Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nephron ; 145(4): 445-450, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33975323

RESUMEN

The MAFB gene encodes an important basic leucine zipper transcription factor that functions in glomerular podocytes, macrophages, and osteoclasts. Recently, MAFB was identified as the gene that was responsible for causing nephropathy with focal segmental glomerulosclerosis (FSGS) with multicentric carpotarsal osteolysis (MCTO) or Duane retraction syndrome (DRS). Here, we describe a patient with nephropathy associated with FSGS who exhibited a novel stop-gain variant in the MAFB gene (NM_005461:c.590C>A (p.Ser197Ter)). The patient's father exhibited proteinuria with FSGS with possible DRS, whereas the patient exhibited nephropathy with FSGS and nearly normal eye movement and hearing function, as well as intact bone structure in the extremities. Conventional oral steroids or immunosuppressive drugs have not demonstrated effectiveness for patients with nephropathy who exhibit pathogenic variants in MAFB, except for a patient with nephropathy with FSGS and MCTO who experienced attenuated proteinuria within the subnephrotic range in response to cyclosporine A (CyA) treatment for at least 4 years. Thus, we attempted administration of CyA in our patient. Unexpectedly, the patient demonstrated good and rapid responses to CyA, including a partial reduction in proteinuria from approximately 2.0 g/g Cr to proteinuria within the subnephrotic range (0.27 g/g Cr) after 13 months of observation. Our findings suggest that CyA may be a suitable treatment option for patients with nephropathy with FSGS who exhibit pathogenic MAFB variants.


Asunto(s)
Ciclosporina/uso terapéutico , Inmunosupresores/uso terapéutico , Factor de Transcripción MafB/inmunología , Adulto , Edad de Inicio , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Fallo Renal Crónico/etiología , Masculino , Trastornos de la Motilidad Ocular/etiología
2.
Eur J Immunol ; 51(4): 824-834, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33169838

RESUMEN

CD28 expression is generally considered to be T lymphocyte specific. We have previously shown CD28 mRNA expression in M-CSF-dependent anti-inflammatory monocyte-derived macrophages (M-MØ), and now demonstrate that CD28 cell surface expression is higher in M-MØ than in GM-CSF-dependent macrophages, and that macrophage CD28 expression is regulated by MAFB and activin A. In vivo, CD28 was found in tumor-associated macrophages and, to a lower extent, in pro-inflammatory synovial fluid macrophages from rheumatoid arthritis patients. Analysis of mouse macrophages confirmed Cd28 expression in bone-marrow derived M-MØ. Indeed, anti-CD28 antibodies triggered ERK1/2 phosphorylation in mouse M-MØ. At the functional level, Cd28KO M-MØ exhibited a significantly higher capacity to activate the OVA-specific proliferation of OT-II CD4+ T cells than WT M-MØ, as well as enhanced LPS-induced IL-6 production. Besides, the Cd28KO M-MØ transcriptome was significantly different from WT M-MØ regarding the expression IFN response, inflammatory response, and TGF-ß signaling related gene sets. Therefore, defective CD28 expression in mouse macrophages associates to changes in gene expression profile, what might contribute to the altered functionality displayed by Cd28KO M-MØ. Thus, CD28 expression appears as a hallmark of anti-inflammatory macrophages and might be a target for immunotherapy.


Asunto(s)
Antígenos CD28/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Linfocitos T/inmunología , Activinas/genética , Activinas/inmunología , Activinas/metabolismo , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Antígenos CD28/genética , Antígenos CD28/metabolismo , Células Cultivadas , Expresión Génica/inmunología , Perfilación de la Expresión Génica/métodos , Humanos , Inflamación/genética , Inflamación/metabolismo , Activación de Linfocitos/genética , Macrófagos/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/inmunología , Factor de Transcripción MafB/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo
3.
Front Immunol ; 11: 603507, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33312178

RESUMEN

Defective IFN production and exacerbated inflammatory and pro-fibrotic responses are hallmarks of SARS-CoV-2 infection in severe COVID-19. Based on these hallmarks, and considering the pivotal role of macrophages in COVID-19 pathogenesis, we hypothesize that the transcription factors MAFB and MAF critically contribute to COVID-19 progression by shaping the response of macrophages to SARS-CoV-2. Our proposal stems from the recent identification of pathogenic lung macrophage subsets in severe COVID-19, and takes into consideration the previously reported ability of MAFB to dampen IFN type I production, as well as the critical role of MAFB and MAF in the acquisition and maintenance of the transcriptional signature of M-CSF-conditioned human macrophages. Solid evidences are presented that link overexpression of MAFB and silencing of MAF expression with clinical and biological features of severe COVID-19. As a whole, we propose that a high MAFB/MAF expression ratio in lung macrophages could serve as an accurate diagnostic tool for COVID-19 progression. Indeed, reversing the macrophage MAFB/MAF expression ratio might impair the exacerbated inflammatory and profibrotic responses, and restore the defective IFN type I production, thus becoming a potential strategy to limit severity of COVID-19.


Asunto(s)
COVID-19/inmunología , Macrófagos/inmunología , Factores de Transcripción Maf/inmunología , Factor de Transcripción MafB/inmunología , SARS-CoV-2/inmunología , COVID-19/genética , COVID-19/virología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/metabolismo , Factores de Transcripción Maf/genética , Factores de Transcripción Maf/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/metabolismo , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad
4.
J Immunol ; 205(3): 776-788, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591394

RESUMEN

Growth hormone (GH), a pleiotropic hormone secreted by the pituitary gland, regulates immune and inflammatory responses. In this study, we show that GH regulates the phenotypic and functional plasticity of macrophages both in vitro and in vivo. Specifically, GH treatment of GM-CSF-primed monocyte-derived macrophages promotes a significant enrichment of anti-inflammatory genes and dampens the proinflammatory cytokine profile through PI3K-mediated downregulation of activin A and upregulation of MAFB, a critical transcription factor for anti-inflammatory polarization of human macrophages. These in vitro data correlate with improved remission of inflammation and mucosal repair during recovery in the acute dextran sodium sulfate-induced colitis model in GH-overexpressing mice. In this model, in addition to the GH-mediated effects on other immune cells, we observed that macrophages from inflamed gut acquire an anti-inflammatory/reparative profile. Overall, these data indicate that GH reprograms inflammatory macrophages to an anti-inflammatory phenotype and improves resolution during pathologic inflammatory responses.


Asunto(s)
Reprogramación Celular/inmunología , Colitis/inmunología , Regulación de la Expresión Génica/inmunología , Hormona del Crecimiento/inmunología , Macrófagos/inmunología , Factor de Transcripción MafB/inmunología , Animales , Bovinos , Reprogramación Celular/genética , Colitis/inducido químicamente , Colitis/genética , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Hormona del Crecimiento/genética , Factor de Transcripción MafB/genética , Ratones , Ratones Transgénicos
5.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32213710

RESUMEN

Monocyte-derived DCs (moDCs) have been implicated in the pathogenesis of autoimmunity, but the molecular pathways determining the differentiation potential of these cells remain unclear. Here, we report that microRNA-148a (miR-148a) serves as a critical regulator for moDC differentiation. First, miR-148a deficiency impaired the moDC development in vitro and in vivo. A mechanism study showed that MAFB, a transcription factor that hampers moDC differentiation, was a direct target of miR-148a. In addition, a promoter study identified that miR-148a could be transcriptionally induced by PU.1, which is crucial for moDC generation. miR-148a ablation eliminated the inhibition of PU.1 on MAFB. Furthermore, we found that miR-148a increased in monocytes from patients with psoriasis, and miR-148a deficiency or intradermal injection of antagomir-148a immensely alleviated the development of psoriasis-like symptoms in a psoriasis-like mouse model. Therefore, these results identify a pivotal role for the PU.1-miR-148a-MAFB circuit in moDC differentiation and suggest a potential therapeutic avenue for autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Células Dendríticas/inmunología , Factor de Transcripción MafB/inmunología , MicroARNs/inmunología , Animales , Autoinmunidad/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Factor de Transcripción MafB/genética , Ratones , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Psoriasis/genética , Psoriasis/inmunología , Transactivadores/genética , Transactivadores/inmunología
6.
Nat Commun ; 8(1): 1700, 2017 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-29167450

RESUMEN

The transcription factor MafB is expressed by monocytes and macrophages. Efferocytosis (apoptotic cell uptake) by macrophages is important for inhibiting the development of autoimmune diseases, and is greatly reduced in Mafb-deficient macrophages. Here, we show the expression of the first protein in the classical complement pathway C1q is important for mediating efferocytosis and is reduced in Mafb-deficient macrophages. The efferocytosis defect in Mafb-deficient macrophages can be rescued by adding serum from wild-type mice, but not by adding serum from C1q-deficient mice. By hemolysis assay we also show that activation of the classical complement pathway is decreased in Mafb-deficient mice. In addition, MafB overexpression induces C1q-dependent gene expression and signals that induce C1q genes are less effective in the absence of MafB. We also show that Mafb-deficiency can increase glomerular autoimmunity, including anti-nuclear antibody deposition. These results show that MafB is an important regulator of C1q.


Asunto(s)
Complemento C1q/metabolismo , Factor de Transcripción MafB/inmunología , Animales , Apoptosis/inmunología , Autoinmunidad , Complemento C1q/deficiencia , Complemento C1q/genética , Vía Clásica del Complemento , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Factor de Transcripción MafB/deficiencia , Factor de Transcripción MafB/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Células RAW 264.7 , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/inmunología
7.
Sci Rep ; 7(1): 7591, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790455

RESUMEN

Macrophages play pivotal roles in the progression and regression of atherosclerosis. Accumulating evidence suggests that macrophage polarization into an anti-inflammatory M2 state is a key characteristic of atherosclerotic plaques undergoing regression. However, the molecular mechanisms underlying this potential association of the M2 polarization with atherosclerosis regression remain poorly understood. Further, human genetic factors that facilitate these anti-atherogenic processes remain largely unknown. We report that the transcription factor MafB plays pivotal roles in promoting macrophage M2 polarization. Further, MafB promotes cholesterol efflux from macrophage foam cells by directly up-regulating its key cellular mediators. Notably, MafB expression is significantly up-regulated in response to various metabolic and immunological stimuli that promote macrophage M2 polarization or cholesterol efflux, and thereby MafB mediates their beneficial effects, in both liver x receptor (LXR)-dependent and independent manners. In contrast, MafB is strongly down-regulated upon elevated pro-inflammatory signaling or by pro-inflammatory and pro-atherogenic microRNAs, miR-155 and miR-33. Using an integrative systems biology approach, we also revealed that M2 polarization and cholesterol efflux do not necessarily represent inter-dependent events, but MafB is broadly involved in both the processes. These findings highlight physiological protective roles that MafB may play against atherosclerosis progression.


Asunto(s)
Aterosclerosis/genética , Macrófagos/inmunología , Factor de Transcripción MafB/genética , MicroARNs/genética , Placa Aterosclerótica/genética , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Secuencia de Bases , Transporte Biológico , Diferenciación Celular , Colesterol/inmunología , Colesterol/metabolismo , Regulación de la Expresión Génica , Humanos , Hidrocarburos Fluorados/farmacología , Interleucina-10/farmacología , Interleucina-4/farmacología , Receptores X del Hígado/genética , Receptores X del Hígado/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Factor de Transcripción MafB/inmunología , Masculino , Ratones , Ratones Transgénicos , MicroARNs/inmunología , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Cultivo Primario de Células , Células RAW 264.7 , Transducción de Señal , Sulfonamidas/farmacología , Células THP-1
8.
J Clin Invest ; 127(7): 2842-2854, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28581443

RESUMEN

The quantity and activation state of adipose tissue macrophages (ATMs) impact the development of obesity-induced metabolic diseases. Appetite-controlling hormones play key roles in obesity; however, our understanding of their effects on ATMs is limited. Here, we have shown that human and mouse ATMs express NPFFR2, a receptor for the appetite-reducing neuropeptide FF (NPFF), and that NPFFR2 expression is upregulated by IL-4, an M2-polarizing cytokine. Plasma levels of NPFF decreased in obese patients and high-fat diet-fed mice and increased following caloric restriction. NPFF promoted M2 activation and increased the proliferation of murine and human ATMs. Both M2 activation and increased ATM proliferation were abolished in NPFFR2-deficient ATMs. Mechanistically, the effects of NPFF involved the suppression of E3 ubiquitin ligase RNF128 expression, resulting in enhanced stability of phosphorylated STAT6 and increased transcription of the M2 macrophage-associated genes IL-4 receptor α (Il4ra), arginase 1 (Arg1), IL-10 (Il10), and alkylglycerol monooxygenase (Agmo). NPFF induced ATM proliferation concomitantly with the increase in N-Myc downstream-regulated gene 2 (Ndrg2) expression and suppressed the transcription of Ifi200 cell-cycle inhibitor family members and MAF bZIP transcription factor B (Mafb), a negative regulator of macrophage proliferation. NPFF thus plays an important role in supporting healthy adipose tissue via the maintenance of metabolically beneficial ATMs.


Asunto(s)
Tejido Adiposo/inmunología , Proliferación Celular , Activación de Macrófagos , Macrófagos/inmunología , Oligopéptidos/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Arginasa/genética , Arginasa/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/inmunología , Masculino , Ratones , Ratones Transgénicos , Oligopéptidos/genética , Proteínas/genética , Proteínas/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología
9.
Nat Med ; 23(6): 723-732, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28394332

RESUMEN

Damage-associated molecular patterns (DAMPs) trigger sterile inflammation after tissue injury, but the mechanisms underlying the resolution of inflammation remain unclear. In this study, we demonstrate that common DAMPs, such as high-mobility-group box 1 (HMGB1), peroxiredoxins (PRXs), and S100A8 and S100A9, were internalized through the class A scavenger receptors MSR1 and MARCO in vitro. In ischemic murine brain, DAMP internalization was largely mediated by MSR1. An elevation of MSR1 levels in infiltrating myeloid cells observed 3 d after experimental stroke was dependent on the transcription factor Mafb. Combined deficiency for Msr1 and Marco, or for Mafb alone, in infiltrating myeloid cells caused impaired clearance of DAMPs, more severe inflammation, and exacerbated neuronal injury in a murine model of ischemic stroke. The retinoic acid receptor (RAR) agonist Am80 increased the expression of Mafb, thereby enhancing MSR1 expression. Am80 exhibited therapeutic efficacy when administered, even at 24 h after the onset of experimental stroke. Our findings uncover cellular mechanisms contributing to DAMP clearance in resolution of the sterile inflammation triggered by tissue injury.


Asunto(s)
Alarminas/inmunología , Encéfalo/inmunología , Infarto de la Arteria Cerebral Media/inmunología , Factor de Transcripción MafB/inmunología , Células Mieloides/inmunología , Receptores Inmunológicos/inmunología , Receptores Depuradores de Clase A/inmunología , Animales , Benzoatos/farmacología , Encéfalo/efectos de los fármacos , Isquemia Encefálica/inmunología , Sistemas CRISPR-Cas , Calgranulina A/inmunología , Calgranulina B/inmunología , Inmunoprecipitación de Cromatina , Proteína HMGB1/inmunología , Inflamación , Factor de Transcripción MafB/efectos de los fármacos , Factor de Transcripción MafB/genética , Ratones , Células Mieloides/metabolismo , Peroxirredoxinas/inmunología , Receptores Inmunológicos/genética , Receptores de Ácido Retinoico/agonistas , Receptores Depuradores de Clase A/efectos de los fármacos , Receptores Depuradores de Clase A/genética , Accidente Cerebrovascular/inmunología , Tetrahidronaftalenos/farmacología
11.
Nat Immunol ; 11(8): 743-50, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20581830

RESUMEN

Viral infection induces type I interferons (IFN-alpha and IFN-beta) that recruit unexposed cells in a self-amplifying response. We report that the transcription factor MafB thwarts auto-amplification by a metastable switch activity. MafB acted as a weak positive basal regulator of transcription at the IFNB1 promoter through activity at transcription factor AP-1-like sites. Interferon elicitors recruited the transcription factor IRF3 to the promoter, whereupon MafB acted as a transcriptional antagonist, impairing the interaction of coactivators with IRF3. Mathematical modeling supported the view that prepositioning of MafB on the promoter allows the system to respond rapidly to fluctuations in IRF3 activity. Higher expression of MafB in human pancreatic islet beta cells might increase cellular vulnerability to viral infections associated with the etiology of type 1 diabetes.


Asunto(s)
Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Factor de Transcripción MafB/inmunología , Virosis/inmunología , Animales , Línea Celular Tumoral , Humanos , Factor 3 Regulador del Interferón/antagonistas & inhibidores , Interferón Tipo I/biosíntesis , Interferón beta/genética , Factor de Transcripción MafB/genética , Ratones , Ratones Noqueados , Modelos Inmunológicos , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/inmunología , Transcripción Genética
12.
J Immunol ; 181(8): 5660-72, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18832725

RESUMEN

Although a considerable number of reports indicate an involvement of the Hox-A10 gene in the molecular control of hemopoiesis, the conclusions of such studies are quite controversial given that they support, in some cases, a role in the stimulation of stem cell self-renewal and myeloid progenitor expansion, whereas in others they implicate this transcription factor in the induction of monocyte-macrophage differentiation. To clarify this issue, we analyzed the biological effects and the transcriptome changes determined in human primary CD34(+) hemopoietic progenitors by retroviral transduction of a full-length Hox-A10 cDNA. The results obtained clearly indicated that this homeogene is an inducer of monocyte differentiation, at least partly acting through the up-regulation of the MafB gene, recently identified as the master regulator of such a maturation pathway. By using a combined approach based on computational analysis, EMSA experiments, and luciferase assays, we were able to demonstrate the presence of a Hox-A10-binding site in the promoter region of the MafB gene, which suggested the likely molecular mechanism underlying the observed effect. Stimulation of the same cells with the vitamin D(3) monocyte differentiation inducer resulted in a clear increase of Hox-A10 and MafB transcripts, indicating the existence of a precise transactivation cascade involving vitamin D(3) receptor, Hox-A10, and MafB transcription factors. Altogether, these data allow one to conclude that the vitamin D(3)/Hox-A10 pathway supports MafB function during the induction of monocyte differentiation.


Asunto(s)
Antígenos CD34 , Diferenciación Celular/inmunología , Colecalciferol/farmacología , Proteínas de Homeodominio/inmunología , Factor de Transcripción MafB/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Vitaminas/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células HL-60 , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Hematopoyesis/inmunología , Proteínas Homeobox A10 , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Humanos , Células K562 , Factor de Transcripción MafB/biosíntesis , Factor de Transcripción MafB/genética , Monocitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Elementos de Respuesta/genética , Elementos de Respuesta/inmunología , Retroviridae , Transducción Genética , Células U937 , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA