Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(11): 113377, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37922310

RESUMEN

ZBP1 senses viral Z-RNAs to induce necroptotic cell death to restrain viral infection. ZBP1 is also thought to recognize host cell-derived Z-RNAs to regulate organ development and tissue inflammation in mice. However, it remains unknown how the host-derived Z-RNAs are formed and how these endogenous Z-RNAs are sensed by ZBP1. Here, we report that oxidative stress strongly induces host cell endogenous Z-RNAs, and the Z-RNAs then localize to stress granules for direct sensing by ZBP1 to trigger necroptosis. Oxidative stress triggers dramatically increase Z-RNA levels in tumor cells, and the Z-RNAs then directly trigger tumor cell necroptosis through ZBP1. Localization of the induced Z-RNAs to stress granules is essential for ZBP1 sensing. Oxidative stress-induced Z-RNAs significantly promote tumor chemotherapy via ZBP1-driven necroptosis. Thus, our study identifies oxidative stress as a critical trigger for Z-RNA formation and demonstrates how Z-RNAs are directly sensed by ZBP1 to trigger anti-tumor necroptotic cell death.


Asunto(s)
Proteínas de Unión al ARN , ARN , Ratones , Animales , ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Necroptosis , Muerte Celular/fisiología
2.
Clin Kidney J ; 14(1): 301-308, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33564432

RESUMEN

BACKGROUND: Tubular injury plays a critical role in the development of diabetic nephropathy (DN), but current DN therapies do not combat tubular injury. This study was conducted to investigate if tumor necrosis factor (TNF)-α inhibition protects against tubular injury in diabetic rats and to examine the associated mechanisms. METHODS: Kidney biopsy tissues were collected and analyzed from 12 patients with DN and 5 control subjects. Streptozotocin (STZ)-induced diabetic rats were treated with a TNF-α inhibitor for 12 weeks. Renal function, albuminuria, histological injury, renal TNF-α messenger RNA (mRNA) and the NOD- (nucleotide-binding), LRR- (domain-like receptor) and pyrin domain-containing protein 3 (NLRP3) inflammasome were assessed. RESULTS: Diabetic patients with tubulointerstitial injury (TIN) presented with higher renal tubular expression of TNF-α mRNA and the NLRP3 inflammasome (P < 0.05). TNF-α inhibition reduced albuminuria, glomerular injury and tubular injury in STZ-induced diabetic rats (P < 0.05). Importantly, TNF-α inhibition significantly reduced the NLRP3 inflammasome in tubules (P < 0.05). Moreover, TNF-α inhibition decreased expression of tubular interleukin (IL)-6 and IL-17A mRNA. CONCLUSIONS: TNF-α inhibition protects against TIN by suppressing the NLRP3 inflammasome in DN rats. Future studies may focus on the clinical protective effects of TNF-α inhibition using prospective observation.

3.
Cancer Cell ; 39(3): 423-437.e7, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33450198

RESUMEN

Lung metastasis is the major cause of breast cancer-related mortality. The neutrophil-associated inflammatory microenvironment aids tumor cells in metastatic colonization in lungs. Here, we show that tumor-secreted protease cathepsin C (CTSC) promotes breast-to-lung metastasis by regulating recruitment of neutrophils and formation of neutrophil extracellular traps (NETs). CTSC enzymatically activates neutrophil membrane-bound proteinase 3 (PR3) to facilitate interleukin-1ß (IL-1ß) processing and nuclear factor κB activation, thus upregulating IL-6 and CCL3 for neutrophil recruitment. In addition, the CTSC-PR3-IL-1ß axis induces neutrophil reactive oxygen species production and formation of NETs, which degrade thrombospondin-1 and support metastatic growth of cancer cells in the lungs. CTSC expression and secretion are associated with NET formation and lung metastasis in human breast tumors. Importantly, targeting CTSC with compound AZD7986 effectively suppresses lung metastasis of breast cancer in a mouse model. Overall, our findings reveal a mechanism of how tumor cells regulate neutrophils in metastatic niches and support CTSC-targeting approaches for cancer treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Catepsina C/metabolismo , Trampas Extracelulares/metabolismo , Neoplasias Pulmonares/metabolismo , Infiltración Neutrófila/fisiología , Neutrófilos/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neutrófilos/patología , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/fisiología
4.
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
5.
Kidney Int ; 97(6): 1219-1229, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32331702

RESUMEN

Cytokines are necessary to trigger the inflammatory response in kidney ischemia/reperfusion injury. Interleukin-17C (IL-17C), a unique member of the IL-17 family, is a cytokine produced by epithelial cells implicated in host defense and autoimmune diseases. However, little is known about the role of IL-17C in acute kidney injury. We investigated this and found that IL-17C was significantly increased in kidney biopsies of patients and mice with acute kidney injury. Exposure to hypoxia induced upregulation of IL-17C in kidney tubular epithelial cells. To further investigate the role of IL-17C, kidney ischemia/reperfusion injury was induced in mice. Inhibition of IL-17C action with a neutralizing antibody or IL-17 receptor E (IL-17RE) knockout attenuated tubular injury, kidney oxidative stress, and kidney inflammation. Mechanistically, both IL-17C neutralization and IL-17RE knockout attenuated TH17 activation and IL-17A expression in kidneys of mice with acute kidney injury. TNF-α and IL-1ß, downstream cytokines of IL-17C, were also reduced in IL-17C antibody pretreated and IL-17RE knockout mice. Additionally, IL-17C knockdown with siRNA decreased hypoxia-induced inflammation in kidney tubular cells and silencing IL-17RE abrogated the effects of IL-17C in kidney tubular cells. Thus, IL-17C may participate in the inflammatory response of acute kidney injury and inhibition of IL-17C or blockade of IL-17 RE may be a novel therapeutic strategy for the treatment of acute kidney injury.


Asunto(s)
Interleucina-17 , Daño por Reperfusión , Animales , Humanos , Isquemia , Riñón , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-17
6.
Immunology ; 159(2): 156-166, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31631335

RESUMEN

Host-microbiota interaction plays fundamental roles in the homeostasis of mucosal immunity. Dysbiosis of intestinal microbiota has been demonstrated to participate in various immune responses and many multifactorial diseases. Study of intestinal microbiota has moved beyond the consequences of dysbiosis to the causal microbiota associated with diseases. However, studies of pulmonary microbiota and its dysbiosis are still in their infancy. Improvement of culture-dependent and -independent techniques has facilitated our understanding of lung microbiota that not only exists in healthy lung tissue but also exerts great impact on immune responses under both physiological and pathological conditions. In this review, we summarize recent progresses of lung microbiota dysbiosis and its impact on the local immune system that determines the balance of tolerance and inflammation. We discuss the causal roles of pulmonary dysbiosis under disease settings, and propose that the interaction between lung microbiota and host is critical for establishing the immune homeostasis in lung.


Asunto(s)
Disbiosis , Pulmón/microbiología , Microbiota , Neumonía/microbiología , Inmunidad Adaptativa , Animales , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Pulmón/inmunología , Pulmón/metabolismo , Neumonía/inmunología , Neumonía/metabolismo
7.
Front Immunol ; 10: 1980, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31481966

RESUMEN

Myeloid derived suppressor cells (MDSC) in the liver microenvironment protects against the inflammation-induced liver injury in fulminant hepatitis (FH). However, the molecular mechanism through which MDSC is recruited into the inflamed liver remain elusive. Here we identified a protein kinase Tpl2 as a critical mediator of MDSC recruitment into liver during the pathogenesis of Propionibacterium acnes/LPS-induced FH. Loss of Tpl2 dramatically suppressed MDSC mobilization into liver, leading to exaggerated local inflammation and increased FH-induced mortality. Mechanistically, although the protective effect of Tpl2 for FH-induced mortality was dependent on the presence of MDSC, Tpl2 neither directly targeted myeloid cells nor T cells to regulate FH pathogenesis, but functioned in hepatocytes to mediate the induction of MDSC-attracting chemokine CXCL1 and CXCL2 through modulating IL-25 (also known as IL-17E) signaling. As a consequence, increased MDSC in the inflamed liver specifically restrained the local proliferation of infiltrated pathogenic CD4+ T cells, and thus protected against the inflammation-induced acute liver failure. Together, our findings established Tpl2 as a critical mediator of MDSC recruitment and highlighted the therapeutic potential of Tpl2 for the treatment of FH.


Asunto(s)
Movimiento Celular/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Necrosis Hepática Masiva/inmunología , Necrosis Hepática Masiva/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Cell Death Differ ; 26(12): 2622-2636, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30944411

RESUMEN

Necroptosis is a recently defined type of programmed cell death with the specific signaling cascade of receptor-interacting protein 1 (RIPK1) and RIPK3 complex to activate the executor MLKL. However, the pathophysiological roles of necroptosis are largely unexplored. Here, we report that fungus triggers myeloid cell necroptosis and this type of cell death contributes to host defense against the pathogen infection. Candida albicans as well as its sensor Dectin-1 activation strongly induced necroptosis in myeloid cells through the RIPK1-RIPK3-MLKL cascade. CARD9, a key adaptor in Dectin-1 signaling, was identified to bridge the RIPK1 and RIPK3 complex-mediated necroptosis pathway. RIPK1 and RIPK3 also potentiated Dectin-1-induced MLKL-independent inflammatory response. Both the MLKL-dependent and MLKL-independent pathways were required for host defense against C. albicans infection. Thus, our study demonstrates a new type of host defense system against fungal infection.


Asunto(s)
Candidiasis/metabolismo , Lectinas Tipo C/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Candida albicans , Candidiasis/patología , Candidiasis/prevención & control , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Necroptosis , Transfección
9.
Immunity ; 50(3): 692-706.e7, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824326

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a severe form of lung fibrosis with a high mortality rate. However, the etiology of IPF remains unknown. Here, we report that alterations in lung microbiota critically promote pulmonary fibrosis pathogenesis. We found that lung microbiota was dysregulated, and the dysregulated microbiota in turn induced production of interleukin-17B (IL-17B) during bleomycin-induced mouse lung fibrosis. Either lung-microbiota depletion or IL-17B deficiency ameliorated the disease progression. IL-17B cooperated with tumor necrosis factor-α to induce expression of neutrophil-recruiting genes and T helper 17 (Th17)-cell-promoting genes. Three pulmonary commensal microbes, which belong to the genera Bacteroides and Prevotella, were identified to promote fibrotic pathogenesis through IL-17R signaling. We further defined that the outer membrane vesicles (OMVs) that were derived from the identified commensal microbes induced IL-17B production through Toll-like receptor-Myd88 adaptor signaling. Together our data demonstrate that specific pulmonary symbiotic commensals can promote lung fibrosis by regulating a profibrotic inflammatory cytokine network.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/microbiología , Interleucina-17/metabolismo , Pulmón/metabolismo , Pulmón/microbiología , Microbiota/fisiología , Animales , Bacteroides/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/metabolismo , Prevotella/metabolismo , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Nat Immunol ; 20(4): 433-446, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30804553

RESUMEN

Cells use mitophagy to remove damaged or unwanted mitochondria to maintain homeostasis. Here we report that the intracellular bacterial pathogen Listeria monocytogenes exploits host mitophagy to evade killing. We found that L. monocytogenes induced mitophagy in macrophages through the virulence factor listeriolysin O (LLO). We discovered that NLRX1, the only Nod-like receptor (NLR) family member with a mitochondrial targeting sequence, contains an LC3-interacting region (LIR) and directly associated with LC3 through the LIR. NLRX1 and its LIR motif were essential for L. monocytogenes-induced mitophagy. NLRX1 deficiency and use of a mitophagy inhibitor both increased mitochondrial production of reactive oxygen species and thereby suppressed the survival of L. monocytogenes. Mechanistically, L. monocytogenes and LLO induced oligomerization of NLRX1 to promote binding of its LIR motif to LC3 for induction of mitophagy. Our study identifies NLRX1 as a novel mitophagy receptor and discovers a previously unappreciated strategy used by pathogens to hijack a host cell homeostasis system for their survival.


Asunto(s)
Listeria monocytogenes/fisiología , Proteínas Mitocondriales/fisiología , Mitofagia , Animales , Autofagia , Toxinas Bacterianas/metabolismo , Línea Celular , Femenino , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Listeriosis/microbiología , Macrófagos/microbiología , Macrófagos/ultraestructura , Masculino , Ratones , Ratones Noqueados , Viabilidad Microbiana , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo , Factores de Virulencia/metabolismo
11.
J Allergy Clin Immunol ; 143(1): 229-244.e9, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29625134

RESUMEN

BACKGROUND: IL-17 plays a pathogenic role in asthma. ST2- inflammatory group 2 innate lymphoid cells (ILC2s) driven by IL-25 can produce IL-17, whereas ST2+ natural ILC2s produce little IL-17. OBJECTIVE: We characterized ST2+IL-17+ ILC2s during lung inflammation and determined the pathogenesis and molecular regulation of ST2+IL-17+ ILC2s. METHODS: Lung inflammation was induced by papain or IL-33. IL-17 production by lung ILC2s from wild-type, Rag1-/-, Rorcgfp/gfp, and aryl hydrocarbon receptor (Ahr)-/- mice was examined by using flow cytometry. Bone marrow transfer experiments were performed to evaluate hematopoietic myeloid differentiation primary response gene-88 (MyD88) signaling in regulating IL-17 production by ILC2s. mRNA expression of IL-17 was analyzed in purified naive ILC2s treated with IL-33, leukotrienes, and inhibitors for nuclear factor of activated T cells, p38, c-Jun N-terminal kinase, or nuclear factor κ light-chain enhancer of activated B cells. The pathogenesis of IL-17+ ILC2s was determined by transferring wild-type or Il17-/- ILC2s to Rag2-/-Il2rg-/- mice, which further induced lung inflammation. Finally, expression of 106 ILC2 signature genes was compared between ST2+IL-17+ ILC2s and ST2+IL-17- ILC2s. RESULTS: Papain or IL-33 treatment boosted IL-17 production from ST2+ ILC2s (referred to by us as ILC217s) but not ST2- ILC2s. Ahr, but not retinoic acid receptor-related orphan receptor γt, facilitated the production of IL-17 by ILC217s. The hematopoietic compartment of MyD88 signaling is essential for ILC217 induction. IL-33 works in synergy with leukotrienes, which signal through nuclear factor of activated T-cell activation to promote IL-17 in ILC217s. Il17-/- ILC2s were less pathogenic in lung inflammation. ILC217s concomitantly expressed IL-5 and IL-13 but expressed little GM-CSF. CONCLUSION: During lung inflammation, IL-33 and leukotrienes synergistically induce ILC217s. ILC217s are a highly pathogenic and unexpected source for IL-17 in lung inflammation.


Asunto(s)
Inmunidad Innata , Interleucina-17/inmunología , Pulmón/inmunología , Linfocitos/inmunología , Neumonía/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Regulación de la Expresión Génica/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-17/genética , Interleucina-33/genética , Interleucina-33/inmunología , Interleucinas/genética , Interleucinas/inmunología , Leucotrienos/genética , Leucotrienos/inmunología , Pulmón/patología , Linfocitos/patología , Ratones , Ratones Noqueados , Papaína/farmacología , Neumonía/genética , Neumonía/patología , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/inmunología
12.
J Exp Med ; 215(11): 2850-2867, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30224386

RESUMEN

Th2 immune response is critical for allergic asthma pathogenesis. Molecular mechanisms for regulating Th2 immunity are still not well understood. Here we report that the ubiquitin-specific protease USP38 is crucial for Th2-mediated allergic asthma. TCR stimulation up-regulated the USP38 level, and USP38 in turn mediated the protein stabilization of JunB, a transcription factor specific for Th2 development. Consequently, USP38 was specifically required for TCR-induced production of Th2 cytokines and Th2 development both in vitro and in vivo, and USP38-deficient mice were resistant to asthma pathogenesis induced by OVA or HDM. Mechanistically, USP38 directly associated with JunB, deubiquitinated Lys-48-linked poly-ubiquitination of JunB, and consequently blocked TCR-induced JunB turnover. USP38 represents the first identified deubiquitinase specifically for Th2 immunity and the associated asthma.


Asunto(s)
Asma/inmunología , Células Th2/inmunología , Factores de Transcripción/inmunología , Proteasas Ubiquitina-Específicas/inmunología , Animales , Asma/genética , Asma/patología , Citocinas/genética , Citocinas/inmunología , Ratones , Ratones Noqueados , Poliubiquitina/genética , Poliubiquitina/inmunología , Estabilidad Proteica , Células Th2/patología , Factores de Transcripción/genética , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética , Ubiquitinación/inmunología
13.
Metabolism ; 83: 18-24, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29336982

RESUMEN

BACKGROUND AND OBJECTIVE: Interleukin 6 (IL-6) has been identified as a key mediator in inflammation, immune responses and glucose metabolism. In this study, we assessed the effects of an IL-6 receptor antibody on diabetic nephropathy in a mouse model of type 2 diabetes mellitus. METHODS: Twelve week old male db/db mice were treated with Tocilizumab (an IL-6 receptor antibody), normal IgG1 control antibody, insulin or normal saline for 12 weeks. Renal injury, inflammation and insulin resistance were assessed. RESULTS: Db/db mice treated with Tocilizumab exhibited reduced proteinuria and glomerular mesangial matrix accumulation compared to db/db + IgG controls. Additionally, Tocilizumab suppressed inflammatory response, oxidative stress and the IL-6 signaling pathway in the diabetic kidneys. It is noteworthy that blockade of IL-6 receptor blunted the activation of NLRP3 inflammasome partly through inhibition of IL-17A. Furthermore, insulin resistance assessed by glucose tolerance test, was ameliorated by Tocilizumab treatment. CONCLUSIONS: The protective effects of an IL-6 receptor blockade against diabetic renal injury may be due to decreased insulin resistance and inhibition of the inflammasome.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/prevención & control , Inflamasomas/efectos de los fármacos , Receptores de Interleucina-6/antagonistas & inhibidores , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Inflamasomas/metabolismo , Resistencia a la Insulina/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-6/inmunología
14.
Gastroenterology ; 154(3): 637-651.e7, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29031499

RESUMEN

BACKGROUND & AIMS: B cells infiltrate tumors, but little is known about how they affect tumor growth and progression. microRNA15A (MIR15A or miRNA15A) and microRNA16-1 (MIR16-1 or miRNA16-1) regulate cell proliferation, apoptosis, and drug resistance. We investigated their involvement in B-cell-mediated immune suppression by colorectal tumors. METHODS: Mice with disruptions of the gene cluster that encodes MIR15A and MIR16-1 (knockout mice), and control (C57BL/B6) mice were given azoxymethane with dextran sodium sulfate (AD) to induce formation of colorectal tumors. Mice were given anti-CD20 to delete B cells, or injections of agomir to increase MIR15A and MIR16-1. Proliferation of CD8+T cells was measured by carboxyfluorescein-succinimidyl-ester analysis. Colon tissues were collected from mice and analyzed by flow cytometry, microRNA (miRNA) sequencing, and for cytokine production. Intestinal epithelial cells (IECs) were isolated and transfected with miRNA mimics, to identify their targets. We analyzed miRNA expression patterns and quantified B cells in colorectal cancer tissue microarrays derived from 90 patients who underwent surgical resection, from July 2006 through April 2008, in Shanghai, China; expression data were compared with clinical outcomes. RESULTS: Tumors that developed in knockout mice following administration of AD were larger and contained greater numbers of B cells than tumors that grew in control mice. Most of the B cells in the tumors were positive for immunoglobulin A (IgA+). IgA+ B cells expressed high levels of immune regulatory molecules (programmed death ligand 1, interleukin 10, and transforming growth factor beta), and repressed the proliferation and activation of CD8+ T cells. Levels of MIR15A and MIR16-1 were reduced in colon tumors from mice, compared with nontumor colon tissue. Incubation of IECs with IL17A reduced expression of MIR15A and MIR16-1. Transgenic expression of MIR15A and MIR16-1 in IECs decreased activation of NF-κB and STAT1 by reducing expression of I-kappaB kinases; this resulted in reduced production of chemokine (C-X-C motif) ligands 9 and 10 and decreased chemotaxis of IgA+ B cells. Tumors in mice injected with AD and agomir grew more slowly than tumors in mice not given in agomir and contained fewer IgA+ B cells. We found a negative correlation between levels of MIR15A and MIR16-1 and numbers of IgA+B cells in human colorectal tumor tissues; high levels of MIR15A and MIR16-1 and low numbers of IgA+B cells were associated with longer survival times of patients. CONCLUSIONS: We found increased levels of MIR15A and MIR16-1 to reduce numbers of IgA+ B cells in colorectal tumor tissues and correlate with increased survival time of patients. In mice that lack MIR15A and MIR16-1, colon tumors grow more rapidly and contain increased numbers of IgA+ B cells. MIR15A and MIR16-1 appear to activate signaling pathways required for B-cell-mediated immune suppression.


Asunto(s)
Linfocitos B Reguladores/metabolismo , Quimiotaxis de Leucocito , Neoplasias Colorrectales/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Escape del Tumor , Animales , Azoximetano , Linfocitos B Reguladores/inmunología , Proliferación Celular , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/inmunología , Quimiocina CXCL9/metabolismo , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Sulfato de Dextran , Regulación Neoplásica de la Expresión Génica , Genotipo , Humanos , Quinasa I-kappa B/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , FN-kappa B/metabolismo , Fenotipo , Factor de Transcripción STAT1/metabolismo , Factores de Tiempo , Carga Tumoral
15.
Sci Rep ; 7(1): 7024, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28765647

RESUMEN

IL-17 is a pro-inflammatory cytokine implicated a variety of autoimmune diseases. We have recently reported that FGF2 cooperates with IL-17 to protect intestinal epithelium during dextran sodium sulfate (DSS)-induced colitis. Here, we report a pathogenic role of the FGF2-IL-17 cooperation in the pathogenesis of autoimmune arthritis. Combined treatment with FGF2 and IL-17 synergistically induced ERK activation as well as the production of cytokines and chemokines in human synovial intimal resident fibroblast-like synoviocytes (FLS). Furthermore, ectopic expression of FGF2 in mouse joints potentiated IL-17-induced inflammatory cytokine and chemokine production in the tissue. In the collagen-induced arthritis (CIA) model, while ectopic expression of FGF2 in vivo exacerbated tissue inflammation and disease symptom in the wild-type controls, the effect was largely blunted in Il17a -/- mice. Taken together, our study suggests that FGF2 cooperates with IL-17 to promote the pathogenesis of autoimmune arthritis by cooperating with IL-17 to induce inflammatory response.


Asunto(s)
Artritis/fisiopatología , Enfermedades Autoinmunes/fisiopatología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Inflamación/fisiopatología , Interleucina-17/metabolismo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Sinoviocitos/metabolismo
16.
Nat Commun ; 8: 15818, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28598443

RESUMEN

Regulatory T (Treg) cells are important for the maintenance of immune homoeostasis and prevention of autoimmune diseases. Epigenetic modifications have been reported to modulate autoimmunity by altering Treg cell fate. Here we show that the H3K4 methyltransferase Ash1l facilitates TGF-ß-induced Treg cell polarization in vitro and protects mice from T cell-mediated colitis in vivo. Ash1l upregulates Smad3 expression by directly targeting Smad3 promoter to increase local H3K4 trimethylation. Furthermore, we identify an lncRNA, namely lnc-Smad3, which interacts with the histone deacetylase HDAC1 and silences Smad3 transcription. After TGF-ß stimulation, activated Smad3 suppresses lnc-Smad3 transcription, thereby recovering the Smad3 promoter accessibility to Ash1l. By revealing the opposite regulatory functions of Ash1l and lnc-Smad3 in Smad3 expression, our data provide insights for the epigenetic control of Treg cell fate to potentially aid in the development of therapeutic intervention for autoimmune diseases.


Asunto(s)
Autoinmunidad , Polaridad Celular , N-Metiltransferasa de Histona-Lisina/inmunología , ARN Largo no Codificante/genética , Proteína smad3/genética , Linfocitos T Reguladores/citología , Linfocitos T/inmunología , Adulto , Anciano , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Silenciador del Gen , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/inmunología , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/inmunología , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Largo no Codificante/inmunología , Proteína smad3/inmunología , Linfocitos T/citología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factor de Crecimiento Transformador beta/inmunología
17.
Nat Commun ; 8: 15508, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28561022

RESUMEN

NOTCH1 signalling contributes to defective remyelination by impairing differentiation of oligodendrocyte progenitor cells (OPCs). Here we report that IL-17 stimulation induces NOTCH1 activation in OPCs, contributing to Th17-mediated demyelinating disease. Mechanistically, IL-17R interacts with NOTCH1 via the extracellular domain, which facilitates the cleavage of NOTHC1 intracellular domain (NICD1). IL-17-induced NOTCH1 activation results in the interaction of IL-17R adaptor Act1 with NICD1, followed by the translocation of the Act1-NICD1 complex into the nucleus. Act1-NICD1 are recruited to the promoters of several NOTCH1 target genes (including STEAP4, a metalloreductase important for inflammation and cell proliferation) that are specifically induced in the spinal cord by Th17 cells. A decoy peptide disrupting the IL-17RA-NOTCH1 interaction inhibits IL-17-induced NOTCH1 activation and attenuates Th17-mediated experimental autoimmune encephalitis (EAE). Taken together, these findings demonstrate critical crosstalk between the IL-17 and NOTCH1 pathway, regulating Th17-induced inflammatory and proliferative genes to promote demyelinating disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/metabolismo , Esclerosis Múltiple/inmunología , Células Precursoras de Oligodendrocitos/fisiología , Receptor Notch1/inmunología , Células Th17/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Astrocitos , Diferenciación Celular/inmunología , Proliferación Celular/fisiología , Técnicas de Cocultivo , Femenino , Células HEK293 , Células HeLa , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/inmunología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Unión Proteica/inmunología , Dominios Proteicos/fisiología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores de Interleucina-17/metabolismo , Remielinización/fisiología , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th17/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
18.
Nat Commun ; 8: 15402, 2017 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-28537251

RESUMEN

The connection between innate and adaptive immunity is best exemplified by antigen presentation. Although antigen-presenting cells (APCs) are required for antigen receptor-mediated T-cell activation, how T-cells feedback to APCs to sustain an antigen-specific immune response is not completely clear. Here we show that CD8+ T-cell (also called cytotoxic T lymphocytes, CTL) feedback activates the NLRP3 inflammasome in APCs in an antigen-dependent manner to promote IL-1ß maturation. Perforin from antigen-specific CTLs is required for NLRP3 inflammasome activation in APCs. Furthermore, such activation of NLRP3 inflammasome contributes to the induction of antigen-specific antitumour immunity and pathogenesis of graft-versus-host diseases. Our study reveals a positive feedback loop between antigen-specific CTLs and APC to amplify adaptive immunity.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Neoplasias/inmunología , Perforina/metabolismo , Linfocitos T Citotóxicos/inmunología , Inmunidad Adaptativa , Animales , Células de la Médula Ósea , Trasplante de Médula Ósea , Línea Celular Tumoral , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Perforina/genética , Perforina/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
EMBO Rep ; 18(4): 586-602, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28219902

RESUMEN

Interleukin 17 (IL-17) is an important inducer of tissue inflammation and is involved in numerous autoimmune diseases. However, how its signal transduction is regulated is not well understood. Here, we report that nuclear Dbf2-related kinase 1 (NDR1) functions as a positive regulator of IL-17 signal transduction and IL-17-induced inflammation. NDR1 deficiency or knockdown inhibits the IL-17-induced phosphorylation of p38, ERK1/2, and p65 and the expression of chemokines and cytokines, whereas the overexpression of NDR1 promotes IL-17-induced signaling independent of its kinase activity. Mechanistically, NDR1 interacts with TRAF3 and prevents its binding to IL-17R, which promotes the formation of an IL-17R-Act1-TRAF6 complex and downstream signaling. Consistent with this, IL-17-induced inflammation is significantly reduced in NDR1-deficient mice, and NDR1 deficiency significantly protects mice from MOG-induced experimental autoimmune encephalomyelitis (EAE) and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis likely by its inhibition of IL-17-mediated signaling pathway. NDR1 expression is increased in the colons of ulcerative colitis (UC) patients. Taken together, these findings suggest that NDR1 is involved in the development of autoimmune diseases.


Asunto(s)
Inflamación/metabolismo , Interleucina-17/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Colitis/genética , Colitis/metabolismo , Colitis/patología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Humanos , Inflamación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Ratones Noqueados , Unión Proteica , Transducción de Señal
20.
Cell Mol Immunol ; 13(4): 418-31, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27018218

RESUMEN

The mucosal immune system serves as our front-line defense against pathogens. It also tightly maintains immune tolerance to self-symbiotic bacteria, which are usually called commensals. Sensing both types of microorganisms is modulated by signalling primarily through various pattern-recognition receptors (PRRs) on barrier epithelial cells or immune cells. After sensing, proinflammatory molecules such as cytokines are released by these cells to mediate either defensive or tolerant responses. The interleukin-17 (IL-17) family members belong to a newly characterized cytokine subset that is critical for the maintenance of mucosal homeostasis. In this review, we will summarize recent progress on the diverse functions and signals of this family of cytokines at different mucosal edges.


Asunto(s)
Inmunidad Mucosa , Interleucina-17/metabolismo , Animales , Humanos , Modelos Biológicos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...