Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
2.
Cell Rep ; 38(8): 110420, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35196494

RESUMEN

Dendritic cells (DCs) induce peripheral T cell tolerance, but cell-intrinsic signaling cascades governing their stable tolerogenesis remain poorly defined. Janus Kinase 1 (JAK1) transduces cytokine-receptor signaling, and JAK inhibitors (Jakinibs), including JAK1-specific filgotinib, break inflammatory cycles in autoimmunity. Here, we report in heterogeneous DC populations of multiple secondary lymphoid organs that JAK1 promotes peripheral T cell tolerance during experimental autoimmune encephalomyelitis (EAE). Mice harboring DC-specific JAK1 deletion exhibit elevated peripheral CD4+ T cell expansion, less regulatory T cells (Tregs), and worse EAE outcomes, whereas adoptive DC transfer ameliorates EAE pathogenesis by inducing peripheral Tregs, programmed cell death ligand 1 (PD-L1) dependently. This tolerogenic program is substantially reduced upon the transfer of JAK1-deficient DCs. DC-intrinsic IFN-γ-JAK1-STAT1 signaling induces PD-L1, which is required for DCs to convert CD4+ T cells into Tregs in vitro and attenuated upon JAK1 deficiency and filgotinib treatment. Thus, DC-intrinsic JAK1 promotes peripheral tolerance, suggesting potential unwarranted DC-mediated effects of Jakinibs in autoimmune diseases.


Asunto(s)
Antígeno B7-H1 , Encefalomielitis Autoinmune Experimental , Janus Quinasa 1 , Linfocitos T Reguladores , Animales , Autoinmunidad , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Células Dendríticas/metabolismo , Tolerancia Inmunológica , Janus Quinasa 1/inmunología , Janus Quinasa 1/metabolismo , Ratones , Tolerancia Periférica
3.
J Clin Invest ; 131(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34730109

RESUMEN

Despite the curative potential of hematopoietic stem cell transplantation (HSCT), conditioning-associated toxicities preclude broader clinical application. Antibody-drug conjugates (ADCs) provide an attractive approach to HSCT conditioning that minimizes toxicity while retaining efficacy. Initial studies of ADC conditioning have largely focused on syngeneic HSCT. However, to treat acute leukemias or induce tolerance for solid organ transplantation, this approach must be expanded to allogeneic HSCT (allo-HSCT). Using murine allo-HSCT models, we show that pharmacologic Janus kinase 1/2 (JAK1/2) inhibition combined with CD45- or cKit-targeted ADCs enables robust multilineage alloengraftment. Strikingly, myeloid lineage donor chimerism exceeding 99% was achievable in fully MHC-mismatched HSCT using this approach. Mechanistic studies using the JAK1/2 inhibitor baricitinib revealed marked impairment of T and NK cell survival, proliferation, and effector function. NK cells were exquisitely sensitive to JAK1/2 inhibition due to interference with IL-15 signaling. Unlike irradiated mice, ADC-conditioned mice did not develop pathogenic graft-versus-host alloreactivity when challenged with mismatched T cells. Finally, the combination of ADCs and baricitinib balanced graft-versus-host disease and graft-versus-leukemia responses in delayed donor lymphocyte infusion models. Our allo-HSCT conditioning strategy exemplifies the promise of immunotherapy to improve the safety of HSCT for treating hematologic diseases.


Asunto(s)
Azetidinas/farmacología , Trasplante de Células Madre Hematopoyéticas , Inmunoconjugados/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de las Cinasas Janus/farmacología , Purinas/farmacología , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Aloinjertos , Animales , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Efecto Injerto vs Leucemia/efectos de los fármacos , Efecto Injerto vs Leucemia/genética , Efecto Injerto vs Leucemia/inmunología , Interleucina-15/genética , Interleucina-15/inmunología , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología
4.
Front Immunol ; 12: 704526, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34497607

RESUMEN

Objective: Baricitinib, a selective inhibitor for janus kinase (JAK) 1 and JAK2, is approved for use in rheumatoid arthritis. Systemic lupus erythematosus (SLE) is recently regarded as a potential candidate targeted by JAK inhibitors because of the relationship between its pathogenesis and JAK/signal transducer and activator of transcription (STAT) pathway-mediated cytokines such as type I interferons. The objective of this study was to determine whether baricitinib could effectively ameliorate SLE using a murine model. Methods: To investigate effects of baricitinib on various autoimmune features, especially renal involvements in SLE, eight-week-old MRL/Mp-Faslpr (MRL/lpr) mice were used as a lupus-prone animal model and treated with baricitinib for eight weeks. Immortalized podocytes and primary podocytes and B cells isolated from C57BL/6 mice were used to determine the in vitro efficacy of baricitinib. Results: Baricitinib remarkably suppressed lupus-like phenotypes of MRL/lpr mice, such as splenomegaly, lymphadenopathy, proteinuria, and systemic autoimmunity including circulating autoantibodies and pro-inflammatory cytokines. It also modulated immune cell populations and effectively ameliorated renal inflammation, leading to the recovery of the expression of structural proteins in podocytes. According to in vitro experiments, baricitinib treatment could mitigate B cell differentiation and restore disrupted cytoskeletal structures of podocytes under inflammatory stimulation by blocking the JAK/STAT pathway. Conclusions: The present study demonstrated that baricitinib could effectively attenuate autoimmune features including renal inflammation of lupus-prone mice by suppressing aberrant B cell activation and podocyte abnormalities. Thus, baricitinib as a selective JAK inhibitor could be a promising therapeutic candidate in the treatment of SLE.


Asunto(s)
Azetidinas/farmacología , Lupus Eritematoso Sistémico , Podocitos , Purinas/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/inmunología , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos MRL lpr , Podocitos/inmunología , Podocitos/patología , Transducción de Señal/efectos de los fármacos
5.
Biochem Pharmacol ; 192: 114690, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34274356

RESUMEN

BACKGROUND: Eosinophilic asthma is increasingly recognized as one of the most severe and difficult-to-treat asthma subtypes. The JAK/STAT pathway is the principal signaling mechanism for a variety of cytokines and growth factors involved in asthma. However, the direct effect of JAK inhibitors on eosinophil effector function has not been addressed thus far. OBJECTIVE: Here we compared the effects of the JAK1/2 inhibitor baricitinib and the JAK3 inhibitor tofacitinib on eosinophil effector function in vitro and in vivo. METHODS: Differentiation of murine bone marrow-derived eosinophils. Migratory responsiveness, respiratory burst, phagocytosis and apoptosis of human peripheral blood eosinophils were assessed in vitro. In vivo effects were investigated in a mouse model of acute house dust mite-induced airway inflammation in BALB/c mice. RESULTS: Baricitinib more potently induced apoptosis and inhibited eosinophil chemotaxis and respiratory burst, while baricitinib and tofacitinib similarly affected eosinophil differentiation and phagocytosis. Of the JAK inhibitors, oral application of baricitinib more potently prevented lung eosinophilia in mice following allergen challenge. However, both JAK inhibitors neither affected airway resistance nor compliance. CONCLUSION: Our data suggest that the JAK1/2 inhibitor baricitinib is even more potent than the JAK3 inhibitor tofacitinib in suppressing eosinophil effector function. Thus, targeting the JAK1/2 pathway represents a promising therapeutic strategy for eosinophilic inflammation as observed in severe eosinophilic asthma.


Asunto(s)
Azetidinas/uso terapéutico , Eosinofilia/tratamiento farmacológico , Eosinófilos/efectos de los fármacos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de las Cinasas Janus/uso terapéutico , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Adulto , Animales , Azetidinas/farmacología , Células Cultivadas , Eosinofilia/inducido químicamente , Eosinofilia/inmunología , Eosinófilos/fisiología , Femenino , Humanos , Janus Quinasa 1/inmunología , Janus Quinasa 2/inmunología , Inhibidores de las Cinasas Janus/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Purinas/farmacología , Pirazoles/farmacología , Pyroglyphidae/inmunología , Sulfonamidas/farmacología , Adulto Joven
7.
Rheumatology (Oxford) ; 60(Suppl 2): ii3-ii10, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33950229

RESUMEN

Several cytokines involved in inflammatory pathologies signal via the Janus kinase-signal transducer and activator of transcription pathway. Four JAKs are known: JAK1, JAK2, JAK3 and TYK2. The specific activation of JAKs and STATs determines the biological effects of each cytokine. JAK1 is involved in the signalling of 'γc' receptor cytokines (IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21), pro-inflammatory cytokines including IL-6, as well as IFN. The critical position of JAK1 downstream of these cytokines suggests that JAK1-selective inhibitors are comparable to non-selective ones, without the unwanted consequences of JAK2- or JAK3-blockade. JAK inhibition has led to a better understanding of the biology of synovial inflammation and bone homeostasis. Moreover, the efficacy of non-selective JAK inhibitors and novel JAK1-selective drugs in RA supports a role for JAK1 in its pathogenesis. JAK1-selective drugs are also showing promise in axial spondyloarthritis, suggesting that they may target additional regulatory pathways that impact cytokines such as TNF and IL-17A, which do not use JAKs. Additionally, evidence now supports a JAK1 predominance in the signalling of IL-6 and oncostatin M, and indirectly, of TNF in synovial fibroblasts, macrophages and endothelial cells. Notably, bone homeostasis is also dependent on cytokines relying on JAK1 signalling to promote receptor activator of NF-κB ligand expression in osteoblasts and T cells, contributing to osteoclastogenesis. Here, the contribution of JAK1 over other kinases is unclear. While beneficial effects of JAK inhibitors on bone erosion are supported by preclinical and clinical data, effects on new bone formation in axial spondyloarthritis requires additional study.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Citocinas/inmunología , Janus Quinasa 1/inmunología , Inhibidores de las Cinasas Janus/uso terapéutico , Espondiloartropatías/tratamiento farmacológico , Artritis Reumatoide/inmunología , Resorción Ósea/inmunología , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2 , Janus Quinasa 3 , Osteogénesis/inmunología , Espondiloartropatías/inmunología , Sinovitis/inmunología , TYK2 Quinasa
8.
J Microbiol Immunol Infect ; 54(3): 501-513, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32033858

RESUMEN

BACKGROUND: The regulation of the balance between inflammatory and anti-inflammatory events during the treatment of pulmonary infection is very important. Soluble Schistosoma egg antigens (SEA) can effectively inhibit the expression of cytokines during hepatic acute inflammation. However, the mechanisms by which these proteins suppress the inflammatory responses in lung cells remain unclear. The purpose of this study was to investigate the ability of SEA to inhibit pulmonary inflammation. METHODS: The effects of SEA were investigated in LPS-treated lung IMR-90 cells. The involvement of the JAK/STAT-1 signaling pathway in these effects was evaluated by employing CBA assays, quantitative polymerase chain reaction, and western blotting experiments. RESULTS: Pretreatment of IMR-90 cells with appropriate concentrations of SEA protected cells against the cytotoxic effects of LPS-induced inflammation in a time-dependent manner. SEA pretreatment significantly attenuated the LPS-induced activation of the JAK/STAT1 signaling pathway, including the upregulation of JAK1/2 and STAT1, as well as the production of inflammatory cytokines. The level of phosphorylated STAT1 gradually declined in response to increasing concentrations of SEA. Based on these findings, we hypothesize that SEA-induced anti-inflammatory effects initiate with the downregulation of the IFN-γ-JAK-STAT1 signaling pathway, resulting in the attenuation of LPS-induced inflammation in IMR-90 cells. CONCLUSION: Our study is the first to demonstrate the anti-inflammatory activity of SEA in an in vitro model of pulmonary inflammation, involving the modulation of JAK/STAT1 signaling. We propose SEA as potential therapeutic or preventive agents for the selective suppression of STAT1 and the control of inflammatory response in lung IMR-90 cells.


Asunto(s)
Antígenos Helmínticos/farmacología , Fibroblastos/efectos de los fármacos , Inflamación/prevención & control , Janus Quinasa 1/metabolismo , Lipopolisacáridos/farmacología , Óvulo/química , Factor de Transcripción STAT1/metabolismo , Schistosoma/inmunología , Transducción de Señal , Animales , Línea Celular , Humanos , Janus Quinasa 1/inmunología , Lipopolisacáridos/metabolismo , Factor de Transcripción STAT1/inmunología , Schistosoma/química
9.
Aging (Albany NY) ; 13(2): 2073-2088, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33323549

RESUMEN

Janus kinase 1 (JAK1) is a member of the JAK family, which plays an essential and non-redundant role in tumorigenesis. However, the potential role of JAK1 in immune infiltration and prognosis of lung adenocarcinoma (LUAD) remains unclear. The mRNA expression and methylation level of JAK1 in LUAD were examined using the Oncomine and The Cancer Genome Atlas (TCGA) databases, respectively. The correlations between JAK1 expression and its methylation level and clinicopathological parameters were analyzed. The Kaplan-Meier plotter database was used to evaluate the prognostic value of JAK1 in LUAD. The signaling pathways associated with JAK1 expression were identified by performing a GSEA. The CIBERSORT and TIMER databases were used to analyze the correlations between JAK1 and tumor-infiltrating immune cells. In addition, the JAK1 expression and proportion of immune cells in LUAD cell lines were analyzed. The JAK1 expression was remarkably decreased in patients with LUAD and significantly correlated with the clinical features of patients with LUAD. The JAK1 methylation level was increased and negatively correlated with its mRNA expression. A decrease in JAK1 expression was correlated with poor prognosis. The results of GSEA showed that cell adhesion, tumorigenesis, and immune-related signaling pathways were mainly enriched. JAK1 was positively associated with tumor-infiltrating immune cells, and the results of CIBERSORT analysis suggested that JAK1 was correlated with monotypes and M1 macrophages. The results of the TIMER database analysis confirmed that JAK1 was closely associated with the gene markers of M1 macrophages. Thus, JAK1 may serve as a potential prognostic biomarker in LUAD and is associated with immune infiltration.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Janus Quinasa 1/genética , Neoplasias Pulmonares/genética , Linfocitos Infiltrantes de Tumor/inmunología , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Adenocarcinoma del Pulmón/inmunología , Carcinogénesis/genética , Adhesión Celular/genética , Bases de Datos Genéticas , Humanos , Janus Quinasa 1/inmunología , Neoplasias Pulmonares/inmunología , Monocitos , Pronóstico , ARN Mensajero/metabolismo
10.
Nat Rev Immunol ; 20(10): 585-586, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32788708
11.
Clin Immunol ; 218: 108517, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32585295

RESUMEN

Approximately 15% of patients with coronavirus disease 2019 (COVID-19) experience severe disease, and 5% progress to critical stage that can result in rapid death. No vaccines or antiviral treatments have yet proven effective against COVID-19. Patients with severe COVID-19 experience elevated plasma levels of pro-inflammatory cytokines, which can result in cytokine storm, followed by massive immune cell infiltration into the lungs leading to alveolar damage, decreased lung function, and rapid progression to death. As many of the elevated cytokines signal through Janus kinase (JAK)1/JAK2, inhibition of these pathways with ruxolitinib has the potential to mitigate the COVID-19-associated cytokine storm and reduce mortality. This is supported by preclinical and clinical data from other diseases with hyperinflammatory states, where ruxolitinib has been shown to reduce cytokine levels and improve outcomes. The urgent need for treatments for patients with severe disease support expedited investigation of ruxolitinib for patients with COVID-19.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/prevención & control , Citocinas/antagonistas & inhibidores , Neumonía Viral/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Síndrome Respiratorio Agudo Grave/prevención & control , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Betacoronavirus/inmunología , COVID-19 , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Citocinas/genética , Citocinas/inmunología , Cálculo de Dosificación de Drogas , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Nitrilos , Pandemias , Neumonía Viral/complicaciones , Neumonía Viral/inmunología , Neumonía Viral/virología , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Pirimidinas , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/complicaciones , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos
12.
Leukemia ; 34(7): 1805-1815, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32518419

RESUMEN

A subgroup of patients with severe COVID-19 suffers from progression to acute respiratory distress syndrome and multiorgan failure. These patients present with progressive hyperinflammation governed by proinflammatory cytokines. An interdisciplinary COVID-19 work flow was established to detect patients with imminent or full blown hyperinflammation. Using a newly developed COVID-19 Inflammation Score (CIS), patients were prospectively stratified for targeted inhibition of cytokine signalling by the Janus Kinase 1/2 inhibitor ruxolitinib (Rux). Patients were treated with efficacy/toxicity guided step up dosing up to 14 days. Retrospective analysis of CIS reduction and clinical outcome was performed. Out of 105 patients treated between March 30th and April 15th, 2020, 14 patients with a CIS ≥ 10 out of 16 points received Rux over a median of 9 days with a median cumulative dose of 135 mg. A total of 12/14 patients achieved significant reduction of CIS by ≥25% on day 7 with sustained clinical improvement in 11/14 patients without short term red flag warnings of Rux-induced toxicity. Rux treatment for COVID-19 in patients with hyperinflammation is shown to be safe with signals of efficacy in this pilot case series for CRS-intervention to prevent or overcome multiorgan failure. A multicenter phase-II clinical trial has been initiated (NCT04338958).


Asunto(s)
Antiinflamatorios/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 2/antagonistas & inhibidores , Neumonía Viral/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , Betacoronavirus/patogenicidad , COVID-19 , Ensayos Clínicos como Asunto , Infecciones por Coronavirus/enzimología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/enzimología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/inmunología , Esquema de Medicación , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Masculino , Persona de Mediana Edad , Nitrilos , Pandemias , Seguridad del Paciente , Neumonía Viral/enzimología , Neumonía Viral/inmunología , Neumonía Viral/virología , Pirimidinas , Estudios Retrospectivos , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/enzimología , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/virología , Resultado del Tratamiento
13.
Biochem Pharmacol ; 178: 114103, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32562787

RESUMEN

Janus kinase (JAK) inhibitors (also termed Jakinibs) constitute a family of small drugs that target various isoforms of JAKs (JAK1, JAK2, JAK3 and/or tyrosine kinase 2 (Tyk2)). They exert anti-inflammatory properties linked, in part, to the modulation of the activation state of pro-inflammatory M1 macrophages. The exact impact of JAK inhibitors on a wider spectrum of activation states of macrophages is however still to be determined, especially in the context of disorders involving concomitant activation of pro-inflammatory M1 macrophages and profibrotic M2 macrophages. This is especially the case in autoimmune pulmonary fibrosis like scleroderma-associated interstitial lung disease (ILD), in which M1 and M2 macrophages play a key pathogenic role. In this study, we directly compared the anti-inflammatory and anti-fibrotic effects of three JAK inhibitors (ruxolitinib (JAK2/1 inhibitor); tofacitinib (JAK3/2 inhibitor) and itacitinib (JAK1 inhibitor)) on five different activation states of primary human monocyte-derived macrophages (MDM). These three JAK inhibitors exert anti-inflammatory properties towards macrophages, as demonstrated by the down-expression of key polarization markers (CD86, MHCII, TLR4) and the limited secretion of key pro-inflammatory cytokines (CXCL10, IL-6 and TNFα) in M1 macrophages activated by IFNγ and LPS or by IFNγ alone. We also highlighted that these JAK inhibitors can limit M2a activation of macrophages induced by IL-4 and IL-13, as notably demonstrated by the down-regulation of the M2a associated surface marker CD206 and of the secretion of CCL18. Moreover, these JAK inhibitors reduced the expression of markers such as CXCL13, MARCO and SOCS3 in alternatively activated macrophages induced by IL-10 and dexamethasone (M2c + dex) or IL-10 alone (M2c MDM). For all polarization states, Jakinibs with inhibitory properties over JAK2 had the highest effects, at both 1 µM or 0.1 µM. Based on these in vitro results, we also explored the effects of JAK2/1 inhibition by ruxolitinib in vivo, on mouse macrophages in a model of HOCl-induced ILD, that mimics scleroderma-associated ILD. In this model, we showed that ruxolitinib significantly prevented the upregulation of pro-inflammatory M1 markers (TNFα, CXCL10, NOS2) and pro-fibrotic M2 markers (Arg1 and Chi3L3). These results were associated with an improvement of skin and pulmonary involvement. Overall, our results suggest that the combined anti-inflammatory and anti-fibrotic properties of JAK2/1 inhibitors could be relevant to target lung macrophages in autoimmune and inflammatory pulmonary disorders that have no efficient disease modifying drugs to date.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Esclerodermia Sistémica/tratamiento farmacológico , Animales , Diferenciación Celular , Quimiocina CXCL13/genética , Quimiocina CXCL13/inmunología , Femenino , Regulación de la Expresión Génica , Ácido Hipocloroso/administración & dosificación , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/genética , Janus Quinasa 3/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Enfermedades Pulmonares Intersticiales/inducido químicamente , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Nitrilos , Cultivo Primario de Células , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Esclerodermia Sistémica/inducido químicamente , Esclerodermia Sistémica/inmunología , Esclerodermia Sistémica/patología , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/inmunología
14.
Molecules ; 25(9)2020 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-32397290

RESUMEN

Exposure to particulate matter (PM) has been known to be one of the risk factors to cause allergic asthma, leading to development of respiratory disease. Banhahubak-tang tablet (BHT), a standardized Korean Medicine, is prescribed for neurasthenia, laryngopharyngitis and asthma. In this study, we investigated therapeutic effects of BHT on airway inflammation in ovalbumin (OVA) and PM smaller than 10 µm (PM10)-induced allergic asthma mice. To establish allergic asthma with airway hyper-responsiveness by PM10, BALB/c mice were sensitized and challenged with OVA and PM10, and orally administered BHT. Histological staining was performed to assess airway remodeling. Serum and bronchoalveolar lavage fluid (BALF) was collected for measuring immunoglobulin levels and counting inflammatory cells, respectively. Expression levels of Janus kinase 1 (JAK1)/signal transducer and activator of transcription 6 (STAT6), pro-inflammatory cytokines and type 2 T-helper (Th2)-related cytokines were analyzed in vivo and in vitro models. Histopathological analysis demonstrated that BHT suppressed inflammatory cell infiltration, mucus hypersecretion and collagen deposition in the airway. BHT administration effectively decreased number of inflammatory cells in BALF. BHT reduced total serum Immunoglobulin E (IgE) and Immunoglobulin G (IgG) levels. In addition, BHT significantly inhibited the phosphorylation of JAK1 and STAT6 expressions. Release of pro-inflammatory cytokines and Th2-related cytokines were down-regulated by BHT. In conclusion, BHT mitigated airway inflammation by down-regulating pro-inflammatory and Th2-related cytokines via JAK1/STAT6 signaling. BHT might be a promising herbal medicine for preventing airway inflammation. Moreover, an intervention study among humans is needed to further evaluate the possible beneficial effects of BHT in allergic asthma.


Asunto(s)
Antiasmáticos/farmacología , Asma , Janus Quinasa 1/inmunología , Factor de Transcripción STAT6/inmunología , Transducción de Señal/efectos de los fármacos , Animales , Antiasmáticos/química , Asma/tratamiento farmacológico , Asma/inmunología , Asma/patología , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Comprimidos , Células Th2/inmunología , Células Th2/patología
15.
Expert Rev Hematol ; 13(5): 519-532, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32249631

RESUMEN

Introduction: The traditional therapeutic modalities to manage SR-acute GVHD have focused on the inhibition of the alloreactive T-cell response, while in the setting of SR-chronic GVHD the focus has been on a combination of T-cell and B-cell targeting strategies. However, new therapeutic modalities have shown promise. The purpose of this review is to summarize the current treatment landscape of SR-acute and chronic GVHD.Areas covered: A systematic search of MEDLINE, EMBASE, and clinicaltrials.gov databases for published articles, abstracts, and clinical trials pertaining to available therapeutic modalities for SR-acute and SR-chronic GVHD was conducted. Also highlighted is a number of ongoing clinical trials in both SR-acute and SR-chronic GVHD with strategies targeting the JAK-1/2 pathway, the Treg:Tcon ratio, the immunomodulation mediated by mesenchymal stem cells, and the gut microbiome, among others. Expert opinion: Ruxolitinib has emerged as the preferred therapeutic modality for SR-acute GVHD, with alpha-1-antitrypsin and extracorporeal photophoresis (ECP) being reasonable alternatives. Ruxolitinib and Ibrutinib are among the preferred options for SR-chronic GVHD, with ECP being a viable alternative particularly if the skin is involved. A number of novel therapeutic modalities, including those enhancing the activity of regulatory T-cells have shown great promise in early phase trials of SR-chronic GVHD.


Asunto(s)
Adenina/análogos & derivados , Enfermedad Injerto contra Huésped/terapia , Fotoféresis , Piperidinas/uso terapéutico , Pirazoles/uso terapéutico , alfa 1-Antitripsina/uso terapéutico , Enfermedad Aguda , Adenina/uso terapéutico , Linfocitos B/inmunología , Linfocitos B/patología , Enfermedad Crónica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunomodulación , Janus Quinasa 1/inmunología , Janus Quinasa 2/inmunología , Nitrilos , Pirimidinas , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
16.
Cell Host Microbe ; 27(3): 454-466.e8, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32075740

RESUMEN

Type I interferons (IFNs-I) fulfil multiple protective functions during pathogenic infections, but they can also cause detrimental effects and enhance immunopathology. Here, we report that IFNs-I promote the dysregulation of iron homeostasis in macrophages during systemic infections with the intracellular pathogen Candida glabrata, leading to fungal survival and persistence. By engaging JAK1, IFNs-I disturb the balance of the transcriptional activator NRF2 and repressor BACH1 to induce downregulation of the key iron exporter Fpn1 in macrophages. This leads to enhanced iron accumulation in the phagolysosome and failure to restrict fungal access to iron pools. As a result, C. glabrata acquires iron via the Sit1/Ftr1 iron transporter system, facilitating fungal intracellular replication and immune evasion. Thus, IFNs-I are central regulators of iron homeostasis, which can impact infection, and restricting iron bioavailability may offer therapeutic strategies to combat invasive fungal infections.


Asunto(s)
Candida glabrata/patogenicidad , Homeostasis , Interferón Tipo I/inmunología , Hierro/fisiología , Macrófagos/microbiología , Adulto , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/inmunología , Candidiasis/inmunología , Proteínas de Transporte de Catión/inmunología , Células Cultivadas , Femenino , Humanos , Evasión Inmune , Janus Quinasa 1/inmunología , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/inmunología , Fagosomas/microbiología , Bazo/inmunología
17.
Front Immunol ; 11: 599564, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33488593

RESUMEN

STING-associated vasculopathy of infantile-onset (SAVI) is one of the newly identified types of interferonopathies. SAVI is caused by heterozygous gain-of-function mutations in the STING1. We herein report for the first time a homozygous variant in the STING1 gene in two siblings that resulted in constitutive activation of STING gene and the SAVI phenotype. Exome sequencing revealed a novel homozygous NM_198282.3: c.841C>T; p.(Arg281Trp) variant in exon 7 of the STING1 gene. The variant segregated in the family to be homozygous in all affected and either heterozygous or wild type in all healthy. Computational structural analysis of the mutants revealed changes in the STING protein structure/function. Elevated serum beta-interferon levels were observed in the patients compared to the control family members. Treatment with Janus kinase inhibitor (JAK-I) Ruxolitinib suppressed the inflammatory process, decreased beta-interferon levels, and stopped the progression of the disease.


Asunto(s)
Alelos , Enfermedades Genéticas Congénitas/inmunología , Homocigoto , Proteínas de la Membrana/genética , Mutación Missense , Hermanos , Enfermedades Vasculares/genética , Adolescente , Sustitución de Aminoácidos , Niño , Femenino , Enfermedades Genéticas Congénitas/tratamiento farmacológico , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Masculino , Proteínas de la Membrana/inmunología , Nitrilos , Pirazoles/administración & dosificación , Pirimidinas , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología , Secuenciación del Exoma
18.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31694946

RESUMEN

Several members of the tripartite motif (TRIM) family of E3 ubiquitin ligases regulate immune pathways, including the antiviral type I interferon (IFN-I) system. Previously, we demonstrated that TRIM6 is involved in IFN-I induction and signaling. In the absence of TRIM6, optimal IFN-I signaling is reduced, allowing increased replication of interferon-sensitive viruses. Despite having evolved numerous mechanisms to restrict the vertebrate host's IFN-I response, West Nile virus (WNV) replication is sensitive to pretreatment with IFN-I. However, the regulators and products of the IFN-I pathway that are important in regulating WNV replication are incompletely defined. Consistent with WNV's sensitivity to IFN-I, we found that in TRIM6 knockout (TRIM6-KO) A549 cells, WNV replication is significantly increased and IFN-I induction and signaling are impaired compared to wild-type (wt) cells. IFN-ß pretreatment was more effective in protecting against subsequent WNV infection in wt cells than TRIM6-KO, indicating that TRIM6 contributes to the establishment of an IFN-induced antiviral response against WNV. Using next-generation sequencing, we identified VAMP8 as a potential factor involved in this TRIM6-mediated antiviral response. VAMP8 knockdown resulted in reduced JAK1 and STAT1 phosphorylation and impaired induction of several interferon-stimulated genes (ISGs) following WNV infection or IFN-ß treatment. Furthermore, VAMP8-mediated STAT1 phosphorylation required the presence of TRIM6. Therefore, the VAMP8 protein is a novel regulator of IFN-I signaling, and its expression and function are dependent on TRIM6 activity. Overall, these results provide evidence that TRIM6 contributes to the antiviral response against WNV and identify VAMP8 as a novel regulator of the IFN-I system.IMPORTANCE WNV is a mosquito-borne flavivirus that poses a threat to human health across large discontinuous areas throughout the world. Infection with WNV results in febrile illness, which can progress to severe neurological disease. Currently, there are no approved treatment options to control WNV infection. Understanding the cellular immune responses that regulate viral replication is important in diversifying the resources available to control WNV. Here, we show that the elimination of TRIM6 in human cells results in an increase in WNV replication and alters the expression and function of other components of the IFN-I pathway through VAMP8. Dissecting the interactions between WNV and host defenses both informs basic molecular virology and promotes the development of host- and virus-targeted antiviral strategies.


Asunto(s)
Inmunidad Innata , Interferón Tipo I/inmunología , Proteínas R-SNARE/inmunología , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Replicación Viral/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología , Células A549 , Eliminación de Gen , Células HEK293 , Humanos , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Fosforilación/genética , Fosforilación/inmunología , Proteínas R-SNARE/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Replicación Viral/genética , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/patología
19.
Mol Immunol ; 117: 29-36, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31733446

RESUMEN

Janus kinase 1 (JAK1) is a member of JAK family of non-receptor protein tyrosine kinases that plays critical roles in transducing cytokine signals via JAK-signal transducer and activator of transcription (STAT) signaling pathway. The importance of JAK1 in innate immunity has been well-studied in mammals and fish, yet in avian remains largely unknown. Here, we cloned the full-length of the duck JAK1 (duJAK1) gene for the first time. DuJAK1 encoded a protein of 1152 amino acids and possessed high amino acid identity with goose and budgerigar JAK1s. The duJAK1 was expressed in all detected tissues, especially high in the thymus and bursa of Fabricius. Overexpression of duJAK1 significantly activated ISRE promoter activity and induced duck viperin, 2', 5'-OAS, MX, PKR and ZAP expression. Knockdown of duJAK1 by small interfering RNA significantly inhibited duck Tembusu virus (DTMUV)-, duck Enteritis virus (DEV)-, poly (I:C)-, poly (dA:dT)- or Sendai virus (SeV)-induced ISRE promoter activation. Furthermore, duJAK1 exhibited antiviral activity against DTMUV infection. These results will help us understand the function of JAK family proteins in duck antiviral immunity.


Asunto(s)
Proteínas Aviares/inmunología , Patos/inmunología , Inmunidad Innata/inmunología , Janus Quinasa 1/inmunología , Animales , Proteínas Aviares/genética , Patos/genética , Inmunidad Innata/genética , Janus Quinasa 1/genética
20.
Immunohorizons ; 3(11): 547-558, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748225

RESUMEN

Cytokines are critical for guiding the differentiation of T lymphocytes to perform specialized tasks in the immune response. Developing strategies to manipulate cytokine-signaling pathways holds promise to program T cell differentiation toward the most therapeutically useful direction. Suppressor of cytokine signaling (SOCS) proteins are attractive targets, as they effectively inhibit undesirable cytokine signaling. However, these proteins target multiple signaling pathways, some of which we may need to remain uninhibited. SOCS3 inhibits IL-12 signaling but also inhibits the IL-2-signaling pathway. In this study, we use computational protein design based on SOCS3 and JAK crystal structures to engineer a mutant SOCS3 with altered specificity. We generated a mutant SOCS3 designed to ablate interactions with JAK1 but maintain interactions with JAK2. We show that this mutant does indeed ablate JAK1 inhibition, although, unexpectedly, it still coimmunoprecipitates with JAK1 and does so to a greater extent than with JAK2. When expressed in CD8 T cells, mutant SOCS3 preserved inhibition of JAK2-dependent STAT4 phosphorylation following IL-12 treatment. However, inhibition of STAT phosphorylation was ablated following stimulation with JAK1-dependent cytokines IL-2, IFN-α, and IL-21. Wild-type SOCS3 inhibited CD8 T cell expansion in vivo and induced a memory precursor phenotype. In vivo T cell expansion was restored by expression of the mutant SOCS3, and this also reverted the phenotype toward effector T cell differentiation. These data show that SOCS proteins can be engineered to fine-tune their specificity, and this can exert important changes to T cell biology.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Factor de Transcripción STAT4/inmunología , Factor de Transcripción STAT5/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/genética , Animales , Diferenciación Celular , Células Cultivadas , Técnicas de Silenciamiento del Gen , Janus Quinasa 1/inmunología , Janus Quinasa 2/inmunología , Ratones , Ratones Endogámicos C57BL , Mutación , Fosforilación , Ingeniería de Proteínas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...