Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(12): 1373-1380, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27695001

RESUMEN

The respiratory tract is heavily populated with innate immune cells, but the mechanisms that control such cells are poorly defined. Here we found that the E3 ubiquitin ligase TRIM29 was a selective regulator of the activation of alveolar macrophages, the expression of type I interferons and the production of proinflammatory cytokines in the lungs. We found that deletion of TRIM29 enhanced macrophage production of type I interferons and protected mice from infection with influenza virus, while challenge of Trim29-/- mice with Haemophilus influenzae resulted in lethal lung inflammation due to massive production of proinflammatory cytokines by macrophages. Mechanistically, we demonstrated that TRIM29 inhibited interferon-regulatory factors and signaling via the transcription factor NF-κB by degrading the adaptor NEMO and that TRIM29 directly bound NEMO and subsequently induced its ubiquitination and proteolytic degradation. These data identify TRIM29 as a key negative regulator of alveolar macrophages and might have important clinical implications for local immunity and immunopathology.


Asunto(s)
Infecciones por Haemophilus/inmunología , Haemophilus influenzae/inmunología , Virus de la Influenza A/inmunología , Macrófagos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Sistema Respiratorio/inmunología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/microbiología , Macrófagos/virología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Proteolisis , Transducción de Señal , Factores de Transcripción/genética , Ubiquitinación
2.
Immunity ; 44(6): 1271-83, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27317259

RESUMEN

T helper 17 (Th17) cells are prominently featured in multiple autoimmune diseases, but the regulatory mechanisms that control Th17 cell responses are poorly defined. Here we found that stimulation of OX40 triggered a robust chromatin remodeling response and produced a "closed" chromatin structure at interleukin-17 (IL-17) locus to inhibit Th17 cell function. OX40 activated the NF-κB family member RelB, and RelB recruited the histone methyltransferases G9a and SETDB1 to the Il17 locus to deposit "repressive" chromatin marks at H3K9 sites, and consequently repressing IL-17 expression. Unlike its transcriptional activities, RelB acted independently of both p52 and p50 in the suppression of IL-17. In an experimental autoimmune encephalomyelitis (EAE) disease model, we found that OX40 stimulation inhibited IL-17 and reduced EAE. Conversely, RelB-deficient CD4(+) T cells showed enhanced IL-17 induction and exacerbated the disease. Our data uncover a mechanism in the control of Th17 cells that might have important clinic implications.


Asunto(s)
Ensamble y Desensamble de Cromatina , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/metabolismo , Esclerosis Múltiple/inmunología , Receptores OX40/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Interleucina-17/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores OX40/genética , Transducción de Señal , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/metabolismo
3.
Nat Immunol ; 13(10): 981-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22842344

RESUMEN

The mechanisms that regulate the T(H)9 subset of helper T cells and diseases mediated by T(H)9 cells remain poorly defined. Here we found that the costimulatory receptor OX40 was a powerful inducer of T(H)9 cells in vitro and T(H)9 cell-dependent airway inflammation in vivo. In polarizing conditions based on transforming growth factor-ß (TGF-ß), ligation of OX40 inhibited the production of induced regulatory T cells and the T(H)17 subset of helper T cells and diverted CD4(+)Foxp3(-) T cells to a T(H)9 phenotype. Mechanistically, OX40 activated the ubiquitin ligase TRAF6, which triggered induction of the kinase NIK in CD4(+) T cells and the noncanonical transcription factor NF-κB pathway; this subsequently led to the generation of T(H)9 cells. Thus, our study identifies a previously unknown mechanism for the induction of T(H)9 cells and may have important clinical implications in allergic inflammation.


Asunto(s)
Ligando OX40/metabolismo , Receptores OX40/metabolismo , Sistema Respiratorio/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos CD4/biosíntesis , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-9/biosíntesis , Interleucina-9/metabolismo , Ratones , FN-kappa B/metabolismo , Ligando OX40/inmunología , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Receptores OX40/inmunología , Transducción de Señal/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Factor 6 Asociado a Receptor de TNF/biosíntesis , Factor 6 Asociado a Receptor de TNF/metabolismo , Transactivadores/inmunología , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Quinasa de Factor Nuclear kappa B
5.
J Biomed Sci ; 29(1): 55, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-35909127

RESUMEN

BACKGROUND: Infections by viruses including severe acute respiratory syndrome coronavirus 2 could cause organ inflammations such as myocarditis, pneumonia and encephalitis. Innate immunity to viral nucleic acids mediates antiviral immunity as well as inflammatory organ injury. However, the innate immune mechanisms that control viral induced organ inflammations are unclear. METHODS: To understand the role of the E3 ligase TRIM18 in controlling viral myocarditis and organ inflammation, wild-type and Trim18 knockout mice were infected with coxsackievirus B3 for inducing viral myocarditis, influenza A virus PR8 strain and human adenovirus for inducing viral pneumonia, and herpes simplex virus type I for inducing herpes simplex encephalitis. Mice survivals were monitored, and heart, lung and brain were harvested for histology and immunohistochemistry analysis. Real-time PCR, co-immunoprecipitation, immunoblot, enzyme-linked immunosorbent assay, luciferase assay, flow cytometry, over-expression and knockdown techniques were used to understand the molecular mechanisms of TRIM18 in regulating type I interferon (IFN) production after virus infection in this study. RESULTS: We find that knockdown or deletion of TRIM18 in human or mouse macrophages enhances production of type I IFN in response to double strand (ds) RNA and dsDNA or RNA and DNA virus infection. Importantly, deletion of TRIM18 protects mice from viral myocarditis, viral pneumonia, and herpes simplex encephalitis due to enhanced type I IFN production in vivo. Mechanistically, we show that TRIM18 recruits protein phosphatase 1A (PPM1A) to dephosphorylate TANK binding kinase 1 (TBK1), which inactivates TBK1 to block TBK1 from interacting with its upstream adaptors, mitochondrial antiviral signaling (MAVS) and stimulator of interferon genes (STING), thereby dampening antiviral signaling during viral infections. Moreover, TRIM18 stabilizes PPM1A by inducing K63-linked ubiquitination of PPM1A. CONCLUSIONS: Our results indicate that TRIM18 serves as a negative regulator of viral myocarditis, lung inflammation and brain damage by downregulating innate immune activation induced by both RNA and DNA viruses. Our data reveal that TRIM18 is a critical regulator of innate immunity in viral induced diseases, thereby identifying a potential therapeutic target for treatment.


Asunto(s)
Encefalitis por Herpes Simple , Miocarditis , Ubiquitina-Proteína Ligasas , Virosis , Animales , Antivirales , Humanos , Inmunidad Innata , Inflamación/genética , Ratones , Miocarditis/genética , Miocarditis/virología , Proteína Fosfatasa 2C , ARN , Ubiquitina-Proteína Ligasas/genética
6.
J Immunol ; 201(1): 183-192, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769269

RESUMEN

The innate immunity is critically important in protection against virus infections, and in the case of RNA viral infections, the signaling mechanisms that initiate robust protective innate immunity without triggering autoimmune inflammation remain incompletely defined. In this study, we found the E3 ligase TRIM29 was specifically expressed in poly I:C-stimulated human myeloid dendritic cells. The induced TRIM29 played a negative role in type I IFN production in response to poly I:C or dsRNA virus reovirus infection. Importantly, the challenge of wild-type mice with reovirus led to lethal infection. In contrast, deletion of TRIM29 protected the mice from this developing lethality. Additionally, TRIM29-/- mice have lower titers of reovirus in the heart, intestine, spleen, liver, and brain because of elevated production of type I IFN. Mechanistically, TRIM29 was shown to interact with MAVS and subsequently induce its K11-linked ubiquitination and degradation. Taken together, TRIM29 regulates negatively the host innate immune response to RNA virus, which could be employed by RNA viruses for viral pathogenesis.


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/biosíntesis , Infecciones por Reoviridae/inmunología , Reoviridae/inmunología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Poli I-C , Factores de Transcripción/genética , Ubiquitinación
7.
J Immunol ; 199(7): 2356-2365, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28842469

RESUMEN

Maintaining immune tolerance requires the production of Foxp3-expressing regulatory T (Treg) cells in the thymus. Activation of NF-κB transcription factors is critically required for Treg cell development, partly via initiating Foxp3 expression. NF-κB activation is controlled by a negative feedback regulation through the ubiquitin editing enzyme A20, which reduces proinflammatory signaling in myeloid cells and B cells. In naive CD4+ T cells, A20 prevents kinase RIPK3-dependent necroptosis. Using mice deficient for A20 in T lineage cells, we show that thymic and peripheral Treg cell compartments are quantitatively enlarged because of a cell-intrinsic developmental advantage of A20-deficient thymic Treg differentiation. A20-deficient thymic Treg cells exhibit reduced dependence on IL-2 but unchanged rates of proliferation and apoptosis. Activation of the NF-κB transcription factor RelA was enhanced, whereas nuclear translocation of c-Rel was decreased in A20-deficient thymic Treg cells. Furthermore, we found that the increase in Treg cells in T cell-specific A20-deficient mice was already observed in CD4+ single-positive CD25+ GITR+ Foxp3- thymic Treg cell progenitors. Treg cell precursors expressed high levels of the tumor necrosis factor receptor superfamily molecule GITR, whose stimulation is closely linked to thymic Treg cell development. A20-deficient Treg cells efficiently suppressed effector T cell-mediated graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, suggesting normal suppressive function. Holding thymic production of natural Treg cells in check, A20 thus integrates Treg cell activity and increased effector T cell survival into an efficient CD4+ T cell response.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T Reguladores/fisiología , Timo/citología , Timo/fisiología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Animales , Apoptosis , Diferenciación Celular , Citometría de Flujo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Enfermedad Injerto contra Huésped/prevención & control , Interleucina-2/inmunología , Activación de Linfocitos , Ratones , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/genética , Transducción de Señal , Trasplante de Células Madre , Timo/inmunología , Factor de Transcripción ReIA/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia
8.
Am J Transplant ; 18(3): 737-744, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29127685

RESUMEN

Ischemia and reperfusion injury (IRI) is an inevitable event in conventional organ transplant procedure and is associated with significant mortality and morbidity post-transplantation. We hypothesize that IRI is avoidable if the blood supply for the organ is not stopped, thus resulting in optimal transplant outcomes. Here we described the first case of a novel procedure called ischemia-free organ transplantation (IFOT) for patients with end-stage liver disease. The liver graft with severe macrovesicular steatosis was donated from a 25-year-old man. The recipient was a 51-year-old man with decompensated liver cirrhosis and hepatocellular carcinoma. The graft was procured, preserved, and implanted under continuous normothermic machine perfusion. The recipient did not suffer post-reperfusion syndrome or vasoplegia after revascularization of the allograft. The liver function test and histological study revealed minimal hepatocyte, biliary epithelium and vascular endothelium injury during preservation and post-transplantation. The inflammatory cytokine levels were much lower in IFOT than those in conventional procedure. Key pathways involved in IRI were not activated after allograft revascularization. No rejection, or vascular or biliary complications occurred. The patient was discharged on day 18 post-transplantation. This marks the first case of IFOT in humans, offering opportunities to optimize transplant outcomes and maximize donor organ utilization.


Asunto(s)
Carcinoma Hepatocelular/cirugía , Isquemia , Cirrosis Hepática/cirugía , Trasplante de Hígado/métodos , Preservación de Órganos , Daño por Reperfusión/prevención & control , Obtención de Tejidos y Órganos/métodos , Adulto , Humanos , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Perfusión , Pronóstico , Donantes de Tejidos/provisión & distribución
9.
Vitam Horm ; 123: 231-247, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37717986

RESUMEN

Liver X receptors α and ß are members of the nuclear receptor family, which comprise a flexible N-terminal domain, a DNA binding domain, a hinge linker, and a ligand binding domain. Liver X receptors are important regulators of cholesterol and lipid homeostasis by controlling the transcription of numerous genes. Key to their transcriptional role is synergetic interaction among the domains. DNA binding domain binds on DNA; ligand binding domain is a crucial switch to control the transcription activity through conformational change caused by ligand binding. The Liver X receptors form heterodimers with retinoid X receptor and then the liganded heterodimer may recruit other necessary transcription components to form an active transcription complex.


Asunto(s)
Receptores X del Hígado , Humanos , Ligandos , Dominios Proteicos
10.
J Exp Med ; 203(8): 1851-8, 2006 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-16864660

RESUMEN

Natural killer (NK) cells are programmed to kill target cells without prior antigen priming. Because of their potent cytolytic activities, NK cells are one of the key cell types involved in dismantling allografts. However, in certain transplant models, NK cells also express potent immunoregulatory properties that promote tolerance induction. The precise mechanism for such striking dichotomy remains unknown. In the present study, we showed in a skin transplant model that the skin allografts contain a subset of antigen-presenting cells (APCs) that can home to the recipient mice. We also showed that such graft-derived APCs are usually destroyed by the host NK cells. But in the absence of NK cells, donor APCs can survive and then migrate to the host lymphoid and extralymphoid sites where they directly stimulate the activation of alloreactive T cells. T cells activated in the absence of NK cells are more resistant to costimulatory blockade treatment, and under such conditions stable skin allograft survival is difficult to achieve. Our study identified a novel role for NK cells in regulating T cell priming in transplant models, and may have important clinical implications in tolerance induction.


Asunto(s)
Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Donantes de Tejidos , Tolerancia al Trasplante/inmunología , Traslado Adoptivo , Animales , Muerte Celular , Supervivencia de Injerto/inmunología , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piel/citología , Linfocitos T/citología , Linfocitos T/inmunología , Trasplante Homólogo/inmunología
11.
Nat Commun ; 12(1): 2681, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976210

RESUMEN

Innate immune cells are critical in protective immunity against viral infections, involved in sensing foreign viral nucleic acids. Here we report that the poly(ADP-ribose) polymerase 9 (PARP9), a member of PARP family, serves as a non-canonical sensor for RNA virus to initiate and amplify type I interferon (IFN) production. We find knockdown or deletion of PARP9 in human or mouse dendritic cells and macrophages inhibits type I IFN production in response to double strand RNA stimulation or RNA virus infection. Furthermore, mice deficient for PARP9 show enhanced susceptibility to infections with RNA viruses because of the impaired type I IFN production. Mechanistically, we show that PARP9 recognizes and binds viral RNA, with resultant recruitment and activation of the phosphoinositide 3-kinase (PI3K) and AKT3 pathway, independent of mitochondrial antiviral-signaling (MAVS). PI3K/AKT3 then activates the IRF3 and IRF7 by phosphorylating IRF3 at Ser385 and IRF7 at Ser437/438 mediating type I IFN production. Together, we reveal a critical role for PARP9 as a non-canonical RNA sensor that depends on the PI3K/AKT3 pathway to produce type I IFN. These findings may have important clinical implications in controlling viral infections and viral-induced diseases by targeting PARP9.


Asunto(s)
Células Dendríticas/enzimología , Proteínas de Neoplasias/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Infecciones por Virus ARN/enzimología , ARN Viral/metabolismo , Animales , Chlorocebus aethiops , Células Dendríticas/virología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Poli(ADP-Ribosa) Polimerasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Infecciones por Virus ARN/virología , Virus ARN/genética , Virus ARN/fisiología , Transducción de Señal , Células THP-1 , Células Vero
14.
J Immunol ; 181(5): 3193-201, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18713990

RESUMEN

OX40 is a member of the TNFR superfamily and has potent T cell costimulatory activities. OX40 also inhibits the induction of Foxp3(+) regulatory T cells (Tregs) from T effector cells, but the precise mechanism of such inhibition remains unknown. In the present study, we found that CD4(+) T effector cells from OX40 ligand-transgenic (OX40Ltg) mice are highly resistant to TGF-beta mediated induction of Foxp3(+) Tregs, whereas wild-type B6 and OX40 knockout CD4(+) T effector cells can be readily converted to Foxp3(+) T cells. We also found that CD4(+) T effector cells from OX40Ltg mice are heterogeneous and contain a large population of CD44(high)CD62L(-) memory T cells. Analysis of purified OX40Ltg naive and memory CD4(+) T effector cells showed that memory CD4(+) T cells not only resist the induction of Foxp3(+) T cells but also actively suppress the conversion of naive CD4(+) T effector cells to Foxp3(+) Tregs. This suppression is mediated by the production of IFN-gamma by memory T cells but not by cell-cell contact and also involves the induction of T-bet. Importantly, memory CD4(+) T cells have a broad impact on the induction of Foxp3(+) Tregs regardless of their origins and Ag specificities. Our data suggest that one of the mechanisms by which OX40 inhibits the induction of Foxp3(+) Tregs is by inducing memory T cells in vivo. This finding may have important clinical implications in tolerance induction to transplanted tissues.


Asunto(s)
Proliferación Celular , Memoria Inmunológica , Ligando OX40/metabolismo , Receptores OX40/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores , Animales , Linfocitos T CD4-Positivos , Factores de Transcripción Forkhead , Interferón gamma/metabolismo , Ratones , Subgrupos de Linfocitos T/metabolismo
15.
Transplantation ; 85(11): 1659-62, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18551075

RESUMEN

The role of OX40 in the islet allograft tolerance, especially in the absence of CD154 costimulation, remains poorly defined. In the present study, we used CD154 deficient mice to critically examine the role of OX40 in the activation of T effector cells and Foxp3+ Tregs and the effect of blocking OX40 on the induction of islet allograft tolerance. We found that blocking OX40 costimulation in CD154 deficient mice induced donor specific tolerance but stimulating OX40 resulted in prompt islet allograft rejection. We also found that OX40 differentially regulates T effector cells and Foxp3+ Tregs, OX40 signaling mediates proliferation of CD154 deficient T effector cells but blocks the induction and suppressor functions of Foxp3+ Tregs. Our data suggest that the role of OX40 in the induction of islet allograft tolerance involves modifying not only the T effector cells but also the Foxp3+ Tregs in CD154 deficient mice.


Asunto(s)
Ligando de CD40/deficiencia , Factores de Transcripción Forkhead/metabolismo , Rechazo de Injerto/metabolismo , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Glicoproteínas de Membrana/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Rechazo de Injerto/patología , Rechazo de Injerto/prevención & control , Trasplante de Islotes Pancreáticos/patología , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ligando OX40 , Trasplante Homólogo , Inhibidores del Factor de Necrosis Tumoral , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral
16.
Curr Opin Organ Transplant ; 13(1): 26-30, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18660703

RESUMEN

PURPOSE OF REVIEW: In the past few years, much effort has been focused on the identification of new pathways, new mechanisms, and new therapeutic targets in transplant models. Understanding of T-cell costimulatory molecules remains one of the highly contested areas of research. In this review, we will focus specifically on OX40, and summarize the latest developments on the role of OX40 in transplant models. RECENT FINDINGS: OX40 regulates multiple aspects of the T-cell response; it delivers a potent costimulatory signal to T effector cells and plays a key role in their survival and proliferation. OX40 also supports the transition of activated T effector cells to memory T cells. Importantly, OX40 signaling inhibits the suppressor functions of forkhead box P3 T regulatory cells and also blocks the induction of new forkhead box P3 T regulatory cells from activated T effector cells. These new findings may have major clinical implications in the induction of transplant tolerance. SUMMARY: The current belief is that tolerance to organ transplants involves the apoptotic deletion of T effector cells and the expansion of graft-protective T regulatory cells. Given our recent understanding of OX40, we believe that targeting the OX40 costimulation is therapeutically important in the induction of transplant tolerance.


Asunto(s)
Receptores OX40/inmunología , Linfocitos T/inmunología , Inmunología del Trasplante , Tolerancia al Trasplante , Animales , Activación de Linfocitos
17.
J Exp Med ; 215(2): 559-574, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29339447

RESUMEN

Th9 cells are prominently featured in allergic lung inflammation, but the mechanism that regulates IL-9 induction in T helper cells remains poorly defined. Here we demonstrate that formation of super-enhancers (SEs) is critical in robust induction of IL-9 and that assembly of the Il9 SEs in Th cells requires OX40-triggered chromatin acetylation. Mechanistically, we found that OX40 costimulation induces RelB expression, which recruits the histone acetyltransferase p300 to the Il9 locus to catalyze H3K27 acetylation. This allows binding of the SE factor Brd4 to organize assembly of the SE complex, which in turn drives robust IL-9 expression and Th9 cell induction. Thus, Th9 cells are strongly induced upon OX40 stimulation, and disruption of SEs abolished Th9 cell induction in vitro and inhibited Th9 cell-mediated allergic airway inflammation in vivo. Together, our data suggest that formation of SEs is essential in IL-9 expression and Th9 cell induction. These findings may have important clinical implications.


Asunto(s)
Inflamación/inmunología , Interleucina-9/biosíntesis , Interleucina-9/genética , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Acetilación , Animales , Asma/etiología , Asma/inmunología , Asma/metabolismo , Elementos de Facilitación Genéticos , Código de Histonas , Humanos , Inflamación/etiología , Inflamación/metabolismo , Ratones , Ratones Transgénicos , Familia de Multigenes , Neumonía/etiología , Neumonía/inmunología , Neumonía/metabolismo , Receptores OX40/metabolismo , Transducción de Señal , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Factor de Transcripción ReIB/metabolismo
18.
Artículo en Zh | WPRIM | ID: wpr-971071

RESUMEN

OBJECTIVES@#To investigate the clinical features of juvenile myelomonocytic leukemia (JMML) and their association with prognosis.@*METHODS@#Clinical and prognosis data were collected from the children with JMML who were admitted from January 2008 to December 2016, and the influencing factors for prognosis were analyzed.@*RESULTS@#A total of 63 children with JMML were included, with a median age of onset of 25 months and a male/female ratio of 3.2∶1. JMML genetic testing was performed for 54 children, and PTPN11 mutation was the most common mutation and was observed in 23 children (43%), among whom 19 had PTPN11 mutation alone and 4 had compound PTPN11 mutation, followed by NRAS mutation observed in 14 children (26%), among whom 12 had NRAS mutation alone and 2 had compound NRAS mutation. The 5-year overall survival (OS) rate was only 22%±10% in these children with JMML. Of the 63 children, 13 (21%) underwent hematopoietic stem cell transplantation (HSCT). The HSCT group had a significantly higher 5-year OS rate than the non-HSCT group (46%±14% vs 29%±7%, P<0.05). There was no significant difference in the 5-year OS rate between the children without PTPN11 gene mutation and those with PTPN11 gene mutation (30%±14% vs 27%±10%, P>0.05). The Cox proportional-hazards regression model analysis showed that platelet count <40×109/L at diagnosis was an influencing factor for 5-year OS rate in children with JMML (P<0.05).@*CONCLUSIONS@#The PTPN11 gene was the most common mutant gene in JMML. Platelet count at diagnosis is associated with the prognosis in children with JMML. HSCT can improve the prognosis of children with JMML.


Asunto(s)
Niño , Humanos , Masculino , Femenino , Preescolar , Leucemia Mielomonocítica Juvenil/terapia , Pronóstico , Pruebas Genéticas , Mutación , Trasplante de Células Madre Hematopoyéticas
19.
Chinese Journal of Pediatrics ; (12): 889-895, 2023.
Artículo en Zh | WPRIM | ID: wpr-1013193

RESUMEN

Objective: To analyze the clinical and molecular diagnostic status of Fanconi anemia (FA) in China. Methods: The General situation, clinical manifestations and chromosome breakage test and genetic test results of 107 pediatric FA cases registered in the Chinese Blood and Marrow Transplantation Registry Group (CBMTRG) and the Chinese Children Blood and Marrow Transplantation Registry Group (CCBMTRG) from August 2009 to January 2022 were analyzed retrospectively. Children with FANCA gene variants were divided into mild and severe groups based on the type of variant, and Wilcoxon-test was used to compare the phenotypic differences between groups. Results: Of the 176 registered FA patients, 69 (39.2%) cases were excluded due to lack of definitive genetic diagnosis results, and the remaining 107 children from 15 hospitals were included in the study, including 70 males and 37 females. The age at transplantation treatment were 6 (4, 9) years. The enrolled children were involved in 10 pathogenic genes, including 89 cases of FANCA gene, 7 cases of FANCG gene, 3 cases of FANCB gene, 2 cases of FANCE gene and 1 case each of FANCC, FANCD1, FANCD2, FANCF, FANCJ, and FANCN gene. Compound heterozygous or homozygous of loss-of-function variants account for 69.2% (72/104). Loss-of-function variants account for 79.2% (141/178) in FANCA gene variants, and 20.8% (37/178) were large exon deletions. Fifty-five children (51.4%) had chromosome breakage test records, with a positive rate of 81.8% (45/55). There were 172 congenital malformations in 80 children.Café-au-Lait spots (16.3%, 28/172), thumb deformities (16.3%,28/172), polydactyly (13.9%, 24/172), and short stature (12.2%, 21/172) were the most common congenital malformations in Chinese children with FA. No significant difference was found in the number of congenital malformations between children with severe (50 cases) and mild FANCA variants (26 cases) (Z=-1.33, P=0.185). Conclusions: FANCA gene is the main pathogenic gene in children with FA, where the detection of its exon deletion should be strengthened clinically. There were no phenotypic differences among children with different types of FANCA variants. Chromosome break test is helpful to determine the pathogenicity of variants, but its accuracy needs to be improved.


Asunto(s)
Masculino , Femenino , Humanos , Niño , Anemia de Fanconi/genética , Rotura Cromosómica , Estudios Retrospectivos , Exones , China/epidemiología
20.
Arch Immunol Ther Exp (Warsz) ; 55(5): 309-14, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18219761

RESUMEN

Memory T cells are an important cell type in the immune system and are vital to protective immunity against invading pathogens. However, a significant fraction of memory T cells is found to be alloreactive in transplant models, i.e. they can readily attack and dismantle allografts in transplant models. As memory T cells are not as easily amenable as naive T cells, memory T cells constitute a potent barrier to the induction of transplant tolerance. The key issues concerning memory T cells in transplantation are related to the tolerability of alloreactive memory T cells and the effects of commonly used immunosuppressive drugs on the memory response in transplant recipients. The real challenge in the future is to selectively tolerize alloreactive memory T cells but spare those involved in protective immunity following organ transplantation. This review will discuss recent advances in our understating of memory T cells in transplant models, with specific emphasis on the problems and challenges in targeting memory T cells in the induction of transplant tolerance.


Asunto(s)
Memoria Inmunológica/inmunología , Subgrupos de Linfocitos T/inmunología , Inmunología del Trasplante , Tolerancia al Trasplante/inmunología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA