promote in situ proliferation and activation of skin T cells in a manner dependent on antigen and the integrin LFA-1. We found that DCs accumulated in perivascular areas and that DC clustering was abrogated by depletion of macrophages. Treatment with interleukin 1α (IL-1α) induced production of the chemokine CXCL2 by dermal macrophages, and DC clustering was suppressed by blockade of either the receptor for IL-1 (IL-1R) or the receptor for CXCL2 (CXCR2). Our findings suggest that the dermal leukocyte cluster is an essential structure for elicitating acquired cutaneous immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Dermatitis por Contacto/inmunología , Piel/inmunología , Animales , Antígeno CD11c/genética , Proliferación Celular , Quimiocina CXCL2/biosíntesis , Femenino , Memoria Inmunológica/inmunología , Interleucina-1alfa/farmacología , Activación de Linfocitos/inmunología , Antígeno-1 Asociado a Función de Linfocito/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Neutrófilos/inmunología , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Piel/patología
2.
Int J Mol Sci ; 25(5)2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38474078

RESUMEN

Carbon ion beams have the unique property of higher linear energy transfer, which causes clustered damage of DNA, impacting the cell repair system. This sometimes triggers apoptosis and the release in the cytoplasm of damaged DNA, leading to type I interferon (IFN) secretion via the activation of the cyclic GMP-AMP synthase-stimulator of interferon genes pathway. Dendritic cells phagocytize dead cancer cells and damaged DNA derived from injured cancer cells, which together activate dendritic cells to present cancer-derived antigens to antigen-specific T cells in the lymph nodes. Thus, carbon ion radiation therapy (CIRT) activates anti-cancer immunity. However, cancer is protected by the tumor microenvironment (TME), which consists of pro-cancerous immune cells, such as regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. The TME is too robust to be destroyed by the CIRT-mediated anti-cancer immunity. Various modalities targeting regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages have been developed. Preclinical studies have shown that CIRT-mediated anti-cancer immunity exerts its effects in the presence of these modalities. In this review article, we provide an overview of CIRT-mediated anti-cancer immunity, with a particular focus on recently identified means of targeting the TME.


Asunto(s)
Radioterapia de Iones Pesados , Células Supresoras de Origen Mieloide , Neoplasias , Humanos , Neoplasias/patología , Linfocitos T Reguladores , ADN , Microambiente Tumoral
3.
Cell Physiol Biochem ; 57(4): 212-225, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37463410

RESUMEN

BACKGROUND/AIMS: Pancreatic cancer has the poorest survival rate among all cancer types. Therefore, it is essential to develop an effective treatment strategy for this cancer. METHODS: We performed carbon ion radiotherapy (CIRT) in human pancreatic cancer cell lines and analyzed their survival, apoptosis, necrosis, and autophagy. To investigate the role of CIRT-induced autophagy, autophagy inhibitors were added to cells prior to CIRT. To evaluate tumor formation, we inoculated CIRT-treated murine pancreatic cancer cells on the flank of syngeneic mice and measured tumor weight. We immunohistochemically measured autophagy levels in surgical sections from patients with pancreatic cancer who received neoadjuvant chemotherapy (NAC) plus CIRT or NAC alone. RESULTS: CIRT reduced the survival fraction of pancreatic cancer cells and induced apoptotic and necrotic alterations, along with autophagy. Preincubation with an autophagy inhibitor accelerated cell death. Mice inoculated with control pancreatic cancer cells developed tumors, while those inoculated with CIRT/autophagy inhibitor-treated cells showed significant evasion. Surgical specimens of NAC-treated patients expressed autophagy comparable to control patients, while those in the NAC plus CIRT group expressed little autophagy and nuclear staining. CONCLUSION: CIRT effectively killed the pancreatic cancer cells by inhibiting their autophagy-inducing abilities.


Asunto(s)
Radioterapia de Iones Pesados , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Neoplasias Pancreáticas/metabolismo , Autofagia , Resultado del Tratamiento , Neoplasias Pancreáticas
4.
Immunity ; 38(6): 1187-97, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23791646

RESUMEN

The small intestine harbors a substantial number of commensal bacteria and is sporadically invaded by pathogens, but the response to these microorganisms is fundamentally different. We identified a discriminatory sensor by using Toll-like receptor 3 (TLR3). Double-stranded RNA (dsRNA) of one major commensal species, lactic acid bacteria (LAB), triggered interferon-ß (IFN-ß) production, which protected mice from experimental colitis. The LAB-induced IFN-ß response was diminished by dsRNA digestion and treatment with endosomal inhibitors. Pathogenic bacteria contained less dsRNA and induced much less IFN-ß than LAB, and dsRNA was not involved in pathogen-induced IFN-ß induction. These results identify TLR3 as a sensor to small intestinal commensal bacteria and suggest that dsRNA in commensal bacteria contributes to anti-inflammatory and protective immune responses.


Asunto(s)
Colitis/prevención & control , Enterococcaceae/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Interferón beta/metabolismo , Lactobacillus/inmunología , Macrófagos/inmunología , Receptor Toll-Like 3/metabolismo , Animales , Células Cultivadas , Colitis/etiología , Colitis/inmunología , Colitis/microbiología , Modelos Animales de Enfermedad , Enterococcaceae/patogenicidad , Femenino , Infecciones por Bacterias Grampositivas/complicaciones , Infecciones por Bacterias Grampositivas/microbiología , Intestinos/inmunología , Intestinos/microbiología , Macrófagos/microbiología , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , ARN Bicatenario/inmunología
5.
Cell Physiol Biochem ; 55(4): 400-412, 2021 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-34214389

RESUMEN

BACKGROUND/AIMS: Postoperative adhesions may induce adverse outcomes in patients. Adhesion formation is initiated by fibrin accumulation at the surgical site which is followed by local neutrophilia and the establishment of neutrophil extracellular traps (NET). Previous reports have suggested that the preventive efficacy of reagents designed to reduce postoperative adhesion is inversely correlated with neutrophilia and NET production. Antithrombin (AT) is a natural inhibitor of thrombin, a key factor in coagulation. Here, we evaluate whether treatment with AT and/or NET inhibitors prevent or reduce postoperative adhesion formation in mice. METHODS: Mice were treated with AT and/or NET inhibitors before and/or after cecum cauterization and their adhesion scores were evaluated on day 7 post-operation. Immunochemistry/ immunofluorescence analyses were also performed and we used GSK484, an inhibitor of peptidyl arginine deiminase 4 (PAD4), as the NET inhibitor. RESULTS: AT or GSK484 partially rescued postoperative adhesion formation in mice. AT prevented thrombin-induced plasminogen activator inhibitor 1 and interleukin-6 expression in mesothelial cells in vitro. However, AT could not prevent neutrophilia or NETs formation around the injured serosa. Finally, we investigated a combination of AT and a PAD4 inhibitor and found that this could inhibit almost all adhesion formation in these animals. Since AT-inactivating proteases are liberated following NET release, they might dampen the biological action of the AT treatment. This suggests that NET inhibitors might allow AT to exert its full action in the surgically injured serosa. CONCLUSION: Combined treatment with AT and GSK484 may effectively attenuate postoperative adhesion production in mice.


Asunto(s)
Antitrombinas/farmacología , Trampas Extracelulares/metabolismo , Adherencias Tisulares , Animales , Ciego/metabolismo , Ciego/patología , Ciego/cirugía , Femenino , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Arginina Deiminasa Proteína-Tipo 4/antagonistas & inhibidores , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Serpina E2/metabolismo , Adherencias Tisulares/metabolismo , Adherencias Tisulares/patología , Adherencias Tisulares/prevención & control
6.
Cell Physiol Biochem ; 54(5): 1041-1053, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-33053302

RESUMEN

BACKGROUND/AIMS: Although adhesion formation is a frequent adverse event following intraperitoneal surgery, efficient prophylactic interventions have not yet been established. We recently reported that blockade of interleukin (IL)-6 prevented postoperative adhesion after cecum cauterization. Intriguingly, this intervention dampened tumor necrosis factor (TNF) induction in the injured serosa. Herein, we addressed whether TNF might be a key target and, if so, how TNF blockade rescued adhesion formation. METHODS: Mice were administered an anti-TNF biologic (etanercept) on days -2 and -1 before and upon cecal cauterization. The adhesion scores were evaluated at day 7 postoperatively. Histological alterations were examined by immunochemistry/immunofluorescence studies. We incubated human neutrophils and mesothelial cell line cells with recombinant TNF in the presence of etanercept and measured transcript levels of cytokines and chemokines by quantitative reverse transcription-polymerase chain reaction (RT-qPCR). RESULTS: Etanercept rescued mice from adhesion formation, accompanied by a robust reduction of neutrophilia in the injured serosa. Immunofluorescence revealed a substantial formation of neutrophil extracellular traps (NETs) with the potential to induce tissue damage and profibrotic responses. In contrast, the etanercept-treated mice lacked NET formation. In addition, etanercept inhibited TNF-induced IL-6, TNF, and neutrophil-recruiting chemokines in neutrophils and mesothelial cells, a major cellular source of myofibroblasts in the adhesion band. CONCLUSION: Prophylactic administration of etanercept might be a potential strategy for preventing postoperative adhesion formation.


Asunto(s)
Cauterización/efectos adversos , Ciego/cirugía , Etanercept/farmacología , Trampas Extracelulares/efectos de los fármacos , Interleucina-6/metabolismo , Adherencias Tisulares/prevención & control , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Antiinflamatorios no Esteroideos/farmacología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Adherencias Tisulares/etiología , Adherencias Tisulares/patología
7.
J Immunol ; 199(3): 1122-1130, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28674179

RESUMEN

CD4+ Th cells play crucial roles in orchestrating immune responses against pathogenic microbes, after differentiating into effector subsets. Recent research has revealed the importance of IFN-γ and IL-17 double-producing CD4+ Th cells, termed Th17/Th1 cells, in the induction of autoimmune and inflammatory diseases. In addition, Th17/Th1 cells are involved in the regulation of infection caused by the intracellular bacterium Mycobacterium tuberculosis in humans. However, the precise mechanism of Th17/Th1 induction during pathogen infection is unclear. In this study, we showed that the inflammasome and Fas-dependent IL-1ß induces Th17/Th1 cells in mice, in response to infection with the pathogenic intracellular bacterium Listeria monocytogenes In the spleens of infected wild-type mice, Th17/Th1 cells were induced, and expressed T-bet and Rorγt. In Pycard-/- mice, which lack the adaptor molecule of the inflammasome (apoptosis-associated speck-like protein containing a caspase recruitment domain), Th17/Th1 induction was abolished. In addition, the Fas-mediated IL-1ß production was required for Th17/Th1 induction during bacterial infection: Th17/Th1 induction was abolished in Fas-/- mice, whereas supplementation with recombinant IL-1ß restored Th17/Th1 induction via IL-1 receptor 1 (IL-1R1), and rescued the mortality of Fas-/- mice infected with Listeria IL-1R1, but not apoptosis-associated speck-like protein containing a caspase recruitment domain or Fas on T cells, was required for Th17/Th1 induction, indicating that IL-1ß stimulates IL-1R1 on T cells for Th17/Th1 induction. These results indicate that IL-1ß, produced by the inflammasome and Fas-dependent mechanisms, contributes cooperatively to the Th17/Th1 induction during bacterial infection. This study provides a deeper understanding of the molecular mechanisms underlying Th17/Th1 induction during pathogenic microbial infections in vivo.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Receptor fas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD , Diferenciación Celular , Interleucina-1beta/administración & dosificación , Listeria monocytogenes/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/inmunología , Bazo/inmunología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Receptor fas/deficiencia , Receptor fas/genética
8.
Biol Pharm Bull ; 42(2): 179-186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30713250

RESUMEN

Daikenchuto (DKT) has been widely used for the treatment of postsurgical ileus in Japan. However, its effect on postsurgical adhesion formation has been obscure. In this study, the effect of DKT on postsurgical adhesion formation induced by cecum cauterization or cecum abrasion in mice was investigated. First, the expression of adhesion-related molecules in damaged ceca was investigated by quantitative (q)RT-PCR. During 24 h after surgery, mRNA expressions of interferon-γ (IFN-γ), plasminogen activator inhibitor-1 (PAI-1), interleukin-17 (IL-17), and Substance P (SP) in cauterized ceca and those of PAI-1 and IL-17 in abraded ceca were significantly up-regulated. Next, the effect of DKT on adhesion formation macroscopically evaluated with adhesion scoring standards. DKT (22.5-67.5 mg/d) was administered orally for 7 d during the perioperative period, and DKT did not reduce adhesion scores in either the cauterization model (control : DKT 67.5 mg/d, 4.8 ± 0.2 : 4.8 ± 0.2) or in the abrasion model (control : DKT 67.5 mg/d, 4.9 ± 0.1 : 4.5 ± 0.3). Histologically, collagen deposition and leukocyte accumulation were found at the adhesion areas of control mice in both models, and DKT supplementation did not alleviate them. Last, effect of DKT on expression of proadhesion moleculs was evaluated. DKT also failed to down-regulate mRNA expression levels of them in damaged ceca of both models. In conclusion, PAI-1 and IL-17 may be key molecules of postsurgical adhesion formation. Collagen deposition and leukocytes accumulation are histological characteristic feature of post-surgical adhesion formation. DKT may not have any preventive effect on postsurgical adhesion formation in mice.


Asunto(s)
Ciego/efectos de los fármacos , Ciego/cirugía , Extractos Vegetales/farmacología , Adherencias Tisulares/tratamiento farmacológico , Animales , Cauterización/métodos , Ciego/metabolismo , Ciego/patología , Colágeno/metabolismo , Femenino , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos BALB C , Panax , Serpina E2/metabolismo , Sustancia P/metabolismo , Zanthoxylum , Zingiberaceae
9.
Int J Mol Sci ; 20(3)2019 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-30717382

RESUMEN

Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4⁺ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.


Asunto(s)
Susceptibilidad a Enfermedades , Interleucina-18/genética , Interleucina-18/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Interleucina-18/antagonistas & inhibidores , Interleucina-33/genética , Interleucina-33/metabolismo , Terapia Molecular Dirigida , Unión Proteica , Receptores de Interleucina-18/metabolismo , Transducción de Señal
10.
Proc Natl Acad Sci U S A ; 111(40): E4254-63, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25246571

RESUMEN

When nucleotide-binding oligomerization domain-like receptors (NLRs) sense cytosolic-invading bacteria, they induce the formation of inflammasomes and initiate an innate immune response. In quiescent cells, inflammasome activity is tightly regulated to prevent excess inflammation and cell death. Many bacterial pathogens provoke inflammasome activity and induce inflammatory responses, including cell death, by delivering type III secreted effectors, the rod component flagellin, and toxins. Recent studies indicated that Shigella deploy multiple mechanisms to stimulate NLR inflammasomes through type III secretion during infection. Here, we show that Shigella induces rapid macrophage cell death by delivering the invasion plasmid antigen H7.8 (IpaH7.8) enzyme 3 (E3) ubiquitin ligase effector via the type III secretion system, thereby activating the NLR family pyrin domain-containing 3 (NLRP3) and NLR family CARD domain-containing 4 (NLRC4) inflammasomes and caspase-1 and leading to macrophage cell death in an IpaH7.8 E3 ligase-dependent manner. Mice infected with Shigella possessing IpaH7.8, but not with Shigella possessing an IpaH7.8 E3 ligase-null mutant, exhibited enhanced bacterial multiplication. We defined glomulin/flagellar-associated protein 68 (GLMN) as an IpaH7.8 target involved in IpaH7.8 E3 ligase-dependent inflammasome activation. This protein originally was identified through its association with glomuvenous malformations and more recently was described as a member of a Cullin ring ligase inhibitor. Modifying GLMN levels through overexpression or knockdown led to reduced or augmented inflammasome activation, respectively. Macrophages stimulated with lipopolysaccharide/ATP induced GLMN puncta that localized with the active form of caspase-1. Macrophages from GLMN(+/-) mice were more responsive to inflammasome activation than those from GLMN(+/+) mice. Together, these results highlight a unique bacterial adaptation that hijacks inflammasome activation via interactions between IpaH7.8 and GLMN.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteínas Musculares/metabolismo , Shigella flexneri/metabolismo , Animales , Antígenos Bacterianos/genética , Apoptosis , Proteínas Bacterianas/genética , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Femenino , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Células Jurkat , Macrófagos/microbiología , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Fluorescente , Proteínas Musculares/genética , Unión Proteica , Shigella flexneri/genética , Shigella flexneri/fisiología , Técnicas del Sistema de Dos Híbridos
11.
Am J Physiol Endocrinol Metab ; 311(5): E881-E890, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702746

RESUMEN

Caspase-1 is a cysteine protease responsible for the processing of the proinflammatory cytokine interleukin-1ß and activated by the formation of inflammasome complexes. Although several investigations have found a link between diet-induced obesity and caspase-1, the relationship remains controversial. Here, we found that mice deficient in caspase-1 were susceptible to high-fat diet-induced obesity with increased adiposity as well as normal lipid and glucose metabolism. Caspase-1 deficiency clearly promoted the infiltration of inflammatory macrophages and increased the production of C-C motif chemokine ligand 2 (CCL2) in the adipose tissue. The dominant cellular source of CCL2 was stromal vascular fraction rather than adipocytes in the adipose tissue. These findings demonstrate a critical role of caspase-1 in macrophage-driven inflammation in the adipose tissue and the development of obesity. These data provide novel insights into the mechanisms underlying inflammation in the pathophysiology of obesity.


Asunto(s)
Tejido Adiposo/inmunología , Caspasa 1/genética , Quimiocina CCL2/inmunología , Macrófagos/inmunología , Obesidad/genética , Adipocitos/inmunología , Adipocitos/patología , Adiponectina/inmunología , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Composición Corporal , Caspasa 1/inmunología , Colesterol/metabolismo , Dieta Alta en Grasa , Citometría de Flujo , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leptina/inmunología , Masculino , Ratones , Ratones Noqueados , Obesidad/inmunología , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Microtomografía por Rayos X
12.
Arterioscler Thromb Vasc Biol ; 35(1): 127-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378412

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is considered a chronic inflammatory disease; however, the molecular basis underlying the sterile inflammatory response involved in the process of AAA remains unclear. We previously showed that the inflammasome, which regulates the caspase-1-dependent interleukin-1ß production, mediates the sterile cardiovascular inflammatory responses. Therefore, we hypothesized that the inflammasome is a key mediator of initial inflammation in AAA formation. APPROACH AND RESULTS: Apoptosis-associated speck-like protein containing a caspase recruitment domain is highly expressed in adventitial macrophages in human and murine AAA tissues. Using an established mouse model of AAA induced by continuous infusion of angiotensin II in Apoe(-/-) mice, NLR family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice were shown to decrease the incidence, maximal diameter, and severity of AAA along with adventitial fibrosis and inflammatory responses significantly, such as inflammatory cell infiltration and cytokine expression in the vessel wall. NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency in Apoe(-/-) mice also reduced elastic lamina degradation and metalloproteinase activation in the early phase of AAA formation. Furthermore, angiotensin II stimulated generation of mitochondria-derived reactive oxygen species in the adventitial macrophages, and this mitochondria-derived reactive oxygen species generation was inhibited by NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain, and caspase-1 deficiency. In vitro experiments revealed that angiotensin II stimulated the NLRP3 inflammasome activation and subsequent interleukin-1ß release in macrophages, and this activation was mediated through an angiotensin type I receptor/mitochondria-derived reactive oxygen species-dependent pathway. CONCLUSIONS: Our results demonstrate the importance of the NLRP3 inflammasome in the initial inflammatory responses in AAA formation, indicating its potential as a novel therapeutic target for preventing AAA progression.


Asunto(s)
Angiotensina II , Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/metabolismo , Inflamasomas/metabolismo , Activación de Macrófagos , Macrófagos/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Anciano , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Apolipoproteínas E , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/deficiencia , Caspasa 1/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo
13.
J Immunol ; 192(9): 4342-51, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696236

RESUMEN

Inflammation plays a key role in the pathophysiology of hepatic ischemia-reperfusion (I/R) injury. However, the mechanism by which hepatic I/R induces inflammatory responses remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by I/R is mediated through a multiple-protein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in hepatic I/R injury and found that hepatic I/R stimuli upregulated the inflammasome-component molecule, nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), but not apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). NLRP3(-/-) mice, but not ASC(-/-) and caspase-1(-/-) mice, had significantly less liver injury after hepatic I/R. NLRP3(-/-) mice showed reduced inflammatory responses, reactive oxygen species production, and apoptosis in I/R liver. Notably, infiltration of neutrophils, but not macrophages, was markedly inhibited in the I/R liver of NLRP3(-/-) mice. Bone marrow transplantation experiments showed that NLRP3 not only in bone marrow-derived cells, but also in non-bone marrow-derived cells contributed to liver injury after I/R. In vitro experiments revealed that keratinocyte-derived chemokine-induced activation of heterotrimeric G proteins was markedly diminished. Furthermore, NLRP3(-/-) neutrophils decreased keratinocyte-derived chemokine-induced concentrations of intracellular calcium elevation, Rac activation, and actin assembly formation, thereby resulting in impaired migration activity. Taken together, NLRP3 regulates chemokine-mediated functions and recruitment of neutrophils, and thereby contributes to hepatic I/R injury independently of inflammasomes. These findings identify a novel role of NLRP3 in the pathophysiology of hepatic I/R injury.


Asunto(s)
Proteínas Portadoras/inmunología , Hígado/inmunología , Neutrófilos/inmunología , Daño por Reperfusión/inmunología , Animales , Apoptosis/inmunología , Western Blotting , Proteínas Portadoras/metabolismo , Quimiotaxis de Leucocito , Citometría de Flujo , Inmunohistoquímica , Inflamasomas/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Immunol ; 190(8): 4245-54, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509366

RESUMEN

The molecular mechanisms of Fas (CD95/Apo-1)-mediated apoptosis are increasingly understood. However, the role of Fas-mediated production of proinflammatory cytokines such as IL-18 and IL-1ß in bacterial infection is unclear. We demonstrate the importance of Fas-mediated signaling in IL-18/IL-1ß production postinfection with Listeria monocytogenes without the contribution of caspase-1 inflammasome. IL-18/IL-1ß production in L. monocytogenes-infected peritoneal exudate cells from Fas-deficient mice was lower than those from wild type mice, indicating that Fas signaling contributes to cytokine production. L. monocytogenes infection induced Fas ligand expression on NK cells, which stimulates Fas expressed on the infected macrophages, leading to the production of IL-18/IL-1ß. This was independent of caspase-1, caspase-11, and nucleotide-binding domain and leucine-rich repeat-containing receptors (NLRs) such as Nlrp3 and Nlrc4, but dependent on apoptosis-associated speck-like protein containing a caspase recruitment domain. Wild type cells exhibited caspase-8 activation, whereas Fas-deficient cells did not. L. monocytogenes-induced caspase-8 activation was abrogated by inhibitor for intracellular reactive oxygen species, N-acetyl-L-cysteine. L. monocytogenes-infected macrophages produced type-I IFNs such as IFN-ß1, which was required for Il18 gene expression. Thus, Fas signaling regulates innate inflammatory cytokine production in L. monocytogenes infection.


Asunto(s)
Citocinas/biosíntesis , Mediadores de Inflamación/fisiología , Listeriosis/inmunología , Listeriosis/patología , Receptor fas/fisiología , Animales , Proteína Ligando Fas/biosíntesis , Femenino , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Mediadores de Inflamación/metabolismo , Interleucina-18/biosíntesis , Interleucina-1beta/biosíntesis , Listeriosis/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Receptor fas/biosíntesis , Receptor fas/metabolismo
15.
Mediators Inflamm ; 2015: 630265, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26549942

RESUMEN

The gene encoding IL-1 was sequenced more than 30 years ago, and many related cytokines, such as IL-18, IL-33, IL-36, IL-37, IL-38, IL-1 receptor antagonist (IL-1Ra), and IL-36Ra, have since been identified. IL-1 is a potent proinflammatory cytokine and is involved in various inflammatory diseases. Other IL-1 family ligands are critical for the development of diverse diseases, including inflammatory and allergic diseases. Only IL-1Ra possesses the leader peptide required for secretion from cells, and many ligands require posttranslational processing for activation. Some require inflammasome-mediated processing for activation and release, whereas others serve as alarmins and are released following cell membrane rupture, for example, by pyroptosis or necroptosis. Thus, each ligand has the proper molecular process to exert its own biological functions. In this review, we will give a brief introduction to the IL-1 family cytokines and discuss their pivotal roles in the development of various liver diseases in association with immune responses. For example, an excess of IL-33 causes liver fibrosis in mice via activation and expansion of group 2 innate lymphoid cells to produce type 2 cytokines, resulting in cell conversion into pro-fibrotic M2 macrophages. Finally, we will discuss the importance of IL-1 family cytokine-mediated molecular and cellular networks in the development of acute and chronic liver diseases.


Asunto(s)
Interleucina-1/fisiología , Hepatopatías/etiología , Animales , Citocinas/biosíntesis , Hepatitis Viral Humana/etiología , Humanos , Inflamación/etiología , Interleucina-18/fisiología , Interleucina-33/fisiología , Macrófagos/inmunología , Ratones , Receptores de Interleucina-1/fisiología
16.
Proc Natl Acad Sci U S A ; 109(9): 3451-6, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22331917

RESUMEN

When animals are infected with helminthic parasites, resistant hosts show type II helper T immune responses to expel worms. Recently, natural helper (NH) cells or nuocytes, newly identified type II innate lymphoid cells, are shown to express ST2 (IL-33 receptor) and produce IL-5 and IL-13 when stimulated with IL-33. Here we show the relevant roles of endogenous IL-33 for Strongyloides venezuelensis infection-induced lung eosinophilic inflammation by using Il33(-/-) mice. Alveolar epithelial type II cells (ATII) express IL-33 in their nucleus. Infection with S. venezuelensis or intranasal administration of chitin increases in the number of ATII cells and the level of IL-33. S. venezuelensis infection induces pulmonary accumulation of NH cells, which, after being stimulated with IL-33, proliferate and produce IL-5 and IL-13. Furthermore, S. venezuelensis infected Rag2(-/-) mice increase the number of ATII cells, NH cells, and eosinophils and the expression of IL-33 in their lungs. Finally, IL-33-stimulated NH cells induce lung eosinophilic inflammation and might aid to expel infected worms in the lungs.


Asunto(s)
Interleucinas/fisiología , Parasitosis Intestinales/complicaciones , Linfocitos/inmunología , Eosinofilia Pulmonar/etiología , Estrongiloidiasis/complicaciones , Animales , Líquido del Lavado Bronquioalveolar/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Interleucina-13/biosíntesis , Interleucina-33 , Interleucina-5/biosíntesis , Interleucinas/biosíntesis , Interleucinas/deficiencia , Interleucinas/genética , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/patología , Larva , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/crecimiento & desarrollo , Nippostrongylus/inmunología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Eosinofilia Pulmonar/inmunología , Infecciones por Strongylida/complicaciones , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/patología , Strongyloides/crecimiento & desarrollo , Strongyloides/inmunología , Estrongiloidiasis/inmunología , Estrongiloidiasis/patología
17.
Hepatology ; 57(1): 362-72, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22936459

RESUMEN

UNLABELLED: Concanavalin A (Con A) treatment induces severe hepatitis in mice in a manner dependent on T cells, interferon (IFN)-gamma, and tumor necrosis factor (TNF). Treatment with the anticoagulant heparin protects against hepatitis, despite healthy production of IFN-γ and TNF. Here, we investigated molecular and cellular mechanisms for hypercoagulation-mediated hepatitis. After Con A challenge, liver of wild-type (WT) mice showed prompt induction of Ifnγ and Tnf, followed by messenger RNA expression of tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1), which initiate blood coagulation and inhibit clot lysis, respectively. Mice developed dense intrahepatic fibrin deposition and massive liver necrosis. In contrast, Ifnγ(-/-) mice and Ifnγ(-/-) Tnf(-/-) mice neither induced Pai1 or Tf nor developed hepatitis. In WT mice TF blockade with an anti-TF monoclonal antibody protected against Con A-induced hepatitis, whereas Pai1(-/-) mice were not protected. Both hepatic macrophages and sinusoidal endothelial cells (ECs) expressed Tf after Con A challenge. Macrophage-depleted WT mice reconstituted with hematopoietic cells, including macrophages deficient in signal transducer and activator of transcription-1 (STAT1) essential for IFN-γ signaling, exhibited substantial reduction of hepatic Tf and of liver injuries. This was also true for macrophage-depleted Stat1(-/-) mice reconstituted with WT macrophages. Exogenous IFN-γ and TNF rendered T-cell-null, Con A-resistant mice deficient in recombination-activating gene 2, highly susceptible to Con A-induced liver injury involving TF. CONCLUSIONS: Collectively, these results strongly suggest that proinflammatory signals elicited by IFN-γ, TNF, and Con A in both hepatic macrophages and sinusoidal ECs are necessary and sufficient for the development of hypercoagulation-mediated hepatitis.


Asunto(s)
Hepatitis Animal/etiología , Interferón gamma , Hígado/metabolismo , Trombofilia/complicaciones , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Concanavalina A , Células Endoteliales/metabolismo , Fibrina/metabolismo , Hepatitis Animal/metabolismo , Hepatitis Animal/patología , Interferón gamma/metabolismo , Hígado/inmunología , Hígado/patología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mitógenos , Necrosis , Inhibidor 1 de Activador Plasminogénico/metabolismo , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Linfocitos T/fisiología , Trombofilia/inducido químicamente , Tromboplastina/metabolismo
18.
Int J Mol Sci ; 15(5): 7711-30, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24802875

RESUMEN

Kupffer cells reside within the liver sinusoid and serve as gatekeepers. They produce pro- and anti-inflammatory cytokines and other biologically important molecules upon the engagement of pattern recognition receptors such as Toll-like receptors. Kupffer cell-ablated mice established by in vivo treatment with clodronate liposomes have revealed many important features of Kupffer cells. In this paper, we review the importance of Kupffer cells in murine acute liver injuries and focus on the following two models: lipopolysaccharide (LPS)-induced liver injury, which is induced by priming with Propionibacterium acnes and subsequent challenge with LPS, and hypercoagulability-mediated acute liver failure such as that in concanavalin A (Con A)-induced hepatitis. Kupffer cells are required for LPS sensitization induced by P. acnes and are a major cellular source of interleukin-18, which induces acute liver injury following LPS challenge. Kupffer cells contribute to Con A-induced acute liver failure by initiating pathogenic, intrasinusoidal thrombosis in collaboration with sinusoidal endothelial cells. The mechanisms underlying these models may shed light on human liver injuries induced by various etiologies such as viral infection and/or abnormal metabolism.


Asunto(s)
Modelos Animales de Enfermedad , Macrófagos del Hígado/patología , Hepatopatías/etiología , Hepatopatías/patología , Hígado/lesiones , Ratones , Animales , Infecciones por Bacterias Grampositivas/complicaciones , Hepatitis/etiología , Hepatitis/patología , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/microbiología , Lipopolisacáridos/inmunología , Hígado/inmunología , Hígado/microbiología , Hígado/patología , Hepatopatías/inmunología , Hepatopatías/microbiología , Ratones/fisiología , Propionibacterium acnes/inmunología , Trombofilia/complicaciones
19.
J Immunol ; 184(9): 5287-97, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20348425

RESUMEN

Vibrio vulnificus and Vibrio cholerae are Gram-negative pathogens that cause serious infectious disease in humans. The beta form of pro-IL-1 is thought to be involved in inflammatory responses and disease development during infection with these pathogens, but the mechanism of beta form of pro-IL-1 production remains poorly defined. In this study, we demonstrate that infection of mouse macrophages with two pathogenic Vibrio triggers the activation of caspase-1 via the NLRP3 inflammasome. Activation of the NLRP3 inflammasome was mediated by hemolysins and multifunctional repeat-in-toxins produced by the pathogenic bacteria. NLRP3 activation in response to V. vulnificus infection required NF-kappaB activation, which was mediated via TLR signaling. V. cholerae-induced NLRP3 activation also required NF-kappaB activation but was independent of TLR stimulation. Studies with purified V. cholerae hemolysin revealed that toxin-stimulated NLRP3 activation was induced by TLR and nucleotide-binding oligomerization domain 1/2 ligand-mediated NF-kappaB activation. Our results identify the NLRP3 inflammasome as a sensor of Vibrio infections through the action of bacterial cytotoxins and differential activation of innate signaling pathways acting upstream of NF-kappaB.


Asunto(s)
Toxinas Bacterianas/farmacología , Proteínas Portadoras/metabolismo , FN-kappa B/fisiología , Proteína Adaptadora de Señalización NOD1/fisiología , Proteína Adaptadora de Señalización NOD2/fisiología , Transducción de Señal/inmunología , Receptores Toll-Like/fisiología , Vibrio cholerae/patogenicidad , Vibrio vulnificus/patogenicidad , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Células de la Médula Ósea/patología , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Caspasa 1/metabolismo , Inmunidad Innata/genética , Inflamación/enzimología , Inflamación/inmunología , Inflamación/microbiología , Interleucina-1beta/metabolismo , Ligandos , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Transducción de Señal/genética , Vibrio cholerae/inmunología , Vibrio vulnificus/inmunología
20.
Am J Respir Cell Mol Biol ; 45(1): 31-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20802088

RESUMEN

Clinical and epidemiological studies have shown the contribution of viral infection to the development of allergic asthma. Many RNA viruses, pathogenic for the respiratory tract, generate double-stranded (ds)RNA during their replication. Typical innate immune responses triggered by dsRNA involve the endosomal and cytoplasmic pathways. The former is mediated by Toll/IL-1R domain-containing adaptor inducing IFN-ß (TRIF), and the latter by IFN-ß promoter stimulator 1 (IPS-1). We explored the effect of polyinocinic polycytidilic acid, a synthetic dsRNA, on the development of an asthma phenotype in mice. Administration of dsRNA during ovalbumin sensitization augmented airway eosinophilia and airway hyperresponsiveness after an antigen challenge, which was associated with enhanced induction of IL-13-producing CD8(+) T cells. The augmentation was induced in IPS-1-deficient mice but not in TRIF-deficient mice. The interactions between dendritic cells (DCs) and T cells are regulated by B7-family costimulatory molecules, including B7-H1 (also known as PD-L1), a putative ligand for programmed death-1 (PD-1). Treatment of bone marrow-derived DCs with dsRNA enhanced B7-H1 expression in a TRIF-dependent manner. Additionally, dsRNA increased B7-H1 expression on DCs in the draining lymph nodes of ovalbumin-sensitized mice. The augmentation of the asthma phenotype was prevented by the treatment of mice with anti-B7-H1 mAb but not with anti-PD-1 mAb. The augmentation was not induced in B7-H1-deficient mice. These results suggest that dsRNA-triggered activation of the innate immune system in sensitization leads to augmentation of the asthma phenotype via IL-13 mainly from CD8(+) T cells. B7-H1 plays a crucial role in the process without requiring interaction with PD-1.


Asunto(s)
Asma/inducido químicamente , Asma/inmunología , Antígeno B7-1/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Glicoproteínas de Membrana/inmunología , Péptidos/inmunología , ARN Bicatenario/efectos adversos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Asma/genética , Asma/metabolismo , Asma/patología , Antígeno B7-1/biosíntesis , Antígeno B7-1/genética , Antígeno B7-H1 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Interleucina-13/biosíntesis , Interleucina-13/genética , Interleucina-13/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ovalbúmina/efectos adversos , Ovalbúmina/farmacología , Péptidos/genética , Fenotipo , Eosinofilia Pulmonar/inducido químicamente , Eosinofilia Pulmonar/genética , Eosinofilia Pulmonar/inmunología , Eosinofilia Pulmonar/patología , ARN Bicatenario/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA