Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Nature ; 618(7964): 365-373, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225978

RESUMO

Birth presents a metabolic challenge to cardiomyocytes as they reshape fuel preference from glucose to fatty acids for postnatal energy production1,2. This adaptation is triggered in part by post-partum environmental changes3, but the molecules orchestrating cardiomyocyte maturation remain unknown. Here we show that this transition is coordinated by maternally supplied γ-linolenic acid (GLA), an 18:3 omega-6 fatty acid enriched in the maternal milk. GLA binds and activates retinoid X receptors4 (RXRs), ligand-regulated transcription factors that are expressed in cardiomyocytes from embryonic stages. Multifaceted genome-wide analysis revealed that the lack of RXR in embryonic cardiomyocytes caused an aberrant chromatin landscape that prevented the induction of an RXR-dependent gene expression signature controlling mitochondrial fatty acid homeostasis. The ensuing defective metabolic transition featured blunted mitochondrial lipid-derived energy production and enhanced glucose consumption, leading to perinatal cardiac dysfunction and death. Finally, GLA supplementation induced RXR-dependent expression of the mitochondrial fatty acid homeostasis signature in cardiomyocytes, both in vitro and in vivo. Thus, our study identifies the GLA-RXR axis as a key transcriptional regulatory mechanism underlying the maternal control of perinatal cardiac metabolism.


Assuntos
Ácidos Graxos , Glucose , Coração , Leite Humano , Ácido gama-Linolênico , Feminino , Humanos , Recém-Nascido , Gravidez , Cromatina/genética , Ácidos Graxos/metabolismo , Ácido gama-Linolênico/metabolismo , Ácido gama-Linolênico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Coração/efeitos dos fármacos , Coração/embriologia , Coração/crescimento & desenvolvimento , Homeostase , Técnicas In Vitro , Leite Humano/química , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptores X de Retinoides/metabolismo , Fatores de Transcrição/metabolismo
2.
Mol Cell ; 81(5): 953-968.e9, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33503407

RESUMO

While the role of transcription factors and coactivators in controlling enhancer activity and chromatin structure linked to gene expression is well established, the involvement of corepressors is not. Using inflammatory macrophage activation as a model, we investigate here a corepressor complex containing GPS2 and SMRT both genome-wide and at the Ccl2 locus, encoding the chemokine CCL2 (MCP-1). We report that corepressors co-occupy candidate enhancers along with the coactivators CBP (H3K27 acetylase) and MED1 (mediator) but act antagonistically by repressing eRNA transcription-coupled H3K27 acetylation. Genome editing, transcriptional interference, and cistrome analysis reveals that apparently related enhancer and silencer elements control Ccl2 transcription in opposite ways. 4C-seq indicates that corepressor depletion or inflammatory signaling functions mechanistically similarly to trigger enhancer activation. In ob/ob mice, adipose tissue macrophage-selective depletion of the Ccl2 enhancer-transcribed eRNA reduces metaflammation. Thus, the identified corepressor-eRNA-chemokine pathway operates in vivo and suggests therapeutic opportunities by targeting eRNAs in immuno-metabolic diseases.


Assuntos
Quimiocina CCL2/genética , Proteínas Correpressoras/genética , Elementos Facilitadores Genéticos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Correpressor 2 de Receptor Nuclear/genética , Obesidade/genética , Elementos Silenciadores Transcricionais , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Animais , Sistemas CRISPR-Cas , Quimiocina CCL2/imunologia , Proteínas Correpressoras/imunologia , Edição de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Histona Acetiltransferases/genética , Histona Acetiltransferases/imunologia , Histonas/genética , Histonas/imunologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Subunidade 1 do Complexo Mediador/genética , Subunidade 1 do Complexo Mediador/imunologia , Camundongos , Camundongos Obesos , Correpressor 2 de Receptor Nuclear/imunologia , Obesidade/imunologia , Obesidade/patologia , Células RAW 264.7 , RNA não Traduzido/genética , RNA não Traduzido/imunologia , Transdução de Sinais
3.
Nucleic Acids Res ; 51(3): 1067-1086, 2023 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-36610795

RESUMO

The Th2 cytokine interleukin 4 (IL4) promotes macrophage differentiation into alternative subtypes and plays important roles in physiology, in metabolic and inflammatory diseases, in cancer and in tissue regeneration. While the regulatory transcription factor networks governing IL4 signaling are already well-characterized, it is currently less understood which transcriptional coregulators are involved and how they operate mechanistically. In this study, we discover that G protein pathway suppressor 2 (GPS2), a core subunit of the HDAC3 corepressor complex assembled by SMRT and NCOR, represses IL4-dependent enhancer activation in mouse macrophages. Our genome-wide and gene-specific characterization revealed that, instead of directly repressing STAT6, chromatin-bound GPS2 cooperates with SMRT and NCOR to antagonize enhancer activation by lysine demethylase 1A (KDM1A, LSD1). Mechanistically, corepressor depletion increased KDM1A recruitment to enhancers linked to IL4-induced genes, accompanied by demethylation of the repressive histone marks H3K9me2/3 without affecting H3K4me1/2, the classic KDM1A substrates for demethylation in other cellular contexts. This in turn caused enhancer and gene activation already in the absence of IL4/STAT6 and sensitized the STAT6-dependent IL4 responsiveness of macrophages. Thus, our work identified with the antagonistic action of a GPS2-containing corepressor complex and the lysine demethylase KDM1A a hitherto unknown epigenetic corepressor-coactivator switching mechanism that governs alternative macrophage activation.


Assuntos
Histona Desmetilases , Animais , Camundongos , Proteínas Correpressoras , Histona Desmetilases/genética , Interleucina-4 , Peptídeos e Proteínas de Sinalização Intracelular , Lisina , Ativação de Macrófagos
5.
Arterioscler Thromb Vasc Biol ; 42(5): 659-676, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35321563

RESUMO

BACKGROUND: Understanding the processes behind carotid plaque instability is necessary to develop methods for identification of patients and lesions with stroke risk. Here, we investigated molecular signatures in human plaques stratified by echogenicity as assessed by duplex ultrasound. METHODS: Lesion echogenicity was correlated to microarray gene expression profiles from carotid endarterectomies (n=96). The findings were extended into studies of human and mouse atherosclerotic lesions in situ, followed by functional investigations in vitro in human carotid smooth muscle cells (SMCs). RESULTS: Pathway analyses highlighted muscle differentiation, iron homeostasis, calcification, matrix organization, cell survival balance, and BCLAF1 (BCL2 [B-cell lymphoma 2]-associated transcription factor 1) as the most significant signatures. BCLAF1 was downregulated in echolucent plaques, positively correlated to proliferation and negatively to apoptosis. By immunohistochemistry, BCLAF1 was found in normal medial SMCs. It was repressed early during atherogenesis but reappeared in CD68+ cells in advanced plaques and interacted with BCL2 by proximity ligation assay. In cultured SMCs, BCLAF1 was induced by differentiation factors and mitogens and suppressed by macrophage-conditioned medium. BCLAF1 silencing led to downregulation of BCL2 and SMC markers, reduced proliferation, and increased apoptosis. Transdifferentiation of SMCs by oxLDL (oxidized low-denisty lipoprotein) was accompanied by upregulation of BCLAF1, CD36, and CD68, while oxLDL exposure with BCLAF1 silencing preserved MYH (myosin heavy chain) 11 expression and prevented transdifferentiation. BCLAF1 was associated with expression of cell differentiation, contractility, viability, and inflammatory genes, as well as the scavenger receptors CD36 and CD68. BCLAF1 expression in CD68+/BCL2+ cells of SMC origin was verified in plaques from MYH11 lineage-tracing atherosclerotic mice. Moreover, BCLAF1 downregulation associated with vulnerability parameters and cardiovascular risk in patients with carotid atherosclerosis. CONCLUSIONS: Plaque echogenicity correlated with enrichment of distinct molecular pathways and identified BCLAF1, previously not described in atherosclerosis, as the most significant gene. Functionally, BCLAF1 seems necessary for survival and transdifferentiation of SMCs into a macrophage-like phenotype. The role of BCLAF1 in plaque vulnerability should be further evaluated.


Assuntos
Aterosclerose , Placa Aterosclerótica , Proteínas Repressoras/metabolismo , Animais , Aterosclerose/diagnóstico por imagem , Aterosclerose/genética , Aterosclerose/metabolismo , Transdiferenciação Celular , Humanos , Lipídeos , Camundongos , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Repressoras/genética , Transcriptoma , Proteínas Supressoras de Tumor/genética , Ultrassonografia
6.
FASEB J ; 33(2): 1631-1643, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30153049

RESUMO

Macrophages play important roles in linking alterations of cholesterol metabolism and inflammation to the development of atherosclerosis. Previous studies have identified several positive and negative crosstalk mechanisms that connect cholesterol efflux and inflammation at the transcriptional level. Of particular relevance is that the expression of ATP-binding cassette transporter A1 ( Abca1), a main regulator of cholesterol efflux, can be induced by oxysterol receptor LXR agonists but also by bacterial endotoxins, such as LPS, that activate TLR4 signaling. However, the extent to which these pathways influence each other has remained incompletely understood. We investigated the possible role of the transcriptional coregulator G protein pathway suppressor 2 (GPS2) in LPS-induced Abca1 expression and cholesterol efflux in mouse and human macrophages. To activate Abca1, GPS2 cooperates with the LPS-inducible NF-κB subunit p65, but not with LXRs nor with corepressor complex subunits that otherwise cooperate with GPS2 to repress proinflammatory gene expression. Overall, our work identifies a regulatory chromatin component of crosstalk mechanisms between cholesterol efflux and inflammation that specifically affects ABCA1. Because GPS2 expression is down-regulated in some humans with obese and type 2 diabetes, the macrophage GPS-2/ABC-A1 pathway could be altered and contribute to atherogenesis.-Huang, Z., Liang, N., Damdimopoulos, A., Fan, R., Treuter, E. G protein pathway suppressor 2 (GPS2) links inflammation and cholesterol efflux by controlling lipopolysaccharide-induced ATP-binding cassette transporter A1 expression in macrophages.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Colesterol/metabolismo , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Animais , Transporte Biológico , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Transdução de Sinais , Células THP-1 , Fator de Transcrição RelA/metabolismo , Transcrição Gênica
7.
Genes Dev ; 24(4): 381-95, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20159957

RESUMO

The orphan receptor LRH-1 and the oxysterol receptors LXRalpha and LXRbeta are established transcriptional regulators of lipid metabolism that appear to control inflammatory processes. Here, we investigate the anti-inflammatory actions of these nuclear receptors in the hepatic acute phase response (APR). We report that selective synthetic agonists induce SUMOylation-dependent recruitment of either LRH-1 or LXR to hepatic APR promoters and prevent the clearance of the N-CoR corepressor complex upon cytokine stimulation. Investigations of the APR in vivo, using LXR knockout mice, indicate that the anti-inflammatory actions of LXR agonists are triggered selectively by the LXRbeta subtype. We further find that hepatic APR responses in small ubiquitin-like modifier-1 (SUMO-1) knockout mice are increased, which is due in part to diminished LRH-1 action at APR promoters. Finally, we provide evidence that the metabolically important coregulator GPS2 functions as a hitherto unrecognized transrepression mediator of interactions between SUMOylated nuclear receptors and the N-CoR corepressor complex. Our study extends the knowledge of anti-inflammatory mechanisms and pathways directed by metabolic nuclear receptor-corepressor networks to the control of the hepatic APR, and implies alternative pharmacological strategies for the treatment of human metabolic diseases associated with inflammation.


Assuntos
Reação de Fase Aguda/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Fígado/imunologia , Receptores Nucleares Órfãos/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/imunologia , Animais , Anti-Inflamatórios/imunologia , Células COS , Chlorocebus aethiops , Feminino , Regulação da Expressão Gênica , Células HeLa , Humanos , Receptores X do Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Mol Cell ; 34(4): 510-8, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19481530

RESUMO

Transcriptional coregulators, rather than ligand signals, are suspected to confer context and pathway specificity to nuclear receptor signaling, but the identity of such specifying coregulators and the underlying molecular mechanisms remain largely enigmatic. Here we address this issue in metabolic oxysterol receptor LXR pathways and describe the selective requirement of GPS2 for ABCG1 cholesterol transporter gene transcription and cholesterol efflux from macrophages. We implicate GPS2 in facilitating LXR recruitment to an ABCG1-specific promoter/enhancer unit upon ligand activation and identify functional links to histone H3K9 demethylation. We further describe fundamental differences between ABCG1 and ABCA1 with regard to GPS2 in relation to other coregulators, which are likely to apply to additional LXR-regulated genes. Our work identifies a coregulator-dependent epigenetic mechanism governing the access of a nuclear receptor to communicating regulatory regions in the genome. The pathway and coregulator selectivity of this mechanism implies pharmacological possibilities for the development of selective LXR agonists.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Epistasia Genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Receptores X do Fígado , Macrófagos/citologia , Macrófagos/metabolismo , Receptores Nucleares Órfãos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Transcrição Gênica , Técnicas do Sistema de Duplo-Híbrido
9.
Handb Exp Pharmacol ; 233: 95-135, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25903414

RESUMO

Within the past two decades, coregulators have emerged as essential chromatin components of metabolic signaling by nuclear receptors and additional metabolite-sensing transcription factors. Intriguingly, coregulators themselves are efficient sensors and effectors of metabolic stimuli that modulate gene expression at different levels, often via post-translational modifications of histones or other factors. There is already evidence that alterations of expression or function of coregulators contributes to metabolic disease by propagating disease-specific epigenomes linked to the dysregulation of transcription and downstream pathways. In this chapter we review the current progress made in understanding the role of coregulators in metabolic pathways, with a particular emphasis on their study in vivo and in the context of metabolic disease.


Assuntos
Doenças Metabólicas/etiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Tecido Adiposo/metabolismo , Animais , Metabolismo Energético , Humanos , Resistência à Insulina , Músculo Esquelético/metabolismo , Especificidade de Órgãos , Processamento de Proteína Pós-Traducional , Transdução de Sinais
10.
Adv Sci (Weinh) ; 11(20): e2307201, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38549193

RESUMO

Macrophages regulate essential aspects of innate immunity against pathogens. In response to microbial components, macrophages activate primary and secondary inflammatory gene programs crucial for host defense. The liver X receptors (LXRα, LXRß) are ligand-dependent nuclear receptors that direct gene expression important for cholesterol metabolism and inflammation, but little is known about the individual roles of LXRα and LXRß in antimicrobial responses. Here, the results demonstrate that induction of LXRα transcription by prolonged exposure to lipopolysaccharide (LPS) supports inflammatory gene expression in macrophages. LXRα transcription is induced by NF-κB and type-I interferon downstream of TLR4 activation. Moreover, LPS triggers a reprogramming of the LXRα cistrome that promotes cytokine and chemokine gene expression through direct LXRα binding to DNA consensus sequences within cis-regulatory regions including enhancers. LXRα-deficient macrophages present fewer binding of p65 NF-κB and reduced histone H3K27 acetylation at enhancers of secondary inflammatory response genes. Mice lacking LXRα in the hematopoietic compartment show impaired responses to bacterial endotoxin in peritonitis models, exhibiting reduced neutrophil infiltration and decreased expansion and inflammatory activation of recruited F4/80lo-MHC-IIhi peritoneal macrophages. Together, these results uncover a previously unrecognized function for LXRα-dependent transcriptional cis-activation of secondary inflammatory gene expression in macrophages and the host response to microbial ligands.


Assuntos
Inflamação , Receptores X do Fígado , Macrófagos , Transcriptoma , Animais , Camundongos , Modelos Animais de Doenças , Inflamação/genética , Inflamação/metabolismo , Lipopolissacarídeos , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Camundongos Endogâmicos C57BL
11.
Sci Adv ; 10(1): eadi2689, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38170777

RESUMO

Defects in adipocyte lipolysis drive multiple aspects of cardiometabolic disease, but the transcriptional framework controlling this process has not been established. To address this, we performed a targeted perturbation screen in primary human adipocytes. Our analyses identified 37 transcriptional regulators of lipid mobilization, which we classified as (i) transcription factors, (ii) histone chaperones, and (iii) mRNA processing proteins. On the basis of its strong relationship with multiple readouts of lipolysis in patient samples, we performed mechanistic studies on one hit, ZNF189, which encodes the zinc finger protein 189. Using mass spectrometry and chromatin profiling techniques, we show that ZNF189 interacts with the tripartite motif family member TRIM28 and represses the transcription of an adipocyte-specific isoform of phosphodiesterase 1B (PDE1B2). The regulation of lipid mobilization by ZNF189 requires PDE1B2, and the overexpression of PDE1B2 is sufficient to attenuate hormone-stimulated lipolysis. Thus, our work identifies the ZNF189-PDE1B2 axis as a determinant of human adipocyte lipolysis and highlights a link between chromatin architecture and lipid mobilization.


Assuntos
Adipócitos , Mobilização Lipídica , Humanos , Adipócitos/metabolismo , Lipólise/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cromatina/genética , Cromatina/metabolismo
12.
J Clin Invest ; 133(20)2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37651203

RESUMO

Lung cancer progression relies on angiogenesis, which is a response to hypoxia typically coordinated by hypoxia-inducible transcription factors (HIFs), but growing evidence indicates that transcriptional programs beyond HIFs control tumor angiogenesis. Here, we show that the redox-sensitive transcription factor BTB and CNC homology 1 (BACH1) controls the transcription of a broad range of angiogenesis genes. BACH1 is stabilized by lowering ROS levels; consequently, angiogenesis gene expression in lung cancer cells, tumor organoids, and xenograft tumors increased substantially following administration of vitamins C and E and N-acetylcysteine in a BACH1-dependent fashion under normoxia. Moreover, angiogenesis gene expression increased in endogenous BACH1-overexpressing cells and decreased in BACH1-knockout cells in the absence of antioxidants. BACH1 levels also increased upon hypoxia and following administration of prolyl hydroxylase inhibitors in both HIF1A-knockout and WT cells. BACH1 was found to be a transcriptional target of HIF1α, but BACH1's ability to stimulate angiogenesis gene expression was HIF1α independent. Antioxidants increased tumor vascularity in vivo in a BACH1-dependent fashion, and overexpressing BACH1 rendered tumors sensitive to antiangiogenesis therapy. BACH1 expression in tumor sections from patients with lung cancer correlated with angiogenesis gene and protein expression. We conclude that BACH1 is an oxygen- and redox-sensitive angiogenesis transcription factor.


Assuntos
Antioxidantes , Fatores de Transcrição de Zíper de Leucina Básica , Neoplasias Pulmonares , Humanos , Antioxidantes/farmacologia , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Hipóxia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Animais , Camundongos
13.
Biochim Biophys Acta ; 1812(8): 909-18, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21172431

RESUMO

Nuclear receptors (NRs) exert crucial functions in controlling metabolism and inflammation by both positively and negatively regulating gene expression. Recent evidence suggests that the transcriptional activities of many NRs can be modulated and even re-directed through post-translational modification by small ubiquitin-related modifiers (SUMO). SUMOylation triggers a plethora of diverse molecular events that can alter both the fate and function of modified NRs at the nongenomic, genomic, and epigenomic level. However, it is the intriguing link of SUMOylation to transcriptional repression, and in particular to transrepression, that has emerged as a common underlying mechanism that impacts on biological processes controlled by NRs. It further appears that the cell-type-specific SUMOylation status of NRs can be regulated by ligands and by signal-dependent crosstalk of post-translational modifications. Given the causal role of altered NR signaling in the development and pathogenesis of human diseases, it is likely that aberrant SUMO conjugation, deconjugation, or interpretation contributes to these alterations. Here, we review the current progress made in both the study and understanding of the molecular mechanisms and consequences of NR SUMOylation and also discuss the physiological and pharmacological implications with a particular focus on transrepression pathways that link metabolism and inflammation. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.


Assuntos
Inflamação/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Sumoilação , Transcrição Gênica , Humanos
14.
BMC Genomics ; 13: 50, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22292898

RESUMO

BACKGROUND: The liver X receptors (LXRs) are oxysterol sensing nuclear receptors with multiple effects on metabolism and immune cells. However, the complete genome-wide cistrome of LXR in cells of human origin has not yet been provided. RESULTS: We performed ChIP-seq in phorbol myristate acetate-differentiated THP-1 cells (macrophage-type) after stimulation with the potent synthetic LXR ligand T0901317 (T09). Microarray gene expression analysis was performed in the same cellular model. We identified 1357 genome-wide LXR locations (FDR < 1%), of which 526 were observed after T09 treatment. De novo analysis of LXR binding sequences identified a DR4-type element as the major motif. On mRNA level T09 up-regulated 1258 genes and repressed 455 genes. Our results show that LXR actions are focused on 112 genomic regions that contain up to 11 T09 target genes per region under the control of highly stringent LXR binding sites with individual constellations for each region. We could confirm that LXR controls lipid metabolism and transport and observed a strong association with apoptosis-related functions. CONCLUSIONS: This first report on genome-wide binding of LXR in a human cell line provides new insights into the transcriptional network of LXR and its target genes with their link to physiological processes, such as apoptosis.The gene expression microarray and sequence data have been submitted collectively to the NCBI Gene Expression Omnibus http://www.ncbi.nlm.nih.gov/geo under accession number GSE28319.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Macrófagos/metabolismo , Receptores Nucleares Órfãos/metabolismo , Apoptose , Sítios de Ligação , Linhagem Celular , Regulação para Baixo , Humanos , Metabolismo dos Lipídeos , Receptores X do Fígado , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Nucleares Órfãos/antagonistas & inibidores , Ligação Proteica , Regulação para Cima
15.
STAR Protoc ; 3(2): 101338, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35496794

RESUMO

Chromosome conformation capture combined with high-throughput sequencing (4C-seq) is a powerful tool to map genomic DNA regions that communicate with a specific locus of interest such as functional single-nucleotide polymorphism (SNPs)-containing regions. This protocol describes detailed steps to perform 4C-seq in mouse macrophage RAW264.7 cells, starting from the primer design based on cistrome and epigenome data, sample processing, and to the bioinformatics analysis. For complete details on the use and execution of this protocol, please refer to Huang et al. (2021).


Assuntos
Epigenoma , Sequenciamento de Nucleotídeos em Larga Escala , Animais , Cromossomos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Macrófagos , Camundongos , Células RAW 264.7
16.
Hum Mol Genet ; 18(15): 2802-12, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19423554

RESUMO

Dyslexia, or specific reading disability, is the unexpected failure in learning to read and write when intelligence and senses are normal. One of the susceptibility genes, DYX1C1, has been implicated in neuronal migration, but little is known about its interactions and functions. As DYX1C1 was suggested to interact with the U-box protein CHIP (carboxy terminus of Hsc70-interacting protein), which also participates in the degradation of estrogen receptors alpha (ERalpha) and beta (ERbeta), we hypothesized that the effects of DYX1C1 might be at least in part mediated through the regulation of ERs. ERs have shown to be important in brain development and cognitive functions. Indeed, we show that DYX1C1 interacts with both ERs in the presence of 17beta-estradiol, as determined by co-localization, co-immunoprecipitation and proximity ligation assays. Protein levels of endogenous ERalpha or exogenous ERbeta were reduced upon over-expression of DYX1C1, resulting in decreased transcriptional responses to 17beta-estradiol. Furthermore, we detected in vivo complexes of DYX1C1 with ERalpha or ERbeta at endogenous levels along neurites of primary rat hippocampal neurons. Taken together, our data suggest that DYX1C1 is involved in the regulation of ERalpha and ERbeta, and may thus affect the brain development and regulate cognitive functions. These findings provide novel insights into the function of DYX1C1 and link neuronal migration and developmental dyslexia to the estrogen-signaling effects in the brain.


Assuntos
Proteínas de Transporte/metabolismo , Dislexia/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Células Cultivadas , Proteínas do Citoesqueleto , Dislexia/genética , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Humanos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/química , Neurônios/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Transporte Proteico , Ratos
17.
Obesity (Silver Spring) ; 29(12): 2013-2025, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34813171

RESUMO

The rising prevalence of obesity over the past decades coincides with the rising awareness that a detailed understanding of both adipose tissue biology and obesity-associated remodeling is crucial for developing therapeutic and preventive strategies. Substantial progress has been made in identifying the signaling pathways and transcriptional networks that orchestrate alterations of adipocyte gene expression linked to diverse phenotypes. Owing to recent advances in epigenomics, we also gained a better appreciation for the fact that different environmental cues can epigenetically reprogram adipocyte fate and function, mainly by altering DNA methylation and histone modification patterns. Intriguingly, it appears that transcription factors and chromatin-modifying coregulator complexes are the key regulatory components that coordinate both signaling-induced transcriptional and epigenetic alterations in adipocytes. In this review, we summarize and discuss current molecular insights into how these alterations and the involved regulatory components trigger adipogenesis and adipose tissue remodeling in response to energy surplus.


Assuntos
Adipócitos , Epigenômica , Adipócitos/metabolismo , Adipogenia/genética , Epigênese Genética , Humanos , Obesidade/genética , Obesidade/metabolismo
18.
Mol Cell Biol ; 27(4): 1407-24, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17145766

RESUMO

SHP has been implicated as a pleiotropic regulator of diverse biological functions by its ability to inhibit numerous nuclear receptors. Recently, we reported that SHP inhibits transcription of CYP7A1, a key gene in bile acid biosynthesis, by recruiting histone deacetylases (HDACs) and a Swi/Snf-Brm complex. To further delineate the mechanism of this inhibition, we have examined whether methylation of histones is also involved and whether a functional interplay between chromatin-modifying enzymes occurs. The histone methyltransferase G9a, but not SUV39, was colocalized with SHP in the nucleus and directly interacted with SHP in vitro. G9a, which was coimmunoprecipitated with hepatic SHP, methylated Lys-9 of histone 3 (H3K9) in vitro. Expression of G9a enhanced inhibition of CYP7A1 transcription by SHP, while a catalytically inactive G9a dominant negative (DN) mutant reversed the SHP inhibition. G9a was recruited to and H3K9 was methylated at the CYP7A1 promoter in a SHP-dependent manner in bile acid-treated HepG2 cells. Expression of the G9a-DN mutant inhibited H3K9 methylation, blocked the recruitment of the Brm complex, and partially reversed CYP7A1 inhibition by bile acids. Inhibition of HDAC activity with trichostatin A blocked deacetylation and methylation of H3K9 at the promoter, and, conversely, inhibition of H3K9 methylation by G9a-DN partially blocked deacetylation. Hepatic expression of G9a-DN in mice fed cholic acid disrupted bile acid homeostasis, resulting in increased bile acid pools and partial de-repression of Cyp7a1 and Cyp8b1. Our studies establish a critical role for G9a methyltransferase, histone deacetylases, and the Swi/Snf-Brm complex in the SHP-mediated inhibition of hepatic bile acid synthesis via coordinated chromatin modification at target genes.


Assuntos
Ácido Cólico/metabolismo , Cromatina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Fígado/enzimologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Acetilação/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Colesterol 7-alfa-Hidroxilase/genética , Ácido Cólico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histonas/química , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Lisina/metabolismo , Masculino , Metilação/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Metiltransferases , Transporte Proteico/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/genética , Esteroide 12-alfa-Hidroxilase/genética , Transcrição Gênica/efeitos dos fármacos
19.
Mol Metab ; 42: 101066, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32798719

RESUMO

OBJECTIVE: Adipogenesis is critical for adipose tissue remodeling during the development of obesity. While the role of transcription factors in the orchestration of adipogenic pathways is already established, the involvement of coregulators that transduce regulatory signals into epigenome alterations and transcriptional responses remains poorly understood. The aim of our study was to investigate which pathways are controlled by G protein pathway suppressor 2 (GPS2) during the differentiation of human adipocytes. METHODS: We generated a unique loss-of-function model by RNAi depletion of GPS2 in human multipotent adipose-derived stem (hMADS) cells. We thoroughly characterized the coregulator depletion-dependent pathway alterations during adipocyte differentiation at the level of transcriptome (RNA-seq), epigenome (ChIP-seq H3K27ac), cistrome (ChIP-seq GPS2), and lipidome. We validated the in vivo relevance of the identified pathways in non-diabetic and diabetic obese patients. RESULTS: The loss of GPS2 triggers the reprogramming of cellular processes related to adipocyte differentiation by increasing the responses to the adipogenic cocktail. In particular, GPS2 depletion increases the expression of BMP4, an important trigger for the commitment of fibroblast-like progenitors toward the adipogenic lineage and increases the expression of inflammatory and metabolic genes. GPS2-depleted human adipocytes are characterized by hypertrophy, triglyceride and phospholipid accumulation, and sphingomyelin depletion. These changes are likely a consequence of the increased expression of ATP-binding cassette subfamily G member 1 (ABCG1) that mediates sphingomyelin efflux from adipocytes and modulates lipoprotein lipase (LPL) activity. We identify ABCG1 as a direct transcriptional target, as GPS2 depletion leads to coordinated changes of transcription and H3K27 acetylation at promoters and enhancers that are occupied by GPS2 in wild-type adipocytes. We find that in omental adipose tissue of obese humans, GPS2 levels correlate with ABCG1 levels, type 2 diabetic status, and lipid metabolic status, supporting the in vivo relevance of the hMADS cell-derived in vitro data. CONCLUSION: Our study reveals a dual regulatory role of GPS2 in epigenetically modulating the chromatin landscape and gene expression during human adipocyte differentiation and identifies a hitherto unknown GPS2-ABCG1 pathway potentially linked to adipocyte hypertrophy in humans.


Assuntos
Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Adipócitos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células 3T3-L1 , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adipócitos/fisiologia , Adipogenia/fisiologia , Tecido Adiposo/metabolismo , Adulto , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Masculino , Camundongos , Obesidade/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo
20.
Cell Rep ; 32(11): 108141, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32937117

RESUMO

Glucose homeostasis is maintained through organ crosstalk that regulates secretion of insulin to keep blood glucose levels within a physiological range. In type 2 diabetes, this coordinated response is altered, leading to a deregulation of beta cell function and inadequate insulin secretion. Reprogramming of white adipose tissue has a central role in this deregulation, but the critical regulatory components remain unclear. Here, we demonstrate that expression of the transcriptional coregulator GPS2 in white adipose tissue is correlated with insulin secretion rate in humans. The causality of this relationship is confirmed using adipocyte-specific GPS2 knockout mice, in which inappropriate secretion of insulin promotes glucose intolerance. This phenotype is driven by adipose-tissue-secreted factors, which cause increased pancreatic islet inflammation and impaired beta cell function. Thus, our study suggests that, in mice and in humans, GPS2 controls the reprogramming of white adipocytes to influence pancreatic islet function and insulin secretion.


Assuntos
Tecido Adiposo Branco/metabolismo , Células Secretoras de Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Adipócitos Brancos/metabolismo , Tecido Adiposo/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Inflamação/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética , Secreção de Insulina/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA