Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 119: 539-553, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38663774

RESUMEN

Interleukin-33 (IL-33), secreted by astrocytes, regulates the synapse development in the spinal cord and hippocampus and suppresses autoimmune disease in the central nervous system (CNS). However, the mechanism of unconventional protein secretion of this cytokine remains unclear. In this study, we found that IFN-γ promotes the active secretion of IL-33 from astrocytes, and the active secretion of IL-33 from cytoplasm to extracellular space was dependent on interaction with transmembrane emp24 domain 10 (TMED10) via the IL-1 like cytokine domain in astrocytes. Knockout of Il-33 or its receptor St2 induced hippocampal astrocyte activation and depressive-like disorder in naive mice, as well as increased spinal cord astrocyte activation and polarization to a neurotoxic reactive subtype and aggravated passive experimental autoimmune encephalomyelitis (EAE). Our results have identified that IL-33 is actively secreted by astrocytes through the unconventional protein secretion pathway facilitated by TMED10 channels. This process helps maintain CNS homeostasis by inhibiting astrocyte activation.

2.
Neurosci Bull ; 38(11): 1303-1314, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35697993

RESUMEN

High mobility group box 1 (HMGB1) has been reported to play an important role in experimental autoimmune encephalomyelitis (EAE). Astrocytes are important components of neurovascular units and tightly appose the endothelial cells of microvessels by their perivascular endfeet and directly regulate the functions of the blood-brain barrier. Astrocytes express more HMGB1 during EAE while the exact roles of astrocytic HMGB1 in EAE have not been well elucidated. Here, using conditional-knockout mice, we found that astrocytic HMGB1 depletion decreased morbidity, delayed the onset time, and reduced the disease score and demyelination of EAE. Meanwhile, there were fewer immune cells, especially pathogenic T cells infiltration in the central nervous system of astrocytic HMGB1 conditional-knockout EAE mice, accompanied by up-regulated expression of the tight-junction protein Claudin5 and down-regulated expression of the cell adhesion molecules ICAM1 and VCAM1 in vivo. In vitro, HMGB1 released from astrocytes decreased Claudin5 while increased ICAM1 and VCAM1 expressed by brain microvascular endothelial cells (BMECs) through TLR4 or RAGE. Taken together, our results demonstrate that HMGB1 derived from astrocytes aggravates EAE by directly influencing the immune cell infiltration-associated functions of BMECs.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Proteína HMGB1 , Ratones , Animales , Encefalomielitis Autoinmune Experimental/metabolismo , Astrocitos/metabolismo , Proteína HMGB1/metabolismo , Células Endoteliales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Barrera Hematoencefálica/metabolismo
3.
Front Immunol ; 12: 584097, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868221

RESUMEN

High mobility group box 1 protein (HMGB1) is known to be a trigger of inflammation in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). However, it may play a different role in some way. Here we investigated the effect of HMGB1 on promoting sonic hedgehog (shh) release from astrocytes as well as the possible signal pathway involved in it. Firstly, shh increased in astrocytes after administration of recombinant HMGB1 or decreased after HMGB1 was blocked when stimulated by homogenate of the onset stage of EAE. Moreover, the expression of HMGB1 receptors, toll-like receptor (TLR) 2 and receptor for advanced glycation end products (RAGE) increased after HMGB1 administration in primary astrocytes. However, the enhancing effect of HMGB1 on shh release from astrocytes was suppressed only after RAGE was knocked out or blocked. Mechanistically, HMGB1 functioned by activating RAGE-mediated JNK, p38, stat3 phosphorylation. Moreover, HMGB1 could induce shh release in EAE. Additionally, intracerebroventricular injection of recombinant shh protein on the onset stage of EAE alleviated the progress of disease and decreased demylination, compared to the mice with normal saline treatment. Overall, HMGB1 promoted the release of shh from astrocytes through signal pathway JNK, p38 and stat3 mediated by receptor RAGE, which may provide new insights of HMGB1 function in EAE.


Asunto(s)
Astrocitos/efectos de los fármacos , Proteína HMGB1/farmacología , Proteínas Hedgehog/metabolismo , Proteínas Recombinantes/farmacología , Animales , Antiinflamatorios/farmacología , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Ácido Glicirrínico/farmacología , Proteína HMGB1/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/prevención & control , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal/efectos de los fármacos
4.
Front Immunol ; 12: 657803, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815420

RESUMEN

The role of IL-33/ST2 signaling in cardiac allograft vasculopathy (CAV) is not fully addressed. Here, we investigated the role of IL-33/ST2 signaling in allograft or recipient in CAV respectively using MHC-mismatch murine chronic cardiac allograft rejection model. We found that recipients ST2 deficiency significantly exacerbated allograft vascular occlusion and fibrosis, accompanied by increased F4/80+ macrophages and CD3+ T cells infiltration in allografts. In contrast, allografts ST2 deficiency resulted in decreased infiltration of F4/80+ macrophages, CD3+ T cells and CD20+ B cells and thus alleviated vascular occlusion and fibrosis of allografts. These findings indicated that allografts or recipients ST2 deficiency oppositely affected cardiac allograft vasculopathy/fibrosis via differentially altering immune cells infiltration, which suggest that interrupting IL-33/ST2 signaling locally or systematically after heart transplantation leads different outcome.


Asunto(s)
Enfermedad Coronaria/etiología , Enfermedad Coronaria/patología , Trasplante de Corazón , Proteína 1 Similar al Receptor de Interleucina-1/deficiencia , Leucocitos/patología , Aloinjertos , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Subgrupos de Linfocitos B/patología , Enfermedad Coronaria/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Rechazo de Injerto , Supervivencia de Injerto , Trasplante de Corazón/efectos adversos , Trasplante de Corazón/métodos , Inmunohistoquímica , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Complicaciones Posoperatorias , Subgrupos de Linfocitos T/metabolismo
5.
Front Immunol ; 12: 641973, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777037

RESUMEN

Cardiac allograft vasculopathy (CAV) charactered with aberrant remodeling and fibrosis usually leads to the loss of graft after heart transplantation. Our previous work has reported that extracellular high-mobility group box 1 (HMGB1) participated in the CAV progression via promoting inflammatory cells infiltration and immune damage. The aim of this study was to investigate the involvement of HMGB1 in the pathogenesis of CAV/fibrosis and potential mechanisms using a chronic cardiac rejection model in mice. We found high levels of transforming growth factor (TGF)-ß1 in cardiac allografts after transplantation. Treatment with HMGB1 neutralizing antibody markedly prolonged the allograft survival accompanied by attenuated fibrosis of cardiac allograft, decreased fibroblasts-to-myofibroblasts conversion, and reduced synthesis and release of TGF-ß1. In addition, recombinant HMGB1 stimulation promoted release of active TGF-ß1 from cardiac fibroblasts and macrophages in vitro, and subsequent phosphorylation of Smad2 and Smad3 which were downstream of TGF-ß1 signaling. These data indicate that HMGB1 contributes to the CAV/fibrosis via promoting the activation of TGF-ß1/Smad signaling. Targeting HMGB1 might become a new therapeutic strategy for inhibiting cardiac allograft fibrosis and dysfunction.


Asunto(s)
Rechazo de Injerto/metabolismo , Proteína HMGB1/metabolismo , Trasplante de Corazón/efectos adversos , Miocardio/patología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Rechazo de Injerto/patología , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Transducción de Señal/fisiología
6.
J Invest Dermatol ; 141(3): 596-606.e7, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32853659

RESUMEN

IL-33 is constitutively expressed in the skin. Psoriasis is a common skin inflammatory disease. The roles of IL-33 in psoriasis have not been well-elucidated. We identified that keratinocytes (KCs) are the predominant cells expressing IL-33 and its receptor, suppression of tumorigenicity 2, in the skin. KCs actively released IL-33 on psoriasis inflammatory stimuli and induced psoriasis-related cytokine, chemokine, and inflammatory molecules genes transcription in KCs in an autocrine manner. IL-33‒specific deficiency in KCs ameliorated imiquimod-induced psoriatic dermatitis. In addition, intradermal injection of recombinant IL-33 alone induced psoriasis-like dermatitis, which is attributed to the transcriptional upregulation of genes enriched in IL-17, TNF, and chemokine signaling pathway in KCs on recombinant IL-33 stimulation. Our data demonstrate that the autocrine circuit of IL-33 in KCs promotes the progression of psoriatic skin inflammation, and IL-33 is a potential therapeutic target for psoriasis.


Asunto(s)
Interleucina-33/metabolismo , Queratinocitos/metabolismo , Psoriasis/inmunología , Adulto , Animales , Comunicación Autocrina/inmunología , Biopsia , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Voluntarios Sanos , Humanos , Imiquimod/inmunología , Inyecciones Intradérmicas , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/administración & dosificación , Interleucina-33/genética , Queratinocitos/inmunología , Masculino , Ratones , Persona de Mediana Edad , Psoriasis/diagnóstico , Psoriasis/genética , Psoriasis/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Índice de Severidad de la Enfermedad , Transducción de Señal/genética , Transducción de Señal/inmunología , Piel/inmunología , Piel/patología , Activación Transcripcional/inmunología , Regulación hacia Arriba/inmunología
7.
Cell Mol Immunol ; 17(5): 519-532, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31853001

RESUMEN

Allergic asthma, a chronic inflammatory airway disease associated with type 2 cytokines, often originates in early life. Immune responses at an early age exhibit a Th2 cell bias, but the precise mechanisms remain elusive. Plasmacytoid dendritic cells (pDCs), which play a regulatory role in allergic asthma, were shown to be deficient in neonatal mice. We report here that this pDC deficiency renders neonatal mice more susceptible to severe allergic airway inflammation than adult mice in an OVA-induced experimental asthma model. Adoptive transfer of pDCs or administration of IFN-α to neonatal mice prevented the development of allergic inflammation in wild type but not in IFNAR1-/- mice. Similarly, adult mice developed more severe allergic inflammation when pDCs were depleted. The protective effects of pDCs were mediated by the pDC-/IFN-α-mediated negative regulation of the secretion of epithelial cell-derived CCL20, GM-CSF, and IL-33, which in turn impaired the recruitment of cDC2 and ILC2 cells to the airway. In asthmatic patients, the percentage of pDCs and the level of IFN-α were lower in children than in adults. These results indicate that impairment of pDC-epithelial cell crosstalk in neonates is a susceptibility factor for the development of allergen-induced allergic airway inflammation.


Asunto(s)
Células Dendríticas/metabolismo , Hipersensibilidad/patología , Inflamación/patología , Interferón-alfa/biosíntesis , Pulmón/patología , Traslado Adoptivo , Animales , Animales Recién Nacidos , Asma/inmunología , Asma/patología , Bronquios/patología , Líquido del Lavado Bronquioalveolar , Recuento de Células , Citocinas/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Hipersensibilidad/inmunología , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/metabolismo , Esputo/citología
8.
Curr Med Sci ; 39(6): 883-889, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31845218

RESUMEN

Malaria remains a global health problem. The relationship between Plasmodium spp. and the gut microbiota as well as the impact of Plasmodium spp. on the gut microbiota in vertebrate hosts is unclear. The aim of the current study was to evaluate the effect of blood-stage Plasmodium parasites on the gut microbiota of mice. The gut microbiota was analyzed by 16S rRNA sequencing and bioinformatic analyses at three stages. The gut microbiota changed during the three phases: the healthy stage, the infection stage, and the cure stage (on the 9th day after malarial elimination). Moreover, the gut microbiota of these infected animals did not recover after malaria infection. There were 254 operational taxonomic units (OTUs) across all three stages, and there were unique strains or OTUs at each stage of the experiment. The percentages of community abundance of 8 OTUs changed significantly (P<0.05). The dominant OTU in both the healthy mice and the mice with malaria was OTU265, while that in the cured mice was OTU234. In addition, the changes in OTU147 were the most noteworthy. Its percentage of community abundance varied greatly, with higher values during malaria than before malaria infection and after malaria elimination. These results indicated that the external environment influenced the gut microbiota after host C57BL/6 mice were infected with blood-stage P. berghei ANKA and that the same was true during and after elimination of blood-stage P. berghei ANKA. In addition, we could not isolate OTU147 for further study. This study identified gut microbiota components that were reconstructed after infection by and elimination of blood-stage P. berghei ANKA in host C57BL/6 mice, and this process was affected by P. berghei ANKA and the external environment of the host.


Asunto(s)
Bacterias/clasificación , Malaria/microbiología , Plasmodium berghei/patogenicidad , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , Barrera Hematotesticular/parasitología , Estudios de Casos y Controles , ADN Ribosómico/genética , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Filogenia
10.
Cell Immunol ; 338: 9-20, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30826001

RESUMEN

The emergence of alloreactive Th17 cells that mediate allograft rejection has provided an impetus to understand the factors affecting the generation of Th17 cells in allograft transplantation. How toll-like receptor 2 (TLR2) signalling regulates the generation of Th17 cells upon alloantigen stimuli remains unclear. In this study, we used a mouse model of cardiac allograft transplantation to investigate whether TLR2 signalling influences the development of Th17 cells. Here, we demonstrate that the TLR2-deficient recipient mice show high Th17 cells, both in spleens and allografts, as well as higher infiltrating inflammatory leukocytes in cardiac allografts compared to wild-type control recipient mice. mRNA expression of IL-17, IL-6, TNF-α, CCR6 and CCL20 within the allografts is markedly increased in TLR2-deficient recipient mice compared to wild-type recipient mice. In addition, TLR2 deficiency leads to upregulation of Signal transducer and activator of transcription 3 (STAT3) phosphorylation in both spleens and allografts. In an in vitro experiment, a mixed lymphocyte reaction was assessed, which further confirmed that TLR2 deficiency leads to a significant increase in the generation of Th17 cells compared with wild-type controls. Furthermore, IL-6 secreted by the dendritic cells of TLR2-deficient mice contributes to driving the generation of these Th17 cells. These results suggest that TLR2 signalling is important in regulating the development of Th17 cells after cardiac allograft transplantation.


Asunto(s)
Aloinjertos/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón , Interleucina-6/metabolismo , Células Th17/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Quimiocina CCL20/genética , Modelos Animales de Enfermedad , Humanos , Interleucina-6/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR6/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Receptor Toll-Like 2/genética , Trasplante Homólogo
11.
Int Immunopharmacol ; 68: 145-155, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30634142

RESUMEN

Gasdermin D (GSDMD), a genetic substrate for inflammatory caspases, plays a central role in pyroptosis of macrophages and release of interleukin­1ß (IL-1ß), but was mainly referred to microbial infection. High mobility group box-1 (HMGB1), served as an alarm molecule during various pathological process, has been widely recognized to be involved in liver ischemia-reperfusion (I/R). Glycyrrhizin, a natural anti-inflammatory and antiviral triterpene in clinical use, was recently referred to have ability to prevent I/R induced liver injury by inhibiting HMGB1 expression and activity. However, the mechanisms responsible for damage amelioration subsequently to HMGB1 inhibition during liver I/R remain enigmatic. This study was designed to explore the functional role and molecular mechanism of glycyrrhizin in the regulation of I/R induced liver injury. We found that liver I/R promotes GSDMD-mediated pyroptotic cell death of Kupffer cells, which was inhibited by glycyrrhizin. Interestingly, endogenous HMGB1, not exogenous one, was involved in hypoxia-reoxygenation (H/R) induced pyroptosis. Moreover, GSDMD knockdown protects kupffer cells against H/R induced pyroptosis in vitro. Here, we report, for the first time, that glycyrrhizin attenuated tissue damage and kupffer cells pyroptosis during liver ischemia-reperfusion injury (LIRI) and identify a previously unrecognized HMGB1- dependent GSDMD- mediated signaling pathway in the mechanism of kupffer cells pyroptosis induced by H/R. Our findings provide the first demonstration of GSDMD-determined pyroptotic cell death responsible for I/R induced release of IL-1ß and this would provide a mandate to better understand the unconventional mechanisms of cytokine release in the sterile innate immune system.


Asunto(s)
Antiinflamatorios , Proteínas Reguladoras de la Apoptosis/fisiología , Ácido Glicirrínico , Proteína HMGB1/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Hepatopatías , Piroptosis/efectos de los fármacos , Daño por Reperfusión , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Ácido Glicirrínico/farmacología , Ácido Glicirrínico/uso terapéutico , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Macrófagos del Hígado/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hepatopatías/tratamiento farmacológico , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Proteínas de Unión a Fosfato , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
12.
Autophagy ; 15(6): 960-975, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30681394

RESUMEN

Macroautophagy/autophagy is a conserved ubiquitous pathway that performs diverse roles in health and disease. Although many key, widely expressed proteins that regulate autophagosome formation followed by lysosomal fusion have been identified, the possibilities of cell-specific elements that contribute to the autophagy fusion machinery have not been explored. Here we show that a macrophage-specific isoform of the vacuolar ATPase protein ATP6V0D2/subunit d2 is dispensable for lysosome acidification, but promotes the completion of autophagy via promotion of autophagosome-lysosome fusion through its interaction with STX17 and VAMP8. Atp6v0d2-deficient macrophages have augmented mitochondrial damage, enhanced inflammasome activation and reduced clearance of Salmonella typhimurium. The susceptibility of atp6v0d2 knockout mice to DSS-induced colitis and Salmonella typhimurium-induced death, highlights the in vivo significance of ATP6V0D2-mediated autophagosome-lysosome fusion. Together, our data identify ATP6V0D2 as a key component of macrophage-specific autophagosome-lysosome fusion machinery maintaining macrophage organelle homeostasis and, in turn, limiting both inflammation and bacterial infection. Abbreviations: ACTB/ß-actin: actin, beta; ATG14: autophagy related 14; ATG16L1: autophagy related 16-like 1 (S. cerevisiae); ATP6V0D1/2: ATPase, H+ transporting, lysosomal V0 subunit D1/2; AIM2: absent in melanoma 2; BMDM: bone marrow-derived macrophage; CASP1: caspase 1; CGD: chronic granulomatous disease; CSF1/M-CSF: colony stimulating factor 1 (macrophage); CTSB: cathepsin B; DSS: dextran sodium sulfate; IL1B: interleukin 1 beta; IL6: interleukin 6; IRGM: immunity-related GTPase family M member; KO: knockout; LAMP1: lysosomal-associated membrane protein 1; LC3: microtubule-associated protein 1 light chain 3; LPS: lipo-polysaccaride; NLRP3: NLR family, pyrin domain containing 3; PYCARD/ASC: PYD and CARD domain containing; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SNAP29: synaptosomal-associated protein 29; SQSTM1/p62: sequestosome 1; STX17: syntaxin 17; TLR: toll-like receptor; TNF: tumor necrosis factor ; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1; VAMP8: vesicle-associated membrane protein 8; WT: wild type; 3-MA: 3-methyladenine.


Asunto(s)
Autofagosomas/metabolismo , Inflamasomas/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/ultraestructura , Autofagia/efectos de los fármacos , Autofagia/genética , Células Cultivadas , Colitis/genética , Colitis/inmunología , Células HEK293 , Humanos , Inflamasomas/genética , Lisosomas/genética , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Fusión de Membrana/efectos de los fármacos , Fusión de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/inmunología , Mitocondrias/ultraestructura , Peritonitis/genética , Peritonitis/inmunología , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , ATPasas de Translocación de Protón Vacuolares/genética
13.
Inflamm Res ; 68(1): 75-91, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30430216

RESUMEN

BACKGROUND: Previous studies showed that CD4+ T cells play a critical role in Con A-induced hepatitis in wild-type mice. However, the role of CD8+ T cells in the setting of Con A-induced hepatitis is enigmatic. The aim of study is to investigate the function of CD8+ T cells in the context of Con-A-induced hepatitis. MATERIALS AND SUBJECTS: Two different mouse models of Con A-induced hepatitis, T cell-transferred Rag2-/- mice and wild-type C57BL/6 mice, were used in the present study. IL-33 gene knockout mice were used to confirm the role of alarmin in Con A-induced hepatitis. RESULTS: Opposing to the previous results obtained in wild-type mice, transferred CD4+ T cells alone into Rag2-knockout mice cannot cause hepatitis upon Con A challenge. In stark contrast, transferred CD8+ T cells play an important role in the pathogenesis of Con A-induced liver injury in T cell-transferred Rag2-deficient mice. Furthermore, we found that hepatocytes injured by perforin-based CD8+ T cell cytotoxicity release the alarmin IL-33. This cytokine promotes ST2+ ILC2 development and the secretion of cytokines IL-5 and IL-13 to mediate liver inflammation triggered by Con A challenge. In addition, these type 2 cytokines, including those originated from CD4+ T cells, result in eosinophils accumulation in liver to exacerbate the liver injury after Con A administration. CONCLUSION: Our data for the first time revealed that CD8+ T cells play an indispensable role in the pathogenesis of Con A-induced liver injury in T cell-transferred Rag2-deficient mice. Therefore, the CD8+ T cell/IL-33/ILC2 axis is a potential therapeutic target for acute immune-mediated liver injury.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Citocinas/inmunología , Proteínas de Unión al ADN/inmunología , Linfocitos/inmunología , Animales , Células Cultivadas , Concanavalina A , Citocinas/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Inmunidad Innata , Ganglios Linfáticos/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología
14.
J Clin Invest ; 129(2): 631-646, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30431439

RESUMEN

Macrophages perform key functions in tissue homeostasis that are influenced by the local tissue environment. Within the tumor microenvironment, tumor-associated macrophages can be altered to acquire properties that enhance tumor growth. Here, we found that lactate, a metabolite found in high concentration within the anaerobic tumor environment, activated mTORC1 that subsequently suppressed TFEB-mediated expression of the macrophage-specific vacuolar ATPase subunit ATP6V0d2. Atp6v0d2-/- mice were more susceptible to tumor growth, with enhanced HIF-2α-mediated VEGF production in macrophages that display a more protumoral phenotype. We found that ATP6V0d2 targeted HIF-2α but not HIF-1α for lysosome-mediated degradation. Blockade of HIF-2α transcriptional activity reversed the susceptibility of Atp6v0d2-/- mice to tumor development. Furthermore, in a cohort of patients with lung adenocarcinoma, expression of ATP6V0d2 and HIF-2α was positively and negatively correlated with survival, respectively, suggesting a critical role of the macrophage lactate/ATP6V0d2/HIF-2α axis in maintaining tumor growth in human patients. Together, our results highlight the ability of tumor cells to modify the function of tumor-infiltrating macrophages to optimize the microenvironment for tumor growth.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ácido Láctico/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos , Proteínas de Neoplasias/metabolismo , Microambiente Tumoral , ATPasas de Translocación de Protón Vacuolares/biosíntesis , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , ATPasas de Translocación de Protón Vacuolares/genética
15.
Biomed Pharmacother ; 110: 692-699, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30553196

RESUMEN

Glycyrrhizin, a triterpenoid compound, has been reported to be an anti-inflammatory agent for the treatment of a variety of inflammatory diseases including hepatitis. However, the mechanism by which glycyrrhizin inhibits inflammation is unclear. Using a Con A-induced hepatitis model in mice, we found that administration of glycyrrhizin ameliorates Con A-induced liver injury, which manifests as reduction in the production of inflammatory cytokines IFN-γ, IL-6 and IL-17, as well as serum alanine aminotransferase (ALT). Blockade of IL-17 dramatically mitigates liver injury resulting from Con A challenge. Interestingly, at both the mRNA and protein levels, the endogenous alarmin inflammatory molecule high-mobility group box 1 (HMGB1) is significantly decreased in mice injected with glycyrrhizin combined with Con A compared to those injected with Con A alone. In contrast, the administration of glycyrrhizin with Con A challenge up-regulates the production of IL-25. Furthermore, an increase in the proportion of protective lymphocyte subset, Gr-1+ CD11b+ (Myeloid-Derived Suppressor Cell, MDSCs), could be induced by increased IL-25 to restrain immune cell activation and favor the resolution of detrimental immune reactions caused by Con A challenge. The results indicate that glycyrrhizin plays a protective role in Con A-induced hepatitis. This protective role is particularly associated with reducing the production of IL-17 and enhancing the expression of IL-25. The present study may provide a new strategy for the treatment of acute hepatitis in the clinical setting.


Asunto(s)
Concanavalina A/toxicidad , Ácido Glicirrínico/uso terapéutico , Hepatitis/tratamiento farmacológico , Hepatitis/metabolismo , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Ácido Glicirrínico/farmacología , Interleucina-17/antagonistas & inhibidores , Interleucinas/agonistas , Masculino , Ratones , Ratones Endogámicos C57BL
16.
Mol Med Rep ; 18(6): 5527-5535, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30365069

RESUMEN

Extracellular high mobility group box 1 (HMGB1) serves a critical role in inflammatory diseases. HMGB1 is released into the extracellular environment mainly by passive release from necrotic cells or active secretion from monocytes/macrophages following stimulation. However, the translocation of actively secreted HMGB1 from the nucleus to the cytoplasm with post­translational modifications such as acetylation is required; HMGB1 is then released into the extracellular space. Whether acetylation influences the extracellular function of HMGB1 remains unknown. In the present study, an optimized method of gene mutation by using well­designed primers in particular, which were employed to identify the mutant gene. The substitution of six lysine residues for glutamines was conducted to mimic acetylated HMGB1 (HMGB1­M) and observe the effects of HMGB1­M on macrophages and dendritic cells (DCs). Tumor necrosis factor (TNF)­α production in RAW 264.7 cells was assessed by ELISA. The phagocytic potential of RAW 264.7 cells, DC maturation and CXCR4 expression were analyzed by flow cytometry. The results of the present study revealed that HMGB1­M increased cytoplasmic translocation. Compared with HMGB1, HMGB1­M increased TNF­α production within RAW 264.7 cells and decreased the mean fluorescence intensity (MFI) of integrin α X, and the percentage and MFI of major histocompatibility complex­II on DCs. HMGB1­M exhibited no significant effects on phagocytosis of macrophages and expression frequency of cluster of differentiation 80 and chemokine receptor type 4 on DCs. These results suggested that HMGB1­M may partly promote inflammation and decrease DC maturation. Thus, the findings of the present study may provide insight into the complex role of HMGB1 in inflammatory diseases.


Asunto(s)
Células Dendríticas/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Acetilación , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Dendríticas/efectos de los fármacos , Femenino , Proteína HMGB1/genética , Proteína HMGB1/farmacología , Humanos , Macrófagos/efectos de los fármacos , Ratones , Mutación , Fenotipo , Plásmidos/genética , Células RAW 264.7 , Proteínas Recombinantes , Factor de Necrosis Tumoral alfa/biosíntesis
17.
Mol Immunol ; 101: 550-563, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30173119

RESUMEN

Interleukin (IL)-33, a member of the IL-1 cytokine family, is highly expressed in central nervous system (CNS), suggesting its potential role in CNS. Although some studies have focused on the role of IL-33 in multiple sclerosis (MS) / experimental autoimmune encephalomyelitis (EAE), an autoimmune disease characterized by demyelination and axonal damage in CNS, the exact role of IL-33 in MS/EAE remains unclear and controversial. Here, we used IL-33 knockout mice to clarify the role of endogenous IL-33 in EAE by simultaneously eliminating its role as a nuclear transcription factor and an extracellular cytokine. We found that the clinical score in IL-33 knockout EAE mice was higher accompanied by more severe demyelination compared with the wild-type (WT) EAE mice. As for the main immune cells participating in EAE in IL-33 knockout mice, pathogenic effector T cells increased both in peripheral immune organs and CNS, while CD4+FOXP3+ regulatory T cells decreased in spleen and lymph nodes, Th2 cells and natural killer (NK) cells decreased in CNS. Additionally, the populations of microglia/macrophages and CD11C+CD11B+ dendritic cells (DCs) increased in CNS of IL-33 knockout mice with EAE, among which iNOS-producing microglia/macrophages increased. Moreover, resident astrocytes/microglia were more activated in IL-33 knockout mice with EAE. In vitro, after blocking the IL-33, the proliferation of primary astrocytes, the production of MCP-1/CCL2 and TNF-α by astrocytes, and the production of TNF-α by primary microglia stimulated by the homogenate of the peak stage of EAE were increased. Our results indicate that IL-33 plays a protective role in EAE and exerts extensive influences on multiple immune cells and neural cells involved in EAE.


Asunto(s)
Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-33/deficiencia , Neuroglía/patología , Linfocitos T/inmunología , Linfocitos T/patología , Animales , Astrocitos/metabolismo , Citocinas , Enfermedades Desmielinizantes/patología , Células Dendríticas/metabolismo , Femenino , Células Asesinas Naturales/inmunología , Ganglios Linfáticos/patología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Modelos Biológicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/patología , Bazo/patología , Células Th2/inmunología
19.
Front Immunol ; 9: 1518, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30013568

RESUMEN

The inflammatory mediator high-mobility group box 1 (HMGB1) plays a critical role in the pathogenesis of human multiple sclerosis (MS) and mouse experimental autoimmune encephalomyelitis (EAE). Glycyrrhizin (GL), a glycoconjugated triterpene extracted from licorice root, has the ability to inhibit the functions of HMGB1; however, GL's function against EAE has not been thoroughly characterized to date. To determine the benefit of GL as a modulator of neuroinflammation, we used an in vivo study to examine GL's effect on EAE along with primary cultured cortical neurons to study the GL effect on HMGB1 release. Treatment of EAE mice with GL from onset to the peak stage of disease resulted in marked attenuation of EAE severity, reduced inflammatory cell infiltration and demyelination, decreased tumor necrosis factor-alpha (TNF-α), IFN-γ, IL-17A, IL-6, and transforming growth factor-beta 1, and increased IL-4 both in serum and spinal cord homogenate. Moreover, HMGB1 levels in different body fluids were reduced, accompanied by a decrease in neuronal damage, activated astrocytes and microglia, as well as HMGB1-positive astrocytes and microglia. GL significantly reversed HMGB1 release into the medium induced by TNF-α stimulation in primary cultured cortical neurons. Taken together, the results indicate that GL has a strong neuroprotective effect on EAE mice by reducing HMGB1 expression and release and thus can be used to treat central nervous system inflammatory diseases, such as MS.

20.
Mol Immunol ; 101: 167-175, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29944986

RESUMEN

Liver sinusoidal endothelial cells (LSECs) are organ resident APCs capable of antigen presentation and subsequent tolerization of T cells under physiological conditions. In this study, we investigated whether LSEC pretreatment with NOD-like receptor (NLR) agonists can switch the cells from a tolerogenic to an immunogenic state and promote the development of T cell immunity. LSECs constitutively express NOD1, NOD2 and RIPK2. Stimulation of LSECs with DAP induced the activation of NF-κB and MAP kinases and upregulated the expression of chemokines (CXCL2/9, CCL2/7/8) and cytokines (IFN-γ, TNF-α and IL-2). Pretreatment of LSECs with DAP induced significantly increased IFN-γ and IL-2-production by HBV-stimulated CD8+ T cells primed by DAP-treated LSECs. Consistently, a significant reduction in the HBV DNA and HBsAg level occurred in mice receiving T cells primed by DAP-treated LSECs. MDP stimulation had no impact on LSECs or HBV-stimulated CD8+ T cells primed with MDP-treated LSECs except for the upregulation of PD-L1. DAP stimulation in vitro could promote LSEC maturation and activate HBV-specific T cell responses. These results are of particular relevance for the regulation of the local innate immune response against HBV infections.


Asunto(s)
Diferenciación Celular , Células Endoteliales/metabolismo , Inmunidad Celular , Hígado/citología , Proteína Adaptadora de Señalización NOD1/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Acetilmuramil-Alanil-Isoglutamina/farmacología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/efectos de los fármacos , Ácido Diaminopimélico/farmacología , Células Endoteliales/efectos de los fármacos , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/fisiología , Inmunidad Celular/efectos de los fármacos , Interleucina-2/biosíntesis , Ligandos , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA