Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 147
Filtrer
1.
Commun Biol ; 7(1): 896, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39043941

RÉSUMÉ

The central nervous system (CNS) includes anatomically distinct macrophage populations including parenchyma microglia and CNS-associated macrophages (CAMs) localized at the interfaces like meninges and perivascular space, which play specialized roles for the maintenance of the CNS homeostasis with the help of precisely controlled gene expressions. However, the transcriptional machinery that determines their cell-type specific states of microglia and CAMs remains poorly understood. Here we show, by myeloid cell-specific deletion of transcription factors, IRF8 and MAFB, that both adult microglia and CAMs utilize IRF8 to maintain their core gene signatures, although the genes altered by IRF8 deletion are different in the two macrophage populations. By contrast, MAFB deficiency robustly affected the gene expression profile of adult microglia, whereas CAMs are almost independent of MAFB. Our data suggest that distinct transcriptional machineries regulate different macrophages in the CNS.


Sujet(s)
Système nerveux central , Facteurs de régulation d'interféron , Macrophages , Facteur de transcription MafB , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Animaux , Macrophages/métabolisme , Facteurs de régulation d'interféron/métabolisme , Facteurs de régulation d'interféron/génétique , Souris , Système nerveux central/métabolisme , Microglie/métabolisme , Souris knockout , Souris de lignée C57BL , Transcription génétique , Régulation de l'expression des gènes
2.
Cell Rep ; 43(4): 113978, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38522069

RÉSUMÉ

Transcription factor MAFB regulates various homeostatic functions of macrophages. This study explores the role of MAFB in brown adipose tissue (BAT) thermogenesis using macrophage-specific Mafb-deficient (Mafbf/f::LysM-Cre) mice. We find that Mafb deficiency in macrophages reduces thermogenesis, energy expenditure, and sympathetic neuron (SN) density in BAT under cold conditions. This phenotype features a proinflammatory environment that is characterized by macrophage/granulocyte accumulation, increases in interleukin-6 (IL-6) production, and IL-6 trans-signaling, which lead to decreases in nerve growth factor (NGF) expression and reduction in SN density in BAT. We confirm MAFB regulation of IL-6 expression using luciferase readout driven by IL-6 promoter in RAW-264.7 macrophage cell lines. Immunohistochemistry shows clustered organization of NGF-producing cells in BAT, which are primarily TRPV1+ vascular smooth muscle cells, as additionally shown using single-cell RNA sequencing and RT-qPCR of the stromal vascular fraction. Treating Mafbf/f::LysM-Cre mice with anti-IL-6 receptor antibody rescues SN density, body temperature, and energy expenditure.


Sujet(s)
Tissu adipeux brun , Basse température , Interleukine-6 , Macrophages , Facteur de transcription MafB , Neurones , Thermogenèse , Animaux , Facteur de transcription MafB/métabolisme , Facteur de transcription MafB/génétique , Tissu adipeux brun/métabolisme , Souris , Macrophages/métabolisme , Neurones/métabolisme , Interleukine-6/métabolisme , Cellules RAW 264.7 , Facteur de croissance nerveuse/métabolisme , Métabolisme énergétique , Mâle , Souris de lignée C57BL
3.
Cell Mol Life Sci ; 81(1): 152, 2024 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-38528207

RÉSUMÉ

Monocyte-derived macrophages play a key pathogenic role in inflammatory diseases. In the case of rheumatoid arthritis (RA), the presence of specific synovial tissue-infiltrating macrophage subsets is associated with either active disease or inflammation resolution. JAK inhibitors (JAKi) are the first targeted synthetic disease-modifying antirheumatic drugs (tsDMARD) approved for treatment of RA with comparable efficacy to biologics. However, the effects of JAKi on macrophage specification and differentiation are currently unknown. We have analyzed the transcriptional and functional effects of JAKi on human peripheral blood monocyte subsets from RA patients and on the differentiation of monocyte-derived macrophages promoted by granulocyte-macrophage colony-stimulating factor (GM-CSF), a factor that drives the development and pathogenesis of RA. We now report that JAKi Upadacitinib restores the balance of peripheral blood monocyte subsets in RA patients and skewed macrophages towards the acquisition of an anti-inflammatory transcriptional and functional profile in a dose-dependent manner. Upadacitinib-treated macrophages showed a strong positive enrichment of the genes that define synovial macrophages associated to homeostasis/inflammation resolution. Specifically, Upadacitinib-treated macrophages exhibited significantly elevated expression of MAFB and MAFB-regulated genes, elevated inhibitory phosphorylation of GSK3ß, and higher phagocytic activity and showed an anti-inflammatory cytokine profile upon activation by pathogenic stimuli. These outcomes were also shared by macrophages exposed to other JAKi (baricitinib, tofacitinib), but not in the presence of the TYK2 inhibitor deucravacitinib. As a whole, our results indicate that JAKi promote macrophage re-programming towards the acquisition of a more anti-inflammatory/pro-resolution profile, an effect that correlates with the ability of JAKi to enhance MAFB expression.


Sujet(s)
Polyarthrite rhumatoïde , Inhibiteurs des Janus kinases , Humains , Inhibiteurs des Janus kinases/pharmacologie , Inhibiteurs des Janus kinases/métabolisme , Inhibiteurs des Janus kinases/usage thérapeutique , Macrophages/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Anti-inflammatoires/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
4.
JCI Insight ; 8(24)2023 Dec 22.
Article de Anglais | MEDLINE | ID: mdl-37917179

RÉSUMÉ

Monocyte-derived macrophages, the major source of pathogenic macrophages in COVID-19, are oppositely instructed by macrophage CSF (M-CSF) or granulocyte macrophage CSF (GM-CSF), which promote the generation of antiinflammatory/immunosuppressive MAFB+ (M-MØ) or proinflammatory macrophages (GM-MØ), respectively. The transcriptional profile of prevailing macrophage subsets in severe COVID-19 led us to hypothesize that MAFB shapes the transcriptome of pulmonary macrophages driving severe COVID-19 pathogenesis. We have now assessed the role of MAFB in the response of monocyte-derived macrophages to SARS-CoV-2 through genetic and pharmacological approaches, and we demonstrate that MAFB regulated the expression of the genes that define pulmonary pathogenic macrophages in severe COVID-19. Indeed, SARS-CoV-2 potentiated the expression of MAFB and MAFB-regulated genes in M-MØ and GM-MØ, where MAFB upregulated the expression of profibrotic and neutrophil-attracting factors. Thus, MAFB determines the transcriptome and functions of the monocyte-derived macrophage subsets that underlie pulmonary pathogenesis in severe COVID-19 and controls the expression of potentially useful biomarkers for COVID-19 severity.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/métabolisme , COVID-19/métabolisme , Macrophages/métabolisme , Macrophages alvéolaires/métabolisme , Marqueurs biologiques/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
5.
Chem Biol Interact ; 380: 110534, 2023 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-37182688

RÉSUMÉ

Induced pluripotent stem cells (iPSCs) have been the focus of cellular therapy studies. The use of iPSCs in regenerative medicine is limited by their tumorigenic potential. This study sought to determine whether iPSCs-derived podocytes attenuate acute kidney injury (AKI) and the molecular mechanism. Inoculation of iPSCs-podocytes significantly promoted the repair of kidney injury in AKI mice, reduced the levels of kidney injury factors Scr, BUN, and urinary NAG, and alleviated the inflammatory response. Histological analysis revealed a significant increase in the number of M2 macrophages and a significant decrease in M1 macrophages in the kidney tissues. Subsequently, the genes and signaling pathways that may be associated with kidney injury repair in mice were analyzed by RNA-seq and bioinformatics prediction. The polarization of M2 macrophages was promoted by MAF bZIP transcription factor B (Mafb)-mediated activation of C-C motif chemokine receptor 5 (Ccr5) and nicotinamide phosphoribosyltransferase (Nampt) signaling pathway. Taken together, these results show that iPSCs-podocytes depend on Mafb to activate the Nampt signaling pathway through transcriptional activation of Ccr5, thereby promoting the repair of AKI caused by ischemia-reperfusion.


Sujet(s)
Atteinte rénale aigüe , Cellules souches pluripotentes induites , Podocytes , Lésion d'ischémie-reperfusion , Souris , Animaux , Cellules souches pluripotentes induites/métabolisme , Nicotinamide phosphoribosyltransferase/métabolisme , Podocytes/métabolisme , Macrophages/métabolisme , Atteinte rénale aigüe/anatomopathologie , Rein/métabolisme , Lésion d'ischémie-reperfusion/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
6.
Cell Mol Life Sci ; 80(4): 96, 2023 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-36930354

RÉSUMÉ

Monocyte-derived macrophages contribute to pathogenesis in inflammatory diseases and their effector functions greatly depend on the prevailing extracellular milieu. Whereas M-CSF primes macrophages for acquisition of an anti-inflammatory profile, GM-CSF drives the generation of T cell-stimulatory and pro-inflammatory macrophages. Liver X Receptors (LXRα and LXRß) are nuclear receptors that control cholesterol metabolism and regulate differentiation of tissue-resident macrophages. Macrophages from rheumatoid arthritis and other inflammatory pathologies exhibit an enriched LXR pathway, and recent reports have shown that LXR activation raises pro-inflammatory effects and impairs the acquisition of the anti-Inflammatory profile of M-CSF-dependent monocyte-derived macrophages (M-MØ). We now report that LXR inhibition prompts the acquisition of an anti-inflammatory gene and functional profile of macrophages generated within a pathological environment (synovial fluid from Rheumatoid Arthritis patients) as well as during the GM-CSF-dependent differentiation of human monocyte-derived macrophages (GM-MØ). Mechanistically, inhibition of LXR results in macrophages with higher expression of the v-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homolog B (MAFB) transcription factor, which governs the macrophage anti-inflammatory profile, as well as over-expression of MAFB-regulated genes. Indeed, gene silencing experiments on human macrophages evidenced that MAFB is required for the LXR inhibitor to enhance the anti-inflammatory nature of human macrophages. As a whole, our results demonstrate that LXR inhibition prompts the acquisition of an anti-inflammatory transcriptional and functional profile of human macrophages in a MAFB-dependent manner, and propose the use of LXR antagonists as potential therapeutic alternatives in macrophage re-programming strategies during inflammatory responses.


Sujet(s)
Polyarthrite rhumatoïde , Facteur de stimulation des colonies de granulocytes et de macrophages , Humains , Facteur de stimulation des colonies de granulocytes et de macrophages/pharmacologie , Facteur de stimulation des colonies de macrophages/génétique , Régulation positive , Macrophages/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Anti-inflammatoires/métabolisme , Récepteurs hépatiques X/génétique , Récepteurs hépatiques X/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
7.
Development ; 150(6)2023 03 15.
Article de Anglais | MEDLINE | ID: mdl-36897571

RÉSUMÉ

Hormone secretion from pancreatic islets is essential for glucose homeostasis, and loss or dysfunction of islet cells is a hallmark of type 2 diabetes. Maf transcription factors are crucial for establishing and maintaining adult endocrine cell function. However, during pancreas development, MafB is not only expressed in insulin- and glucagon-producing cells, but also in Neurog3+ endocrine progenitor cells, suggesting additional functions in cell differentiation and islet formation. Here, we report that MafB deficiency impairs ß cell clustering and islet formation, but also coincides with loss of neurotransmitter and axon guidance receptor gene expression. Moreover, the observed loss of nicotinic receptor gene expression in human and mouse ß cells implied that signaling through these receptors contributes to islet cell migration/formation. Inhibition of nicotinic receptor activity resulted in reduced ß cell migration towards autonomic nerves and impaired ß cell clustering. These findings highlight a novel function of MafB in controlling neuronal-directed signaling events required for islet formation.


Sujet(s)
Diabète de type 2 , Cellules à insuline , Ilots pancréatiques , Souris , Adulte , Animaux , Humains , Glucagon/génétique , Glucagon/métabolisme , Diabète de type 2/métabolisme , Ilots pancréatiques/métabolisme , Insuline/métabolisme , Pancréas/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
8.
Int J Mol Sci ; 24(3)2023 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-36768908

RÉSUMÉ

Hydrocephalus has been observed in rats with spontaneous hypertension (SHRs). It has been demonstrated that activation of the oxidative stress related protein retinoic acid receptor alpha (RARα) has neuroprotective impacts. Our investigation aims to determine the potential role and mechanism of RARα in hydrocephalus. The RARα-specific agonist (Am80) and RARα inhibitor (AGN196996) were used to investigate the role of RARα in cerebrospinal fluid (CSF) secretion in the choroid plexus of SHRs. Evaluations of CSF secretion, ventricular volume, Western blotting, and immunofluorescent staining were performed. Hydrocephalus and CSF hypersecretion were identified in SHRs but not in Wistar-Kyoto rats, occurring at the age of 7 weeks. The RARα/MAFB/MSR1 pathway was also activated in SHRs. Therapy with Am80 beginning in week 5 decreased CSF hypersecretion, hydrocephalus development, and pathological changes in choroid plexus alterations by week 7. AGN196996 abolished the effect of Am80. In conclusion, activation of the RARα attenuated CSF hypersecretion to inhibit hydrocephalus development via regulating the MAFB/MSR1 pathway. RARα may act as a possible therapeutic target for hydrocephalus.


Sujet(s)
Hydrocéphalie , Hypertension artérielle , Animaux , Rats , Plexus choroïde/métabolisme , Hydrocéphalie/métabolisme , Hypertension artérielle/métabolisme , Facteur de transcription MafB/métabolisme , Protéines oncogènes/métabolisme , Rats de lignée SHR , Rats de lignée WKY , Récepteurs éboueurs de classe A/métabolisme
9.
Intern Med ; 62(1): 11-19, 2023 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-35249929

RÉSUMÉ

The increasing number of patients with chronic kidney disease (CKD) is being recognized as an emerging global health problem. Recently, it has become clear that injury and loss of glomerular visceral epithelial cells, known as podocytes, is a common early event in many forms of CKD. Podocytes are highly specialized epithelial cells that cover the outer layer of the glomerular basement membrane. They serve as the final barrier to urinary protein loss through the formation and maintenance of specialized foot-processes and an interposed slit-diaphragm. We previously reported that the transcription factor MafB regulates the podocyte slit diaphragm protein production and transcription factor Tcf21. We showed that the forced expression of MafB was able to prevent CKD. In this review, we discuss recent advances and offer an updated overview of the functions of podocyte-specific transcription factors in kidney biology, aiming to present new perspectives on the progression of CKD and respective therapeutic strategies.


Sujet(s)
Podocytes , Insuffisance rénale chronique , Humains , Facteurs de transcription/génétique , Membrane basale glomérulaire , Cellules épithéliales , Insuffisance rénale chronique/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
10.
Front Immunol ; 13: 1050494, 2022.
Article de Anglais | MEDLINE | ID: mdl-36578483

RÉSUMÉ

Cre-LoxP system has been widely used to induce recombination of floxed genes of interest. Currently available macrophage promoter-specific Cre recombinase mice strains have various limitations that warrants the testing of additional Cre strains. V-maf musculoaponeurotic fibrosarcoma oncogene family, protein b -Cre (Mafb-Cre) mice label macrophages in most organs such as spleen, small intestine, lung, bone marrow, and peritoneal cavity. However, whether Mafb-Cre recombinase targets the gene recombination in alveolar macrophage remains untested. Here, we utilized MafbCre/WTR26mTmG/WT strain that expresses mTOM protein in all the cells of mouse body except for those that express Mafb-Cre-regulated mEGFP. We performed fluorescent microscopy and flow cytometry to analyze mTOM and mEGFP expression in alveolar macrophages from MafbCre/WTR26mTmG/WT mice. Our analyses revealed that the Mafb-Cre is active in only ~40% of the alveolar macrophages in an age-independent manner. While Mafb- (mTOM+/mEGFP-) and Mafb+ (mEGFP+) alveolar macrophages exhibit comparable expression of CD11b and CD11c surface markers, the surface expression of MHCII is elevated in the Mafb+ (mEGFP+) macrophages. The bone marrow-derived macrophages from MafbCre/WTR26mTmG/WT mice are highly amenable to Cre-LoxP recombination in vitro. The bone marrow depletion and reconstitution experiment revealed that ~98% of alveolar macrophages from MafbCre/WTR26mTmG/WT → WT chimera are amenable to the Mafb-Cre-mediated recombination. Finally, the Th2 stimulation and ozone exposure to the MafbCre/WTR26mTmG/WT mice promote the Mafb-Cre-mediated recombination in alveolar macrophages. In conclusion, while the Mafb-/Mafb+ dichotomy thwarts the use of Mafb-Cre for the induction of floxed alleles in the entire alveolar macrophage population, this strain provides a unique tool to induce gene deletion in alveolar macrophages that encounter Th2 microenvironment in the lung airspaces.


Sujet(s)
Macrophages alvéolaires , Macrophages , Souris , Animaux , Macrophages alvéolaires/métabolisme , Macrophages/métabolisme , Poumon/métabolisme , Régions promotrices (génétique) , Protéines/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme
11.
J Orthop Surg Res ; 17(1): 428, 2022 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-36153605

RÉSUMÉ

BACKGROUND: Circular RNAs are involved in various cellular processes of bone diseases by acting as miRNA sponges to regulate gene expression levels, including osteosarcoma (OS). This research concentrated on the molecular mechanism of circ_0051079 in OS progression. METHODS: Reverse transcription-quantitative polymerase chain reaction assay was used for expression detection of circ_0051079, microRNA-1286 (miR-1286), and musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB). Cell Counting Kit-8 assay and Edu assay were used for cell proliferation analysis. Cell apoptosis was evaluated using flow cytometry. Western blot was performed to measure protein levels. Migration and invasion were assessed via transwell assay. Interaction of circ_0051079/miR-1286 or miR-1286/MAFB was explored through a dual-luciferase reporter assay. In vivo research was carried out via tumor xenograft assay and immunohistochemistry staining. RESULTS: Circ_0051079 expression was upregulated in OS. Downregulation of circ_0051079 reduced OS cell proliferation, migration, invasion, and accelerated apoptosis. Circ_0051079 interacted with miR-1286, and the tumor-inhibitory function of si-circ_0051079 was abolished by miR-1286 inhibition in OS cells. MAFB served as a target for miR-1286. OS cell progression was suppressed by miR-1286 overexpression via downregulating MAFB. Circ_0051079/miR-1286 resulted in expression change of MAFB in OS cells. Silencing circ_0051079 inhibited tumor growth in vivo via regulating the miR-1286/MAFB axis. CONCLUSION: The collective results elucidated that circ_0051079 contributed to OS progression via miR-1286-mediated upregulation of MAFB, confirming the interaction of circ_0051079/miR-1286/MAFB axis in OS.


Sujet(s)
Tumeurs osseuses , Facteur de transcription MafB , microARN , Ostéosarcome , ARN circulaire , Tumeurs osseuses/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Humains , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , microARN/génétique , Oncogènes , Ostéosarcome/anatomopathologie , ARN circulaire/génétique , Régulation positive/génétique
12.
Curr Neurovasc Res ; 19(3): 245-254, 2022.
Article de Anglais | MEDLINE | ID: mdl-35927915

RÉSUMÉ

BACKGROUND: Ischemic brain injury often results in irreversible pyroptosis of neurons. Sevoflurane (Sevo) post-treatment exerts an alleviative role in neuroinflammation. OBJECTIVES: This work evaluated the mechanism of Sevo post-treatment in oxygen-glucose deprivation (OGD)-induced pyroptosis of rat hippocampal neurons. METHODS: Rat hippocampal neuron cell line H19-7 cells were treated with OGD, followed by posttreatment of 2% Sevo. The expression patterns of Mafb ZIP Transcription Factor B (Mafb) and dual- specificity phosphatase 14 (DUSP14) were determined via quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting methods. H19-7 cell viability and the release of lactate dehydrogenase (LDH) were examined via the cell counting kit-8 and LDH assay kits. Levels of pyroptosis-related proteins and cytokines NOD-like receptor family, pyrin domain containing 3 (NLRP3), N-term cleaved Gasdermin-D (GSDMD-N), cleaved-caspase-1, interleukin (IL)-1ß, and IL-18 were also examined. The binding relation between Mafb and the DUSP14 promoter was detected. Besides, the roles of Mafb/DUSP14 in OGD-induced pyroptosis of rat hippocampal neurons were investigated through functional rescue experiments. RESULTS: Mafb and DUSP14 expression levels were decreased in OGD-induced hippocampal neurons. Sevo post-treatment up-regulated Mafb and DUSP14, facilitated H19-7 cell viability, inhibited LDH release, and reduced levels of NLRP3, GSDMD-N, cleaved-caspase-1, IL-1ß, and IL-18. Mafb increased DUSP14 expression via binding to the DUSP14 promoter. Repressing Mafb or DUSP14 exacerbated pyroptosis of hippocampal neurons. CONCLUSION: Sevo post-treatment increased Mafb and DUSP14 expressions, which repressed OGDinduced pyroptosis of hippocampal neurons.


Sujet(s)
Interleukine-18 , Pyroptose , Rats , Animaux , Pyroptose/physiologie , Interleukine-18/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Oxygène/métabolisme , Glucose/métabolisme , Sévoflurane , Caspase-1/métabolisme , Neurones/métabolisme , Hippocampe/métabolisme , Protéines oncogènes/métabolisme , Facteur de transcription MafB/métabolisme
13.
Exp Cell Res ; 416(1): 113158, 2022 07 01.
Article de Anglais | MEDLINE | ID: mdl-35430273

RÉSUMÉ

Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant diseases in the world. Although the somatic alterations have been fully identified, there are still no targeted drugs at present. Our previous studies revealed that loss of grand H3K27me3 domains mediated transcriptional activation of a series of genes in ESCC. Among them, we focus on the investigation of MAFB, as its high expression is associated with a poor prognosis in ESCC. Functional assays show that knockdown of MAFB significantly suppresses cell growth, migration and invasion. Mechanistic investigation demonstrates that MAFB exerts its function by upregulating IGFBP6. Our findings suggest that MAFB may play a tumor-promoting role and may act as a potential therapeutic target for ESCC.


Sujet(s)
Carcinome épidermoïde , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Humains , Carcinome épidermoïde/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Tumeurs de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie , Régulation de l'expression des gènes tumoraux/génétique , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Invasion tumorale/génétique , Phénotype , Protéine-6 de liaison aux IGF
14.
Front Immunol ; 13: 839796, 2022.
Article de Anglais | MEDLINE | ID: mdl-35237277

RÉSUMÉ

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a life-threatening subtype of stroke with high rates of mortality. In the early stages of SAH, neuroinflammation is one of the important mechanisms leading to brain injury after SAH. In various central nervous system diseases, activation of RARα receptor has been proven to demonstrate neuroprotective effects. This study aimed to investigate the anti-inflammatory effects of RARα receptor activation after SAH. METHODS: Internal carotid artery puncture method used to established SAH model in Sprague-Dawley rats. The RARα specific agonist Am80 was injected intraperitoneally 1 hour after SAH. AGN196996 (specific RARα inhibitor), Msr1 siRNA and LY294002 (PI3K-Akt inhibitor) were administered via the lateral ventricle before SAH. Evaluation SAH grade, neurological function score, blood-brain barrier permeability. BV2 cells and SH-SY5Y cells were co-cultured and stimulated by oxyhemoglobin to establish an in vitro model of SAH. RT-PCR, Western blotting, and immunofluorescence staining were used to investigate pathway-related proteins, microglia activation and inflammatory response. Results: The expression of RARα, Mafb, and Msr1 increased in rat brain tissue after SAH. Activation of the RARα receptor with Am80 improved neurological deficits and attenuated brain edema, blood brain barrier permeability. Am80 increased the expression of Mafb and Msr1, and reduced neuroinflammation by enhancing the phosphorylation of Akt and by inhibiting the phosphorylation of NF-κB. AGN196996, Msr1 siRNA, and LY294002 reversed the therapeutic effects of Am80 by reducing the expression of Msr1 and the phosphorylation of Akt. In vitro model of SAH, Am80 promoted M1-to-M2 phenotypic polarization in microglia and suppressed the nuclear transcription of NF-κB. CONCLUSION: Activation of the RARα receptor attenuated neuroinflammation by promoting M1-to-M2 phenotypic polarization in microglia and regulating the Mafb/Msr1/PI3K-Akt/NF-κB pathway. RARα might serve as a potential target for SAH therapy.


Sujet(s)
Facteur de transcription NF-kappa B , Hémorragie meningée , Animaux , Facteur de transcription MafB/métabolisme , Microglie/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Maladies neuro-inflammatoires , Protéines oncogènes , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Petit ARN interférent/métabolisme , Rats , Rat Sprague-Dawley , Transduction du signal , Hémorragie meningée/traitement médicamenteux , Hémorragie meningée/métabolisme
15.
Andrologia ; 54(6): e14411, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35220623

RÉSUMÉ

Mafb plays a significant role in the development and differentiation of various organs, tissues and cells. Nevertheless, its role in the control of external genital cell proliferation and function in the mechanism of hypospadias remains unknown. In this study, the expression of Mafb in foreskin fibroblasts was inhibited by siRNA. The Cell Count Kit-8 (CCK-8) assay showed cell proliferation increased after transfection, and the number of cells entered the S phase significantly increased via flow cytometry. Both mRNA and protein levels of cyclin E, cyclin-dependent kinase 2 (CDK2) and proliferating cell nuclear antigen (PCNA) were significantly upregulated in the siRNA group. Meanwhile, twenty-five prepuce tissue samples were collected from hypospadias repair surgery. These samples were divided into two groups: the severe and mild groups. Normal prepuce tissue specimens were obtained during circumcision as the normal control. The upregulated expression of cyclin E, CDK2 and PCNA and downregulated Mafb expression were observed in the hypospadias group. This study reveals for the first time that the reduction in Mafb promotes the foreskin fibroblast proliferation. Thus, downregulated Mafb expression may cause hypospadias by upregulating CDK2, cyclin E and PCNA. These findings can shed new light on the embryonic development of the urethra.


Sujet(s)
Prépuce , Hypospadias , Facteur de transcription MafB , Prolifération cellulaire , Cycline E/génétique , Cycline E/métabolisme , Kinase-2 cycline-dépendante/génétique , Kinase-2 cycline-dépendante/métabolisme , Fibroblastes/métabolisme , Prépuce/métabolisme , Humains , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Mâle , Antigène nucléaire de prolifération cellulaire/génétique , Antigène nucléaire de prolifération cellulaire/métabolisme , Petit ARN interférent/métabolisme , Régulation positive
16.
Acta Physiol (Oxf) ; 234(2): e13761, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34978761

RÉSUMÉ

AIMS: Reduced expression of exocytotic genes is associated with functional defects in insulin exocytosis contributing to impaired insulin secretion and type 2 diabetes (T2D) development. MAFA and MAFB transcription factors regulate ß-cell physiology, and their gene expression is reduced in T2D ß cells. We investigate if loss of MAFA and MAFB in human ß cells contributes to T2D progression by regulating genes required for insulin exocytosis. METHODS: Three approaches were performed: (1) RNAseq analysis with the focus on exocytosis-related genes in MafA-/- mouse islets, (2) correlational analysis between MAFA, MAFB and exocytosis-related genes in human islets and (3) MAFA and MAFB silencing in human islets and EndoC-ßH1 cells followed by functional in vitro studies. RESULTS: The expression of 30 exocytosis-related genes was significantly downregulated in MafA-/- mouse islets. In human islets, the expression of 29 exocytosis-related genes correlated positively with MAFA and MAFB. Eight exocytosis-related genes were downregulated in MafA-/- mouse islets and positively correlated with MAFA and MAFB in human islets. From this analysis, the expression of RAB3A, STXBP1, UNC13A, VAMP2, NAPA, NSF, STX1A and SYT7 was quantified after acute MAFA or MAFB silencing in EndoC-ßH1 cells and human islets. MAFA and MAFB silencing resulted in impaired insulin secretion and reduced STX1A, SYT7 and STXBP1 (EndoC-ßH1) and STX1A (human islets) mRNA expression. STX1A and STXBP1 protein expression was also impaired in islets from T2D donors which lack MAFA expression. CONCLUSION: Our data indicate that STXBP1 and STX1A are important MAFA/B-regulated exocytosis genes which may contribute to insulin exocytosis defects observed in MAFA-deficient human T2D ß cells.


Sujet(s)
Diabète de type 2 , Cellules à insuline , Animaux , Diabète de type 2/génétique , Diabète de type 2/métabolisme , Exocytose , Humains , Insuline/métabolisme , Sécrétion d'insuline , Cellules à insuline/métabolisme , Grandes protéines des facteurs de transcription Maf/génétique , Grandes protéines des facteurs de transcription Maf/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Souris
17.
Nucleic Acids Res ; 50(1): 108-126, 2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-34893889

RÉSUMÉ

Glucocorticoids (GCs) exert potent anti-inflammatory effects in immune cells through the glucocorticoid receptor (GR). Dendritic cells (DCs), central actors for coordinating immune responses, acquire tolerogenic properties in response to GCs. Tolerogenic DCs (tolDCs) have emerged as a potential treatment for various inflammatory diseases. To date, the underlying cell type-specific regulatory mechanisms orchestrating GC-mediated acquisition of immunosuppressive properties remain poorly understood. In this study, we investigated the transcriptomic and epigenomic remodeling associated with differentiation to DCs in the presence of GCs. Our analysis demonstrates a major role of MAFB in this process, in synergy with GR. GR and MAFB both interact with methylcytosine dioxygenase TET2 and bind to genomic loci that undergo specific demethylation in tolDCs. We also show that the role of MAFB is more extensive, binding to thousands of genomic loci in tolDCs. Finally, MAFB knockdown erases the tolerogenic properties of tolDCs and reverts the specific DNA demethylation and gene upregulation. The preeminent role of MAFB is also demonstrated in vivo for myeloid cells from synovium in rheumatoid arthritis following GC treatment. Our results imply that, once directly activated by GR, MAFB plays a critical role in orchestrating the epigenomic and transcriptomic remodeling that define the tolerogenic phenotype.


Sujet(s)
Cellules dendritiques/immunologie , Épigenèse génétique , Tolérance immunitaire , Facteur de transcription MafB/métabolisme , Récepteurs aux glucocorticoïdes/métabolisme , Adulte , Cellules cultivées , Méthylation de l'ADN , Protéines de liaison à l'ADN/métabolisme , Dioxygenases/métabolisme , Femelle , Humains , Facteur de transcription MafB/génétique , Mâle , Adulte d'âge moyen
18.
Int Immunol ; 34(3): 159-172, 2022 02 23.
Article de Anglais | MEDLINE | ID: mdl-34734243

RÉSUMÉ

Type I IFNs (IFN-α and IFN-ß), immunomodulatory cytokines secreted from activated plasmacytoid dendritic cells (pDCs), contribute to the innate defense against pathogenic infections and the pathogenesis of the autoimmune disease psoriasis vulgaris. A previous study has shown that an E26 transformation-specific (Ets) family transcription factor Spi-B can transactivate the type I IFN promoter in synergy with IFN regulatory factor (IRF)-7 and is required for type I IFN production in pDCs. However, the mechanism of negative regulation of type I IFNs by pDCs remains unknown. In this study, we report that a basic leucine zipper (bZip) transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MafB) suppresses the induction of type I IFNs in pDCs. The elevated expression of MafB inhibited the transactivation of type I IFN genes in a dose-dependent manner. At the molecular level, MafB interacted with the Ets domain of Spi-B and interfered with IRF-7-Spi-B complexation. Decreased MafB mRNA expression and degradation of MafB protein in the early phase of immune responses led to the enhancement of type I IFNs in pDCs. In vivo studies indicated that MafB is involved in resistance against imiquimod-induced psoriasis-like skin inflammation. Overall, these findings demonstrate that MafB acts as a negative regulator of type I IFN induction in pDCs and plays an important role in maintaining immune homeostasis.


Sujet(s)
Interféron de type I , Psoriasis , Cellules dendritiques , Humains , Interféron de type I/métabolisme , Interféron alpha/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Régions promotrices (génétique)
19.
JCI Insight ; 6(18)2021 09 22.
Article de Anglais | MEDLINE | ID: mdl-34428183

RÉSUMÉ

Islet-enriched transcription factors (TFs) exert broad control over cellular processes in pancreatic α and ß cells, and changes in their expression are associated with developmental state and diabetes. However, the implications of heterogeneity in TF expression across islet cell populations are not well understood. To define this TF heterogeneity and its consequences for cellular function, we profiled more than 40,000 cells from normal human islets by single-cell RNA-Seq and stratified α and ß cells based on combinatorial TF expression. Subpopulations of islet cells coexpressing ARX/MAFB (α cells) and MAFA/MAFB (ß cells) exhibited greater expression of key genes related to glucose sensing and hormone secretion relative to subpopulations expressing only one or neither TF. Moreover, all subpopulations were identified in native pancreatic tissue from multiple donors. By Patch-Seq, MAFA/MAFB-coexpressing ß cells showed enhanced electrophysiological activity. Thus, these results indicate that combinatorial TF expression in islet α and ß cells predicts highly functional, mature subpopulations.


Sujet(s)
Cellules à glucagon/métabolisme , Cellules à insuline/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Adulte , Phénomènes électrophysiologiques , Expression des gènes , Cellules à glucagon/physiologie , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Humains , Insuline/métabolisme , Cellules à insuline/physiologie , Grandes protéines des facteurs de transcription Maf/génétique , Grandes protéines des facteurs de transcription Maf/métabolisme , Facteur de transcription MafB/génétique , Facteur de transcription MafB/métabolisme , Adulte d'âge moyen , Analyse de séquence d'ARN , Analyse sur cellule unique , Transcriptome , Jeune adulte
20.
Cells Dev ; 167: 203725, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34324991

RÉSUMÉ

Cardiac neural crest cells arise in the caudal hindbrain and then migrate to the heart through the pharyngeal arches. These cells contribute to the formation of the heart, including the outflow tract, and are unique to this neural crest population. MafB is a transcription factor expressed specifically in early migrating cardiac neural crest cells as well as in rhombomeres (r) 5 and 6. Here, we identified the regulatory region in the chicken genome controlling the expression of endogenous MafB transcripts and used these essential elements to express MafB in the cardiac neural crest in reporter assays. A reporter driven by this regulatory region was employed to trace the migration of these cells into the pharyngeal arches. This regulatory region demonstrated transcriptional activity in the cardiac neural crest but not in other neural crest cell subpopulations, such as the cranial and trunk cells. This study provides insights into the gene regulatory mechanisms that specify cardiac neural crest cells among neural crest cell populations.


Sujet(s)
Poulets/génétique , Régulation de l'expression des gènes au cours du développement , Facteur de transcription MafB/génétique , Myocarde/métabolisme , Crête neurale/embryologie , Crête neurale/métabolisme , Séquences d'acides nucléiques régulatrices/génétique , Animaux , Protéines aviaires/métabolisme , Région branchiale/métabolisme , Mouvement cellulaire/génétique , Séquence conservée/génétique , ADN intergénique/génétique , Embryon non mammalien/métabolisme , Développement embryonnaire/génétique , Génome , Protéines à fluorescence verte/métabolisme , Facteur de transcription MafB/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE