Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 80(2): 279-295.e8, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33065020

RESUMO

The PTEN tumor suppressor controls cell death and survival by regulating functions of various molecular targets. While the role of PTEN lipid-phosphatase activity on PtdIns(3,4,5)P3 and inhibition of PI3K pathway is well characterized, the biological relevance of PTEN protein-phosphatase activity remains undefined. Here, using knockin (KI) mice harboring cancer-associated and functionally relevant missense mutations, we show that although loss of PTEN lipid-phosphatase function cooperates with oncogenic PI3K to promote rapid mammary tumorigenesis, the additional loss of PTEN protein-phosphatase activity triggered an extensive cell death response evident in early and advanced mammary tumors. Omics and drug-targeting studies revealed that PI3Ks act to reduce glucocorticoid receptor (GR) levels, which are rescued by loss of PTEN protein-phosphatase activity to restrain cell survival. Thus, we find that the dual regulation of GR by PI3K and PTEN functions as a rheostat that can be exploited for the treatment of PTEN loss-driven cancers.


Assuntos
Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , PTEN Fosfo-Hidrolase/metabolismo , Receptores de Glucocorticoides/metabolismo , Animais , Carcinogênese , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Dexametasona/farmacologia , Feminino , Humanos , Isoenzimas/metabolismo , Camundongos , Modelos Biológicos , Mutação/genética , Organoides/patologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Estabilidade Proteica , Proteoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
Neurobiol Dis ; 156: 105422, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34126164

RESUMO

Synthetic glucocorticoids (sGCs) such as dexamethasone (DEX), while used to mitigate inflammation and disease progression in premature infants with severe bronchopulmonary dysplasia (BPD), are also associated with significant adverse neurologic effects such as reductions in myelination and abnormalities in neuroanatomical development. Ciclesonide (CIC) is a sGC prodrug approved for asthma treatment that exhibits limited systemic side effects. Carboxylesterases enriched in the lower airways convert CIC to the glucocorticoid receptor (GR) agonist des-CIC. We therefore examined whether CIC would likewise activate GR in neonatal lung but have limited adverse extra-pulmonary effects, particularly in the developing brain. Neonatal rats were administered subcutaneous injections of CIC, DEX or vehicle from postnatal days 1-5 (PND1-PND5). Systemic effects linked to DEX exposure, including reduced body and brain weight, were not observed in CIC treated neonates. Furthermore, CIC did not trigger the long-lasting reduction in myelin basic protein expression in the cerebral cortex nor cerebellar size caused by neonatal DEX exposure. Conversely, DEX and CIC were both effective at inducing the expression of select GR target genes in neonatal lung, including those implicated in lung-protective and anti-inflammatory effects. Thus, CIC is a promising, novel candidate drug to treat or prevent BPD in neonates given its activation of GR in neonatal lung and limited adverse neurodevelopmental effects. Furthermore, since sGCs such as DEX administered to pregnant women in pre-term labor can adversely affect fetal brain development, the neurological-sparing properties of CIC, make it an attractive alternative for DEX to treat pregnant women severely ill with respiratory illness, such as with asthma exacerbations or COVID-19 infections.


Assuntos
Cerebelo/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Glucocorticoides , Pulmão/efeitos dos fármacos , Pregnenodionas/farmacologia , Pró-Fármacos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Dexametasona/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Básica da Mielina/biossíntese , Tamanho do Órgão/efeitos dos fármacos , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
3.
Respir Res ; 19(1): 155, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-30126423

RESUMO

BACKGROUND: Prenatal glucocorticoid treatment decreases alveolar tissue volumes and facilitates fetal lung maturation, however the mechanisms responsible are largely unknown. This study examines whether changes in versican levels or sulphation patterns of chondroitin sulphate (CS) side chains, are associated with glucocorticoid-induced reductions in peri-alveolar tissue volumes. METHODS: Lung tissue was collected from 1) fetal sheep at 131 ± 0.1 days gestational age (GA) infused with cortisol (122-131d GA) to prematurely induce a pre-parturient-like rise in circulating cortisol, 2) fetal sheep at 143d GA bilaterally adrenalectomised (ADX) at 112d GA to remove endogenous cortisol and 3) fetal sheep at 124d GA in which bolus doses (2 × 11.4 mg) of betamethasone were administered to the pregnant ewe. The level and distribution of versican and CS glycosaminoglycans (GAG) were determined using immunohistochemistry (IHC). Fluorophore assisted carbohydrate electrophoresis (FACE) was used to determine changes in CS sulphation patterns. RESULTS: Cortisol infusion significantly decreased chondrotin-6-sulphate levels (C-6-S) to 16.4 ± 0.7 AU, compared with saline-infused fetuses (18.9 ± 0.7 AU: p = 0.04) but did not significantly alter the level of versican or chondroitin-4-sulphate (C-4-S). ADX significantly increased the level of C-4-S (28.2 ± 2.2 AU), compared with sham-operated fetuses (17.8 ± 2.0 AU; p = 0.006) without altering versican or C-6-S levels. Betamethasone significantly decreased versican, C-4-S and C-6-S in the fetal sheep lung (19.2 ± 0.9 AU, 24.9 ± 1.4 AU and 23.2 ± 1.0 AU, respectively), compared with saline-exposed fetuses (24.3 ± 0.4 AU, p = 0.0004; 33.3±0.6 AU, p = 0.0003; 29.8±1.3 AU, 0.03, respectively). CONCLUSIONS: These results indicate that glucocorticoids alter versican levels and CS side chain microstructure in alveolar lung tissue. Betamethasone appears to have a greater impact on versican and CS side chains than cortisol.


Assuntos
Sulfatos de Condroitina/biossíntese , Desenvolvimento Fetal/fisiologia , Glucocorticoides/farmacologia , Pulmão/metabolismo , Proteoglicanas/biossíntese , Versicanas/biossíntese , Animais , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Feto , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Gravidez , Ovinos
4.
Dev Dyn ; 244(2): 99-109, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25523132

RESUMO

BACKGROUND: Trop2 was first discovered as a biomarker of invasive trophoblast cells. Since then most research has focused on its role in tumourigenesis because it is highly expressed in the vast majority of human tumours and animal models of cancer. It is also highly expressed in stem cells and in many organs during development. RESULTS: We review the multifaceted role of Trop2 during development and tumourigenesis, including its role in regulating cell proliferation and migration, self-renewal, and maintenance of basement membrane integrity. We discuss the evolution of Trop2 and its related protein Epcam (Trop1), including their distinct roles. Mutation of Trop2 leads to gelatinous drop-like corneal dystrophy, whereas over-expression of Trop2 in human tumours promotes tumour aggressiveness and increases mortality. Although Trop2 expression is sufficient to promote tumour growth, the surprising discovery that Trop2-null mice have an increased risk of tumour development has highlighted the complexity of Trop2 signaling. Recently, studies have begun to identify the mechanisms underlying TROP2's functions, including regulated intramembrane proteolysis or specific interactions with integrin b1 and claudin proteins. CONCLUSIONS: Understanding the mechanisms underlying TROP2 signaling will clarify its role during development, aid in the development of better cancer treatments and unlock a promising new direction in regenerative medicine.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Distrofias Hereditárias da Córnea/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/metabolismo , Transdução de Sinais/genética , Animais , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Movimento Celular/genética , Proliferação de Células/genética , Conexinas/genética , Conexinas/metabolismo , Distrofias Hereditárias da Córnea/genética , Humanos , Camundongos , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Proteína beta-1 de Junções Comunicantes
5.
Am J Respir Cell Mol Biol ; 50(2): 419-28, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24053134

RESUMO

Endogenous glucocorticoid (GC) hormones, signaling via the GC receptor (GR), are essential for normal lung development, and synthetic GCs are routinely used to treat respiratory disorders of very preterm babies. Germline GR knockout (GR(-/-)) mice show immature lung morphology and severe lung cellular hyperplasia during embryogenesis and die at birth due to respiratory failure. Two recent studies have reported contradictory results regarding the necessity for GR expression in specific lung germ layers during respiratory maturation. We further investigate in detail the lung phenotype in mice with a conditional deletion of GR in the endothelium, mesenchyme, and lung epithelium. We show that loss of GR in the mesenchyme alone produces a retarded lung phenotype almost identical to that of germline GR(-/-) mice, including severe postnatal lethality and lung cell hyperplasia. Loss of GR in lung epithelial cells and the endothelium had no gross effect on survival or lung morphology, but loss of epithelial GR caused increased cell proliferation in multiple compartments. Mesenchymal GR loss also produced increased epithelial cell proliferation, implying the existence of GC-regulated germ layer cross-talk. Protein levels of GR-mediated cell cycle regulators, including the cyclin-dependent kinase inhibitor p21(CIP1) and the growth factor midkine, were unaffected by mesenchymal GR deletion, yet expression of the extracellular matrix proteoglycan versican was up-regulated in the distal lung on loss of mesenchymal GR. These results show that GR-mediated signaling from the mesenchyme regulates respiratory maturation and ultimately survival at birth and that a key GR-repressed transcriptional target in lung mesenchymal cells is versican.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/metabolismo , Pulmão/citologia , Mesoderma/citologia , Receptores de Glucocorticoides/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Citocinas/farmacologia , Modelos Animais de Doenças , Epitélio/metabolismo , Glucocorticoides/genética , Pulmão/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Midkina , Receptores de Glucocorticoides/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
6.
Immunol Cell Biol ; 92(2): 148-55, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24217809

RESUMO

Studies using glucocorticoid receptor exon 2-disrupted knockout (GR2KO) mice provided strong evidence against an obligatory role for glucocorticoid receptor (GR) signalling in T-cell selection. These mice express a truncated form of the GR that is incapable of transmitting a range of glucocorticoid (GC)-induced signals, including GC-induced thymocyte death. However, one study that suggested that truncated GR function is preserved in the context of GR-mediated repression of T-cell activation-induced genes, challenged earlier conclusions derived from the use of these mice. Because GR versus T-cell receptor (TCR) signalling cross-talk is the means by which GCs are hypothesized to have a role in T-cell selection, we reassessed the utility of GR2KO mice to study the role of the GR in this process. Here, we show that GR-mediated repression of TCR signalling is impaired in GR2KO T cells in terms of TCR-induced activation, proliferation and cytokine production. GC-induced apoptosis was largely abolished in peripheral T cells, and induction of the GC-responsive molecule, interleukin-7 receptor, was also severely reduced in GR2KO thymocytes. Together, these data strongly re-affirm conclusions derived from earlier studies of these mice that the GR is not obligatory for normal T-cell selection.


Assuntos
Seleção Clonal Mediada por Antígeno/fisiologia , Éxons , Glucocorticoides/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Glucocorticoides/imunologia , Transdução de Sinais/imunologia , Animais , Glucocorticoides/genética , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/genética , Receptores de Glucocorticoides/genética , Transdução de Sinais/genética , Timo/imunologia
7.
Immunol Cell Biol ; 92(10): 825-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25047641

RESUMO

Glucocorticoids (GCs) are powerful inhibitors of inflammation and immunity. Although glucocorticoid-induced cell death (GICD) is an important part of GCs actions, the cell types and molecular mechanisms involved are not well understood. Untranslated exon 1A3 of the human glucocorticoid receptor (GR) gene is a major determinant of GICD in GICD-sensitive human cancer cell lines, operating to dynamically upregulate GR levels in response to GCs. We measured the GICD sensitivity of freshly isolated peripheral blood mononuclear cells and thymocytes to dexamethasone in vitro, relating this to GR exon 1A3 expression. A clear GICD sensitivity hierarchy was detected: B cells>thymocytes/natural killer (NK) cells>peripheral T cells. Within thymocyte populations, GICD sensitivity decreased with maturation. Interestingly, NK cell subsets were differentially sensitive to GICD, with CD16(+)CD56(int) (cytotoxic) NK cells being highly resistant to GICD, whereas CD16(-)CD56(hi) (cytokine producing) NK cells were highly sensitive (similar to B cells). B-cell GICD was rescued by co-culture with interleukin-4. Strikingly, although no significant increases in GR protein were observed during 48 h of culture of GICD-sensitive and -resistant cells alike, GR 1A3 expression was increased over pre-culture levels in a manner directly proportional to the GICD sensitivity of each cell type. Accordingly, this is the first evidence that the GR exon 1A3 promoter is differentially regulated during thymic development and maturation of human T cells. Furthermore, human peripheral blood B cells are exquisitely GICD-sensitive in vitro, giving new insight into how GCs may downregulate immunity. Collectively, these data show that GR 1A3 expression is tied with GICD sensitivity in human lymphocytes, underscoring the potential for GR 1A3 expression to be used as a biomarker for sensitivity to GICD.


Assuntos
Apoptose/fisiologia , Glucocorticoides/toxicidade , Linfócitos/efeitos dos fármacos , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/genética , Técnicas de Cocultura , Regulação para Baixo , Expressão Gênica , Humanos , Interleucina-4/imunologia , Linfócitos/imunologia , Regulação para Cima
8.
BMC Pulm Med ; 14: 59, 2014 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-24712406

RESUMO

BACKGROUND: The lungs of very preterm human babies display deficits in alveolarization and vascularization as a result of the clinical use of high oxygen treatment (leading to hyperoxia) required to decrease the risk of mortality. Detailed analyses of the persistence of the respiratory deficits following this treatment and means to restore a normal state have not been investigated in full detail. In this study, high oxygen administration to neonatal mouse lungs was established as a proxy to hyperoxia in human preterm infant lungs, to better characterize the associated deficits and thus provide a means to assist in the development of treatments in the future. METHODS: Ninety percent oxygen was administered to newborn mice for four consecutive days. The effects of this treatment upon alveolarization and vascularization were investigated by morphometric, histochemical, immunohistochemical and protein analyses at day five (D5), D28 and D56 postpartum. RESULTS: Relative to control untreated lungs, septation of hyperoxic lungs was significantly reduced and airspaces were significantly enlarged at all stages examined. Furthermore, compared to controls, the number of secondary septa per tissue area was significantly reduced at D5, significantly increased at D28 and then the same as controls at D56. Analysis of vascularization parameters indicated a reduction in mature blood vessel number and the amount of Pecam1 at D5. Both of these parameters returned to control levels by D28. CONCLUSIONS: This study suggests that administration of high oxygen to underdeveloped lungs has a transient reductive effect on secondary septal number and pulmonary vascularization and a significant long-term reduction in alveolarization persisting into adulthood. This model can be used for future research of premature lung disease therapies in humans, addressing these short term septal and vascular and long term alveolar deficits, specifically relating to injury by hyperoxia.


Assuntos
Hiperóxia/fisiopatologia , Pulmão/irrigação sanguínea , Pulmão/crescimento & desenvolvimento , Oxigênio/administração & dosagem , Animais , Animais Recém-Nascidos , Feminino , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/crescimento & desenvolvimento , Fatores de Tempo
9.
Am J Physiol Lung Cell Mol Physiol ; 305(7): L508-21, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23893297

RESUMO

Proliferation and migration of fibroblasts are vital for fetal lung development. However, the regulatory mechanisms are poorly understood. We have previously shown that TROP2 gene expression is closely associated with fetal lung cell proliferation in vivo and that TROP2 knockdown decreases proliferation of fetal lung fibroblasts in culture. We hypothesized that the Trop2 protein also regulates the morphology and motility of fetal lung fibroblasts. Fibroblasts isolated from fetal rat lungs (gestational age embryonic day 19) adopted a myofibroblast-like morphology in culture. Trop2 protein was localized to lamellipodia. TROP2 siRNA significantly decreased: TROP2 mRNA levels by 77%, the proportion of cells containing Trop2 protein by 70%, and cell proliferation by 50%. TROP2 siRNA also decreased the degree of motility as determined by the number of gridlines that cells moved across (2.2 ± 0.2 vs. 3.2 ± 0.2; P < 0.001). TROP2 knockdown altered cell morphology, causing a notable absence of lamellipodia and abnormal localization of components of the cell migration apparatus, and it reduced phosphorylated ERK1 and ERK2 levels. In contrast, TROP2 overexpression significantly increased: TROP2 mRNA levels by 40-fold, cell proliferation by 40%, the proportion of cells that were motile by 20%, and the number of gridlines that cells moved across (2.1 ± 0.2 vs. 1.6 ± 0.1; P < 0.001). Our data suggest that Trop2 regulates cell proliferation and motility and that it does so by regulating the ERK pathway and several critical components of the cell migration apparatus.


Assuntos
Antígenos de Neoplasias/metabolismo , Movimento Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Pulmão/embriologia , Proteínas Oncogênicas/metabolismo , Pseudópodes/fisiologia , Animais , Antígenos de Neoplasias/genética , Proliferação de Células , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Pulmão/citologia , Proteínas Oncogênicas/genética , Fosforilação , Gravidez , Pseudópodes/metabolismo , Interferência de RNA , RNA Mensageiro , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley
10.
PLoS Genet ; 6(10): e1001170, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20975950

RESUMO

Zn²(+)-finger proteins comprise one of the largest protein superfamilies with diverse biological functions. The ATM substrate Chk2-interacting Zn²(+)-finger protein (ASCIZ; also known as ATMIN and ZNF822) was originally linked to functions in the DNA base damage response and has also been proposed to be an essential cofactor of the ATM kinase. Here we show that absence of ASCIZ leads to p53-independent late-embryonic lethality in mice. Asciz-deficient primary fibroblasts exhibit increased sensitivity to DNA base damaging agents MMS and H2O2, but Asciz deletion knock-down does not affect ATM levels and activation in mouse, chicken, or human cells. Unexpectedly, Asciz-deficient embryos also exhibit severe respiratory tract defects with complete pulmonary agenesis and severe tracheal atresia. Nkx2.1-expressing respiratory precursors are still specified in the absence of ASCIZ, but fail to segregate properly within the ventral foregut, and as a consequence lung buds never form and separation of the trachea from the oesophagus stalls early. Comparison of phenotypes suggests that ASCIZ functions between Wnt2-2b/ß-catenin and FGF10/FGF-receptor 2b signaling pathways in the mesodermal/endodermal crosstalk regulating early respiratory development. We also find that ASCIZ can activate expression of reporter genes via its SQ/TQ-cluster domain in vitro, suggesting that it may exert its developmental functions as a transcription factor. Altogether, the data indicate that, in addition to its role in the DNA base damage response, ASCIZ has separate developmental functions as an essential regulator of respiratory organogenesis.


Assuntos
Proteínas de Transporte/fisiologia , Reparo do DNA/fisiologia , Pulmão/embriologia , Proteínas Nucleares/fisiologia , Organogênese/fisiologia , Animais , Western Blotting , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Senescência Celular , Dano ao DNA , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Genótipo , Humanos , Peróxido de Hidrogênio/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidantes/farmacologia , Fatores de Tempo , Traqueia/embriologia , Fatores de Transcrição , Raios Ultravioleta
11.
J Endocrinol ; 259(1)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37486000

RESUMO

Primary aldosteronism, or Conn syndrome, is the most common endocrine cause of hypertension. It is associated with a higher risk of cardiovascular, metabolic and renal diseases, as well as a lower quality of life than for hypertension due to other causes. The multi-systemic effects of primary aldosteronism can be attributed to aldosterone-mediated activation of the mineralocorticoid receptor in a range of tissues. In this review, we explore the signalling pathways of the mineralocorticoid receptor, with a shift from the traditional focus on the regulation of renal sodium-potassium exchange to a broader understanding of its role in the modulation of tissue inflammation, fibrosis and remodelling. The appreciation of primary aldosteronism as a multi-system disease with tissue-specific pathophysiology may lead to more vigilant testing and earlier institution of targeted interventions.


Assuntos
Hiperaldosteronismo , Hipertensão , Humanos , Hiperaldosteronismo/complicações , Receptores de Mineralocorticoides/metabolismo , Qualidade de Vida , Antagonistas de Receptores de Mineralocorticoides , Aldosterona/metabolismo , Hipertensão/complicações
12.
Vitam Horm ; 123: 285-312, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37717989

RESUMO

The cellular response to the adrenal steroid aldosterone is mediated by the mineralocorticoid receptor (MR), a member of the nuclear receptor superfamily of ligand-dependent transcription factors. The MR binds more than one physiological ligand with binding at the MR determined by pre-receptor metabolism of glucocorticoid ligands by 11ß hydroxysteroid dehydrogenase type 2. The MR has a wide tissue distribution with multiple roles beyond the classical role in electrolyte homeostasis including cardiovascular function, immune cell signaling, neuronal fate and adipocyte differentiation. The MR has three principal functional domains, an N-terminal ligand domain, a central DNA binding domain and a C-terminal, ligand binding domain, with structures having been determined for the latter two domains but not for the whole receptor. MR signal-transduction can be best viewed as a series of interactions which are determined by the conformation conferred on the receptor by ligand binding. This conformation then determines subsequent intra- and inter-molecular interactions. These interactions include chromatin, coregulators and other transcription factors, and additional less well characterized cytoplasmic non-genomic effects via crosstalk with other signaling pathways. This chapter will provide a review of MR structure and function, and an analysis of the critical interactions involved in MR-mediated signal transduction, which contribute to ligand- and tissue-specificity. Understanding the relevant mechanisms for selective MR signaling in terms of these interactions opens the possibility of novel therapeutic approaches for the treatment of MR-mediated diseases.


Assuntos
Aldosterona , Receptores de Mineralocorticoides , Humanos , Ligantes , Transdução de Sinais , Relação Estrutura-Atividade
13.
Nat Commun ; 14(1): 7096, 2023 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-37925459

RESUMO

Disrupted circadian rhythms have been linked to an increased risk of hypertension and cardiovascular disease. However, many studies show inconsistent findings and are not sufficiently powered for targeted subgroup analyses. Using the UK Biobank cohort, we evaluate the association between circadian rhythm-disrupting behaviours, blood pressure (SBP, DBP) and inflammatory markers in >350,000 adults with European white British ancestry. The independent U-shaped relationship between sleep length and SBP/DBP is most prominent with a low inflammatory status. Poor sleep quality and permanent night shift work are also positively associated with SBP/DBP. Although fully adjusting for BMI in the linear regression model attenuated effect sizes, these associations remain significant. Two-sample Mendelian Randomisation (MR) analyses support a potential causal effect of long sleep, short sleep, chronotype, daytime napping and sleep duration on SBP/DBP. Thus, in the current study, we present a positive association between circadian rhythm-disrupting behaviours and SBP/DBP regulation in males and females that is largely independent of age.


Assuntos
Jornada de Trabalho em Turnos , Distúrbios do Início e da Manutenção do Sono , Adulto , Masculino , Feminino , Humanos , Pressão Sanguínea/fisiologia , Bancos de Espécimes Biológicos , Sono/fisiologia , Ritmo Circadiano/fisiologia , Inflamação , Reino Unido
14.
Am J Physiol Lung Cell Mol Physiol ; 301(4): L478-89, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21743029

RESUMO

The factors regulating growth of the developing lung are poorly understood, although the degree of fetal lung expansion is critical. The oncogene Trop2 (trophoblast antigen 2) is upregulated during accelerated fetal lung growth, and we hypothesized that it may regulate normal fetal lung growth. We investigated Trop2 expression in the fetal and neonatal sheep lung during accelerated and delayed lung growth induced by alterations in fetal lung expansion, as well as in response to glucocorticoids. Trop2 expression was measured using real-time PCR and localized spatially using in situ hybridization and immunofluorescence. During normal lung development, Trop2 expression was higher at 90 days gestational age (GA; 4.0 ± 0.8) than at 128 days GA (1.0 ± 0.1), decreased to 0.5 ± 0.1 at 142 days GA (full term ∼147 days GA), and was positively correlated to lung cell proliferation rates (r = 0.953, P < 0.005). Trop2 expression was regulated by fetal lung expansion, but not by glucocorticoids. It was increased nearly threefold by 36 h of increased fetal lung expansion (P < 0.05) and was reduced to ∼55% of control levels by reduced fetal lung expansion (P < 0.05). Trop2 expression was associated with lung cell proliferation during normal and altered lung growth, and the TROP2 protein colocalized with Ki-67-positive cells in the fetal lung. TROP2 was predominantly localized to fibroblasts and type II alveolar epithelial cells. Trop2 small interfering RNA decreased Trop2 expression by ∼75% in cultured fetal rat lung fibroblasts and decreased their proliferation by ∼50%. Cell viability was not affected. This study demonstrates that TROP2 regulates lung cell proliferation during development.


Assuntos
Células Epiteliais Alveolares/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hidrocortisona/farmacologia , Pulmão , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Ovinos/genética , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/efeitos dos fármacos , Animais , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Feto , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Imunofluorescência , Inativação Gênica/efeitos dos fármacos , Hibridização in Situ Fluorescente , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Pulmão/metabolismo , Tamanho do Órgão , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Ovinos/embriologia , Ovinos/metabolismo
15.
Clin Exp Pharmacol Physiol ; 37(11): 1044-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20659135

RESUMO

1. In the present study, we have (i) measured basal blood pressure by telemetry in wild-type (WT) and glucocorticoid receptor knockout (GRKO) mice; (ii) investigated whether or not adrenocorticotrophic hormone (ACTH) can induce hypertension in GRKO mice; and (iii) investigated the effect of mineralocortocoid receptor blockade on the cardiovascular physiology of GRKO mice. 2. Male WT and GRKO mice were treated with ACTH (2mg/kg per day s.c.) or spironolactone (100mg/kg per day s.c.) for 1-2weeks. Blood pressure (BP) was measured using a radiotelemetry system. Urinary Na:K, blood glucose concentrations and haematocrit were also measured during the treatment period. 3. Baseline systolic blood pressure (SBP) was higher in GRKO mice (126±4 mmHg, mean±SEM, n=11) than WT mice (114±2mmHg, n=10; P<0.05). There was no significant difference in baseline haematocrit, blood glucose and urine Na:K ratio in WT and GRKO mice. ACTH raised SBP in WT (135±8mmHg, n=8; P<0.05), but not in GRKO mice (113±9mmHg, n=6). Spironolactone treatment did not alter SBP. 4. Basal SBP was higher in GRKO than WT mice; ACTH raised blood pressure in WT, but not GRKO mice; and spironolactone did not alter BP in GRKO or WT mice. These data, together with a previous study showing both ACTH and corticosterone are increased in GRKO mice, show that the GR is required for the development of ACTH-induced hypertension in mice. Increased endogenous ACTH levels in this model might contribute to the increased basal SBP in GRKO mice, possibly through residual fragments of GR. Mineralocorticoid receptors do not appear to play a critical role in maintaining BP in glucocorticoid receptor deficient mice.


Assuntos
Hormônio Adrenocorticotrópico/farmacologia , Hipertensão/induzido quimicamente , Receptores de Glucocorticoides/fisiologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/genética , Hipertensão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antagonistas de Receptores de Mineralocorticoides , Tamanho do Órgão/efeitos dos fármacos , Receptores de Glucocorticoides/genética , Espironolactona/farmacologia , Timo/efeitos dos fármacos , Timo/metabolismo
16.
J Endocrinol ; 246(2): 123-134, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32464598

RESUMO

MR activation in macrophages is critical for the development of cardiac inflammation and fibrosis. We previously showed that MR activation modifies macrophage pro-inflammatory signalling, changing the cardiac tissue response to injury via both direct gene transcription and JNK/AP-1 second messenger pathways. In contrast, MR-mediated renal electrolyte homeostasis is critically determined by DNA-binding-dependent processes. Hence, ascertaining the relative contribution of MR actions via DNA binding or alternative pathways on macrophage behaviour and cardiac inflammation may provide therapeutic opportunities which separate the cardioprotective effects of MR antagonists from their undesirable renal potassium-conserving effects. We developed new macrophage cell lines either lacking MR or harbouring a mutant MR incapable of DNA binding. Western blot analysis demonstrated that MR DNA binding is required for lipopolysaccharide (LPS), but not phorbol 12-myristate-13-acetate (PMA), induction of the MAPK/pJNK pathway in macrophages. Quantitative RTPCR for pro-inflammatory and pro-fibrotic targets revealed subsets of LPS- and PMA-induced genes that were either enhanced or repressed by the MR via actions that do not always require direct MR-DNA binding. Analysis of the MR target gene and profibrotic factor MMP12 identified promoter elements that are regulated by combined MR/MAPK/JNK signalling. Evaluation of cardiac tissue responses to an 8-day DOC/salt challenge in mice selectively lacking MR DNA-binding in macrophages demonstrated levels of inflammatory markers equivalent to WT, indicating non-DNA binding-dependent MR signalling in macrophages is sufficient for DOC/salt-induced tissue inflammation. Our data demonstrate that the MR regulates a macrophage pro-inflammatory phenotype and cardiac tissue inflammation, partially via pathways that do not require DNA binding.


Assuntos
Fibrose/metabolismo , Inflamação/metabolismo , Miocárdio/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animais , Western Blotting , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Masculino , Metaloproteinase 12 da Matriz/metabolismo , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo
17.
J Mol Endocrinol ; 64(3): 155-164, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31958317

RESUMO

Glucocorticoid (GC) signaling via the glucocorticoid receptor (GR) is essential for lung maturation in mammals. Previous studies using global or conditional mouse model knockouts of the GR gene have established that GR-mediated signaling in the interstitial mesenchyme of the fetal lung is critical for normal lung development. Screens for downstream GC-targets in conditional mesenchymal GR deficient mouse lung (GRmesKO) identified Versican (Vcan), an important extracellular matrix component and cell proliferation regulator, as a potential GR-regulated target. We show that, of the five major VCAN isoforms, the VCAN-V1 isoform containing the GAGß domain is the predominant VCAN isoform in the fetal mouse lung distal mesenchyme at both E16.5 and E18.5, whereas the GAGα-specific VCAN-V2 isoform was only localized to the smooth muscle surrounding proximal airways. Both Vcan-V1 mRNA and protein levels were strongly overexpressed in the GRmesKO lung at E18.5. Finally, we investigated the GC regulation of the ECM protease ADAMTS 12 and showed that Adamts 12 mRNA levels were markedly reduced at E18.5 in GRmesKO fetal mouse lung and were strongly induced by both cortisol and betamethasone in cultures of primary rat fetal lung fibroblasts. ADAMTS12 protein immunoreactivity was also strongly increased in the distal lung at E18.5, after dexamethasone treatment in utero. In summary, glucocorticoid signaling via GR represses GAGß domain-containing VCAN isoforms in distal lung mesenchyme in vivo by repressing Vcan gene expression and, in part, by inducing the ECM protease ADAMTS12, thereby contributing to the control of ECM remodelling and lung cell proliferation prior to birth.


Assuntos
Glucocorticoides/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Versicanas/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Embrião de Mamíferos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Versicanas/metabolismo
18.
Respir Res ; 10: 19, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19284536

RESUMO

BACKGROUND: Bronchopulmonary dysplasia (BPD) is closely associated with ventilator-induced lung injury (VILI) in very preterm infants. The greatest risk of VILI may be in the immediate period after birth, when the lungs are surfactant deficient, still partially filled with liquid and not uniformly aerated. However, there have been very few studies that have examined this immediate post-birth period and identified the initial injury-related pathways that are activated. We aimed to determine if the early response genes; connective tissue growth factor (CTGF), cysteine rich-61 (CYR61) and early growth response 1 (EGR1), were rapidly induced by VILI in preterm lambs and whether ventilation with different tidal volumes caused different inflammatory cytokine and early response gene expression. METHODS: To identify early markers of VILI, preterm lambs (132 d gestational age; GA, term approximately 147 d) were resuscitated with an injurious ventilation strategy (V(T) 20 mL/kg for 15 min) then gently ventilated (5 mL/kg) for 15, 30, 60 or 120 min (n = 4 in each). To determine if early response genes and inflammatory cytokines were differentially regulated by different ventilation strategies, separate groups of preterm lambs (125 d GA; n = 5 in each) were ventilated from birth with a V(T) of 5 (VG5) or 10 mL/kg (VG10) for 135 minutes. Lung gene expression levels were compared to levels prior to ventilation in age-matched control fetuses. RESULTS: CTGF, CYR61 and EGR1 lung mRNA levels were increased approximately 25, 50 and 120-fold respectively (p < 0.05), within 30 minutes of injurious ventilation. VG5 and VG10 caused significant increases in CTGF, CYR61, EGR1, IL1- , IL-6 and IL-8 mRNA levels compared to control levels. CTGF, CYR61, IL-6 and IL-8 expression levels were higher in VG10 than VG5 lambs; although only the IL-6 and CYR61 mRNA levels reached significance. CONCLUSION: CTGF, CYR61 and EGR1 may be novel early markers of lung injury and mechanical ventilation from birth using relatively low tidal volumes may be less injurious than using higher tidal volumes.


Assuntos
Pulmão/metabolismo , Nascimento Prematuro , Respiração Artificial/efeitos adversos , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Animais , Animais Recém-Nascidos , Fator de Crescimento do Tecido Conjuntivo/genética , Proteína Rica em Cisteína 61/genética , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/genética , Marcadores Genéticos , Idade Gestacional , Interleucinas/genética , Pulmão/fisiopatologia , RNA Mensageiro/metabolismo , Respiração Artificial/métodos , Ovinos , Volume de Ventilação Pulmonar , Fatores de Tempo , Regulação para Cima , Lesão Pulmonar Induzida por Ventilação Mecânica/etiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle
19.
Semin Fetal Neonatal Med ; 24(3): 170-175, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31147162

RESUMO

Steroids are complex lipophilic molecules that have many actions in the body to regulate cellular, tissue and organ functions across the life-span. Steroid hormones such as cortisol, aldosterone, estradiol and testosterone are synthesised from cholesterol in specialised endocrine cells in the adrenal gland, ovary and testis, and released into the circulation when required. Steroid hormones move freely into cells to activate intracellular nuclear receptors that function as multi-domain ligand-dependent transcriptional regulators in the cell nucleus. Activated nuclear receptors modify expression of hundreds to thousands of specific target genes in the genome. Steroid hormone actions in the fetus include developmental roles in the respiratory system, brain, and cardiovascular system. The synthetic glucocorticoid steroid betamethasone is used antenatally to reduce the complications of preterm birth. Development of novel selective partial glucocorticoid receptor agonists may provide improved therapies to treat the respiratory complications of preterm birth and spare the deleterious effects of postnatal glucocorticoids in other organs.


Assuntos
Esteroides/fisiologia , Desenvolvimento Fetal/fisiologia , Glucocorticoides/farmacologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/embriologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Esteroides/fisiologia , Transdução de Sinais/fisiologia , Esteroides/biossíntese
20.
Endocrinology ; 160(8): 1868-1884, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31107524

RESUMO

Preterm birth is characterized by severe lung immaturity that is frequently treated antenatally or postnatally with the synthetic steroid betamethasone. The underlying cellular targets and pathways stimulated by betamethasone in the fetal lung are poorly defined. In this study, betamethasone was compared with corticosterone in steroid-treated primary cultures of fetal rat lung fibroblasts stimulated for 6 hours and analyzed by whole-cell transcriptome sequencing and glucocorticoid (GC) receptor (GR) chromatin immunoprecipitation sequencing (ChIP-Seq) analysis. Strikingly, betamethasone stimulated a much stronger transcriptional response compared with corticosterone for both induced and repressed genes. A total of 483 genes were significantly stimulated by betamethasone or corticosterone, with 476 stimulated by both steroids, indicating a strong overlap in regulation. Changes in mRNA levels were confirmed by quantitative PCR for eight induced and repressed target genes. Pathway analysis identified cell proliferation and cytoskeletal/cell matrix remodeling pathways as key processes regulated by both steroids. One target, transglutaminase 2 (Tgm2), was localized to fetal lung mesenchymal cells. Tgm2 mRNA and protein levels were strongly increased in fibroblasts by both steroids. Whole-genome GR ChIP-Seq analysis with betamethasone identified GC response element-binding sites close to the previously characterized GR target genes Per1, Dusp1, Fkbp5, and Sgk1 and near the genes identified by transcriptome sequencing encoding Crispld2, Tgm2, Hif3α, and Kdr, defining direct genomic induction of expression in fetal lung fibroblasts via the GR. These results demonstrate that betamethasone stimulates specific genes and cellular pathways controlling cell proliferation and extracellular matrix remodeling in lung mesenchymal fibroblasts, providing a basis for betamethasone's treatment efficacy in preterm birth.


Assuntos
Betametasona/farmacologia , Fibroblastos/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Mesoderma/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Corticosterona/farmacologia , Feminino , Perfilação da Expressão Gênica , Pulmão/citologia , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/fisiologia , Transdução de Sinais , Transglutaminases/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA