Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Endocrinology ; 161(5)2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32243503

RESUMO

It has recently been shown that the loss of the Hippo signaling effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in adrenocortical steroidogenic cells impairs the postnatal maintenance of the adrenal gland. To further explore the role of Hippo signaling in mouse adrenocortical cells, we conditionally deleted the key Hippo kinases large tumor suppressor homolog kinases 1 and -2 (Lats1 and Lats2, two kinases that antagonize YAP and TAZ transcriptional co-regulatory activity) in steroidogenic cells using an Nr5a1-cre strain (Lats1flox/flox;Lats2flox/flox;Nr5a1-cre). We report here that developing adrenocortical cells adopt characteristics of myofibroblasts in both male and female Lats1flox/flox;Lats2flox/flox;Nr5a1-cre mice, resulting in a loss of steroidogenic gene expression, adrenal failure and death by 2 to 3 weeks of age. A marked accumulation of YAP and TAZ in the nuclei of the myofibroblast-like cell population with an accompanying increase in the expression of their transcriptional target genes in the adrenal glands of Lats1flox/flox;Lats2flox/flox;Nr5a1-cre animals suggested that the myofibroblastic differentiation could be attributed in part to YAP and TAZ. Taken together, our results suggest that Hippo signaling is required to maintain proper adrenocortical cell differentiation and suppresses their differentiation into myofibroblast-like cells.


Assuntos
Córtex Suprarrenal/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Organogênese/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/genética , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/deficiência , Transdução de Sinais/genética , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Proteínas Supressoras de Tumor/deficiência
2.
Adv Anat Embryol Cell Biol ; 230: 1-70, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30543033

RESUMO

The observation of two precursor groups of the early stem cells (Groups I and II) leads to the realization that a first amount of fetal stem cells (Group I) migrate from the AMG (Aortal-Mesonephric-Gonadal)-region into the aorta and its branching vessels. A second group (Group II) gains quite a new significance during human development. This group presents a specific developmental step which is found only in the human. This continuation of the early development along a different way indicates a general alteration of the stem cell biology. This changed process in the stem cell scene dominates the further development of the human stem cells. It remains unclear where this phylogenetic step first appears. By far not all advanced mammals show this second group of stem cells and their axonal migration. Essentially only primates seem to be involved in this special development.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/ultraestrutura , Gônadas/citologia , Gônadas/embriologia , Células APUD/citologia , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiologia , Córtex Suprarrenal/ultraestrutura , Medula Suprarrenal/citologia , Medula Suprarrenal/embriologia , Medula Suprarrenal/fisiologia , Aorta/citologia , Aorta/embriologia , Aorta/ultraestrutura , Sistema Nervoso Autônomo/citologia , Sistema Nervoso Autônomo/embriologia , Sistema Nervoso Autônomo/fisiologia , Orientação de Axônios/fisiologia , Movimento Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Gônadas/fisiologia , Gônadas/ultraestrutura , Desenvolvimento Humano/fisiologia , Humanos , Microscopia Eletrônica , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/fisiologia , Pâncreas/citologia , Pâncreas/crescimento & desenvolvimento , Pâncreas/ultraestrutura , Paragânglios Cromafins/citologia , Paragânglios Cromafins/fisiologia , Paragânglios Cromafins/ultraestrutura , Teratoma/embriologia , Teratoma/fisiopatologia
3.
Gene ; 679: 219-231, 2018 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-30189268

RESUMO

In mammalian cells TPP1, encoded by the Acd gene, is a key component of the shelterin complex, which is required for telomere length maintenance and telomere protection. In mice, a hypomorphic mutation in Acd causes the adrenocortical dysplasia (acd) phenotype, which includes limb and body axis anomalies, and perinatal lethality. p53 deficiency partially rescues limb and body axis anomalies in acd mutant embryos, but not perinatal lethality, implicating p53-independent mechanisms in the acd phenotype. Loss of function of most shelterin proteins results in early embryonic lethality. Thus, study of the hypomorphic acd allele provides a unique opportunity to understand telomere dysfunction at an organismal level. The aim of this study was to identify transcriptome alterations in acd mutant and acd, p53 double mutant embryos to understand the p53-dependent and -independent factors that contribute to the mutant phenotypes in the context of the whole organism. Genes involved in developmental processes, cell cycle, metabolic pathways, tight junctions, axon guidance and signaling pathways were regulated by p53-driven mechanisms in acd mutant embryos, while genes functioning in immune response, and RNA processing were altered independently of p53 in acd, p53 double mutant embryos. To our best of knowledge, this is the first study revealing detailed transcriptomic alterations, reflecting novel p53-dependent and -independent pathways contributing to the acd phenotype. Our data confirm the importance of cell cycle and DNA repair pathways, and suggest novel links between telomere dysfunction and immune system regulation and the splicing machinery. Given the broad applicability of telomere maintenance in growth, development, and genome stability, our data will also provide a rich resource for others studying telomere maintenance and DNA damage responses in mammalian model systems.


Assuntos
Anormalidades Múltiplas/genética , Córtex Suprarrenal/anormalidades , Perfilação da Expressão Gênica/métodos , Proteínas de Ligação a Telômeros/genética , Proteína Supressora de Tumor p53/genética , Anormalidades Múltiplas/embriologia , Córtex Suprarrenal/embriologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Instabilidade Genômica , Camundongos , Mutação , Fenótipo , Complexo Shelterina , Transdução de Sinais
4.
Clinics (Sao Paulo) ; 73(suppl 1): e473s, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30208164

RESUMO

This review summarizes key knowledge regarding the development, growth, and growth disorders of the adrenal cortex from a molecular perspective. The adrenal gland consists of two distinct regions: the cortex and the medulla. During embryological development and transition to the adult adrenal gland, the adrenal cortex acquires three different structural and functional zones. Significant progress has been made in understanding the signaling and molecules involved during adrenal cortex zonation. Equally significant is the knowledge obtained regarding the action of peptide factors involved in the maintenance of zonation of the adrenal cortex, such as peptides derived from proopiomelanocortin processing, adrenocorticotropin and N-terminal proopiomelanocortin. Findings regarding the development, maintenance and growth of the adrenal cortex and the molecular factors involved has improved the scientific understanding of disorders that affect adrenal cortex growth. Hypoplasia, hyperplasia and adrenocortical tumors, including adult and pediatric adrenocortical adenomas and carcinomas, are described together with findings regarding molecular and pathway alterations. Comprehensive genomic analyses of adrenocortical tumors have shown gene expression profiles associated with malignancy as well as methylation alterations and the involvement of miRNAs. These findings provide a new perspective on the diagnosis, therapeutic possibilities and prognosis of adrenocortical disorders.


Assuntos
Doenças do Córtex Suprarrenal/fisiopatologia , Córtex Suprarrenal/crescimento & desenvolvimento , Desenvolvimento Embrionário/fisiologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiologia , Humanos
5.
Clinics ; 73(supl.1): e473s, 2018. tab, graf
Artigo em Inglês | LILACS | ID: biblio-952822

RESUMO

This review summarizes key knowledge regarding the development, growth, and growth disorders of the adrenal cortex from a molecular perspective. The adrenal gland consists of two distinct regions: the cortex and the medulla. During embryological development and transition to the adult adrenal gland, the adrenal cortex acquires three different structural and functional zones. Significant progress has been made in understanding the signaling and molecules involved during adrenal cortex zonation. Equally significant is the knowledge obtained regarding the action of peptide factors involved in the maintenance of zonation of the adrenal cortex, such as peptides derived from proopiomelanocortin processing, adrenocorticotropin and N-terminal proopiomelanocortin. Findings regarding the development, maintenance and growth of the adrenal cortex and the molecular factors involved has improved the scientific understanding of disorders that affect adrenal cortex growth. Hypoplasia, hyperplasia and adrenocortical tumors, including adult and pediatric adrenocortical adenomas and carcinomas, are described together with findings regarding molecular and pathway alterations. Comprehensive genomic analyses of adrenocortical tumors have shown gene expression profiles associated with malignancy as well as methylation alterations and the involvement of miRNAs. These findings provide a new perspective on the diagnosis, therapeutic possibilities and prognosis of adrenocortical disorders.


Assuntos
Humanos , Córtex Suprarrenal/crescimento & desenvolvimento , Doenças do Córtex Suprarrenal/fisiopatologia , Desenvolvimento Embrionário/fisiologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiologia
6.
J Clin Endocrinol Metab ; 102(9): 3349-3359, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28911133

RESUMO

Context: The human fetal adrenal (HFA) is an integral component of the fetoplacental unit and important for the maintenance of pregnancy. Low kisspeptin levels during pregnancy are associated with miscarriage, and kisspeptin and its receptor are expressed in the HFA. However, the role of kisspeptin in fetal adrenal function remains unknown. Objective: To determine the role of kisspeptin in the developing HFA. Design: Experiments using H295R and primary HFA cells as in vitro models of the fetal adrenal. Association of plasma kisspeptin levels with HFA size in a longitudinal clinical study. Setting: Academic research center and tertiary fetal medicine unit. Participants: Thirty-three healthy pregnant women were recruited at their 12-week routine antenatal ultrasound scan. Main Outcome Measures: The spatiotemporal expression of Kiss1R in the HFA. The production of dehydroepiandrosterone sulfate (DHEAS) from HFA cells after kisspeptin treatment, alone or in combination with adrenocorticotropic hormone or corticotropin-releasing hormone. Fetal adrenal volume (FAV) and kisspeptin levels at four antenatal visits (∼20, 28, 34, and 38 weeks' gestation). Results: Expression of Kiss1R was present in the HFA from 8 weeks after conception to term and was shown in the inner fetal zone. Kisspeptin significantly increased DHEAS production in H295R and second-trimester HFA cells. Serial measurements of kisspeptin confirmed a correlation with FAV growth in the second trimester, independent of sex or estimated fetal weight. Conclusions: Kisspeptin plays a key role in the regulation of the HFA and thus the fetoplacental unit, particularly in the second trimester of pregnancy.


Assuntos
Córtex Suprarrenal/embriologia , Glândulas Suprarrenais/embriologia , Desenvolvimento Fetal/fisiologia , Kisspeptinas/sangue , Córtex Suprarrenal/crescimento & desenvolvimento , Glândulas Suprarrenais/crescimento & desenvolvimento , Hormônio Adrenocorticotrópico/metabolismo , Adulto , Análise de Variância , Biomarcadores/sangue , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Humanos , Gravidez , Resultado da Gravidez , Segundo Trimestre da Gravidez , Estudos Prospectivos , Ultrassonografia Pré-Natal , Adulto Jovem
8.
Reprod Sci ; 22(8): 932-41, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25656500

RESUMO

This study was designed to determine the role of the MEK/ERK1/2 and PI3K/Akt pathways in cortisol production and endothelial nitric oxide synthase (eNOS) phosphorylation (peNOS) in the ovine fetal adrenal in response to long-term hypoxia (LTH). Pregnant ewes were maintained at high altitude (3820 m) for the last 100 days of gestation (dGa). At 138 to 142 dGa, fetal adrenal cortical cells (FACs) were collected from LTH and age-matched normoxic fetuses. Cortisol production and peNOS were measured in response to pretreatment with the MEK/ERK1/2 pathway inhibitor UO126 (UO) and adrenocorticotropic hormone (ACTH) stimulation. UO126 reduced ACTH-stimulated cortisol in both normoxic and LTH FACs. UO126 alone or in combination with ACTH reduced peNOS in the normoxic group, while ACTH alone or ACTH + UO inhibited peNOS in LTH FACs. Additionally, cortisol was measured in response to pretreatment with UO and treatment with 22R-hydroxycholesterol (22R-OHC) or water-soluble cholesterol (WSC) with and without ACTH stimulation. UO126 had no effect on 22R-OHC-treated cells, but reduced cortisol in cells treated with WSC and/or ACTH. Cortisol and peNOS were also measured in response to pretreatment with PI3K/Akt pathway inhibitor Wortmannin (WT) and ACTH stimulation. Wortmannin further increased cortisol under ACTH-stimulated conditions and, like ACTH, reduced peNOS in LTH but not normoxic FACs. Together, these data suggest that in LTH FACs MEK/ERK1/2 does not regulate peNOS but that UO acts downstream from eNOS, possibly at cholesterol transport, to affect cortisol production in LTH FACs, while the PI3K/Akt pathway, along with ACTH, regulates peNOS and plays a role in the fetal adaptation to LTH in FACs.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/enzimologia , Hormônio Adrenocorticotrópico/farmacologia , Hipóxia Fetal/enzimologia , Hidrocortisona/biossíntese , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Adaptação Fisiológica , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiopatologia , Altitude , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/embriologia , Hipóxia Fetal/fisiopatologia , Idade Gestacional , Hidroxicolesteróis/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ovinos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
Compr Physiol ; 5(1): 293-326, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25589272

RESUMO

The adrenal gland consists of two distinct parts, the cortex and the medulla. Molecular mechanisms controlling differentiation and growth of the adrenal gland have been studied in detail using mouse models. Knowledge also came from investigations of genetic disorders altering adrenal development and/or function. During embryonic development, the adrenal cortex acquires a structural and functional zonation in which the adrenal cortex is divided into three different steroidogenic zones. Significant progress has been made in understanding adrenal zonation. Recent lineage tracing experiments have accumulated evidence for a centripetal differentiation of adrenocortical cells from the subcapsular area to the inner part of the adrenal cortex. Understanding of the mechanism of adrenocortical cancer (ACC) development was stimulated by knowledge of adrenal gland development. ACC is a rare cancer with a very poor overall prognosis. Abnormal activation of the Wnt/ß-catenin as well as the IGF2 signaling plays an important role in ACC development. Studies examining rare genetic syndromes responsible for familial ACT have played an important role in identifying genetic alterations in these tumors (like TP53 or CTNNB1 mutations as well as IGF2 overexpression). Recently, genomic analyses of ACT have shown gene expression profiles associated with malignancy as well as chromosomal and methylation alterations in ACT and exome sequencing allowed to describe the mutational landscape of these tumors. This progress leads to a new classification of these tumors, opening new perspectives for the diagnosis and prognostication of ACT. This review summarizes current knowledge of adrenocortical development, growth, and tumorigenesis.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Córtex Suprarrenal/crescimento & desenvolvimento , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/fisiologia , Neoplasias do Córtex Suprarrenal/diagnóstico , Animais , Biomarcadores Tumorais/metabolismo , Desenvolvimento Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Síndromes Neoplásicas Hereditárias/diagnóstico , Síndromes Neoplásicas Hereditárias/genética , Regeneração/fisiologia
10.
Endocrinology ; 155(12): 4774-84, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25247468

RESUMO

We have previously shown that estrogen selectively suppresses growth of the fetal zone of the baboon fetal adrenal cortex, which produces the C19-steroid precursors, eg, dehydroepiandrosterone sulfate, which are aromatized to estrogen within the placenta. In the present study, we determined whether fetal adrenal expression of cell cycle regulators are altered by estrogen and thus provide a mechanism by which estrogen regulates fetal adrenocortical development. Cyclin D1 mRNA levels in the whole fetal adrenal were increased 50% (P < .05), and the number of cells in the fetal adrenal definitive zone expressing cyclin D1 protein was increased 2.5-fold (P < .05), whereas the total number of cells in the fetal zone and fetal serum dehydroepiandrosterone sulfate levels were elevated 2-fold (P < .05) near term in baboons in which fetal serum estradiol levels were decreased by 95% (P < .05) after maternal administration of the aromatase inhibitor letrozole and restored to normal by concomitant administration of letrozole plus estradiol throughout second half of gestation. However, fetal adrenocortical expression of cyclin D2, the cyclin-dependent kinase (Cdk)-2, Cdk4, and Cdk6, and Cdk regulatory proteins p27(Kip1) and p57(Kip2) were not changed by letrozole or letrozole plus estradiol administration. We suggest that estrogen controls the growth of the fetal zone of the fetal adrenal by down-regulating cyclin D1 expression and thus proliferation of progenitor cells within the definitive zone that migrate to the fetal zone. We propose that estrogen restrains growth and function of the fetal zone via cyclin D1 to maintain estrogen levels in a physiological range during primate pregnancy.


Assuntos
Córtex Suprarrenal/embriologia , Ciclina D1/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Estrogênios/fisiologia , Córtex Suprarrenal/metabolismo , Animais , Inibidores da Aromatase , Proteínas de Transporte/sangue , Sulfato de Desidroepiandrosterona/sangue , Estradiol/sangue , Feminino , Desenvolvimento Fetal , Peso Fetal , Hidrocortisona/sangue , Antígeno Ki-67/metabolismo , Letrozol , Masculino , Nitrilas , Papio anubis , Placenta/metabolismo , Gravidez , Triazóis
11.
J Clin Endocrinol Metab ; 99(7): E1209-16, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24712566

RESUMO

BACKGROUND: The sonic hedgehog (SHH) pathway plays a key role in rodent adrenal cortex development and is involved in tumorigenesis in several human tissues, but data in human adrenal glands are limited. OBJECTIVES: The objectives of the study were to analyze the involvement of the SHH pathway in human adrenal development and tumorigenesis and the effects of SHH inhibition on an adrenocortical tumor (ACT) cell line. PATIENTS AND METHODS: Expression of SHH pathway components was evaluated by immunohistochemistry in 51 normal adrenals (33 fetal) and 34 ACTs (23 pediatric) and by quantitative PCR in 81 ACTs (61 pediatric) and 19 controls (10 pediatric). The effects of SHH pathway inhibition on gene expression and cell viability in the NCI-H295A adrenocortical tumor cell line after cyclopamine treatment were analyzed. RESULTS: SHH pathway proteins were present in fetal and postnatal normal adrenals and showed distinct patterns of spatiotemporal expression throughout development. Adult adrenocortical carcinomas presented with higher expression of PTCH1, SMO, GLI3, and SUFU compared with normal adult adrenal cortices. Conversely, pediatric ACTs showed lower mRNA expression of SHH, PTCH1, SMO, GLI1, and GLI3 compared with normal pediatric adrenal cortices. In vitro treatment with cyclopamine resulted in decreased GLI3, SFRP1, and CTNNB1 mRNA expression and ß-catenin staining as well as decreased cell viability. CONCLUSIONS: The SHH pathway is active in human fetal and postnatal adrenals, up-regulated in adult adrenocortical carcinomas, and down-regulated in pediatric ACTs. SHH pathway antagonism impaired cell viability. The SHH pathway is deregulated in ACTs and might provide a new target therapy to be explored.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/crescimento & desenvolvimento , Carcinoma Adrenocortical/genética , Proteínas Hedgehog/genética , Córtex Suprarrenal/metabolismo , Neoplasias do Córtex Suprarrenal/metabolismo , Carcinoma Adrenocortical/metabolismo , Adulto , Estudos de Casos e Controles , Células Cultivadas , Criança , Desenvolvimento Embrionário/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Recém-Nascido , Gravidez , Estudos Retrospectivos , Transdução de Sinais/genética
12.
Curr Top Dev Biol ; 106: 239-312, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24290352

RESUMO

The adrenal gland controls a plethora of crucial physiological functions, and dysfunction is associated with severe morbidity. Because of the vital importance of appropriate adrenal function, the development and function of the gland have been intensively studied, and these investigations have revealed fascinating developmental origins and a remarkable remodeling and regenerative capacity in the adult. This chapter, focusing on the adrenal cortex, will describe our current understanding of the development and maintenance of the adrenal gland, which has been advanced over recent years by the use of sophisticated genetic models in the study of both normal function and disease. This work has shed light on the transcriptional networks and signaling pathways involved in development and maintenance of the gland and in its pathology; these are discussed in the light of the wealth of physiological information gathered in studies of human and rodent adrenal development and function.


Assuntos
Córtex Suprarrenal/embriologia , Doenças das Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/embriologia , Modelos Biológicos , Córtex Suprarrenal/crescimento & desenvolvimento , Córtex Suprarrenal/metabolismo , Corticosteroides/metabolismo , Doenças das Glândulas Suprarrenais/genética , Glândulas Suprarrenais/crescimento & desenvolvimento , Glândulas Suprarrenais/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Transdução de Sinais/genética
13.
Development ; 140(22): 4522-32, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24131628

RESUMO

The lineage relationships of fetal adrenal cells and adrenal capsular cells to the differentiated adrenal cortex are not fully understood. Existing data support a role for each cell type as a progenitor for cells of the adult cortex. This report reveals that subsets of capsular cells are descendants of fetal adrenocortical cells that once expressed Nr5a1. These fetal adrenocortical cell descendants within the adrenal capsule express Gli1, a known marker of progenitors of steroidogenic adrenal cells. The capsule is also populated by cells that express Tcf21, a known inhibitor of Nr5a1 gene expression. We demonstrate that Tcf21-expressing cells give rise to Nr5a1-expressing cells but only before capsular formation. After the capsule has formed, capsular Tcf21-expressing cells give rise only to non-steroidogenic stromal adrenocortical cells, which also express collagen 1a1, desmin and platelet-derived growth factor (alpha polypeptide) but not Nr5a1. These observations integrate prior observations that define two separate origins of adult adrenocortical steroidogenic cells (fetal adrenal cortex and/or the adrenal capsule). Thus, these observations predict a unique temporal and/or spatial role of adult cortical cells that arise directly from either fetal cortical cells or from fetal cortex-derived capsular cells. Last, the data uncover the mechanism by which two populations of fetal cells (fetal cortex derived Gli1-expressing cells and mesenchymal Tcf21-expressing mesenchymal cells) participate in the establishment of the homeostatic capsular progenitor cell niche of the adult cortex.


Assuntos
Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Linhagem da Célula , Feto/citologia , Células-Tronco/citologia , Esteroides/metabolismo , Envelhecimento/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Homeostase , Camundongos , Modelos Biológicos , Fator Esteroidogênico 1/metabolismo , Células Estromais , Fatores de Transcrição/metabolismo , Proteína GLI1 em Dedos de Zinco
14.
Mol Cell Endocrinol ; 371(1-2): 182-8, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23376610

RESUMO

Developmental signalling pathways are implicated in the formation and maintenance of the adrenal gland, but their roles are currently not well defined. In recent years it has emerged that Sonic hedgehog (Shh) and Wnt/ß catenin signalling are crucial for the growth and development of the adrenal cortex. Here we demonstrate that Fibroblast growth factor receptor (Fgfr) 2 isoforms IIIb and IIIc are expressed mainly in the adrenal subcapsule during embryogenesis and that specific deletion of the Fgfr2 IIIb isoform impairs adrenal development, causing reduced adrenal growth and impaired expression of SF1 and steroidogenic enzymes. The hypoplastic adrenals also have thicker, disorganised capsules which retain Gli1 expression but no longer express Dlk1. Fgfr2 ligands were detected in both the capsule and the cortex, suggesting the importance of signalling between the capsule and the cortex in adrenal development.


Assuntos
Córtex Suprarrenal/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Isoformas de Proteínas/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Proteínas de Ligação ao Cálcio , Feminino , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Fatores de Transcrição Kruppel-Like/biossíntese , Masculino , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais , Fator Esteroidogênico 1/biossíntese , Fator Esteroidogênico 1/genética , Proteína GLI1 em Dedos de Zinco
15.
Am J Physiol Regul Integr Comp Physiol ; 304(8): R636-43, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23427082

RESUMO

This study assessed the role of the extracellular signal-regulated kinase (ERK) signaling pathway on the previously observed enhanced cortisol secretion in response to adrenocorticotropic hormone (ACTH) treatment in fetal adrenocortical cells (FACs) from long-term hypoxic (LTH) ovine fetuses. Ewes were maintained at high altitude (3,820 m) from ~40 to 138-141 days gestation when FACs were collected and challenged with either ACTH (10 nM) or 8-bromoadenosine 3',5'-cyclic monophosphate (8-bromo-cAMP, 10 mM) in the presence or absence of the mitogen-activated protein kinase/extracellular signal-regulated protein kinase (MEK)/ERK inhibitor UO126 (10 µM). FACs from age-matched normoxic fetuses served as controls. Media and FACs were collected at selected time intervals after ACTH or 8-bromo-cAMP stimulation for cortisol measurement and Western analysis of ERK1/2 and phospho-ERK1 and -2 (pERK1/2). After ACTH or 8-bromo-cAMP treatment, cortisol production was greater in the LTH group compared with control (P < 0.05). UO126 reduced ACTH and 8-bromo-cAMP-mediated cortisol output in both groups (P < 0.01 vs. ACTH or 8-bromo-cAMP alone). Under basal conditions, ERK1/2 and pERK1/2 were not different between LTH and normoxic fetuses. In response to ACTH or 8-bromo-cAMP treatment, ERK1/2 were not different between groups; however, pERK1/2 were elevated in the LTH FACs compared with normoxic control FACs. ERK1/2 phosphorylation declined following ACTH treatment in the control group, but UO126 had no effect on ERK1/2 compared with untreated levels. Both ACTH and 8-bromo-cAMP treatment resulted in a decline of protein levels. UO126 pretreatment virtually eliminated pERK1/2 expression. We conclude that basal ERK signaling in FACs is necessary for normal cortisol production and sustained pERK in LTH adrenals enhances cortisol production.


Assuntos
Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/metabolismo , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Hidrocortisona/biossíntese , Sistema de Sinalização das MAP Quinases/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Hormônio Adrenocorticotrópico/farmacologia , Animais , Western Blotting , Butadienos/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Gravidez , Ovinos
16.
Exp Toxicol Pathol ; 64(1-2): 25-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20580540

RESUMO

UNLABELLED: The (As) arsenic exposure is a risk factor for causing disturbances in the endocrine organs. OBJECTIVE: To evaluate if sub-chronic As exposure during the pre- and postnatal development causes disturbances in the puberty. Moreover, determine adverse effects of As on the ovarian follicle and adrenocortical cell maturation. METHODS: Females adult Wistar rats were exposed to sodium arsenite at 3 ppm calculated as As in drinking water from mating, gestation. Following the birth, the female offspring continued exposured to As via lactation. Weaned pups received the same As treatment as mothers, until they were 1-4 months (mo) old. At these ages, blood sampling and tissue harvest were done. The tissues were fixed in situ with 4% paraformaldehyde in phosphate buffer. After the perfusion the ovaries, uterus, adrenal glands were harvested, dissected out, weighted. The ovaries and the adrenal glands were processed to paraffin and sectioned at 5 µM and stained with hematoxylin and eosin for light microscopy. STATISTICAL ANALYSIS: Comparisons between groups were made by unpaired t-test or nonparametric Mann-Whitney test as appropriate. RESULTS: 100% As treated rats at 1 mo of age were at diestrous stage, with low estradiol E2. As treatment caused disturbances in the morphology of the ovarian cell consisting in DNA damage evidenced by picknotic chromatin, cariorexis, significant cytoplasmic vacuolization and also vasculature damaged. Arrest in follicle maturation was also present. CONCLUSIONS: We found that the onset of puberty in the As treated rats was 1 mo delayed since vagina was still closed, the vaginal smear showed that they were at diestrus stage with plasma low E2 levels.


Assuntos
Envelhecimento/efeitos dos fármacos , Arsenitos/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Maturidade Sexual/efeitos dos fármacos , Compostos de Sódio/toxicidade , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/crescimento & desenvolvimento , Envelhecimento/sangue , Animais , Arsenitos/farmacocinética , Estradiol/sangue , Ciclo Estral/sangue , Ciclo Estral/efeitos dos fármacos , Feminino , Lactação , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/sangue , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Progesterona/sangue , Ratos , Ratos Wistar , Compostos de Sódio/farmacocinética , Vagina/efeitos dos fármacos , Vagina/embriologia , Vagina/crescimento & desenvolvimento
17.
Fetal Pediatr Pathol ; 31(1): 30-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22026750

RESUMO

Complications act as stress-inducers during pregnancy so the fetus can develop functional compensatory mechanisms or morphologic changes. The cases analyzed are with congenital malformations or acute stress; chronic included cases with ascending infection (AI) and perinatal hypoxia/anoxia (PHA). The hematoxylin-eosin (H&E) was done to analyze the vacuolization, and the immunohistochemistry to the phagocytosis. The discreet standard of vacuolization was observed in 52.6% of the cases, 22.1% moderate, and 25.3% severe. The number of macrophages was higher in PHA. Changes in these organs are closely related to the cause of death and to the period during which the harmful agent.


Assuntos
Infecções Bacterianas/patologia , Morte Fetal , Doenças Fetais/patologia , Mortalidade Infantil , Complicações Infecciosas na Gravidez/patologia , Estresse Fisiológico , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/metabolismo , Córtex Suprarrenal/patologia , Adulto , Infecções Bacterianas/metabolismo , Infecções Bacterianas/mortalidade , Contagem de Células , Anormalidades Congênitas , Feminino , Doenças Fetais/mortalidade , Hipóxia Fetal/metabolismo , Hipóxia Fetal/mortalidade , Hipóxia Fetal/patologia , Idade Gestacional , Humanos , Recém-Nascido , Infecções , Macrófagos/patologia , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/mortalidade , Nascimento Prematuro , Timo/patologia , Vacúolos/patologia
18.
Toxicol Appl Pharmacol ; 257(3): 328-37, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21971485

RESUMO

Steroidogenic acute regulatory protein (StAR) mediates the rate-limiting step in the synthesis of steroid hormones, essential to fetal development. We have reported that the StAR expression in fetal adrenal is inhibited in a rat model of nicotine-induced intrauterine growth retardation (IUGR). Here using primary human fetal adrenal cortex (pHFAC) cells and a human fetal adrenal cell line NCI-H295A, we show that nicotine inhibits StAR expression and cortisol production in a dose- and time-dependent manner, and prolongs the inhibitory effect on cells proliferating over 5 passages after termination of nicotine treatment. Methylation detection within the StAR promoter region uncovers a single site CpG methylation at nt -377 that is sensitive to nicotine treatment. Nicotine-induced alterations in frequency of this point methylation correlates well with the levels of StAR expression, suggesting an important role of the single site in regulating StAR expression. Further studies using bioinformatics analysis and siRNA approach reveal that the single CpG site is part of the Pax6 binding motif (CGCCTGA) in the StAR promoter. The luciferase activity assays validate that Pax6 increases StAR gene expression by binding to the glucagon G3-like motif (CGCCTGA) and methylation of this site blocks Pax6 binding and thus suppresses StAR expression. These data identify a nicotine-sensitive CpG site at the Pax6 binding motif in the StAR promoter that may play a central role in regulating StAR expression. The results suggest an epigenetic mechanism that may explain how nicotine contributes to onset of adult diseases or disorders such as metabolic syndrome via fetal programming.


Assuntos
Ilhas de CpG/genética , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Nicotina/toxicidade , Agonistas Nicotínicos/toxicidade , Fatores de Transcrição Box Pareados/genética , Fosfoproteínas/genética , Proteínas Repressoras/genética , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/metabolismo , Motivos de Aminoácidos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucagon/genética , Humanos , Hidrocortisona/metabolismo , Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Fator de Transcrição PAX6 , Regiões Promotoras Genéticas , Fatores de Tempo
19.
J Pediatr Hematol Oncol ; 33(4): e149-53, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21516013

RESUMO

The germline R337H mutation in the TP53 gene is considered to be responsible for the increased incidence of adrenocortical tumors (ACTs) in children from Brazil. High level production of hormones in ACTs (>95%) cause virilization alone (60%), Cushing syndrome (<5%), the mixed type (30%), or other rarer manifestations. ACT probably develops owing to events occurring during the final stages of intrauterine life based on the very common early onset of signs and symptoms shortly after birth. In this study, we determined by immunohistochemistry and enzyme assays whether placental alkaline phosphatase (PLAP) is expressed in pediatric ACTs. Immunohistochemical analysis revealed positive p53 expression in 88% of the tested ACTs (29 of 33). PLAP was detected at a slightly lower frequency based on immunohistochemical (17 of 33, 51%) and enzyme activity analyses (9 of 16, 56%). In conclusion, probably at a certain time point during adrenocortical development (end of gestation to early postnatal period), some fetal zone cells survive owing to defective apoptosis and develop into childhood ACT, maintaining some characteristics of the embryonal period, such as PLAP expression. Further studies of PLAP should investigate the functional role, if any, of PLAP in such tumors.


Assuntos
Neoplasias do Córtex Suprarrenal/metabolismo , Neoplasias do Córtex Suprarrenal/patologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/metabolismo , Fosfatase Alcalina/metabolismo , Isoenzimas/metabolismo , Neoplasias do Córtex Suprarrenal/genética , Apoptose/fisiologia , Criança , Feminino , Proteínas Ligadas por GPI/metabolismo , Mutação em Linhagem Germinativa , Humanos , Imuno-Histoquímica , Masculino , Proteína Supressora de Tumor p53/genética
20.
Mol Cell Endocrinol ; 336(1-2): 117-22, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21094676

RESUMO

Sonic hedgehog signaling was recently demonstrated to play an important role in murine adrenal cortex development. The organization of the rat adrenal differs from that of the mouse, with the zona glomerulosa and zona fasciculata separated by an undifferentiated zone in the rat, but not in the mouse. In the present study we aimed to determine the mRNA expression patterns of Sonic hedgehog and the hedgehog signaling pathway components Patched-1 and Gli1 in the developing and adult rat adrenal. Sonic hedgehog expression was detected at the periphery of the cortex in cells lacking CYP11B1 and CYP11B2 expression, while signal-receiving cells were localized in the overlying capsule mesenchyme. Using combined in situ hybridization and immunohistochemistry we found that the cells expressing Sonic hedgehog lie between the CYP11B2 and CYP11B1 layers, and thus Sonic hedgehog expression defines one cell population of the undifferentiated zone.


Assuntos
Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Envelhecimento/metabolismo , Proteínas Hedgehog/metabolismo , Córtex Suprarrenal/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Receptores Patched , Receptor Patched-1 , Transporte Proteico , Ratos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteína GLI1 em Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA