Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JHEP Rep ; 5(2): 100615, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36687468

RESUMO

Background & Aims: Histological assessment of liver biopsies is the gold standard for diagnosis of non-alcoholic steatohepatitis (NASH), the progressive form of non-alcoholic fatty liver disease (NAFLD), despite its well-established limitations. Therefore, non-invasive biomarkers that can offer an integrated view of the liver are needed to improve diagnosis and reduce sampling bias. Hepatic stellate cells (HSCs) are central in the development of hepatic fibrosis, a hallmark of NASH. Secreted HSC-specific proteins may, therefore, reflect disease state in the NASH liver and serve as non-invasive diagnostic biomarkers. Methods: We performed RNA-sequencing on liver biopsies from a histologically characterised cohort of obese patients (n = 30, BMI >35 kg/m2) to identify and evaluate HSC-specific genes encoding secreted proteins. Bioinformatics was used to identify potential biomarkers and their expression at single-cell resolution. We validated our findings using single-molecule fluorescence in situ hybridisation (smFISH) and ELISA to detect mRNA in liver tissue and protein levels in plasma, respectively. Results: Hepatic expression of SPARC-related modular calcium-binding protein 2 (SMOC2) was increased in NASH compared to no-NAFLD (p.adj <0.001). Single-cell RNA-sequencing data indicated that SMOC2 was primarily expressed by HSCs, which was validated using smFISH. Finally, plasma SMOC2 was elevated in NASH compared to no-NAFLD (p <0.001), with a predictive accuracy of AUROC 0.88. Conclusions: Increased SMOC2 in plasma appears to reflect HSC activation, a key cellular event associated with NASH progression, and may serve as a non-invasive biomarker of NASH. Impact and implications: Non-alcoholic fatty liver disease (NAFLD) and its progressive form, non-alcoholic steatohepatitis (NASH), are the most common forms of chronic liver diseases. Currently, liver biopsies are the gold standard for diagnosing NAFLD. Blood-based biomarkers to complement liver biopsies for diagnosis of NAFLD are required. We found that activated hepatic stellate cells, a cell type central to NAFLD pathogenesis, upregulate expression of the secreted protein SPARC-related modular calcium-binding protein 2 (SMOC2). SMOC2 was elevated in blood samples from patients with NASH and may hold promise as a blood-based biomarker for the diagnosis of NAFLD.

2.
Nat Struct Mol Biol ; 29(12): 1148-1158, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36482255

RESUMO

Enhancers play a central role in the spatiotemporal control of gene expression and tend to work in a cell-type-specific manner. In addition, they are suggested to be major contributors to phenotypic variation, evolution and disease. There is growing evidence that enhancer dysfunction due to genetic, structural or epigenetic mechanisms contributes to a broad range of human diseases referred to as enhanceropathies. Such mechanisms often underlie the susceptibility to common diseases, but can also play a direct causal role in cancer or Mendelian diseases. Despite the recent gain of insights into enhancer biology and function, we still have a limited ability to predict how enhancer dysfunction impacts gene expression. Here we discuss the major challenges that need to be overcome when studying the role of enhancers in disease etiology and highlight opportunities and directions for future studies, aiming to disentangle the molecular basis of enhanceropathies.


Assuntos
Elementos Facilitadores Genéticos , Epigênese Genética , Humanos , Elementos Facilitadores Genéticos/genética
3.
J Neurosci ; 41(33): 7148-7159, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34210784

RESUMO

Following stroke, the survival of neurons and their ability to reestablish connections is critical to functional recovery. This is strongly influenced by the balance between neuronal excitation and inhibition. In the acute phase of experimental stroke, lethal hyperexcitability can be attenuated by positive allosteric modulation of GABAA receptors (GABAARs). Conversely, in the late phase, negative allosteric modulation of GABAAR can correct the suboptimal excitability and improves both sensory and motor recovery. Here, we hypothesized that octadecaneuropeptide (ODN), an endogenous allosteric modulator of the GABAAR synthesized by astrocytes, influences the outcome of ischemic brain tissue and subsequent functional recovery. We show that ODN boosts the excitability of cortical neurons, which makes it deleterious in the acute phase of stroke. However, if delivered after day 3, ODN is safe and improves motor recovery over the following month in two different paradigms of experimental stroke in mice. Furthermore, we bring evidence that, during the subacute period after stroke, the repairing cortex can be treated with ODN by means of a single hydrogel deposit into the stroke cavity.SIGNIFICANCE STATEMENT Stroke remains a devastating clinical challenge because there is no efficient therapy to either minimize neuronal death with neuroprotective drugs or to enhance spontaneous recovery with neurorepair drugs. Around the brain damage, the peri-infarct cortex can be viewed as a reservoir of plasticity. However, the potential of wiring new circuits in these areas is restrained by a chronic excess of GABAergic inhibition. Here we show that an astrocyte-derived peptide, can be used as a delayed treatment, to safely correct cortical excitability and facilitate sensorimotor recovery after stroke.


Assuntos
Inibidor da Ligação a Diazepam/uso terapêutico , Agonistas de Receptores de GABA-A/uso terapêutico , Neurônios/efeitos dos fármacos , Neuropeptídeos/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Receptores de GABA-A/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Adulto , Animais , Astrócitos/metabolismo , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Inibidor da Ligação a Diazepam/deficiência , Inibidor da Ligação a Diazepam/fisiologia , Implantes de Medicamento , Potenciais Somatossensoriais Evocados , Feminino , Agonistas de Receptores de GABA-A/farmacologia , Humanos , Hidrogéis , Infarto da Artéria Cerebral Média/tratamento farmacológico , Trombose Intracraniana/tratamento farmacológico , Trombose Intracraniana/etiologia , Luz , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/toxicidade , Neurônios/fisiologia , Neuropeptídeos/deficiência , Neuropeptídeos/fisiologia , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/deficiência , Fragmentos de Peptídeos/fisiologia , Ratos , Rosa Bengala/efeitos da radiação , Rosa Bengala/toxicidade , Método Simples-Cego , Acidente Vascular Cerebral/etiologia
4.
Nat Rev Mol Cell Biol ; 22(7): 465-482, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33837369

RESUMO

Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.


Assuntos
Diferenciação Celular/genética , Redes Reguladoras de Genes , Células-Tronco Mesenquimais/citologia , Transcrição Gênica/genética , Animais , Linhagem da Célula , Plasticidade Celular , Epigênese Genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Nat Genet ; 52(11): 1227-1238, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33020665

RESUMO

Adipocyte differentiation is driven by waves of transcriptional regulators that reprogram the enhancer landscape and change the wiring of the promoter interactome. Here, we use high-throughput chromosome conformation enhancer capture to interrogate the role of enhancer-to-enhancer interactions during differentiation of human mesenchymal stem cells. We find that enhancers form an elaborate network that is dynamic during differentiation and coupled with changes in enhancer activity. Transcription factors (TFs) at baited enhancers amplify TF binding at target enhancers, a phenomenon we term cross-interaction stabilization of TFs. Moreover, highly interconnected enhancers (HICE) act as integration hubs orchestrating differentiation by the formation of three-dimensional enhancer communities, inside which, HICE, and other enhancers, converge on phenotypically important gene promoters. Collectively, these results indicate that enhancer interactions play a key role in the regulation of enhancer function, and that HICE are important for both signal integration and compartmentalization of the genome.


Assuntos
Linhagem da Célula/genética , Elementos Facilitadores Genéticos , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Adipogenia/genética , Células Cultivadas , Redes Reguladoras de Genes , Humanos , Osteoblastos/citologia , Osteogênese/genética , Fatores de Transcrição/metabolismo
6.
Nutrients ; 12(10)2020 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993128

RESUMO

Long-chain n-3 polyunsaturated fatty acids (n-3 LC-PUFAs) are collectively recognized triglyceride-lowering agents, and their preventive action is likely mediated by changes in gene expression. However, as most studies employ fish oil, which contains a mixture of n-3 LC-PUFAs, the docosahexaenoic acid (DHA)-specific transcriptional effects on lipid metabolism are still unclear. The aim of the present study was to further elucidate the DHA-induced transcriptional effects on lipid metabolism in the liver, and to investigate the effects of co-administration with other bioactive compounds having effects on lipid metabolism. To this purpose, HepG2 cells were treated for 6 or 24 h with DHA, the short-chain fatty acid propionate (PRO), and protocatechuic acid (PCA), the main human metabolite of cyanidin-glucosides. Following supplementation, we mapped the global transcriptional changes. PRO and PCA alone had a very slight effect on the transcriptome; on the contrary, supplementation of DHA highly repressed the steroid and fatty acid biosynthesis pathways, this transcriptional modulation being not affected by co-supplementation. Our results confirm that DHA effect on lipid metabolism are mediated at least in part by modulation of the expression of specific genes. PRO and PCA could contribute to counteracting dyslipidemia through other mechanisms.


Assuntos
Células Cultivadas/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Hepatócitos/efeitos dos fármacos , Hidroxibenzoatos/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Propionatos/administração & dosagem , Ácidos Graxos/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Óleos de Peixe/metabolismo , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Transcriptoma
7.
Int J Mol Sci ; 21(11)2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32492936

RESUMO

Chronic exposure of pancreatic ß-cells to elevated nutrient levels impairs their function and potentially induces apoptosis. Like in other cell types, AMPK is activated in ß-cells under conditions of nutrient deprivation, while little is known on AMPK responses to metabolic stresses. Here, we first reviewed recent studies on the role of AMPK activation in ß-cells. Then, we investigated the expression profile of AMPK pathways in ß-cells following metabolic stresses. INS-1E ß-cells and human islets were exposed for 3 days to glucose (5.5-25 mM), palmitate or oleate (0.4 mM), and fructose (5.5 mM). Following these treatments, we analyzed transcript levels of INS-1E ß-cells by qRT-PCR and of human islets by RNA-Seq; with a special focus on AMPK-associated genes, such as the AMPK catalytic subunits α1 (Prkaa1) and α2 (Prkaa2). AMPKα and pAMPKα were also evaluated at the protein level by immunoblotting. Chronic exposure to the different metabolic stresses, known to alter glucose-stimulated insulin secretion, did not change AMPK expression, either in insulinoma cells or in human islets. Expression profile of the six AMPK subunits was marginally modified by the different diabetogenic conditions. However, the expression of some upstream kinases and downstream AMPK targets, including K-ATP channel subunits, exhibited stress-specific signatures. Interestingly, at the protein level, chronic fructose treatment favored fasting-like phenotype in human islets, as witnessed by AMPK activation. Collectively, previously published and present data indicate that, in the ß-cell, AMPK activation might be implicated in the pre-diabetic state, potentially as a protective mechanism.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose , Regulação Enzimológica da Expressão Gênica , Ilhotas Pancreáticas/enzimologia , Adulto , Animais , Glicemia/análise , Feminino , Frutose/metabolismo , Perfilação da Expressão Gênica , Homeostase , Humanos , Insulina/metabolismo , Insulinoma/enzimologia , Masculino , Pessoa de Meia-Idade , Ácido Oleico/análise , Ácido Palmítico/análise , Fenótipo , RNA-Seq , Ratos , Estresse Fisiológico
8.
Hepatology ; 72(6): 2119-2133, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32145072

RESUMO

BACKGROUND AND AIMS: Hepatic sinusoidal cells are known actors in the fibrogenic response to injury. Activated hepatic stellate cells (HSCs), liver sinusoidal endothelial cells, and Kupffer cells are responsible for sinusoidal capillarization and perisinusoidal matrix deposition, impairing vascular exchange and heightening the risk of advanced fibrosis. While the overall pathogenesis is well understood, functional relations between cellular transitions during fibrogenesis are only beginning to be resolved. At single-cell resolution, we here explored the heterogeneity of individual cell types and dissected their transitions and crosstalk during fibrogenesis. APPROACH AND RESULTS: We applied single-cell transcriptomics to map the heterogeneity of sinusoid-associated cells in healthy and injured livers and reconstructed the single-lineage HSC trajectory from pericyte to myofibroblast. Stratifying each sinusoidal cell population by activation state, we projected shifts in sinusoidal communication upon injury. Weighted gene correlation network analysis of the HSC trajectory led to the identification of core genes whose expression proved highly predictive of advanced fibrosis in patients with nonalcoholic steatohepatitis (NASH). Among the core members of the injury-repressed gene module, we identified plasmalemma vesicle-associated protein (PLVAP) as a protein amply expressed by mouse and human HSCs. PLVAP expression was suppressed in activated HSCs upon injury and may hence define hitherto unknown roles for HSCs in the regulation of microcirculatory exchange and its breakdown in chronic liver disease. CONCLUSIONS: Our study offers a single-cell resolved account of drug-induced injury of the mammalian liver and identifies key genes that may serve important roles in sinusoidal integrity and as markers of advanced fibrosis in human NASH.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/patologia , Células Endoteliais/patologia , Redes Reguladoras de Genes , Cirrose Hepática/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Biópsia , Capilares/citologia , Capilares/patologia , Tetracloreto de Carbono/administração & dosagem , Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Feminino , Veias Hepáticas/citologia , Veias Hepáticas/patologia , Humanos , Fígado/irrigação sanguínea , Fígado/patologia , Cirrose Hepática/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , RNA-Seq , Análise de Célula Única
9.
Cell Rep ; 29(9): 2756-2769.e6, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31775043

RESUMO

B cell development depends on the coordinated expression and cooperation of several transcription factors. Here we show that the transcription factor ETS-related gene (ERG) is crucial for normal B cell development and that its deletion results in a substantial loss of bone marrow B cell progenitors and peripheral B cells, as well as a skewing of splenic B cell populations. We find that ERG-deficient B lineage cells exhibit an early developmental block at the pre-B cell stage and proliferate less. The cells fail to express the immunoglobulin heavy chain due to inefficient V-to-DJ recombination, and cells that undergo recombination display a strong bias against incorporation of distal V gene segments. Furthermore, antisense transcription at PAX5-activated intergenic repeat (PAIR) elements, located in the distal region of the Igh locus, depends on ERG. These findings show that ERG serves as a critical regulator of B cell development by ensuring efficient and balanced V-to-DJ recombination.


Assuntos
Linfócitos B/metabolismo , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Humanos , Regulador Transcricional ERG/metabolismo
10.
Nat Genet ; 51(4): 716-727, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30833796

RESUMO

Mesenchymal (stromal) stem cells (MSCs) constitute populations of mesodermal multipotent cells involved in tissue regeneration and homeostasis in many different organs. Here we performed comprehensive characterization of the transcriptional and epigenomic changes associated with osteoblast and adipocyte differentiation of human MSCs. We demonstrate that adipogenesis is driven by considerable remodeling of the chromatin landscape and de novo activation of enhancers, whereas osteogenesis involves activation of preestablished enhancers. Using machine learning algorithms for in silico modeling of transcriptional regulation, we identify a large and diverse transcriptional network of pro-osteogenic and antiadipogenic transcription factors. Intriguingly, binding motifs for these factors overlap with SNPs related to bone and fat formation in humans, and knockdown of single members of this network is sufficient to modulate differentiation in both directions, thus indicating that lineage determination is a delicate balance between the activities of many different transcription factors.


Assuntos
Adipogenia/genética , Osteogênese/genética , Fator de Células-Tronco/genética , Fatores de Transcrição/genética , Células A549 , Adipócitos/fisiologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Células-Tronco Mesenquimais/fisiologia , Osteoblastos/fisiologia , Polimorfismo de Nucleotídeo Único/genética
11.
Sci Rep ; 9(1): 2324, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30787418

RESUMO

Non-alcoholic steatohepatitis (NASH) signified by hepatic steatosis, inflammation, hepatocellular injury, and fibrosis is a growing cause of chronic liver disease, cirrhosis, and hepatocellular carcinoma. Hepatic fibrosis resulting from accumulation of extracellular matrix proteins secreted by hepatic myofibroblasts plays an important role in disease progression. Activated hepatic stellate cells (HSCs) have been identified as the primary source of myofibroblasts in animal models of hepatotoxic liver injury; however, so far HSC activation and plasticity have not been thoroughly investigated in the context of NASH-related fibrogenesis. Here we have determined the time-resolved changes in the HSC transcriptome during development of Western diet- and fructose-induced NASH in mice, a NASH model recapitulating human disease. Intriguingly, HSC transcriptional dynamics are highly similar across disease models pointing to HSC activation as a point of convergence in the development of fibrotic liver disease. Bioinformatic interrogation of the promoter sequences of activated genes combined with loss-of-function experiments indicates that the transcriptional regulators ETS1 and RUNX1 act as drivers of NASH-associated HSC plasticity. Taken together, our results implicate HSC activation and transcriptional plasticity as key aspects of NASH pathophysiology.


Assuntos
Regulação da Expressão Gênica , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/genética , Transcrição Gênica , Animais , Plasticidade Celular , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Dieta Ocidental , Comportamento Alimentar , Frutose , Humanos , Cirrose Hepática/complicações , Cirrose Hepática/patologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/patologia , Proteína Proto-Oncogênica c-ets-1/metabolismo , Fatores de Tempo , Transcriptoma/genética
12.
Methods Mol Biol ; 1794: 335-352, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29855970

RESUMO

Chromatin immunoprecipitation (ChIP) is a powerful technique allowing for investigation of protein-DNA interactions in living cells. Here, we provide a detailed step-by-step protocol for ChIP and highlight important considerations, challenges and pitfalls often encountered in the ChIP procedure. Furthermore, we present data of key quality control (QC) steps and exemplify material performance validation on transcription factor ChIP to provide a QC guide for setting up ChIP. Finally, we provide guidelines for scaling of the ChIP procedure to ChIP sequencing (ChIP-seq) and discuss important considerations associated with this.


Assuntos
Imunoprecipitação da Cromatina/métodos , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas/metabolismo , Fatores de Transcrição/metabolismo , Células Cultivadas , Cromatina/genética , DNA/genética , Proteínas de Ligação a DNA/genética , Humanos , Células-Tronco Mesenquimais/citologia , Proteínas/genética , Fatores de Transcrição/genética
13.
Cell Mol Life Sci ; 75(11): 2075-2091, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29264673

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by a progressive loss of dopamine (DA) neurons through apoptotic, inflammatory and oxidative stress mechanisms. The octadecaneuropeptide (ODN) is a diazepam-binding inhibitor (DBI)-derived peptide, expressed by astrocytes, which protects neurons against oxidative cell damages and apoptosis in an in vitro model of PD. The present study reveals that a single intracerebroventricular injection of 10 ng ODN 1 h after the last administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) prevented the degeneration of DA neurons induced by the toxin in the substantia nigra pars compacta of mice, 7 days after treatment. ODN-mediated neuroprotection was associated with a reduction of the number of glial fibrillary acidic protein-positive reactive astrocytes and a strong inhibition of the expression of pro-inflammatory genes such as interleukins 1ß and 6, and tumor necrosis factor-α. Moreover, ODN blocked the inhibition of the anti-apoptotic gene Bcl-2, and the stimulation of the pro-apoptotic genes Bax and caspase-3, induced by MPTP in the substantia nigra pars compacta. ODN also decreased or even in some cases abolished MPTP-induced oxidative damages, overproduction of reactive oxygen species and accumulation of lipid oxidation products in DA neurons. Furthermore, DBI knockout mice appeared to be more vulnerable than wild-type animals to MPTP neurotoxicity. Taken together, these results show that the gliopeptide ODN exerts a potent neuroprotective effect against MPTP-induced degeneration of nigrostriatal DA neurons in mice, through mechanisms involving downregulation of neuroinflammatory, oxidative and apoptotic processes. ODN may, thus, reduce neuronal damages in PD and other cerebral injuries involving oxidative neurodegeneration.


Assuntos
Inibidor da Ligação a Diazepam/uso terapêutico , Neuropeptídeos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Espécies Reativas de Oxigênio/metabolismo
14.
Endocrinology ; 158(5): 1231-1251, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323980

RESUMO

Triglycerides are stored in specialized organelles called lipid droplets. Numerous proteins have been shown to be physically associated with lipid droplets and govern their function. Previously, the protein hypoxia-inducible lipid droplet-associated (HILPDA) was localized to lipid droplets and was suggested to inhibit triglyceride lipolysis in hepatocytes. We confirm the partial localization of HILPDA to lipid droplets and show that HILPDA is highly abundant in adipose tissue, where its expression is controlled by the peroxisome proliferator-activated receptor γ and by ß-adrenergic stimulation. Levels of HILPDA markedly increased during 3T3-L1 adipocyte differentiation. Nevertheless, silencing of Hilpda using small interfering RNA or overexpression of Hilpda using adenovirus did not show a clear impact on 3T3-L1 adipogenesis. Following ß-adrenergic stimulation, the silencing of Hilpda in adipocytes did not significantly alter the release of nonesterified fatty acids (NEFA) and glycerol. By contrast, adenoviral-mediated overexpression of Hilpda modestly attenuated the release of NEFA from adipocytes following ß-adrenergic stimulation. In mice, adipocyte-specific inactivation of Hilpda had no effect on plasma levels of NEFA and glycerol after fasting, cold exposure, or pharmacological ß-adrenergic stimulation. In addition, other relevant metabolic parameters were unchanged by adipocyte-specific inactivation of Hilpda. Taken together, we find that HILPDA is highly abundant in adipose tissue, where its levels are induced by peroxisome proliferator-activated receptor γ and ß-adrenergic stimulation. In contrast to the reported inhibition of lipolysis by HILPDA in hepatocytes, our data do not support an important direct role of HILPDA in the regulation of lipolysis in adipocytes in vivo and in vitro.


Assuntos
Tecido Adiposo/metabolismo , Lipólise/genética , Proteínas de Neoplasias/fisiologia , Células 3T3-L1 , Adipócitos/fisiologia , Adipogenia/genética , Animais , Feminino , Gotículas Lipídicas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Nutr Metab (Lond) ; 13: 4, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26788115

RESUMO

BACKGROUND: Previous studies suggest that intake of specific bioactive compounds may have beneficial clinical effects on adipose tissue partly due to their anti-inflammatory and insulin-sensitizing properties. With the overall aim to contribute to better understanding of the mechanisms of selected bioactive nutrients on fat metabolism, we investigated their role on human white adipocyte function. METHODS: The influence of the omega-3-fatty acid docosahexaenoic acid (DHA), the anthocyanin (AC) cyanidin-3-glucoside and its metabolite protocatechuic acid, and the beta-glucan metabolite propionic acid (PI) on adipokine secretion, fatty acid metabolism (lipolysis/lipogenesis) and adipocyte differentiation (lipid accumulation) was studied in human fat cells differentiated in vitro. To investigate possible synergistic, additive or antagonistic effects, DHA was also combined with AC or PI. RESULTS: Each compound, alone or together with DHA, suppressed basal adipocyte lipolysis compared to control treated cells. DHA alone attenuated the secretion of pro-inflammatory adipokines such as chemerin, interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1/CCL2), whereas AC suppressed only the latter two. Treatment with PI decreased IL-6, tumour necrosis factor alpha (TNFα) and adiponectin secretion. A combination of DHA and AC decreased TNFα secretion and increased insulin-stimulated lipogenesis. No effect was found on adipocyte differentiation. At the selected concentrations, none of the compounds was found to be cytotoxic. CONCLUSION: The studied bioactive food compounds or their metabolites have beneficial effects in human primary fat cells measured as decreased basal lipolytic activity and secretion of inflammatory markers. A minor effect was also observed on insulin-stimulated glucose uptake albeit only with the combination of DHA and AC. Taken together, our results may link the reported health benefits of the selected bioactives on metabolic disorders such as insulin resistance, hypertension and dyslipidemia to effects on white adipocytes.

16.
Cell Rep ; 13(9): 2000-13, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26628366

RESUMO

Cold exposure greatly alters brown adipose tissue (BAT) gene expression and metabolism to increase thermogenic capacity. Here, we used RNA sequencing and mass-spectrometry-based lipidomics to provide a comprehensive resource describing the molecular signature of cold adaptation at the level of the transcriptome and lipidome. We show that short-term (3-day) cold exposure leads to a robust increase in expression of several brown adipocyte genes related to thermogenesis as well as the gene encoding the hormone irisin. However, pathway analysis shows that the most significantly induced genes are those involved in glycerophospholipid synthesis and fatty acid elongation. This is accompanied by significant changes in the acyl chain composition of triacylglycerols (TAGs) as well as subspecies-selective changes of acyl chains in glycerophospholipids. These results indicate that cold adaptation of BAT is associated with significant and highly species-selective remodeling of both TAGs and glycerophospholipids.


Assuntos
Tecido Adiposo Marrom/metabolismo , Glicerofosfolipídeos/metabolismo , Animais , Análise por Conglomerados , Temperatura Baixa , Fibronectinas/metabolismo , Glutationa/metabolismo , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , RNA/metabolismo , Análise de Sequência de RNA , Termogênese , Fatores de Tempo , Transcriptoma
17.
Artigo em Inglês | MEDLINE | ID: mdl-26432526

RESUMO

The ability to modify the transcriptional program in response to external signals provides a way for mammalian cells to alter their biological fate and properties, thereby adapting to changes in the environment. Adipocytes are excellent examples of differentiated cells that possess a striking transcriptional plasticity when exposed to physiological and metabolic stimuli. In our work, we have focused on understanding the processes responsible for modulating the genomic programming in response to different external signals. Thus, we have shown that browning of human adipocytes with rosiglitazone, an antidiabetic agonist of the key adipocyte transcription factor peroxisome proliferator-activated receptor γ (PPARγ), involves redistribution of PPARγ binding to form browning-selective PPARγ super-enhancers that drive expression of key browning genes. These include genes encoding transcriptional regulators, such as Krüppel-like factor 11 (KLF11) that are essential for modulating the genomic program in white adipocytes to induce browning. Furthermore, we have shown that acute suppression of adipocyte genes by the proinflammatory cytokine, tumor necrosis factor (TNF), involves redistribution of cofactors to enhancers activated by the master inflammatory regulator, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Interestingly, this redistribution occurs selectively from enhancers with high-cofactor occupancies, thereby predominantly affecting super-enhancers and their associated genes. We propose that this is a general mechanism contributing to transcriptional repression associated with activation of signal-dependent transcription factors.


Assuntos
Adipócitos/metabolismo , Reprogramação Celular/genética , Regulação da Expressão Gênica , Adipócitos/efeitos dos fármacos , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Marrons/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adipócitos Brancos/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Ciclo Celular/metabolismo , Plasticidade Celular , Técnicas de Reprogramação Celular , Humanos , Hipoglicemiantes/farmacologia , NF-kappa B/metabolismo , PPAR gama/agonistas , Proteínas Repressoras/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
18.
Genome Res ; 25(9): 1281-94, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26113076

RESUMO

The proinflammatory cytokine tumor necrosis factor (TNF) plays a central role in low-grade adipose tissue inflammation and development of insulin resistance during obesity. In this context, nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) is directly involved and required for the acute activation of the inflammatory gene program. Here, we show that the major transactivating subunit of NFκB, v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA), is also required for acute TNF-induced suppression of adipocyte genes. Notably, this repression does not involve RELA binding to the associated enhancers but rather loss of cofactors and enhancer RNA (eRNA) selectively from high-occupancy sites within super-enhancers. Based on these data, we have developed models that, with high accuracy, predict which enhancers and genes are repressed by TNF in adipocytes. We show that these models are applicable to other cell types where TNF represses genes associated with super-enhancers in a highly cell-type-specific manner. Our results propose a novel paradigm for NFκB-mediated repression, whereby NFκB selectively redistributes cofactors from high-occupancy enhancers, thereby specifically repressing super-enhancer-associated cell identity genes.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica/efeitos dos fármacos , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Proteínas de Ciclo Celular , Diferenciação Celular , Reprogramação Celular/genética , Humanos , Subunidade 1 do Complexo Mediador/metabolismo , Proteínas Nucleares/metabolismo , Especificidade de Órgãos/genética , Ligação Proteica , Transporte Proteico , Fator de Transcrição RelA/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma
19.
Mol Biosyst ; 11(5): 1260-70, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25720604

RESUMO

LNA-containing DNA aptamers against CD73 (human ecto-5'-nucleotidase), a protein frequently overexpressed in solid tumours, were isolated by SELEX. A pre-defined stem-loop library, containing LNA in the forward primer region, was enriched with CD73 binding sequences through six rounds of SELEX with recombinant his-tagged CD73 immobilised on anti-his plates. Enriched pools isolated from rounds one, three and six were subjected to next-generation sequencing and analysed for enrichment using custom bioinformatics software. The software identified aptamer sequences via the primers and then performed several steps including sequence unification, clustering and alignment to identify enriched sequences. Three enriched sequences were synthesised for further analysis, two of which showed sequence similarities. These sequences exhibited binding to the recombinant CD73 with KD values of 10 nM and 3.5 nM when tested by surface plasmon resonance. Truncated variants of these aptamers and variants where the LNA nucleotides were substituted for the DNA equivalent also exhibited affinity for the recombinant CD73 in the low nanomolar range. In enzyme inhibition assays with recombinant CD73 the aptamer sequences were able to decrease the activity of the protein. However, the aptamers exhibited no binding to cellular CD73 by flow cytometry analysis likely since the epitope recognised by the aptamer was not available for binding on the cellular protein.


Assuntos
5'-Nucleotidase/antagonistas & inibidores , 5'-Nucleotidase/metabolismo , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Proteínas Nucleares/química , Fosfoproteínas/química , Proteínas Recombinantes , 5'-Nucleotidase/química , Sequência de Bases , Linhagem Celular , Biologia Computacional/métodos , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Conformação de Ácido Nucleico , Ligação Proteica , Ressonância de Plasmônio de Superfície
20.
Mol Cell Biol ; 33(7): 1303-16, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23339868

RESUMO

Angiopoietin-like protein 4 (ANGPTL4/FIAF) has been proposed as a circulating mediator between the gut microbiota and fat storage. Here, we show that transcription and secretion of ANGPTL4 in human T84 and HT29 colon adenocarcinoma cells is highly induced by physiological concentrations of short-chain fatty acids (SCFA). SCFA induce ANGPTL4 by activating the nuclear receptor peroxisome proliferator activated receptor γ (PPARγ), as demonstrated using PPARγ antagonist, PPARγ knockdown, and transactivation assays, which show activation of PPARγ but not PPARα and PPARδ by SCFA. At concentrations required for PPARγ activation and ANGPTL4 induction in colon adenocarcinoma cells, SCFA do not stimulate PPARγ in mouse 3T3-L1 and human SGBS adipocytes, suggesting that SCFA act as selective PPARγ modulators (SPPARM), which is supported by coactivator peptide recruitment assay and structural modeling. Consistent with the notion that fermentation leads to PPAR activation in vivo, feeding mice a diet rich in inulin induced PPAR target genes and pathways in the colon. We conclude that (i) SCFA potently stimulate ANGPTL4 synthesis in human colon adenocarcinoma cells and (ii) SCFA transactivate and bind to PPARγ. Our data point to activation of PPARs as a novel mechanism of gene regulation by SCFA in the colon, in addition to other mechanisms of action of SCFA.


Assuntos
Adenocarcinoma/metabolismo , Angiopoietinas/biossíntese , Neoplasias do Colo/metabolismo , Ácidos Graxos Voláteis/metabolismo , PPAR gama/metabolismo , Células 3T3-L1 , Adenocarcinoma/genética , Adipogenia/genética , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Linhagem Celular Tumoral , Colo/metabolismo , Neoplasias do Colo/genética , Células HT29 , Humanos , Inulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/agonistas , PPAR gama/genética , Transcrição Gênica , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA