Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 13: 906381, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35712256

RESUMO

Anti-Müllerian hormone (AMH) is a distinctive biomarker of the immature Sertoli cell. AMH expression, triggered by specific transcription factors upon fetal Sertoli cells differentiation independently of gonadotropins or sex steroids, drives Müllerian duct regression in the male, preventing the development of the uterus and Fallopian tubes. AMH continues to be highly expressed by Sertoli until the onset of puberty, when it is downregulated to low adult levels. FSH increases testicular AMH output by promoting immature Sertoli cell proliferation and individual cell expression. AMH secretion also showcases a differential regulation exerted by intratesticular levels of androgens and estrogens. In the fetus and the newborn, Sertoli cells do not express the androgen receptor, and the high androgen concentrations do not affect AMH expression. Conversely, estrogens can stimulate AMH production because estrogen receptors are present in Sertoli cells and aromatase is stimulated by FSH. During childhood, sex steroids levels are very low and do not play a physiological role on AMH production. However, hyperestrogenic states upregulate AMH expression. During puberty, testosterone inhibition of AMH expression overrides stimulation by estrogens and FSH. The direct effects of sex steroids on AMH transcription are mediated by androgen receptor and estrogen receptor α action on AMH promoter sequences. A modest estrogen action is also mediated by the membrane G-coupled estrogen receptor GPER. The understanding of these complex regulatory mechanisms helps in the interpretation of serum AMH levels found in physiological or pathological conditions, which underscores the importance of serum AMH as a biomarker of intratesticular steroid concentrations.


Assuntos
Hormônio Antimülleriano , Testículo , Androgênios/fisiologia , Hormônio Antimülleriano/fisiologia , Biomarcadores , Estrogênios/fisiologia , Hormônio Foliculoestimulante/fisiologia , Humanos , Masculino , Receptores Androgênicos/fisiologia , Testículo/crescimento & desenvolvimento , Testosterona/fisiologia
2.
Sci Rep ; 10(1): 15062, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934281

RESUMO

Anti-Müllerian hormone (AMH) is secreted by Sertoli cells of the testes from early fetal life until puberty, when it is downregulated by androgens. In conditions like complete androgen insensitivity syndrome (CAIS), AMH downregulation does not occur and AMH increases at puberty, due in part to follicle-stimulating hormone (FSH) effect. However, other conditions like Peutz-Jeghers syndrome (PJS), characterised by low FSH, also have increased AMH. Because both CAIS and PJS may present as hyperoestrogenic states, we tested the hypothesis that oestradiol (E2) upregulates AMH expression in peripubertal Sertoli cells and explored the molecular mechanisms potentially involved. The results showed that E2 is capable of inducing an upregulation of endogenous AMH and of the AMH promoter activity in the prepubertal Sertoli cell line SMAT1, signalling through ERα binding to a specific ERE sequence present on the hAMH promoter. A modest action was also mediated through the membrane oestrogen receptor GPER. Additionally, the existence of ERα expression in Sertoli cells in patients with CAIS was confirmed by immunohistochemistry. The evidence presented here provides biological plausibility to the hypothesis that testicular AMH production increases in clinical conditions in response to elevated oestrogen levels.


Assuntos
Síndrome de Resistência a Andrógenos/metabolismo , Hormônio Antimülleriano/metabolismo , Receptor alfa de Estrogênio/biossíntese , Proteínas de Neoplasias/biossíntese , Síndrome de Peutz-Jeghers/metabolismo , Elementos de Resposta , Células de Sertoli/metabolismo , Síndrome de Resistência a Andrógenos/patologia , Animais , Linhagem Celular , Criança , Pré-Escolar , Estradiol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Síndrome de Peutz-Jeghers/patologia , Células de Sertoli/patologia
3.
Mol Reprod Dev ; 87(1): 66-77, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31755607

RESUMO

Meiosis begins at puberty and relies on several factors, including androgens and retinoic acid in the mouse testis. CYP26B1 degrades retinoic acid in the testis during prenatal development preventing meiosis initiation. Given the concurrence of meiotic entry and completion of Sertoli cell maturation in response to androgens at puberty in the mouse, we proposed that CYP26B1 is downregulated by androgens in the Sertoli cell during this period. By immunohistochemistry, we showed that CYP26B1 declines in Sertoli cells after birth. However, luciferase reporter assays and quantitative reverse transcription-polymerase chain reaction performed in the prepubertal mouse Sertoli cell line SMAT1 revealed no changes in Cyp26b1 expression in response to androgen treatment. Furthermore, studies carried out using primary Sertoli cells of 10-day-old mice showed no changes in either Cyp26b1 or CYP26B1 expression in response to androgen treatment. In summary, the hereby reported decline in CYP26B1 expression in Sertoli cells towards pubertal onset does not appear to be caused by a direct inhibitory effect of androgens on Sertoli cells in the mouse.


Assuntos
Androgênios/farmacologia , Regulação para Baixo/efeitos dos fármacos , Ácido Retinoico 4 Hidroxilase/genética , Ácido Retinoico 4 Hidroxilase/metabolismo , Células de Sertoli/metabolismo , Androgênios/metabolismo , Animais , Animais Recém-Nascidos , Sítios de Ligação , Linhagem Celular , Regulação para Baixo/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Gônadas/embriologia , Masculino , Meiose/efeitos dos fármacos , Meiose/genética , Camundongos , Gravidez , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transfecção , Tretinoína/metabolismo
4.
Hum Mol Genet ; 28(19): 3211-3218, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31238341

RESUMO

The persistent Müllerian duct syndrome (PMDS) is a 46,XY disorder of sexual development characterized by the persistence of Müllerian duct derivatives, uterus and tubes, in otherwise normally masculinized males. The condition, transmitted as a recessive autosomal trait, is usually due to mutations in either the anti-Müllerian hormone (AMH) gene or its main receptor. Many variants of these genes have been described, all targeting the coding sequences. We report the first case of PMDS due to a regulatory mutation. The AMH promoter contains two binding sites for steroidogenic factor 1 (SF1), one at -102 and the other at -228. Our patient carries a single base deletion at -225, significantly decreasing its capacity for binding SF1, as measured by the electrophoresis mobility shift assay. Furthermore, by linking the AMH promoter to the luciferase gene, we show that the transactivation capacity of the promoter is significantly decreased by the mutation, in contrast to the disruption of the -102 binding site. To explain the difference in impact we hypothesize that SF1 could partially overcome the lack of binding to the -102 binding site by interacting with a GATA4 molecule linked to a nearby response element. We show that disruption of both the -102 SF1 and the -84 GATA response elements significantly decreases the transactivation capacity of the promoter. In conclusion, we suggest that the distance between mutated SF1 sites and potentially rescuing GATA binding motifs might play a role in the development of PMDS.


Assuntos
Hormônio Antimülleriano/química , Hormônio Antimülleriano/metabolismo , Transtorno 46,XY do Desenvolvimento Sexual/genética , Mutação , Fatores de Processamento de RNA/metabolismo , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Hormônio Antimülleriano/genética , Sítios de Ligação/genética , Linhagem Celular , Criança , Pré-Escolar , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Regiões Promotoras Genéticas , Ligação Proteica
5.
Biol Reprod ; 99(6): 1303-1312, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29985989

RESUMO

Testicular anti-Müllerian hormone (AMH) production is inhibited by androgens around pubertal onset, as observed under normal physiological conditions and in patients with precocious puberty. In agreement, AMH downregulation is absent in patients with androgen insensitivity. The molecular mechanisms underlying the negative regulation of AMH by androgens remain unknown. Our aim was to elucidate the mechanisms through which androgens downregulate AMH expression in the testis. A direct negative effect of androgens on the transcriptional activity of the AMH promoter was found using luciferase reporter assays in the mouse prepubertal Sertoli cell line SMAT1. A strong inhibition of AMH promoter activity was seen in the presence of both testosterone and DHT and of the androgen receptor. By site-directed mutagenesis and chromatin immunoprecipitation assays, we showed that androgen-mediated inhibition involved the binding sites for steroidogenic factor 1 (SF1) present in the proximal promoter of the AMH gene. In this study, we describe for the first time the mechanism behind AMH inhibition by androgens, as seen in physiological and pathological conditions in males. Inhibition of AMH promoter activity by androgens could be due to protein-protein interactions between the ligand-bound androgen receptor and SF1 or by blockage of SF1 binding to its sites on the AMH promoter.


Assuntos
Androgênios/farmacologia , Hormônio Antimülleriano/metabolismo , Células de Sertoli/fisiologia , Fator Esteroidogênico 1/metabolismo , Animais , Hormônio Antimülleriano/genética , Linhagem Celular , Imunoprecipitação da Cromatina , Regulação para Baixo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Regiões Promotoras Genéticas , Receptores Androgênicos/metabolismo , Fator Esteroidogênico 1/genética , Transcriptoma
6.
Int J Pediatr Endocrinol ; 2016: 20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27799946

RESUMO

In pediatric patients, basal testosterone and gonadotropin levels may be uninformative in the assessment of testicular function. Measurement of serum anti-Müllerian hormone (AMH) has become increasingly widespread since it provides information about the activity of the male gonad without the need for dynamic tests, and also reflects the action of FSH and androgens within the testis. AMH is secreted in high amounts by Sertoli cells from fetal life until the onset of puberty. Basal AMH expression is not dependent on gonadotropins or sex steroids; however, FSH further increases and testosterone inhibits AMH production. During puberty, testosterone induces Sertoli cell maturation, and prevails over FSH on AMH regulation. Therefore, AMH production decreases. Serum AMH is undetectable in patients with congenital or acquired anorchidism, or with complete gonadal dysgenesis. Low circulating levels of AMH may reflect primary testicular dysfunction, e.g. in certain patients with cryptorchidism, monorchidism, partial gonadal dysgenesis, or central hypogonadism. AMH is low in boys with precocious puberty, but it increases to prepubertal levels after successful treatment. Conversely, serum AMH remains at high, prepubertal levels in boys with constitutional delay of puberty. Serum AMH measurements are useful, together with testosterone determination, in the diagnosis of patients with ambiguous genitalia: both are low in patients with gonadal dysgenesis, including ovotesticular disorders of sex development, testosterone is low but AMH is in the normal male range or higher in patients with disorders of androgen synthesis, and both hormones are normal or high in patients with androgen insensitivity. Finally, elevation of serum AMH above normal male prepubertal levels may be indicative of rare cases of sex-cord stromal tumors or Sertoli cell-limited disturbance in the McCune Albright syndrome.

7.
Reprod Toxicol ; 58: 203-12, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26498383

RESUMO

We analyzed the potential role of oxidative stress induced by mono (2-ethylhexyl) phthalate (MEHP) in adherent cell junction protein expression of prepubertal rat Sertoli cells (SC) in vitro. Five-day SC cultures were treated with MEHP (200µM) for 24h and compared to cells in basal conditions. Western blot and immunofluorescent (IF) analyses showed that MEHP induced increase of N-cadherin and catenin expression, modifying its distribution. Concomitantly, Cx-43 expression decreased significantly and delocalization of the IF signal for tight junction proteins (occludin, claudin-11 and ZO-1) occurred. Indicative of oxidative stress, MEHP induced in SC an increase of lipoperoxides, a decrease in glutathione (GSH) levels and a concomitant increase in Glutathione S-Transferases (GST) activity. Antioxidant N-acetyl-cysteine (1mM) treatment prevented GSH decrease and N-cadherin and α-catenin up-regulation induced by MEHP. Our data suggest that oxidative stress signaling is a mechanism involved in adherent cell junctions disruption induced by MEHP in SC cultures.


Assuntos
Dietilexilftalato/análogos & derivados , Junções Intercelulares/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Células de Sertoli/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Caderinas/metabolismo , Forma Celular/efeitos dos fármacos , Células Cultivadas , Claudina-1/metabolismo , Conexina 43/metabolismo , Citoproteção , Dietilexilftalato/toxicidade , Glutationa/metabolismo , Glutationa Transferase/metabolismo , Junções Intercelulares/metabolismo , Junções Intercelulares/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Ocludina/metabolismo , Ratos Sprague-Dawley , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Proteína da Zônula de Oclusão-1/metabolismo , alfa Catenina/metabolismo
8.
Curr Opin Endocrinol Diabetes Obes ; 20(3): 224-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23549308

RESUMO

PURPOSE OF REVIEW: Biomarkers of prepubertal testicular function have become widely available only in recent years. The aim of this review is to update the knowledge on key biomarkers used to assess hypogonadism in boys. RECENT FINDINGS: Sertoli cells are the most representative cells of the prepubertal testis. Anti-Müllerian hormone and inhibin B are essential biomarkers of Sertoli cell function. Also, INSL3 arises as an additional marker of Leydig cell dysfunction. SUMMARY: The widespread use of these biomarkers has enhanced our knowledge on the pathophysiology and diagnosis of prepubertal male hypogonadism. Beyond their well known germ-cell toxicity, oncologic treatments may also affect Sertoli cell function. Pathophysiology is not the same in all aneuploidies leading to infertility: while hypogonadism is not evident until mid-puberty in Klinefelter syndrome, it is established in early infancy in Down syndrome. In Noonan syndrome, the occurrence of primary hypogonadism depends on the existence of cryptorchidism, and Prader-Willi syndrome may present with either primary or combined forms of hypogonadism. Prepubertal testicular markers have also provided insights into the effects of environmental disruptors on gonadal function from early life, and helped dissipate concerns about testicular function in boys born preterm or small for gestational age or conceived by assisted reproductive technique procedures.


Assuntos
Desenvolvimento Infantil , Espermatogênese , Testículo/crescimento & desenvolvimento , Hormônio Antimülleriano/metabolismo , Biomarcadores/metabolismo , Criança , Transtornos do Desenvolvimento Sexual/etiologia , Transtornos do Desenvolvimento Sexual/metabolismo , Transtornos do Desenvolvimento Sexual/patologia , Humanos , Lactente , Inibinas/metabolismo , Insulina/metabolismo , Células Intersticiais do Testículo/citologia , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia , Masculino , Proteínas/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Testículo/citologia , Testículo/metabolismo , Testículo/patologia
9.
Am J Physiol Endocrinol Metab ; 301(3): E539-47, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21693691

RESUMO

In Sertoli cells, anti-Müllerian hormone (AMH) expression is upregulated by FSH via cyclic AMP (cAMP), although no classical cAMP response elements exist in the AMH promoter. The response to cAMP involves NF-κB and AP2; however, targeted mutagenesis of their binding sites in the AMH promoter do not completely abolish the response. In this work we assessed whether SOX9, SF1, GATA4, and AP1 might represent alternative pathways involved in cAMP-mediated AMH upregulation, using real-time RT-PCR (qPCR), targeted mutagenesis, luciferase assays, and immunocytochemistry in the Sertoli cell line SMAT1. We also explored the signaling cascades potentially involved. In qPCR experiments, Amh, Sox9, Sf1, and Gata4 mRNA levels increased after SMAT1 cells were incubated with cAMP. Blocking PKA abolished the effect of cAMP on Sox9, Sf1, and Gata4 expression, inhibiting PI3K/PKB impaired the effect on Sf1 and Gata4, and reducing MEK1/2 and p38 MAPK activities curtailed Gata4 increase. SOX9 and SF1 translocated to the nucleus after incubation with cAMP. Mutations of the SOX9 or SF1 sites, but not of GAT4 or AP1 sites, precluded the response of a 3,063-bp AMH promoter to cAMP. In conclusion, in the Sertoli cell line SMAT1 cAMP upregulates SOX9, SF1, and GATA4 expression and induces SOX9 and SF1 nuclear translocation mainly through PKA, although other kinases may also participate. SOX9 and SF1 binding to the AMH promoter is essential to increase the activity of the AMH promoter in response to cAMP.


Assuntos
Hormônio Antimülleriano/metabolismo , AMP Cíclico/metabolismo , Fatores de Transcrição SOX9/metabolismo , Células de Sertoli/metabolismo , Fator Esteroidogênico 1/metabolismo , Hormônio Antimülleriano/genética , Linhagem Celular , AMP Cíclico/genética , Proteínas de Ligação a DNA , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Expressão Gênica , Humanos , Masculino , Regiões Promotoras Genéticas , Fatores de Processamento de RNA , Fatores de Transcrição SOX9/genética , Transdução de Sinais/fisiologia , Fator Esteroidogênico 1/genética , Fatores de Transcrição , Regulação para Cima
10.
Int J Androl ; 32(3): 218-25, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18042181

RESUMO

Migration of developing germ cells from the basal to the adluminal compartment of the seminiferous epithelium requires extensive tissue restructuring, resulting in the production of reactive oxygen species. Sertoli cells are involved in this process. Glutathione (GSH), produced by Sertoli cells, has an essential role in cell protection against oxidative stress. Intracellular GSH content is maintained by de novo synthesis, involving glutamate-cysteine ligase catalytic (GCLC) and modulatory (GCLM) subunits, and by recycling from oxidized GSH, catalysed by glutathione reductase (GR). To assess whether follicle-stimulating hormone (FSH) and basic fibroblast growth factor (bFGF) modulate GSH production in Sertoli cells by regulating the expression of GCLC, GCLM and/or GR, we performed in vitro studies using rat Sertoli cells in primary culture. FSH and bFGF stimulation increased Sertoli cell GSH levels after 24 h incubation. The simultaneous addition of FSH and bFGF did not produce any further effect. GCLM expression was upregulated by FSH and bFGF 6 h. At 24 h, only the FSH-mediated effect was still observed. FSH and bFGF also upregulated GR expression. In conclusion, our results show that FSH and bFGF increase GSH levels in Sertoli cells through stimulation of the de novo synthesis and recycling by upregulating GCLM and GR expression respectively. Therefore, protection of germ cells against oxidative stress seems to be regulated by hormones and germ cell-released growth factors capable of influencing the production of Sertoli cell GSH.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Hormônio Foliculoestimulante/farmacologia , Glutationa/biossíntese , Células de Sertoli/metabolismo , Animais , Células Cultivadas , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Peróxido de Hidrogênio/farmacologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Células de Sertoli/efeitos dos fármacos , Regulação para Cima
11.
Int J Androl ; 26(5): 310-7, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14511220

RESUMO

The aim of the study was to determine whether arachidonic acid (AA) is involved in the regulation of Sertoli cell lactate production and if this fatty acid participates in follicle-stimulating hormone (FSH) regulation of Sertoli cell function. In a first set of experiments the effect of AA and porcine pancreas phospholipase A2 (PLA2) on lactate production, glucose uptake, lactate dehydrogenase (LDH) activity and LDH A mRNA levels in Sertoli cell cultures obtained from 20-day-old rats was evaluated. In a second set of experiments the effect of two PLA2 inhibitors--quinacrine (Q) and AACOCF3--on FSH stimulation of the above-mentioned parameters of Sertoli cell function was investigated. Treatment with PLA2 and AA increased Sertoli cell lactate production. The observed action of exogenously added PLA2 involved its catalytic properties responsible for AA release. PLA2 and AA treatments also stimulated Sertoli cell glucose uptake, LDH activity and LDH A mRNA levels. In order to determine whether AA participates in FSH regulation of Sertoli cell lactate production cells were incubated with FSH in the absence or presence of the PLA2 inhibitors Q and AACOCF3. Both drugs partially inhibited the ability of FSH to stimulate lactate production, glucose uptake and LDH activity. The present investigation suggests that AA is involved in the regulation of lactate production, glucose transport, LDH activity and LDH A mRNA levels. In addition, these results suggest that cytosolic PLA2 and AA may participate in FSH-regulation of Sertoli cell energetic metabolism.


Assuntos
Ácido Araquidônico/fisiologia , Hormônio Foliculoestimulante/fisiologia , Ácido Láctico/biossíntese , Células de Sertoli/metabolismo , Animais , Ácido Araquidônico/farmacologia , Células Cultivadas , Hormônio Foliculoestimulante/farmacologia , Glucose/metabolismo , L-Lactato Desidrogenase/metabolismo , Masculino , Fosfolipases A/metabolismo , Fosfolipases A/farmacologia , Fosfolipases A2 , Ratos , Ratos Sprague-Dawley , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...