Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
EMBO Rep ; 23(9): e54195, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35801407

RESUMO

Nuclear receptors are transcription factors with important functions in a variety of physiological and pathological processes. Targeting glucocorticoid receptor (GR) activity using glucocorticoids is a cornerstone in the treatment of patients with T cell acute lymphoblastic leukemia (T-ALL), and resistance to GC-induced cell death is associated with poor outcome and a high risk for relapse. Next to ligand-binding, heterodimerization with other transcription factors presents an important mechanism for the regulation of GR activity. Here, we describe a GC-induced direct association of the Liver Receptor Homolog-1 (LRH-1) with the GR in the nucleus, which results in reciprocal inhibition of transcriptional activity. Pharmacological and molecular interference with LRH-1 impairs proliferation and survival in T-ALL and causes a profound sensitization to GC-induced cell death, even in GC-resistant T-ALL. Our data illustrate that direct interaction between GR and LRH-1 critically regulates glucocorticoid sensitivity in T-ALL opening up new perspectives for developing innovative therapeutic approaches to treat GC-resistant T-ALL.


Assuntos
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Receptores Citoplasmáticos e Nucleares , Receptores de Glucocorticoides , Apoptose , Glucocorticoides/farmacologia , Humanos , Erros Inatos do Metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Glucocorticoides/deficiência , Receptores de Glucocorticoides/genética , Fatores de Transcrição
2.
Acta Pharmacol Sin ; 45(8): 1571-1581, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38632319

RESUMO

Liver receptor homolog-1 (LRH-1), a member of the nuclear receptor superfamily, is a ligand-regulated transcription factor that plays crucial roles in metabolism, development, and immunity. Despite being classified as an 'orphan' receptor due to the ongoing debate surrounding its endogenous ligands, recent researches have demonstrated that LRH-1 can be modulated by various synthetic ligands. This highlights the potential of LRH-1 as an attractive drug target for the treatment of inflammation, metabolic disorders, and cancer. In this review, we provide an overview of the structural basis, functional activities, associated diseases, and advancements in therapeutic ligand research targeting LRH-1.


Assuntos
Descoberta de Drogas , Receptores Citoplasmáticos e Nucleares , Humanos , Animais , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Ligantes , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(8): 2467-72, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25675535

RESUMO

Colorectal cancers (CRCs) account for nearly 10% of all cancer deaths in industrialized countries. Recent evidence points to a central role for the nuclear receptor liver receptor homolog-1 (LRH-1) in intestinal tumorigenesis. Interaction of LRH-1 with the Wnt/ß-catenin pathway, highly active in a critical subpopulation of CRC cells, underscores the importance of elucidating LRH-1's role in this disease. Reduction of LRH-1 diminishes tumor burden in murine models of CRC; however, it is not known whether LRH-1 is required for tumorigenesis, for proliferation, or for both. In this work, we address this question through shRNA-mediated silencing of LRH-1 in established CRC cell lines. LRH-1 mRNA knockdown results in significantly impaired proliferation in a cell line highly expressing the receptor and more modest impairment in a cell line with moderate LRH-1 expression. Cell-cycle analysis shows prolongation of G0/G1 with LRH-1 silencing, consistent with LRH-1 cell-cycle influences in other tissues. Cluster analysis of microarray gene expression demonstrates significant genome wide alterations with major effects in cell-cycle regulation, signal transduction, bile acid and cholesterol metabolism, and control of apoptosis. This study demonstrates a critical proproliferative role for LRH-1 in established colon cancer cell lines. LRH-1 exerts its effects via multiple signaling networks. Our results suggest that selected CRC patients could benefit from LRH-1 inhibitors.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Receptores Citoplasmáticos e Nucleares/genética , Células CACO-2 , Ciclo Celular/genética , Proliferação de Células , Técnicas de Silenciamento de Genes , Células HT29 , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodutibilidade dos Testes
4.
Biochem Biophys Res Commun ; 489(3): 299-304, 2017 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-28552526

RESUMO

Liver receptor homolog-1 (LRH-1) is a nuclear receptor that plays an important role in the regulation of bile acid biosynthesis, cholesterol reverse transport, steroidogenesis, and exocrine pancreatic enzyme production. In the current study, previously published data from a genome wide analysis of LRH-1 binding in the liver were re-analyzed to identify new LRH-1 targets and propose new roles for LRH-1 in the liver. Superoxide dismutase 2 (Sod2) was identified, which contains putative LRH-1 binding sites in the proximal promoter. When hepatocytes were treated with the LRH-1 agonist RJW101, Sod2 expression was dramatically increased and reactive oxygen species (ROS) production, which was induced by a high concentration of palmitate, was significantly reduced. A LRH-1 binding site was mapped to -288/-283 in the Sod2 promoter, which increased Sod2 promoter activity in response to LRH-1 and its agonist. LRH-1 binding to this site was confirmed using a chromatin immunoprecipitation assay. These results suggest that Sod2 is a target gene of LRH-1, and that LRH-1 agonists can mediate a reduction in ROS production and oxidative stress driven by an excess of fatty acids, as exhibited in nonalcoholic fatty liver disease.


Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Superóxido Dismutase/metabolismo , Animais , Células Cultivadas , Hepatócitos/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/genética , Superóxido Dismutase/genética , Transcriptoma
5.
Biochem Biophys Res Commun ; 473(4): 980-986, 2016 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-27049310

RESUMO

MicroRNAs (miRNAs) that negatively regulate gene expression have emerged as novel therapeutic tools for cancer treatment. In this study, we investigated the potential role of Liver receptor homolog-1 (LRH-1), a novel oncogene, in non-small-cell lung cancer (NSCLC), and examined the regulation of LRH-1 by miRNAs. We found that LRH-1 was highly overexpressed in NSCLC cell lines. Knockdown of LRH-1 by small interfering RNA significantly inhibited NSCLC cell growth and invasion. miR-376c directly targeted the 3'-untranslated region (UTR) of LRH-1 and negatively regulated LRH-1 expression, as detected by dual-luciferase reporter assay, real-time quantitative polymerase chain reaction and Western blot analysis. Further data showed that miR-376c expression was inversely correlated with LRH-1 expression in clinical cancer samples. Overexpression of miR-376c could inhibit NSCLC cell growth and invasion as well as Wnt signaling. In contrast, depletion of miR-376c exhibited the opposite effects. Moreover, these effects of miR-376c overexpression were partially abrogated by overexpression of LRH-1. Taken together, these results indicate that LRH-1 is involved in regulating the growth and invasion of NSCLC cells and that miR-376c inhibits NSCLC cell growth and invasion by targeting LRH-1, providing a novel insight into the potential for development of anti-cancer drugs for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , MicroRNAs/fisiologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Via de Sinalização Wnt , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular , Linhagem Celular , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Invasividade Neoplásica , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo
6.
Stem Cells ; 33(5): 1390-404, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25546009

RESUMO

We previously demonstrated that coexpressing retinoic acid (RA) receptor gamma and liver receptor homolog-1 (LRH1 or NR5A2) with OCT4, MYC, KLF4, and SOX2 (4F) rapidly reprograms mouse embryonic fibroblast cells (MEFs) into induced pluripotent stem cells (iPSCs). Here, we further explore the role of RA in reprogramming and report that the six factors (6F) efficiently and directly reprogram MEFs into integration-free iPSCs in defined medium (N2B27) in the absence of feeder cells. Through genetic and chemical approaches, we find that RA signalling is essential, in a highly dose-sensitive manner, for MEF reprogramming. The removal of exogenous RA from N2B27, the inhibition of endogenous RA synthesis or the expression of a dominant-negative form of RARA severely impedes reprogramming. By contrast, supplementing N2B27 with various retinoids substantially boosts reprogramming. In addition, when coexpressed with LRH1, RA receptors (RARs) can promote reprogramming in the absence of both exogenous and endogenously synthesized RA. Remarkably, the reprogramming of epiblast stem cells into embryonic stem cell-like cells also requires low levels of RA, which can modulate Wnt signalling through physical interactions of RARs with ß-catenin. These results highlight the important functions of RA signalling in reprogramming somatic cells and primed stem cells to naïve pluripotency. Stem Cells 2015;33:1390-1404.


Assuntos
Reprogramação Celular , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Camadas Germinativas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Animais , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Ligantes , Camundongos , Fatores de Transcrição , Tretinoína/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Receptor gama de Ácido Retinoico
7.
J Struct Biol ; 192(3): 342-348, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416531

RESUMO

The nuclear receptor LRH-1 (Liver Receptor Homolog-1, NR5A2) is a transcription factor that regulates gene expression programs critical for many aspects of metabolism and reproduction. Although LRH-1 is able to bind phospholipids, it is still considered an orphan nuclear receptor (NR) with an unknown regulatory hormone. Our prior cellular and structural studies demonstrated that the signaling phosphatidylinositols PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3) bind and regulate SF-1 (Steroidogenic Factor-1, NR5A1), a close homolog of LRH-1. Here, we describe the crystal structure of human LRH-1 ligand binding domain (LBD) bound by PIP3 - the first phospholipid with a head group endogenous to mammals. We show that the phospholipid hormone binds LRH-1 with high affinity, stabilizing the receptor LBD. While the hydrophobic PIP3 tails (C16/C16) are buried inside the LRH-1 ligand binding pocket, the negatively charged PIP3 head group is presented on the receptor surface, similar to the phosphatidylinositol binding mode observed in the PIP3-SF-1 structure. Thus, data presented in this work reinforce our earlier findings demonstrating that signaling phosphatidylinositols regulate the NR5A receptors LRH-1 and SF-1.


Assuntos
Fosfatidilinositóis/química , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/ultraestrutura , Fator Esteroidogênico 1/ultraestrutura , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Receptor Nuclear Órfão DAX-1/química , Humanos , Modelos Moleculares , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Fator Esteroidogênico 1/química
8.
Biochim Biophys Acta ; 1839(5): 406-14, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24705138

RESUMO

Pluripotent stem cells maintain their pluripotency and undifferentiated status through a network of transcription factors. Liver receptor homolog-1 (Lrh-1) is one of these, and regulates the expression of other important transcription factors such as Oct-3/4 and Nanog. In early embryo and embryonic stem (ES) cells, Lrh-1 is transcribed using a unique promoter. In this study, we investigated the transcriptional regulation of embryonic Lrh-1 using ES and embryonal carcinoma F9 cells. Reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays demonstrated that Sox2 and Gabp proteins bind to the promoter region of embryonic Lrh-1, and are necessary for its activation. The Sox2 site showed strong promoter activity and affinity for protein binding. Upon differentiation into the parietal endoderm by retinoic acid and cAMP, Lrh-1 promoter activity and transcripts were markedly reduced within 24 h. At the same time, Sox2 and Gabp binding to the promoter region of Lrh-1 were decreased, followed by a reduction of their expression. These results indicate that embryonic Lrh-1 expression is regulated by both Sox2 and Gabp. Our study presents new insights into the transcription factor network of pluripotent stem cells.


Assuntos
Células-Tronco Embrionárias/fisiologia , Fator de Transcrição de Proteínas de Ligação GA/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição SOXB1/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Regulação da Expressão Gênica , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transcrição Gênica , Transfecção
9.
Biochem Biophys Res Commun ; 461(3): 450-5, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-25869073

RESUMO

Liver receptor homolog-1 (LRH-1) plays an important role in the onset and progression of many cancer types. However, the role of LRH-1 in osteosarcoma has not been well investigated. In this study, the critical role of LRH-1 in osteosarcoma cells was described. Quantitative polymerase chain reaction and Western blot analysis results revealed that LRH-1 was highly overexpressed in osteosarcoma cells. LRH-1 was knocked down by small interfering RNA (siRNA), and this phenomenon significantly inhibited osteosarcoma cell proliferation. Bioinformatics analysis results showed that LRH-1 contained putative binding sites of microRNA-451 (miR-451); this result was further validated through a dual-luciferase activity reporter assay. miR-451 was overexpressed in osteosarcoma cells through transfection of miR-451 mimics; miR-451 overexpression then significantly inhibited LRH-1 expression and cell proliferation. The loss of LRH-1 by siRNA or miR-451 mimics significantly impaired Wnt/ß-catenin activity, leading to G0/G1 cell cycle arrest. Results showed that LRH-1 is implicated in osteosarcoma. Therefore, miR-451-induced suppression of LRH-1 can be a novel therapy to treat osteosarcoma.


Assuntos
Proliferação de Células , MicroRNAs/fisiologia , Osteossarcoma/patologia , Receptores Citoplasmáticos e Nucleares/genética , Linhagem Celular Tumoral , Inativação Gênica , Humanos
10.
J Anat ; 227(4): 541-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26241668

RESUMO

In the last decade, the study of human sperm anatomy, at molecular level, has revealed the presence of several nuclear protein receptors. In this work, we examined the expression profile and the ultrastructural localization of liver receptor homolog-1 (LRH-1) in human spermatozoa. We evidenced the presence of the receptor by Western blotting and real time-RT-PCR. Furthermore, we used immunogold electron microscopy to investigate the sperm anatomical regions containing LRH-1. The receptor was mainly located in the sperm head, whereas its expression was reduced in the neck and across the tail. Interestingly, we observed the presence of LRH-1 in different stages of testicular germ cell development by immunohistochemistry. In somatic cells, it has been suggested that the LRH-1 pathway is tightly linked with estrogen signaling and the important role of estradiol has been widely studied in sperm cells. To assess the significance of LRH-1 in male gametes and to deepen understanding of the role of estrogens in these cells, we investigated important sperm features such as motility, survival and capacitation. Spermatozoa were treated with 10 nm estradiol and the inhibition of LRH-1 reversed the estradiol stimulatory action. From our data, we discovered that human spermatozoa can be considered a new site of expression for LRH-1, evidencing its role in sperm motility, survival and cholesterol efflux. Furthermore, we may presume that in spermatozoa the LRH-1 effects are closely integrated with the estrogen signaling, supporting LRH-1 as a downstream effector of the estradiol pathway on some sperm functions.


Assuntos
Estrogênios/metabolismo , Regulação da Expressão Gênica , RNA/genética , Receptores Citoplasmáticos e Nucleares/genética , Espermatozoides/metabolismo , Western Blotting , Diferenciação Celular , Sobrevivência Celular , Humanos , Imuno-Histoquímica , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Receptores Citoplasmáticos e Nucleares/biossíntese , Transdução de Sinais , Motilidade dos Espermatozoides , Espermatozoides/citologia
11.
Zoolog Sci ; 32(4): 323-30, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26245218

RESUMO

Steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1) belong to the nuclear receptor superfamily and are categorized as orphan receptors. In addition to other nuclear receptors, these play roles in various physiological phenomena by regulating the transcription of target genes. Both factors share very similar structures and exhibit common functions. Of these, the roles of SF-1 and LRH-1 in steroidogenesis are the most important, especially that of SF-1, which was originally discovered and named to reflect such roles. SF-1 and LRH-1 are essential for steroid hormone production in gonads and adrenal glands through the regulation of various steroidogenesis-related genes. As SF-1 is also necessary for the development of gonads and adrenal glands, it is also considered a master regulator of steroidogenesis. Recent studies have clearly demonstrated that LRH-1 also represents another master regulator of steroidogenesis, which similarly to SF-1, can induce differentiation of non-steroidogenic stem cells into steroidogenic cells. Here, we review the functions of both factors in these steroidogenesis-related phenomena.


Assuntos
Receptores Citoplasmáticos e Nucleares/fisiologia , Fator Esteroidogênico 1/metabolismo , Esteroides/biossíntese , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator Esteroidogênico 1/genética , Esteroides/metabolismo
12.
Biochem Biophys Res Commun ; 438(3): 533-9, 2013 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-23911789

RESUMO

The expression of orphan nuclear receptor Liver Receptor Homolog-1 (LRH-1) is elevated in breast cancer and promotes proliferation, migration and invasion in vitro. LRH-1 expression is regulated by oestrogen (E2), with LRH-1 mRNA transcript levels higher in oestrogen receptor α (ERα) positive (ER+) breast cancer cells compared to ER- cells. However, the presence of LRH-1 protein in ER- cells suggests discordance between mRNA transcript levels and protein expression. To understand this, we investigated the impact of mRNA and protein stability in determining LRH-1 protein levels in breast cancer cells. LRH-1 transcript levels were significantly higher in ER+ versus ER- breast cancer cells lines; however LRH-1 protein was expressed at similar levels. We found LRH-1 mRNA and protein was more stable in ER- compared to ER+ cell lines. The tumor-specific LRH-1 variant isoform, LRH-1v4, which is highly responsive to E2, showed increased mRNA stability in ER- versus ER+ cells. In addition, in MCF-7 and T47-D cell lines, LRH-1 total mRNA stability was reduced with E2 treatment, this effect mediated by ERα. Our data demonstrates that in ER- cells, increased mRNA and protein stability contribute to the abundant protein expression levels. Expression and immunolocalisation of LRH-1 in ER- cells as well as ER- tumors suggests a possible role in the development of ER- tumors. The modulation of LRH-1 bioactivity may therefore be beneficial as a treatment option in both ER- and ER+ breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Estabilidade Proteica/efeitos dos fármacos , Estabilidade de RNA/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Isoformas de Proteínas/genética , Estabilidade de RNA/genética , RNA Mensageiro/metabolismo
13.
Mol Oncol ; 17(8): 1545-1566, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36861295

RESUMO

Control of tumour development and growth by the immune system critically defines patient fate and survival. What regulates the escape of colorectal tumours from destruction by the immune system remains currently unclear. Here, we investigated the role of intestinal synthesis of glucocorticoids in the tumour development during an inflammation-induced mouse model of colorectal cancer. We demonstrate that the local synthesis of immunoregulatory glucocorticoids has dual roles in the regulation of intestinal inflammation and tumour development. In the inflammation phase, LRH-1/Nr5A2-regulated and Cyp11b1-mediated intestinal glucocorticoid synthesis prevents tumour development and growth. In established tumours, however, tumour-autonomous Cyp11b1-mediated glucocorticoid synthesis suppresses anti-tumour immune responses and promotes immune escape. Transplantation of glucocorticoid synthesis-proficient colorectal tumour organoids into immunocompetent recipient mice resulted in rapid tumour growth, whereas transplantation of Cyp11b1-deleted and glucocorticoid synthesis-deficient tumour organoids was characterized by reduced tumour growth and increased immune cell infiltration. In human colorectal tumours, high expression of steroidogenic enzymes correlated with the expression of other immune checkpoints and suppressive cytokines, and negatively correlated with overall patients' survival. Thus, LRH-1-regulated tumour-specific glucocorticoid synthesis contributes to tumour immune escape and represents a novel potential therapeutic target.


Assuntos
Neoplasias Colorretais , Glucocorticoides , Humanos , Camundongos , Animais , Glucocorticoides/farmacologia , Esteroide 11-beta-Hidroxilase/metabolismo , Intestinos , Inflamação , Neoplasias Colorretais/genética
14.
J Endocr Soc ; 6(8): bvac095, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35854979

RESUMO

Context: Adiponectin is an adipokine mainly secreted by adipocytes that regulates the metabolism of lipids and glucose. Liver receptor homolog-1 (LRH-1), also named NR5A2, is a nuclear receptor that regulates lipid metabolism and homeostasis. Objective: The purpose of this study was to compare adiponectin and LRH-1 messenger RNA (mRNA) expression in adipose tissue and LRH-1 expression in skeletal muscle between men and women at baseline and to study the effects of aerobic exercise (AEX) training or weight loss (WL) on their expression. Methods: This hospital and university setting study included 62 overweight and obese men (n = 23) and women (n = 39) older than 45 years, of whom 41 completed 6 months of WL (n = 21) or AEX (n = 20). Outcomes included abdominal and gluteal adipose tissue and skeletal muscle gene expression. Results: Adiponectin and LRH-1 mRNA expression in adipose tissue and LRH-1 mRNA expression in skeletal muscle is higher in women than in men (P < .05). Adiponectin mRNA expression in gluteal and abdominal adipose tissue did not change significantly after AEX or WL. LRH-1 mRNA expression increased both in adipose tissue and skeletal muscle after AEX (P < .05) and the change in muscle LRH-1 was different between the groups (P < .05). Adiponectin was positively correlated to LRH-1 in adipose tissue (P < .001). The change in maximal oxygen consumption related to the change in LRH-1 mRNA (r = 0.43; P = .01). Conclusion: LRH-1, as a nuclear reporter, may activate adiponectin mRNA expression in adipose tissue and increases after AEX.

15.
Virology ; 571: 52-58, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35509137

RESUMO

ß-catenin regulates HBV transcription in cell culture and viral biosynthesis in vivo in the transgenic mouse model of chronic HBV infection. Therefore, it is important to understand which transcription factor activities are coactivated by ß-catenin to enhance HBV biosynthesis. The effect of ß-catenin expression in the context of nuclear receptor-mediated HBV transcription was evaluated initially in the human embryonic kidney cell line, HEK293T. Reporter gene and viral replication assays revealed that ß-catenin can coactivate HBV transcription through some, most predominantly liver receptor homolog 1 (LRH1), but not all nuclear receptors known to activate viral biosynthesis. Similarly, ß-catenin activated nuclear receptor-mediated HBV transcription and replication in the human hepatoma cell line, Huh7, primarily through its effect on the farnesoid X receptor α (FXRα). These data indicate that ß-catenin can enhance nuclear receptor-mediated HBV biosynthesis, but the relative importance of various transcription factors is dependent upon the precise cellular environment.


Assuntos
Vírus da Hepatite B , Hepatite B , Animais , Células HEK293 , Vírus da Hepatite B/fisiologia , Humanos , Camundongos , Receptores Citoplasmáticos e Nucleares/genética , Transcrição Viral , Replicação Viral , beta Catenina/genética , beta Catenina/metabolismo
16.
J Int Med Res ; 50(6): 3000605221105163, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35748030

RESUMO

OBJECTIVE: To investigate the regulation of local aldosterone synthesis by physiological stimulants in the murine gut. METHODS: Male mice were fed for 14 days with normal, high (1.6%) or low (0.01%) sodium diets. Tissue liver receptor homolog-1 and aldosterone in the colon and caecum were detected using an enzyme-linked immunosorbent assay (ELISA). Released corticosterone and aldosterone in tissue incubation experiments after stimulation with angiotensin II (Ang II) and dibutyryl-cAMP (DBA; the second messenger of adrenocorticotropic hormone) were assayed using an ELISA. Tissue aldosterone synthase (CYP11B2) protein levels were measured using an ELISA and Western blots. RESULTS: In incubated colon tissues, aldosterone synthase levels were increased by a low-sodium diet; and by Ang II and DBA in the normal diet group. Release of aldosterone into the incubation buffer was increased from the colon by a low-sodium diet and decreased by a high-sodium diet in parallel with changes in aldosterone synthase levels. In mice fed a normal diet, colon incubation with both Ang II and DBA increased the release of aldosterone as well as its precursor corticosterone. CONCLUSION: Local aldosterone synthesis in the large intestine is stimulated by a low-sodium diet, dibutyryl-cAMP and Ang II similar to the adrenal glands.


Assuntos
Aldosterona , Citocromo P-450 CYP11B2 , Angiotensina II/farmacologia , Animais , Corticosterona , Humanos , Intestino Grosso/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Sódio
17.
Endocrine ; 74(2): 413-420, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34129175

RESUMO

PURPOSE: Polycystic ovary syndrome (PCOS) is considered one of the most common endocrine disorders with heterogeneity. There are also reports that liver receptor homolog 1 [LRH-1 or nuclear receptor subfamily 5 group A member 2] plays an important role in the reproductive system. But up to now, there are no reports related to the link with PCOS and LRH-1. In this study, we aimed to detect the LRH-1 expression in the ovarian granulosa cell (GC) of PCOS patients and explore the potential relationship between LRH-1 and PCOS. METHODS: In all, 146 follicular fluid samples were collected in this study, including 72 from PCOS patients and 74 from control patients who underwent intracytoplasmic sperm injection or in vitro fertilization-embryo transfer. The ovarian GCs were extracted from the patient's follicular fluid by magnetic-activated cell sorting method, and real-time quantitative PCR was used to measure the expression of LRH-1 in ovarian GCs. Then we analyzed the correlation between the expression level of LRH-1 and the clinical characteristics of the patient by using Pearson Correlation analysis. RESULTS: The expression of LRH-1 was significantly higher in PCOS patients ovarian GCs than that in the control patients [(1.38 ± 0.47) vs (1.03 ± 0.32), t = 5.327, p < 0.0001], and it was positively correlated with antral follicles counting (r = 0.3607, p < 0.0001) and the serum anti-Mullerian hormone (r = 0.2662, p = 0.0012), luteotropic hormone (r = 0.2518, p = 0.0022), testosterone (r = 0.2794, p = 0.0006) in all patients. No statistical significance between LRH-1 and body mass index, follicle-stimulating hormone, homeostasis model assessment of insulin resistance, dehydroepiandrosterone sulfate, progesterone. CONCLUSIONS: Compared with the control group, we found that LRH-1 was highly expressed in the ovarian GCs of PCOS patients. Our study has revealed the relationship between the LRH-1 expression and PCOS, which suggested that LRH-1 may play an important role in ovulation disorders. While this finding provided new ideas for the study of pathogenesis, it also provided a theoretical basis for the clinical diagnosis and treatment for PCOS.


Assuntos
Células da Granulosa/metabolismo , Síndrome do Ovário Policístico , Receptores Citoplasmáticos e Nucleares , Hormônio Antimülleriano , Feminino , Líquido Folicular , Humanos , Síndrome do Ovário Policístico/genética , Receptores Citoplasmáticos e Nucleares/genética
18.
Cancers (Basel) ; 13(4)2021 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-33672730

RESUMO

The process of self-renewal in normal intestinal epithelium is characterized by a fine balance between proliferation, differentiation, migration, and cell death. When even one of these aspects escapes the normal control, cellular proliferation and differentiation are impaired, with consequent onset of tumorigenesis. In humans, colorectal cancer (CRC) is the main pathological manifestation of this derangement. Nowadays, CRC is the world's fourth most deadly cancer with a limited survival after treatment. Several conditions can predispose to CRC development, including dietary habits and pre-existing inflammatory bowel diseases. Given their extraordinary ability to interact with DNA, it is widely known that nuclear receptors play a key role in the regulation of intestinal epithelium, orchestrating the expression of a series of genes involved in developmental and homeostatic pathways. In particular, the nuclear receptor Liver Receptor Homolog-1 (LRH-1), highly expressed in the stem cells localized in the crypts, promotes intestine cell proliferation and renewal in both direct and indirect DNA-binding manner. Furthermore, LRH-1 is extensively correlated with diverse intestinal inflammatory pathways. These evidence shed a light in the dynamic intestinal microenvironment in which increased regenerative epithelial cell turnover, mutagenic insults, and chronic DNA damages triggered by factors within an inflammatory cell-rich microenvironment act synergistically to favor cancer onset and progression.

19.
Front Oncol ; 10: 583566, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33194722

RESUMO

BACKGROUND: Liver receptor homolog 1 (LRH1) plays a vital role in several human cancers, but its role in ovarian cancer (OC) remains unclear. We aimed to explore the functions of LRH1 and its clinical relevance. METHODS: LRH1 expression was evaluated by immunohistochemistry and reverse transcription quantitative polymerase chain reaction (RT-qPCR). The effects of LRH1 on tumor cell proliferation, migration and epithelial-mesenchymal transition (EMT) were evaluated in vitro. Furthermore, bioinformatics analysis was applied to predict the functions of LRH1. RESULTS: RT-qPCR showed that LRH1 mRNA expression was higher in the invasive lesions (P < 0.05). LRH1 overexpression was extremely related with elevated International Federation of Gynecology and Obstetrics (FIGO) stage (P = 0.001), lymph node metastasis (P = 0.011), peritoneal metastasis (P = 0.001), and platinum resistance (P = 0.037). Furthermore, LRH1 expression was an independent prognostic index for disease-free survival in patients with OC (P = 0.041). LRH1 overexpression (P = 0.011), FIGO stage (P < 0.001), and ascites (P = 0.015) independently affected peritoneal metastasis in patients with OC. LRH1 knockdown significantly inhibited the proliferation, migration, and EMT of human OC cells (P < 0.05); however, it reversed cisplatin resistance. Bioinformatics analysis indicated that the functions of LRH1 were associated with the PRC1 complex, nuclear ubiquitin ligase complex, and Polycomb-group (PcG) proteins. CONCLUSIONS: This study provides evidence of the predictive value of LRH1 on peritoneal metastasis and poor outcome and highlights the potential role of LRH1 as a biomarker for the targeted therapy of OC. Furthermore, LRH1 promotes OC cell proliferation, migration, and EMT in vitro, and its functions may be associated with PRC1 complex, nuclear ubiquitin ligase complex, and PcG proteins.

20.
Front Immunol ; 10: 1438, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31316505

RESUMO

Glucocorticoids (GCs) are steroid hormones predominantly produced in the adrenal glands in response to physiological cues and stress. Adrenal GCs mediate potent anti-inflammatory and immunosuppressive functions. Accumulating evidence in the past two decades has demonstrated other extra-adrenal organs and tissues capable of synthesizing GCs. This review discusses the role and regulation of GC synthesis in the intestinal epithelium in the regulation of normal immune homeostasis, inflammatory diseases of the intestinal mucosa, and the development of intestinal tumors.


Assuntos
Glândulas Suprarrenais/imunologia , Glucocorticoides/imunologia , Homeostase/imunologia , Fenômenos do Sistema Imunitário , Mucosa Intestinal/imunologia , Glândulas Suprarrenais/metabolismo , Animais , Citocinas/imunologia , Citocinas/metabolismo , Glucocorticoides/biossíntese , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/imunologia , Neoplasias Intestinais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA