Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Hepatol ; 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38583492

RESUMO

BACKGROUND & AIMS: Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS: We performed single-nucleus RNA-sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent haploinsufficient strains for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS: We observed that ageing in wild-type mice results in nuclei polyploidy and marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue-wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbation. ConclusionsOur results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome.

2.
Cell Rep ; 42(10): 113305, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37864798

RESUMO

Oxytocin-expressing paraventricular hypothalamic neurons (PVNOT neurons) integrate afferent signals from the gut, including cholecystokinin (CCK), to adjust whole-body energy homeostasis. However, the molecular underpinnings by which PVNOT neurons orchestrate gut-to-brain feeding control remain unclear. Here, we show that mice undergoing selective ablation of PVNOT neurons fail to reduce food intake in response to CCK and develop hyperphagic obesity on a chow diet. Notably, exposing wild-type mice to a high-fat/high-sugar (HFHS) diet recapitulates this insensitivity toward CCK, which is linked to diet-induced transcriptional and electrophysiological aberrations specifically in PVNOT neurons. Restoring OT pathways in diet-induced obese (DIO) mice via chemogenetics or polypharmacology sufficiently re-establishes CCK's anorexigenic effects. Last, by single-cell profiling, we identify a specialized PVNOT neuronal subpopulation with increased κ-opioid signaling under an HFHS diet, which restrains their CCK-evoked activation. In sum, we document a (patho)mechanism by which PVNOT signaling uncouples a gut-brain satiation pathway under obesogenic conditions.


Assuntos
Ocitocina , Núcleo Hipotalâmico Paraventricular , Camundongos , Animais , Ocitocina/farmacologia , Núcleo Hipotalâmico Paraventricular/metabolismo , Analgésicos Opioides/farmacologia , Neurônios/metabolismo , Saciação , Colecistocinina/metabolismo
3.
Mamm Genome ; 31(5-6): 170-180, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32270277

RESUMO

Cellular heterogeneity is revolutionizing the way to study, monitor and dissect complex diseases. This has been possible with the technological and computational advances associated to single-cell genomics and epigenomics. Deeper understanding of cell-to-cell variation and its impact on tissue function will open new avenues for early disease detection, accurate diagnosis and personalized treatments, all together leading to the next generation of health care. This review focuses on the recent discoveries that single-cell genomics and epigenomics have facilitated in the context of human health. It highlights the potential of single-cell omics to further advance the development of personalized treatments and precision medicine in cancer, diabetes and chronic age-related diseases. The promise of single-cell technologies to generate new insights about the differences in function between individual cells is just emerging, and it is paving the way for identifying biomarkers and novel therapeutic targets to tackle age, complex diseases and understand the effect of life style interventions and environmental factors.


Assuntos
Envelhecimento/genética , Diabetes Mellitus/genética , Epigênese Genética , Epigenômica/métodos , Neoplasias/genética , Análise de Célula Única/métodos , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Células Cultivadas , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Interação Gene-Ambiente , Variação Genética , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Organoides/metabolismo , Organoides/patologia , Medicina de Precisão/tendências
4.
BMJ Open ; 7(3): e014931, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28302638

RESUMO

INTRODUCTION: For the optimal management of children with cystic fibrosis, there are currently no efficient tools for the precise adjustment of pancreatic enzyme replacement therapy, either for advice on appropriate dietary intake or for achieving an optimal nutrition status. Therefore, we aim to develop a mobile application that ensures a successful nutritional therapy in children with cystic fibrosis. METHODS AND ANALYSIS: A multidisciplinary team of 12 partners coordinate their efforts in 9 work packages that cover the entire so-called 'from laboratory to market' approach by means of an original and innovative co-design process. A cohort of 200 patients with cystic fibrosis aged 1-17 years are enrolled. We will develop an innovative, clinically tested mobile health application for patients and health professionals involved in cystic fibrosis management. The mobile application integrates the research knowledge and innovative tools for maximising self-management with the aim of leading to a better nutritional status, quality of life and disease prognosis. Bringing together different and complementary areas of knowledge is fundamental for tackling complex challenges in disease treatment, such as optimal nutrition and pancreatic enzyme replacement therapy in cystic fibrosis. Patients are expected to benefit the most from the outcomes of this innovative project. ETHICS AND DISSEMINATION: The project is approved by the Ethics Committee of the coordinating organisation, Hospital Universitari La Fe (Ref: 2014/0484). Scientific findings will be disseminated via journals and conferences addressed to clinicians, food scientists, information and communications technology experts and patients. The specific dissemination working group within the project will address the wide audience communication through the website (http://www.mycyfapp.eu), the social networks and the newsletter.


Assuntos
Proteção da Criança , Fibrose Cística/terapia , Avaliação de Programas e Projetos de Saúde/métodos , Autogestão/métodos , Telemedicina/métodos , Adolescente , Criança , Pré-Escolar , Europa (Continente) , Feminino , Humanos , Lactente , Masculino
5.
ACS Chem Biol ; 12(3): 654-663, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28059499

RESUMO

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan member of the nuclear receptor family of transcription factors whose activities are modulated upon binding of small molecules into an hydrophobic ligand-binding pocket (LBP). Although the LBP of COUP-TFII is filled with aromatic amino-acid side chains, alternative modes of ligand binding could potentially lead to regulation of the orphan receptor. Here, we screened a synthetic and natural compound library in a yeast one-hybrid assay and identified 4-methoxynaphthol as an inhibitor of COUP-TFII. This synthetic inhibitor was able to counteract processes either positively or negatively regulated by COUP-TFII in different mammalian cell systems. Hence, we demonstrate that the true orphan receptor COUP-TFII can be targeted by small chemicals which could be used to study the physiological functions of COUP-TFII or to counteract detrimental COUP-TFII activities in various pathological conditions.


Assuntos
Fator II de Transcrição COUP/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Sítios de Ligação , Fator II de Transcrição COUP/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Camundongos
6.
J Biol Chem ; 281(40): 29840-9, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16891307

RESUMO

Hepatocyte nuclear factor 4alpha (HNF4alpha) plays critical roles during liver development and in the transcriptional regulation of many hepatic genes in adult liver. Here we have demonstrated that in human hepatoma HepG2 cells, HNF4alpha is expressed at levels as high as in human liver but its activity on target genes is very low or absent. We have discovered that the low expression of key coactivators (PGC1alpha, SRC1, SRC2, and PCAF) might account for the lack of function of HNF4alpha in HepG2 cells. Among them, PGC1alpha and SRC1 are the two most important HNF4alpha coactivators as revealed by reporter assays with an Apo-CIII promoter construct. Moreover, the expression of these two coactivators was found to be down-regulated in all human hepatomas investigated. Overexpression of SRC1 and PGC1alpha by recombinant adenoviruses led to a significant up-regulation of well characterized HNF4alpha-dependent genes (ApoCIII, ApoAV, PEPCK, AldoB, OTC, and CYP7A1) and forced HepG2 cells toward a more differentiated phenotype as demonstrated by increased ureogenic rate. The positive effect of PGC1alpha was seen to be dependent on HNF4alpha. Finally, insulin treatment of human hepatocytes and HepG2 cells caused repression of PGC1alpha and a concomitant down-regulation of ApoCIII, PEPCK, AldoB, and OTC. Altogether, our results suggest that SRC1, and notably PGC1alpha, are key coactivators for the proper function of HNF4alpha in human liver and for an integrative control of multiple hepatic genes involved in metabolism and homeostasis. The down-regulation of key HNF4alpha coactivators could be a determinant factor for the dedifferentiation of human hepatomas.


Assuntos
Carcinoma Hepatocelular/metabolismo , Regulação para Baixo/fisiologia , Proteínas de Choque Térmico/fisiologia , Fator 4 Nuclear de Hepatócito/antagonistas & inibidores , Histona Acetiltransferases/fisiologia , Neoplasias Hepáticas/metabolismo , Fatores de Transcrição/fisiologia , Adulto , Idoso , Carcinoma Hepatocelular/patologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Histona Acetiltransferases/antagonistas & inibidores , Histona Acetiltransferases/genética , Homeostase/fisiologia , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Coativador 1 de Receptor Nuclear , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética
7.
Mol Pharmacol ; 70(5): 1681-92, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16882880

RESUMO

Hepatocyte nuclear factor 4alpha (HNF4alpha) is a key transcription factor for the constitutive expression of cytochromes P450 (P450s) in the liver. However, human hepatoma HepG2 cells show a high level of HNF4alpha but express only marginal P450 levels. We found that the HNF4alpha-mediated P450 transcription in HepG2 is impaired by the low level of coactivators peroxisomal proliferator activated receptor-gamma coactivator 1alpha (PGC1alpha) and steroid receptor coactivator 1 (SRC1). Reporter assays with a chimeric CYP2C9-LUC construct demonstrated that the sole transfection of coactivators induced luciferase activity in HepG2 cells. In HeLa cells however, CYP2C9-LUC activity only significantly increased when coactivators were cotransfected with HNF4alpha. A deletion mutant lacking the two proximal HNF4alpha binding sites in the CYP2C9 promoter did not respond to PGC1alpha or SRC1, demonstrating that coactivators were acting through HNF4alpha response elements. Adenovirus-mediated transfection of PGC1alpha in human hepatoma cells caused a significant dose-dependent increase in CYP2C9, CYP1A1, and CYP1A2 and in the positive control CYP7A1. PGC1alpha also showed a moderate activating effect on CYP3A4, CYP3A5, and CYP2D6. Adenoviral transfection of SRC1 had a lessened effect on P450 genes. Chromatin immunoprecipitation assay demonstrated in vivo binding of HNF4alpha and PGC1alpha to HNF4alpha response sequences in the CYP2C9 promoter and to three new regulatory regions in the common 23.3 kilobase spacer sequence of the CYP1A1/2 cluster. Insulin treatment of HepG2 and human hepatocytes caused repression of PGC1alpha and a concomitant down-regulation of P450s. Our results establish the importance of coactivators PGC1alpha and SRC1 for the hepatic expression of human P450s and uncover a new HNF4alpha-dependent regulatory mechanism to constitutively control the CYP1A1/2 cluster.


Assuntos
Hidrocarboneto de Aril Hidroxilases/genética , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Proteínas de Choque Térmico/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Histona Acetiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Adenoviridae/metabolismo , Células Cultivadas , Citocromo P-450 CYP2C9 , Feminino , Células HeLa , Proteínas de Choque Térmico/genética , Fator 4 Nuclear de Hepatócito/genética , Hepatócitos/efeitos dos fármacos , Histona Acetiltransferases/genética , Humanos , Insulina/farmacologia , Masculino , Pessoa de Meia-Idade , Coativador 1 de Receptor Nuclear , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ligação Proteica/efeitos dos fármacos , Elementos de Resposta/genética , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Transfecção
8.
Expert Opin Drug Metab Toxicol ; 2(2): 183-212, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16866607

RESUMO

Gaining knowledge on the metabolism of a drug, the enzymes involved and its inhibition or induction potential is a necessary step in pharmaceutical development of new compounds. Primary human hepatocytes are considered a cellular model of reference, as they express the majority of drug-metabolising enzymes, respond to enzyme inducers and are capable of generating in vitro a metabolic profile similar to what is found in vivo. However, hepatocytes show phenotypic instability and have a restricted accessibility. Different alternatives have been explored in the past recent years to overcome the limitations of primary hepatocytes. These include immortalisation of adult or fetal human hepatic cells by means of transforming tumour virus genes, oncogenes, conditionally immortalised hepatocytes, and cell fusion. New strategies are currently being used to upregulate the expression of drug-metabolising enzymes in cell lines or to derive hepatocytes from progenitor cells. This paper reviews the features of liver-derived cell lines, their suitability for drug metabolism studies as well as the state-of-the-art of the strategies pursued in order to generate metabolically competent hepatic cell lines.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Hepatócitos/metabolismo , Xenobióticos/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Linhagem Celular , Linhagem Celular Transformada , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Reprodutibilidade dos Testes , Xenobióticos/farmacocinética , Xenobióticos/farmacologia
9.
Mol Pharmacol ; 67(6): 2088-101, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15778453

RESUMO

CCAAT/enhancer-binding proteins (C/EBPs) are key transcription factors involved in the constitutive expression of several cytochrome P450 genes in the liver. Their concentration and activity change in several pathophysiological conditions. For instance, during inflammation, released cytokines induce repressive C/EBPbeta-liver inhibitory protein (LIP), which antagonizes constitutive C/EBP transactivators [C/EBPalpha and C/EBPbeta-liver activating protein (LAP)], down-regulating genes such as CYP3A4. However, the mechanism by which hepatic C/EBP factors modulate transcription of the CYP3A4 gene is not known. To elucidate the mechanism of action, we cotransfected luciferase reporter vectors, containing 5'-flanking deletions of the CYP3A4 gene, along with expression vectors for C/EBPbeta-LAP, C/EBPbeta-LIP, and C/EBPalpha, in hepatic (HepG2) and nonhepatic (HeLa) cells. Analysis of the -3557 to -6954 base pair (bp) region demonstrated the existence of a 288-bp sequence at -5.95 kilobases (kb), which showed maximal response to C/EBPbeta-LAP ( approximately 30-fold increase in HepG2 cells). Coexpression of LAP with increasing amounts of LIP reduced the activating effect by approximately 70%. Site-directed mutagenesis of predicted C/EBPbeta binding sites demonstrated the presence of four functional C/EBPbeta-responsive motifs within this distal flanking region. Further experiments using chromatin immunoprecipitation proved the binding of endogenous C/EBPbeta to the -5.95-kilobase enhancer of the CYP3A4 gene in human hepatocytes. Expression of recombinant LAP and LIP by means of adenoviral vectors resulted in their binding to this region, which was followed by activation/repression of CYP3A4. Together, our results uncover a new distal enhancer site in the CYP3A4 gene where C/EBPbeta-LAP binds and activates transcription, whereas the truncated form, C/EBPbeta-LIP, antagonizes LAP activity and causes gene repression.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/metabolismo , Isoformas de Proteínas/fisiologia , Proteínas Repressoras/genética , Transativadores/genética , Transcrição Gênica/fisiologia , Animais , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/genética , Linhagem Celular , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/genética , Humanos , Insetos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/fisiologia , Transativadores/antagonistas & inibidores , Transativadores/fisiologia
10.
Altern Lab Anim ; 32 Suppl 1A: 65-74, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23577436

RESUMO

Knowledge of metabolism, enzymes so far involved, and potential enzyme-inhibiting or enzyme-inducing properties of new compounds is a key issue in drug development. Primary cultured hepatocytes, cytochrome P450 (CYP)-engineered cells and hepatoma cell lines are currently being used for this purpose, but only primary cultures can produce a metabolic profile of a drug similar to that found in vivo and can respond to inducers. Because of their limited accessibility, alternatives to replace human hepatocytes are currently being explored, including the immortalisation of hepatocytes by using different strategies (i.e. SV40 T-large antigen, conditionally immortalised hepatocytes, transfection with c-myc, cH-ras, N-ras oncogenes, transgenic animals over-expressing growth factors or oncogenes and cre-lox recombination/excision). However, none of the resulting cells has the desirable phenotypic characteristics to replace primary cultures in drug metabolisms studies. We investigated why these differentiated human hepatomas do not express CYP genes and found that the levels of certain key transcription factors clearly differ from those found in hepatocytes. It was then conceivable that re-expression of one (or more) of these transcription factors could lead to an efficient transcription of CYP genes. The feasibility of this hypothesis was demonstrated by genetic engineering of Hep G2 cells with liver-enriched transcription factors followed by the analysis of the expression of the most relevant human CYPs.


Assuntos
Carcinoma Hepatocelular/patologia , Diferenciação Celular , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450/genética , Humanos , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/metabolismo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA