Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vitam Horm ; 124: 297-339, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38408801

RESUMO

Mouse models have been widely used in the study of adrenal gland development and diseases. The X-zone is a unique structure of the mouse adrenal gland and lineage-tracing studies show that the X-zone is a remnant of the fetal adrenal cortex. Although the X-zone is considered analogous to the fetal zone in the human adrenal cortex, the functional significance of the X-zone has remained comparatively more obscure. The X-zone forms during the early postnatal stages of adrenal development and regresses later in a remarkable sexually dimorphic fashion. The formation and regression of the X-zone can be different in mice with different genetic backgrounds. Mouse models with gene mutations, hormone/chemical treatments, and/or gonadectomy can also display an aberrant development of the X-zone or alternatively a dysregulated X-zone regression. These models have shed light on the molecular mechanisms regulating the development and regression of these unique adrenocortical cells. This review paper briefly describes the development of the adrenal gland including the formation and regression processes of the X-zone. It also summarizes and lists mouse models that demonstrate different X-zone phenotypes.


Assuntos
Neoplasias do Córtex Suprarrenal , Córtex Suprarrenal , Camundongos , Humanos , Animais , Glândulas Suprarrenais
2.
Commun Biol ; 6(1): 1253, 2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-38081939

RESUMO

Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor ß (TRß) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRß binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRß-deficiency blunts T3-regulated gene expression, indicating that TRß confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.


Assuntos
Cromatina , Hormônios Tireóideos , Camundongos , Animais , Cromatina/genética , Cromatina/metabolismo , Hormônios Tireóideos/metabolismo , Tri-Iodotironina/farmacologia , Tri-Iodotironina/metabolismo , Hipófise/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/metabolismo
3.
Int J Mol Sci ; 24(2)2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36674444

RESUMO

Steroid hormones are synthesized through enzymatic reactions using cholesterol as the substrate. In steroidogenic cells, the required cholesterol for steroidogenesis can be obtained from blood circulation or synthesized de novo from acetate. One of the key enzymes that control cholesterol synthesis is 24-dehydrocholesterol reductase (encoded by DHCR24). In humans and rats, DHCR24 is highly expressed in the adrenal gland, especially in the zona fasciculata. We recently reported that DHCR24 was expressed in the mouse adrenal gland's inner cortex and also found that thyroid hormone treatment significantly upregulated the expression of Dhcr24 in the mouse adrenal gland. In the present study, we showed the cellular expression of DHCR24 in mouse adrenal glands in early postnatal stages. We found that the expression pattern of DHCR24 was similar to the X-zone marker gene 20αHSD in most developmental stages. This finding indicates that most steroidogenic adrenocortical cells in the mouse adrenal gland do not synthesize cholesterol locally. Unlike the 20αHSD-positive X-zone regresses during pregnancy, some DHCR24-positive cells remain present in parous females. Conditional knockout mice showed that the removal of Dhcr24 in steroidogenic cells did not affect the overall development of the adrenal gland or the secretion of corticosterone under acute stress. Whether DHCR24 plays a role in conditions where a continuous high amount of corticosterone production is needed requires further investigation.


Assuntos
Corticosterona , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Humanos , Camundongos , Feminino , Ratos , Animais , Corticosterona/metabolismo , Glândulas Suprarrenais/metabolismo , Zona Fasciculada/metabolismo , Colesterol/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética
4.
Endocr Connect ; 11(8)2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35904237

RESUMO

Glucocorticoids have short- and long-term effects on adrenal gland function and development. RNA sequencing (RNA-seq) was performed to identify early transcriptomic responses to the synthetic glucocorticoid, dexamethasone (Dex), in vitro and in vivo. In total, 1711 genes were differentially expressed in the adrenal glands of the 1-h Dex-treated mice. Among them, only 113 were also considered differentially expressed genes (DEGs) in murine adrenocortical Y-1 cells treated with Dex for 1 h. Gene ontology analysis showed that the upregulated DEGs in the adrenal gland of the 1-h Dex-treated mice were highly associated with the development of neuronal cells, suggesting the adrenal medulla had a rapid response to Dex. Interestingly, only 4.3% of Dex-responsive genes in the Y-1 cell line under Dex treatment for 1 h were differentially expressed under Dex treatment for 24 h. The heatmaps revealed that most early responsive DEGs in Y-1 cells during 1 h of treatment exhibited a transient response. The expression of these genes under treatment for 24 h returned to basal levels similar to that during control treatment. In summary, this research compared the rapid transcriptomic effects of Dex stimulation in vivo and in vitro. Notably, adrenocortical Y-1 cells had a transient early response to Dex treatment. Furthermore, the DEGs had a minimal overlap in the 1-h Dex-treated group in vivo and in vitro.

5.
Thyroid ; 32(4): 459-471, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35044245

RESUMO

Background: The human adrenal cortex undergoes several rapid remodeling steps during its lifetime. In rodents, similar remodeling occurs postnatally in the "X-zone" layer through unknown mechanisms. Furthermore, little is known regarding the impact of thyroid hormone (TH) on adrenal glands in humans. Methods: To investigate the impact of TH on adrenal pathophysiology, we created two genetic murine models mimicking human nonautoimmune hypothyroidism and hyperthyroidism. Moreover, we analyzed serum thyrotropin (TSH) and steroid hormone concentrations in patients diagnosed with congenital hypothyroidism and premature adrenarche (PA). Results: We found that TH receptor beta-mediated hypertrophy of the X-zone significantly elevated the adrenal weights of hyperthyroid women. In the hypothyroid model, the X-zone was poorly developed in both sexes. Moreover, large reciprocal changes in the expression levels of genes that regulate adrenal cortical function were observed with both models. Unexpectedly, up- and downregulation of several genes involved in catecholamine synthesis were detected in the adrenal glands of the hypothyroid and hyperthyroid models, respectively. Furthermore, TSH and adrenal steroid concentrations correlated positively in pediatric patients with congenital hypothyroidism and PA. Conclusions: Our results revealed that congenital hypothyroidism and hyperthyroidism functionally affect adrenal gland development and related steroidogenic activity, as well as the adrenal medulla.


Assuntos
Hipotireoidismo Congênito , Hipertireoidismo , Animais , Criança , Hipotireoidismo Congênito/genética , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Hormônios Tireóideos , Tireotropina
6.
J Vis Exp ; (178)2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34958080

RESUMO

Cellular heterogeneity poses challenges to understanding the function of complex tissues at a transcriptome level. Using cell-type-specific RNAs avoids potential pitfalls caused by the heterogeneity of tissues and unleashes the powerful transcriptome analysis. The protocol described here demonstrates how to use the Translating Ribosome Affinity Purification (TRAP) method to isolate ribosome-bound RNAs from a small amount of EGFP-expressing cells in a complex tissue without cell sorting. This protocol is suitable for isolating cell-type-specific RNAs using the recently available NuTRAP mouse model and could also be used to isolate RNAs from any EGFP-expressing cells.


Assuntos
Perfilação da Expressão Gênica , Ribossomos , Animais , Perfilação da Expressão Gênica/métodos , Camundongos , RNA Ribossômico , Transcriptoma
7.
Endocrinology ; 162(12)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599818

RESUMO

Glucose is a major energy source for growth. At birth, neonates must change their energy source from maternal supply to its own glucose production. The mechanism of this transition has not been clearly elucidated. To evaluate the possible roles of steroids in this transition, here we examine the defects associated with energy production of a mouse line that cannot synthesize steroids de novo due to the disruption of its Cyp11a1 (cytochrome P450 family 11 subfamily A member 1) gene. The Cyp11a1 null embryos had insufficient blood insulin and failed to store glycogen in the liver since embryonic day 16.5. Their blood glucose dropped soon after maternal deprivation, and the expression of hepatic gluconeogenic and glycogenic genes were reduced. Insulin was synthesized in the mutant fetal pancreas but failed to be secreted. Maternal glucocorticoid supply rescued the amounts of blood glucose, insulin, and liver glycogen in the fetus but did not restore expression of genes for glycogen synthesis, indicating the requirement of de novo glucocorticoid synthesis for glycogen storage. Thus, our investigation of Cyp11a1 null embryos reveals that the energy homeostasis is established before birth, and fetal steroids are required for the regulation of glycogen synthesis, hepatic gluconeogenesis, and insulin secretion at the fetal stage.


Assuntos
Enzima de Clivagem da Cadeia Lateral do Colesterol/fisiologia , Desenvolvimento Embrionário/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Esteroides/biossíntese , Esteroides/farmacologia , Animais , Animais Recém-Nascidos , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Dexametasona/farmacologia , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Metabolismo Energético/genética , Feminino , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Glicogênio/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo
8.
Endocrinology ; 161(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32697836

RESUMO

The sex-specific prevalence of adrenal diseases has been known for a long time. However, the reason for the high prevalence of these diseases in females is not completely understood. Mouse studies have shown that the adult adrenal gland is sexually dimorphic at different levels such as transcriptome, histology, and cell renewal. Here we used RNA-seq to show that in prepubertal mice, male and female adrenal glands were not only sexually dimorphic but also responded differently to the same external stimulus. We previously reported that thyroid hormone receptor ß1 (TRß1) in the adrenal gland is mainly expressed in the inner cortex and the fate of this TRß1-expressing cell population can be changed by thyroid hormone (triiodothyronine; T3) treatment. In the present study, we found that adrenal glands in prepubertal mice were sexually dimorphic at the level of the transcriptome. Under T3 treatment, prepubertal females had 1162 genes differentially expressed between the saline and T3 groups, whereas in males of the same age, only 512 genes were T3-responsive. Immunostaining demonstrated that several top sexually dimorphic T3-responsive genes, including Cyp2f2 and Dhcr24, were specifically expressed in the adrenal inner cortex, precisely in an area partially overlapping with the X-zone. Under T3 treatment, a unique cortical layer that surrounds the adrenal X-zone expanded significantly, forming a distinct layer peculiar to females. Our findings identified novel marker genes for the inner adrenal cortex, indicating there are different sub-zones in the zona fasciculata. The results also highlight the sex-specific response to thyroid hormone in the mouse adrenal gland.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Tireóideos/farmacologia , Zona Fasciculada/efeitos dos fármacos , Zona Fasciculada/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Feminino , Perfilação da Expressão Gênica , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA-Seq , Caracteres Sexuais , Distribuição Tecidual/efeitos dos fármacos
9.
J Vis Exp ; (156)2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32150172

RESUMO

Immunostaining is widely used in biomedical research to show the cellular expression pattern of a given protein. Multiplex immunostaining allows labeling using multiple primary antibodies. To minimize antibody cross-reactivity, multiplex immunostaining using indirect staining requires unlabeled primary antibodies from different host species. However, the appropriate combination of different species antibodies is not always available. Here, we describe a method of using unlabeled primary antibodies from the same host species (e.g., in this case both antibodies are from rabbit) for multiplex immunofluorescence on formalin-fixed paraffin-embedded (FFPE) mouse adrenal sections. This method uses the same procedure and reagents used in the antigen retrieval step to strip the activity of the previously stained primary antibody complex. Slides were stained with the first primary antibody using a general immunostaining protocol followed by a binding step with a biotinylated secondary antibody. Then, an avidin-biotin-peroxidase signal development method was used with fluorophore-tyramide as the substrate. The immunoactivity of the first primary antibody complex was stripped through immersion in a microwaved boiling sodium citrate solution for 8 min. The insoluble fluorophore-tyramide deposition remained on the sample, which allowed the slide to be stained with other primary antibodies. Although this method eliminates most false positive signals, some background from antibody cross-reactivity may remain. If the samples are enriched with endogenous biotin, a peroxidase-conjugated secondary antibody may be used to replace the biotinylated secondary antibody to avoid the false positive from recovered endogenous biotin.


Assuntos
Glândulas Suprarrenais/metabolismo , Anticorpos Monoclonais/imunologia , Corantes Fluorescentes/química , Técnicas Imunoenzimáticas/métodos , Micro-Ondas , Tiramina/análogos & derivados , 3-Hidroxiesteroide Desidrogenases/imunologia , Glândulas Suprarrenais/imunologia , Animais , Biotinilação , Sistema Enzimático do Citocromo P-450/imunologia , Imunofluorescência , Humanos , Camundongos , Peroxidase/metabolismo , Coelhos , Coloração e Rotulagem , Tiramina/metabolismo
10.
J Endocrinol ; 241(1): R51-R63, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30817316

RESUMO

The X-zone is a transient cortical region enriched in eosinophilic cells located in the cortical-medullary boundary of the mouse adrenal gland. Similar to the X-zone, the fetal zone in human adrenals is also a transient cortical compartment, comprising the majority of the human fetal adrenal gland. During adrenal development, fetal cortical cells are gradually replaced by newly formed adult cortical cells that develop into outer definitive zones. In mice, the regression of this fetal cell population is sexually dimorphic. Many mouse models with mutations associated with endocrine factors have been reported with X-zone phenotypes. Increasing findings indicate that the cell fate of this aged cell population of the adrenal cortex can be manipulated by many hormonal and nonhormonal factors. This review summarizes the current knowledge of this transient adrenocortical zone with an emphasis on genes and signaling pathways that affect X-zone cells.


Assuntos
Córtex Suprarrenal/metabolismo , Glândulas Suprarrenais/metabolismo , Medula Suprarrenal/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/embriologia , Medula Suprarrenal/citologia , Medula Suprarrenal/embriologia , Animais , Apoptose/genética , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos
11.
Endocrinology ; 156(6): 2338-48, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25774556

RESUMO

The development of the adrenal cortex involves the formation and then subsequent regression of immature or fetal inner cell layers as the mature steroidogenic outer layers expand. However, controls over this remodeling, especially in the immature inner layer, are incompletely understood. Here we identify an inner cortical cell population that expresses thyroid hormone receptor-ß1 (TRß1), one of two receptor isoforms encoded by the Thrb gene. Using mice with a Thrb(b1) reporter allele that expresses lacZ instead of TRß1, ß-galactosidase was detected in the inner cortex from early stages. Expression peaked at juvenile ages in an inner zone that included cells expressing 20-α-hydroxysteroid dehydrogenase, a marker of the transient, so-called X-zone in mice. The ß-galactosidase-positive zone displayed sexually dimorphic regression in males after approximately 4 weeks of age but persisted in females into adulthood in either nulliparous or parous states. T3 treatment promoted hypertrophy of inner cortical cells, induced some markers of mature cortical cells, and, in males, delayed the regression of the TRß1-positive zone, suggesting that TRß1 could partly divert the differentiation fate and counteract male-specific regression of inner zone cells. TRß1-deficient mice were resistant to these actions of T3, supporting a functional role for TRß1 in the inner cortex.


Assuntos
Córtex Suprarrenal/citologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Mutantes , Receptores beta dos Hormônios Tireóideos/genética , Tri-Iodotironina/farmacologia
12.
Endocrinology ; 153(10): 4749-56, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22962254

RESUMO

During pregnancy, fetal glucocorticoid is derived from both maternal supply and fetal secretion. We have created mice with a disruption of the Cyp11a1 gene resulting in loss of fetal steroid secretion but preserving the maternal supply. Cyp11a1null embryos have appreciable although lower amounts of circulating corticosterone, the major mouse glucocorticoid, suggesting that transplacental corticosterone is a major source of corticosterone in fetal circulation. These embryos thus provide a means to examine the effect of fetal glucocorticoids. The adrenal in Cyp11a1 null embryos was disorganized with abnormal mitochondria and oil accumulation. The adrenal medullary cells did not express phenylethanolamine N-methyltransferase and synthesized no epinephrine. Cyp11a1 null embryos had decreased diencephalon Hsd11b1, increased diencephalon Crh, and increased pituitary Pomc expression, leading to higher adrenocorticotropin level in the plasma. These data indicate blunted feedback suppression despite reasonable amounts of circulating corticosterone. Thus, the corticosterone synthesized in situ by the fetus is required for negative feedback suppression of the hypothalamus-pituitary-adrenal axis and for catecholamine synthesis in adrenal medulla.


Assuntos
Medula Suprarrenal/metabolismo , Hormônio Adrenocorticotrópico/biossíntese , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Corticosterona/biossíntese , Retroalimentação Fisiológica/fisiologia , Hipotálamo/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Medula Suprarrenal/crescimento & desenvolvimento , Hormônio Adrenocorticotrópico/sangue , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Epinefrina/biossíntese , Feminino , Sistema Hipotálamo-Hipofisário/metabolismo , Masculino , Troca Materno-Fetal , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Feniletanolamina N-Metiltransferase/genética , Feniletanolamina N-Metiltransferase/metabolismo , Hipófise/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Gravidez , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
13.
Mol Cell Endocrinol ; 361(1-2): 165-71, 2012 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-22580128

RESUMO

Functions of adrenal medulla, particularly synthesis of catecholamine, are under the control of glucocorticoids produced by the cortex. To further investigate whether development/differentiation of the adrenal medulla is associated with proper organization of the adrenal cortex, we examined development of the medulla in four different mouse models with various defects in the adrenal cortex. By using the Sf1/Cre mouse line that inactivates/activates genes in Steroidogenic factor 1 (SF1)-positive cells of the fetal adrenal cortex, we produced mice that exhibit either (1) cortex hypoplasia, (2) progressive degeneration of fetal adrenal cortex, (3) cortex dysgenesis, or (4) cortex-medulla disorganization. The formation of phenylethanolamine N-methyltransferase (PNMT)-positive medulla in all models indicates that differentiation of adrenal medulla is independent of the growth of adrenal cortex. However, the misplaced/dysgenic medulla in embryos where ß-catenin expression is altered, suggests that the ß-catenin pathway in the adrenal cortical cells plays an indirect role in controlling proper organization of the adrenal medulla.


Assuntos
Córtex Suprarrenal/patologia , Medula Suprarrenal/patologia , Diferenciação Celular , Córtex Suprarrenal/metabolismo , Medula Suprarrenal/metabolismo , Animais , RNA Helicases DEAD-box/metabolismo , Proteínas Hedgehog/metabolismo , Integrases/metabolismo , Camundongos , Camundongos Knockout , Modelos Biológicos , Fenótipo , Ribonuclease III/metabolismo , Fator Esteroidogênico 1/metabolismo , beta Catenina/deficiência , beta Catenina/metabolismo
14.
Mol Reprod Dev ; 77(6): 489-96, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20422709

RESUMO

Adrenal, testis, and ovary are steroidogenic organs derived from a common primordium that consists of steroidogenic factor 1 (SF1)-positive precursor cells. SF1 not only defines the steroidogenic lineages in these organs but also controls their differentiation. Recent evidence implicates the Hedgehog (Hh) signaling pathway as a downstream regulator of SF1 in the appearance of steroidogenic cells in these organs. The Hh signaling pathway serves as a common crosstalk component, yet has evolved diverse functions in the expansion and differentiation of the steroidogenic cells in a tissue-specific manner. The purpose of this review is to compare and contrast the different roles of Hh signaling in these three organs during development.


Assuntos
Glândulas Suprarrenais , Proteínas Hedgehog/metabolismo , Ovário , Transdução de Sinais/fisiologia , Testículo , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/metabolismo , Animais , Linhagem da Célula , Feminino , Proteínas Hedgehog/genética , Masculino , Ovário/citologia , Ovário/embriologia , Ovário/metabolismo , Testículo/citologia , Testículo/embriologia , Testículo/metabolismo
15.
Endocrinology ; 151(3): 1119-28, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20118198

RESUMO

The adrenal capsule is postulated to harbor stem/progenitor cells, the progenies of which contribute to the growth of adrenocortex. We discovered that cells in the adrenal capsule are positive for Ptch1 and Gli1, genes indicative of responsiveness to the stimulation of Hedgehog (Hh) ligands. On the other hand, Sonic hedgehog (Shh), one of the mammalian Hh ligands, is expressed in the adrenocortex underneath the adrenal capsule, possibly acting upon the Hh-Responsive capsule. To investigate the functional significance of Shh in adrenal growth, we ablated Shh in an adrenocortex-specific manner using the Steroidogenic factor 1-Cre mouse. Loss of Shh in the adrenocortex led to reduced proliferation of capsular cells and a 50-75% reduction in adrenocortex thickness and adrenal size. The remaining adrenocortex underwent proper zonation and was able to synthesize steroids, indicating that Shh is dispensable for differentiation of adrenocortex. When these animals reached adulthood, their adrenocortex did not undergo compensatory growth in response to a high level of plasma ACTH, and the size of the adrenal remained significantly smaller than the control adrenal. Using a genetic lineage-tracing model, we further demonstrated that the Hh-responding cells in the adrenal capsule migrated centripetally into the adrenocortex. Our results not only provide the genetic evidence to support that the adrenal capsule contributes to the growth of adrenocortex in both fetal and adult life but also identify a novel role of Shh in this process.


Assuntos
Córtex Suprarrenal/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Células-Tronco/fisiologia , Fator Esteroidogênico 1/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/embriologia , Córtex Suprarrenal/enzimologia , Corticosteroides/biossíntese , Animais , Diferenciação Celular , Linhagem da Célula , Feminino , Proteínas Hedgehog/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamanho do Órgão , Gravidez , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...