Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Biol Rep ; 50(4): 3927-3933, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36680623

RESUMO

BACKGROUND: The classification of the sub-family Schizothoracinae has been debatable due to the overlap in morphological characters. There are discrepancies between classical taxonomy and molecular taxonomy, as well. In the present study, mitochondrial genes CO-I and Cyt b were sequenced to elucidate the phylogenetic status of three species of the genus Schizothorax. METHODS AND RESULTS: In total, 29 samples of three species viz., S. plagiostomus, S. progastus, and S. richardsonii, were collected from rivers of Uttarakhand, India. For phylogenetic analyses, 40 sequences of CO-I and 41 sequences of Cyt b of Schizothoracinae species were downloaded from NCBI. The highest genetic divergence based on CO-I (16.08%) is between S. plagiostomus and Ptychobarbus dipogon, while the lowest divergence (0.00%) is between 10 pairs of species. The highest divergence based on Cyt b (19.43%), is between S. niger and Gymnocypris eckloni, while the lowest divergence (0.00%) is between four pairs of species. The divergence (0.00% for CO-I and 2.38% for Cyt b) between S. chongi and S. kozlovi, seems a case of convergent molecular evolution of the CO-I gene and in this case, CO-I alone cannot be used to differentiate these two species. CONCLUSION: The simultaneous use of two molecular markers along with morphomeristic data is a better strategy for the classification of the sub-family Schizothoracinae. These results will be a resource dataset for determining the taxonomical status of Schizothoracine species and will help in the conservation and commercial production of these commercially important fish species.


Assuntos
Cyprinidae , Citocromos b , Animais , Cyprinidae/genética , Citocromos b/genética , DNA Mitocondrial/genética , Filogenia , Truta/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética
2.
Cell Mol Life Sci ; 79(3): 136, 2022 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-35181820

RESUMO

Sertoli cells (Sc) are the sole target of follicle-stimulating hormone (FSH) in the testis and attain functional maturation post-birth to significantly augment germ cell (Gc) division and differentiation at puberty. Despite having an operational microRNA (miRNA) machinery, limited information is available on miRNA-mediated regulation of Sc maturation and male fertility. We have shown before that miR-92a-3p levels decline in pubertal rat Sc. In response to FSH treatment, the expressions of FSH Receptor, Claudin11 and Klf4 were found to be elevated in pubertal rat Sc coinciding with our finding of FSH-induced decline in miR-92a-3p levels. To investigate the association of miR-92a-3p and spermatogenesis, we generated transgenic mice where such pubertal decline of miR-92a-3p was prevented by its overexpression in pubertal Sc under proximal Rhox5 promoter, which is known to be activated specifically at puberty, in Sc. Our in vivo observations provided substantial evidence that FSH-induced decline in miR-92a-3p expression during Sc maturation acts as an essential prerequisite for the pubertal onset of spermatogenesis. Elevated expression of miR-92a-3p in post-pubertal testes results into functionally compromised Sc, leading to impairment of the blood-testis barrier formation and apoptosis of pre-meiotic Gc, ultimately culminating into infertility. Collectively, our data suggest that regulation of miR-92a-3p expression is crucial for Sc-mediated induction of active spermatogenesis at puberty and regulation of male fertility.


Assuntos
Diferenciação Celular , Fertilidade , Hormônio Foliculoestimulante/farmacologia , Células Germinativas/citologia , MicroRNAs/genética , Células de Sertoli/citologia , Testículo/citologia , Animais , Feminino , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Hormônios/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Wistar , Receptores do FSH/genética , Receptores do FSH/metabolismo , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Maturidade Sexual , Espermatogênese , Testículo/efeitos dos fármacos , Testículo/metabolismo
3.
J Fish Biol ; 103(2): 292-304, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37184405

RESUMO

Schizothorax plagiostomus, commonly known as snow trout, is a popular food fish found in parts of Central Asia and the Indo-Himalayan region. Despite such a broad range of distribution and potential financial value, it is a highly neglected cold-water ichthyofauna. Furthermore, an alarming decline in Schizothoracine population has been reported in the recent past due to climate change and uncontrolled anthropogenic interference. In this study, the seasonal variations in ovarian architecture and development were examined in adult S. plagiostomus from Garhwal Himalayan region, Uttarakhand, India. Ovarian-somatic index ranged from 16.86 ± 0.29 to 0.31 ± 0.56, with a maximal value in September and a minimal value in April. Ovarian histology revealed the abundance of primary growth oocytes in resting and preparatory stages; primary/secondary vitellogenic oocytes with numerous cortical alveoli were predominant in the developing stage of pre-spawning ovaries; secondary/tertiary vitellogenic oocytes were conspicuous in actively spawning ovaries; and atretic follicles/oocytes were discernible during the regressing stage of spent ovaries. Scanning electron microscopy of mature ova (mean diameter 2.003 ± 0.01 mm) prominently showed the structure micropyle (mean diameter 12.93 ± 3.38 µm). Fecundity analyses suggested that September was the principal breeding season, whereas residual spawning occurred with fresh rain in late winter during February-March. Collectively, this is the first comprehensive qualitative and quantitative report of the seasonal variations in the ovarian development and function for S. plagiostomus. These data may provide valuable information towards the captive breeding programme as well as conservation and management for Schizothoracine fishes in normal and altered climatic conditions.


Assuntos
Cyprinidae , Truta , Feminino , Animais , Estações do Ano , Oócitos , Ovário , Folículo Ovariano
4.
Biochem Biophys Res Commun ; 569: 100-105, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34237428

RESUMO

Follicle Stimulating Hormone (FSH) acts via FSH-Receptor (FSH-R) by employing cAMP as the dominant secondary messenger in testicular Sertoli cells (Sc) to support spermatogenesis. Binding of FSH to FSH-R, results the recruitment of the intracellular GTP binding proteins, either stimulatory Gαs or inhibitory Gαi that in turn regulate cAMP production in Sc. The cytosolic concentration of cAMP being generated by FSH-R thereafter critically determines the downstream fate of the FSH signalling. The pleiotropic action of FSH due to differential cAMP output during functional maturation of Sc has been well studied. However, the developmental and cellular regulation of the Gα proteins associated with FSH-R is poorly understood in Sc. In the present study, we report the differential transcriptional modulation of the Gα subunit genes by FSH mediated cAMP signalling in neonatal and pubertal rat Sc. Our data suggested that unlike in neonatal Sc, both the basal and FSH/forskolin induced expression of Gαs, Gαi-1, Gαi-2 and Gαi-3 transcripts was significantly (p < 0.05) up-regulated in pubertal Sc. Further investigations involving treatment of Sc with selective Gαi inhibitor pertussis toxin, confirmed the elevated expression of Gi subunits in pubertal Sc. Collectively our results indicated that the high level of Gαi subunits serves as a negative regulator to optimize cAMP production in pubertal Sc.


Assuntos
AMP Cíclico/metabolismo , Hormônio Foliculoestimulante/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Células Cultivadas , Colforsina/farmacologia , Hormônio Foliculoestimulante/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Toxina Pertussis/farmacologia , Ligação Proteica , Ratos Wistar , Receptores do FSH/genética , Receptores do FSH/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Maturidade Sexual/fisiologia , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética
5.
Mol Hum Reprod ; 26(10): 760-772, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32687199

RESUMO

The alarming decline in sperm count has become a global concern in the recent decades. The division and differentiation of male germ cells (Gc) into sperm are governed by Sertoli cells (Sc) upon their functional maturation during puberty. However, the roles of genes regulating pubertal maturation of Sc have not been fully determined. We have observed that Tetraspanin 8 (Tspan8) is down-regulated in Sc during puberty in rats. However, there has been no in vivo evidence for a causal link between the down-regulation of Tspan8 expression and the onset of spermatogenesis as yet. To investigate this, we generated a novel transgenic (Tg) rat, in which the natural down-regulation of Tspan8 was prevented specifically in Sc from puberty up to adulthood. Adult Tg male rats showed around 98% reduction in sperm count despite having a similar level of serum testosterone (T) as the controls. Functional maturation of Sc was impaired as indicated by elevated levels of Amh and low levels of Kitlg and Claudin11 transcripts. The integrity of the blood testis barrier was compromised due to poor expression of Gja1 and Gc apoptosis was discernible. This effect was due to a significant rise in both Mmp7 and phospho P38 MAPK in Tg rat testis. Taken together, we demonstrated that the natural down-regulation of Tspan8 in Sc during puberty is a prerequisite for establishing male fertility. This study divulges one of the aetiologies of certain forms of idiopathic male infertility where somatic cell defect, but not hormonal deficiency, is responsible for impaired spermatogenesis.


Assuntos
Infertilidade Masculina/genética , Células de Sertoli/metabolismo , Maturidade Sexual/genética , Tetraspaninas/genética , Animais , Regulação para Baixo/genética , Feminino , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/metabolismo , Masculino , Gravidez , Ratos , Ratos Transgênicos , Ratos Wistar , Testículo/metabolismo , Tetraspaninas/metabolismo
6.
Mol Reprod Dev ; 86(11): 1505-1530, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31368614

RESUMO

The term "Puberty", socially known as "Adolescence" is the transitional period from juvenile life to adulthood with functional maturation of gonads and genital organs. In this process, some remarkable developmental changes occur in morphology, physiology, and behavior leading to reproductive competence. Despite sufficient levels of gonadotropins (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]), robust spermatogenesis is not initiated during infancy in primates due to the immaturity of testicular Sertoli cells. Recent studies suggest that developmental competence augmenting functional activities of receptors for androgen and FSH is acquired by Sertoli cells somewhere during the prolonged hypo-gonadotropic juvenile period. This juvenile phase is terminated with the re-awakening of hypothalamic Kisspeptin/Neurokinin B/Dynorphin neurons which induce the release of the gonadotropin-releasing hormone leading to reactivation of the hypothalamo-pituitary-testicular axis at puberty. During this period of pubertal development, FSH and LH facilitate further maturation of testicular cells (Sertoli cells and Leydig cells) triggering robust differentiation of the spermatogonial cells, ensuing the spermatogenic onset. This review aims to precisely address the evolving concepts of the pubertal regulation of hormone production with the corresponding cooperation of testicular cells for the initiation of robust spermatogenesis, which can be truly called "testicular puberty."


Assuntos
Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Células Intersticiais do Testículo/metabolismo , Hormônio Luteinizante/metabolismo , Células de Sertoli/metabolismo , Maturidade Sexual/fisiologia , Espermatogênese/fisiologia , Animais , Células Intersticiais do Testículo/citologia , Masculino , Primatas , Células de Sertoli/citologia
7.
Cell Tissue Res ; 368(2): 389-396, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27139181

RESUMO

In response to various hormonal (follicle-stimulating hormone [FSH] and testosterone [T]) and biochemical inputs, testicular Sertoli cells (Sc) produce factors that regulate spermatogenesis. A number of FSH- and T-responsive Sc-specific genes, necessary for spermatogenesis, have been identified to date. However, the hormone-induced in vitro expression pattern of most of these genes is reported to be inconsistent at various time points in primary rat Sc cultures. As a matter of convenience, cultured Sc are constantly exposed to hormones for a few hours to days in the reported literature, although Sc are exposed to pulsatile FSH and T in vivo. The major aim of the present study is to evaluate the advantage, if any, of the in vitro administration of pulsatile hormone (FSH and T in combination) treatment on gene expression of cultured Sc as compared with that of constant hormone treatment. Pulsatile treatment (a 30-min hormonal exposure every 3 h) mimicking the in vivo condition reveals a more prominent effect of hormones in augmenting gene expression as compared with constant treatment. Our results indicate that the expressions of Stem cell factor (Scf, only responsive to FSH), Claudin11 (only responsive to T) and Transferrin (both FSH- and T-responsive) mRNAs are significantly higher at 12 h upon pulsatile treatment than upon constant hormonal treatment. Maximal expression of relevant genes because of pulsatile treatment with hormones suggests that this protocol provides a more suitable premise for assessing hormone-induced gene expression in isolated Sc than one involving constant exposure to hormones.


Assuntos
Células de Sertoli/metabolismo , Animais , Células Cultivadas , Claudinas/genética , Claudinas/metabolismo , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Células de Sertoli/efeitos dos fármacos , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Testosterona/farmacologia , Transferrina/genética , Transferrina/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(23): E2384-93, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24912173

RESUMO

Organogenesis of the testis is initiated when expression of Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. The cells in the early bipotential gonad undergo de novo organization to form testis cords that enclose germ cells inside tubules lined by epithelial Sertoli cells. Although Sertoli cells are a driving force in the de novo formation of testis cords, recent studies in mouse showed that reorganization of the vasculature and of interstitial cells also play critical roles in testis cord morphogenesis. However, the mechanism driving reorganization of the vasculature during fetal organogenesis remained unclear. Here we demonstrate that fetal macrophages are associated with nascent gonadal and mesonephric vasculature during the initial phases of testis morphogenesis. Macrophages mediate vascular reorganization and prune errant germ cells and somatic cells after testis architecture is established. We show that gonadal macrophages are derived from primitive yolk-sac hematopoietic progenitors and exhibit hallmarks of M2 activation status, suggestive of angiogenic and tissue remodeling functions. Depletion of macrophages resulted in impaired vascular reorganization and abnormal cord formation. These findings reveal a previously unappreciated role for macrophages in testis morphogenesis and suggest that macrophages are an intermediary between neovascularization and organ architecture during fetal organogenesis.


Assuntos
Macrófagos/metabolismo , Morfogênese , Testículo/irrigação sanguínea , Testículo/embriologia , Animais , Receptor 1 de Quimiocina CX3C , Linhagem da Célula , Feto/irrigação sanguínea , Feto/citologia , Feto/embriologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Células Mieloides/metabolismo , Técnicas de Cultura de Órgãos , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Testículo/citologia , Fatores de Tempo , Saco Vitelino/metabolismo
9.
Reprod Sci ; 31(4): 942-965, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38036863

RESUMO

Spermatogenesis is a complex process of germ cell division and differentiation that involves extensive cross-talk between the developing germ cells and the somatic testicular cells. Defective endocrine signaling and/or intrinsic defects within the testes can adversely affect spermatogenic progression, leading to subfertility/infertility. In recent years, male infertility has been recognized as a global public health concern, and research over the last few decades has elucidated the complex etiology of male infertility. Congenital reproductive abnormalities, genetic mutations, and endocrine/metabolic dysfunction have been demonstrated to be involved in infertility/subfertility in males. Furthermore, acquired factors like exposure to environmental toxicants and lifestyle-related disorders such as illicit use of psychoactive drugs have been shown to adversely affect spermatogenesis. Despite the large body of available scientific literature on the etiology of male infertility, a substantial proportion of infertility cases are idiopathic in nature, with no known cause. The inability to treat such idiopathic cases stems from poor knowledge about the complex regulation of spermatogenesis. Emerging scientific evidence indicates that defective functioning of testicular Sertoli cells (Sc) may be an underlying cause of infertility/subfertility in males. Sc plays an indispensable role in regulating spermatogenesis, and impaired functional maturation of Sc has been shown to affect fertility in animal models as well as humans, suggesting abnormal Sc as a potential underlying cause of reproductive insufficiency/failure in such cases of unexplained infertility. This review summarizes the major causes of infertility/subfertility in males, with an emphasis on infertility due to dysregulated Sc function.


Assuntos
Infertilidade Masculina , Testículo , Animais , Masculino , Humanos , Infertilidade Masculina/genética , Células de Sertoli , Espermatogênese/genética , Fertilidade
10.
Front Endocrinol (Lausanne) ; 15: 1394812, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39055054

RESUMO

Spermatogenesis is a multi-step biological process where mitotically active diploid (2n) spermatogonia differentiate into haploid (n) spermatozoa via regulated meiotic programming. The alarming rise in male infertility has become a global concern during the past decade thereby demanding an extensive profiling of testicular gene expression. Advancements in Next-Generation Sequencing (NGS) technologies have revolutionized our empathy towards complex biological events including spermatogenesis. However, despite multiple attempts made in the past to reveal the testicular transcriptional signature(s) either with bulk tissues or at the single-cell, level, comprehensive reviews on testicular transcriptomics and associated disorders are limited. Notably, technologies explicating the genome-wide gene expression patterns during various stages of spermatogenic progression provide the dynamic molecular landscape of testicular transcription. Our review discusses the advantages of single-cell RNA-sequencing (Sc-RNA-seq) over bulk RNA-seq concerning testicular tissues. Additionally, we highlight the cellular heterogeneity, spatial transcriptomics, dynamic gene expression and cell-to-cell interactions with distinct cell populations within the testes including germ cells (Gc), Sertoli cells (Sc), Peritubular cells (PTc), Leydig cells (Lc), etc. Furthermore, we provide a summary of key finding of single-cell transcriptomic studies that have shed light on developmental mechanisms implicated in testicular disorders and male infertility. These insights emphasize the pivotal roles of Sc-RNA-seq in advancing our knowledge regarding testicular transcriptional landscape and may serve as a potential resource to formulate future clinical interventions for male reproductive health.


Assuntos
Infertilidade Masculina , Análise de Célula Única , Testículo , Transcriptoma , Masculino , Humanos , Testículo/metabolismo , Testículo/patologia , Infertilidade Masculina/genética , Infertilidade Masculina/patologia , Infertilidade Masculina/metabolismo , Animais , Espermatogênese/genética , Perfilação da Expressão Gênica
11.
Front Endocrinol (Lausanne) ; 14: 1110572, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37124741

RESUMO

Spermatogenesis is a multi-step process of male germ cell (Gc) division and differentiation which occurs in the seminiferous tubules of the testes under the regulation of gonadotropins - Follicle Stimulating Hormone (FSH) and Luteinising hormone (LH). It is a highly coordinated event regulated by the surrounding somatic testicular cells such as the Sertoli cells (Sc), Leydig cells (Lc), and Peritubular myoid cells (PTc). FSH targets Sc and supports the expansion and differentiation of pre-meiotic Gc, whereas, LH operates via Lc to produce Testosterone (T), the testicular androgen. T acts on all somatic cells e.g.- Lc, PTc and Sc, and promotes the blood-testis barrier (BTB) formation, completion of Gc meiosis, and spermiation. Studies with hypophysectomised or chemically ablated animal models and hypogonadal (hpg) mice supplemented with gonadotropins to genetically manipulated mouse models have revealed the selective and synergistic role(s) of hormones in regulating male fertility. We here have briefly summarized the present concept of hormonal control of spermatogenesis in rodents and primates. We also have highlighted some of the key critical questions yet to be answered in the field of male reproductive health which might have potential implications for infertility and contraceptive research in the future.


Assuntos
Espermatogênese , Testículo , Masculino , Camundongos , Animais , Espermatogênese/fisiologia , Células de Sertoli/fisiologia , Gonadotropinas , Hormônio Foliculoestimulante/fisiologia , Hormônio Luteinizante , Mamíferos
12.
Am J Physiol Endocrinol Metab ; 303(7): E886-98, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22850685

RESUMO

FSH and Testosterone (T) regulate spermatogenesis via testicular Sertoli cells (Sc), which bear receptors for these hormones. Despite sufficient circulating levels of FSH and T postnatally, predominant appearance of spermatogonia B and spermatocytes is not discernible until 11 and 18 days of postnatal age, respectively, in rat testes. In an attempt to explore the underlying causes, we cultured Sc from neonatal (5- and 9-day-old) and prepubertal (12- and 19-day-old) rat testes and compared the status of FSH receptor (FSH-R) and androgen receptor (AR) signaling. Protein and mRNA levels of FSH-R and AR remained uniform in cultured Sc from all age groups. Androgen binding ability of AR was similar, and T-induced nuclear localization of AR was discernible in Sc from all age groups. Binding of FSH to FSH-R, subsequent production of cAMP, and mRNA of stem cell factor (SCF) and glial cell line-derived neurotrophic factor (GDNF), known to be essential for the robust differentiation of repopulating spermatogonia, were significantly augmented in prepubertal Sc compared with those in neonatal Sc. However, treatment of neonatal Sc with cholera toxin or forskolin, which stimulate cAMP production bypassing FSH-R, demonstrated a concomitant rise in SCF and GDNF mRNA expression, which was similar to the FSH-mediated rise observed in prepubertal Sc. These observations suggested that, during prepubertal Sc maturation, the ability of FSH-R to respond to FSH is significantly augmented and is associated with the robust differentiation of repopulating spermatogonia, and such a switch in Sc from FSH-resistant to FSH-responsive mode during prepubertal development may underlie the initiation of robust spermatogenesis.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Receptores do FSH/metabolismo , Células de Sertoli/fisiologia , Espermatogênese/fisiologia , Testículo/crescimento & desenvolvimento , Testículo/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Toxina da Cólera/farmacologia , Colforsina/farmacologia , AMP Cíclico/biossíntese , Hormônio Foliculoestimulante/sangue , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Masculino , Ratos , Ratos Wistar , Receptores Androgênicos/análise , Receptores do FSH/análise , Células de Sertoli/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Fator de Células-Tronco/biossíntese , Testículo/efeitos dos fármacos , Testosterona/sangue
13.
Hum Reprod ; 27(8): 2515-25, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22669085

RESUMO

BACKGROUND: In humans, as well as in other higher primates, the infantile testis is exposed to an adult-like hormonal milieu, but spermatogenesis is not initiated at this stage of primate development. In the present study, we examined the molecular basis of this intriguing infertile state of the primate testis. METHODS: The integrity of androgen receptor (AR) and FSH receptor (FSHR) signaling pathways in primary cultures of Sertoli cells (Scs) harvested from azoospermic infant and spermatogenic pubertal monkey testes were investigated under identical in vitro hormonal conditions. In order to synchronously harvest Scs from early pubertal testis, the activation of testicular puberty was timed experimentally by prematurely initiating gonadotrophin secretion in juvenile animals with an intermittent infusion of gonadotrophin-releasing hormone. RESULTS: While qRT-PCR demonstrated that AR and FSHR mRNA expression in Scs from infant and pubertal testes were comparable, androgen-binding and FSH-mediated cAMP production by infant Scs was extremely low. Compromised AR and FSHR signaling in infant Scs was further supported by the finding that testosterone (T) and FSH failed to augment the expression of the T responsive gene, claudin 11, and the FSH responsive genes, inhibin-ßB, stem cell factor (SCF) and glial cell line-derived neurotrophic factor (GDNF) in Scs harvested at this stage of development. CONCLUSION: These results indicate that compromised AR and FSHR signaling pathways in Scs underlie the inability of the infant primate testis to respond to an endogenous hormonal milieu that later in development, at the time puberty, stimulates the initiation of spermatogenesis. This finding may have relevance to some forms of idiopathic infertility in men.


Assuntos
Androgênios/metabolismo , Azoospermia/metabolismo , Hormônio Foliculoestimulante/metabolismo , Testículo/crescimento & desenvolvimento , Animais , Ligantes , Macaca mulatta , Masculino , RNA Mensageiro/metabolismo , Receptores Androgênicos/metabolismo , Receptores do FSH/metabolismo , Células de Sertoli/metabolismo , Espermatogênese , Testículo/metabolismo , Testosterona/metabolismo , Fatores de Tempo
14.
Front Endocrinol (Lausanne) ; 13: 1086276, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36686449

RESUMO

Leydig cells (Lc) reside in the interstitial compartment of the testis and are the target of Luteinising hormone (LH) for Testosterone (T) production, thus critically regulates male fertility. Classical histological studies have identified two morphologically different populations of Lc during testicular development [fetal (FLc) and adult (ALc)]. Recent progress in ex vivo cell/organ culture, genome-wide analysis, genetically manipulated mouse models, lineage tracing, and single-cell RNA-seq experiments have revealed the diverse cellular origins with differential transcriptomic and distinct steroidogenic outputs of these populations. FLc originates from both coelomic epithelium and notch-active Nestin-positive perivascular cells located at the gonad-mesonephros borders, and get specified as Nr5a1 (previously known as Ad4BP/SF-1) expressing cells by embryonic age (E) 12.5 days in fetal mouse testes. These cells produce androstenedione (precursor of T, due to lack of HSD17ß3 enzyme) and play critical a role in initial virilization and patterning of the male external genitalia. However, in neonatal testis, FLc undergoes massive regression/dedifferentiation and gradually gets replaced by T-producing ALc. Very recent studies suggest a small fraction (5-20%) of FLc still persists in adult testis. Both Nestin-positive perivascular cells and FLc are considered to be the progenitor populations for ALc. This minireview article summarizes the current understanding of Lc development in fetal and adult testes highlighting their common or diverse cellular (progenitor/stem) origins with respective functional significance in both rodents and primates. (227 words).


Assuntos
Células Intersticiais do Testículo , Testículo , Camundongos , Animais , Masculino , Nestina , Diferenciação Celular , Testosterona , Primatas
15.
Front Cell Dev Biol ; 10: 833759, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372365

RESUMO

Germ cells (Gc) propagate the genetic information to subsequent generations. Diploid (2n) Gc get transformed to specialized haploid (n) gametes by mitotic and meiotic divisions in adult gonads. Retinoic acid (RA), an active derivative of vitamin A (retinol), plays a critical role in organ morphogenesis and regulates the meiotic onset in developing Gc. Unlike ovaries, fetal testes express an RA-degrading enzyme CYP26B1, and thereby, male Gc fail to enter into meiosis and instead get arrested at G0/G1 stage, termed as gonocytes/pro-spermatogonia by embryonic (E) 13.5 days. These gonocytes are transformed into spermatogonial stem/progenitor cells after birth (1-3 days of neonatal age). During post-natal testicular maturation, the differentiating spermatogonia enter into the meiotic prophase under the influence RA, independent of gonadotropic (both FSH and LH) support. The first pulse of RA ensures the transition of undifferentiated type A spermatogonia to differentiated A1 spermatogonia and upregulates STRA8 expression in Gc. Whereas, the second pulse of RA induces the meiotic prophase by augmenting MEIOSIN expression in differentiated spermatogonia B. This opinion article briefly reviews our current understanding on the RA-driven spermatogonial differentiation in murine testes.

16.
Sci Rep ; 9(1): 11458, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31391487

RESUMO

An alarming decline in sperm count of men from several countries has become a major concern for the world community. Hormones act on testicular Sertoli cells (Sc) to regulate male fertility by governing the division and differentiation of germ cells (Gc). However, there is a limited knowledge about Sc specific gene(s) regulating the spermatogenic output of the testis. Sclerostin domain-containing 1 protein (Sostdc1) is a dual BMP/Wnt regulator is predominantly expressed in the Sc of infant testes which hardly show any sign of spermatogenesis. In order to investigate the role of Sostdc1 in spermatogenic regulation, we have generated transgenic (Tg) rats which induced persistent expression of Sostdc1 in mature Sc causing reduced sperm counts. Although Sc specific Sostdc1 did not affect the function of either Sc or Leydig cells (Lc) in the adult testis of Tg rat, we observed a selective augmentation of the BMP target genes via activated phospho smad 1/5/8 signaling in Gc leading to apoptosis. Here, for the first time, we have demonstrated that Sostdc1 is a negative regulator of spermatogenesis, and provided substantial evidence that down regulation of Sostdc1 during puberty is critically essential for quantitatively and qualitatively normal spermatogenesis governing male fertility.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Oligospermia/patologia , Células de Sertoli/metabolismo , Espermatogênese/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/fisiologia , Biópsia , Proteínas Morfogenéticas Ósseas/metabolismo , Estudos de Casos e Controles , Conjuntos de Dados como Assunto , Regulação para Baixo/fisiologia , Células Intersticiais do Testículo/metabolismo , Masculino , Modelos Animais , Oligospermia/genética , Ratos , Ratos Transgênicos , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia , Testículo/citologia , Testículo/patologia , Análise Serial de Tecidos
17.
Mol Cell Endocrinol ; 482: 70-80, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30579957

RESUMO

The synergistic actions of Testosterone (T) and FSH via testicular Sertoli cells (Sc) regulate male fertility. We have previously reported that the actions of these hormones (T and FSH) in infant monkey testes are restricted only to the expansion of Sc and spermatogonial cells. The robust differentiation of male Germ cells (Gc) occurs after pubertal maturation of testis. The present study was aimed to investigate the molecular basis of the synergy between T and FSH action in pubertal primate (Macaca mulatta) Sc. Using primary Sc culture, we here have demonstrated that T (but not FSH) downregulated AMH and Inhibin-ß-B (INHBB) mRNAs in pubertal Sc. We also found that, prolonged stimulation of T in pubertal Sc significantly elevated the expression of genes involved in FSH signaling pathway like FSH-Receptor (FSHR), GNAS and RIC8B, and this was associated with a rise in cAMP production. T also augmented FSH induced expression of genes like SCF, GDNF, ABP and Transferrin (TF) in pubertal Sc. We therefore conclude that T acts in synergy with FSH signaling in pubertal Sc. Such a coordinated network of hormonal signaling in Sc may facilitate the timely onset of the first spermatogenic wave in pubertal primates and is responsible for quantitatively and qualitatively normal spermatogenesis.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Receptores do FSH/genética , Receptores do FSH/metabolismo , Células de Sertoli/citologia , Testosterona/farmacologia , Animais , Células Cultivadas , Subunidades beta de Inibinas/genética , Macaca mulatta , Masculino , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Maturidade Sexual , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
18.
PLoS One ; 13(1): e0191201, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29342173

RESUMO

Sertoli cells (Sc) are unique somatic cells of testis that are the target of both FSH and testosterone (T) and regulate spermatogenesis. Although Sc of neonatal rat testes are exposed to high levels of FSH and T, robust differentiation of spermatogonial cells becomes conspicuous only after 11-days of postnatal age. We have demonstrated earlier that a developmental switch in terms of hormonal responsiveness occurs in rat Sc at around 12 days of postnatal age during the rapid transition of spermatogonia A to B. Therefore, such "functional maturation" of Sc, during pubertal development becomes prerequisite for the onset of spermatogenesis. However, a conspicuous difference in robust hormone (both T and FSH) induced gene expression during the different phases of Sc maturation restricts our understanding about molecular events necessary for the spermatogenic onset and maintenance. Here, using microarray technology, we for the first time have compared the differential transcriptional profile of Sc isolated and cultured from immature (5 days old), maturing (12 days old) and mature (60 days old) rat testes. Our data revealed that immature Sc express genes involved in cellular growth, metabolism, chemokines, cell division, MAPK and Wnt pathways, while mature Sc are more specialized expressing genes involved in glucose metabolism, phagocytosis, insulin signaling and cytoskeleton structuring. Taken together, this differential transcriptome data provide an important resource to reveal the molecular network of Sc maturation which is necessary to govern male germ cell differentiation, hence, will improve our current understanding of the etiology of some forms of idiopathic male infertility.


Assuntos
Células de Sertoli/metabolismo , Testículo/crescimento & desenvolvimento , Testículo/metabolismo , Animais , Apoptose/genética , Diferenciação Celular/efeitos dos fármacos , Quimiocinas/genética , Citocinas/genética , Citoesqueleto/genética , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/farmacologia , Perfilação da Expressão Gênica , Substâncias de Crescimento/genética , Sistema de Sinalização das MAP Quinases/genética , Masculino , Proteínas Monoméricas de Ligação ao GTP/genética , Análise de Sequência com Séries de Oligonucleotídeos , Fagocitose/genética , Ratos , Ratos Wistar , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Espermatogênese/fisiologia , Testículo/efeitos dos fármacos , Testosterona/metabolismo , Testosterona/farmacologia
20.
Endocrinology ; 156(3): 1143-55, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25549048

RESUMO

FSH acts via testicular Sertoli cells (Sc) bearing FSH receptor (FSH-R) for regulating male fertility. Despite an adult-like FSH milieu in infant boys and monkeys, spermatogenesis is not initiated until the onset of puberty. We used infant and pubertal monkey Sc to reveal the molecular basis underlying developmental differences of FSH-R signaling in them. Unlike pubertal Sc, increasing doses of FSH failed to augment cAMP production by infant Sc. The expression of Gαs subunit and Ric8b, which collectively activate adenylyl cyclase (AC) for augmenting cAMP production and gene transcription, were significantly low in infant Sc. However, forskolin, which acts directly on AC bypassing FSH-R, augmented cAMP production and gene transcription uniformly in both infant and pubertal Sc. FSH-induced Gαs mRNA expression was higher in pubertal Sc. However, Gαi-2 expression was down-regulated by FSH in pubertal Sc, unlike infant Sc. FSH failed, but forskolin or 8-Bromoadenosine 3',5'-cyclic monophosphate treatment to infant Sc significantly augmented the expression of transferrin, androgen binding protein, inhibin-ß-B, stem cell factor, and glial-derived neurotropic factor, which are usually up-regulated by FSH in pubertal Sc during spermatogenic onset. This suggested that lack of FSH mediated down-regulation of Gαi-2 expression and limited expression of Gαs subunit as well as Ric8b may underlie limited FSH responsiveness of Sc during infancy. This study also divulged that intracellular signaling events downstream of FSH-R are in place and can be activated exogenously in infant Sc. Additionally, this information may help in the proper diagnosis and treatment of infertile individuals having abnormal G protein-coupled FSH-R.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Macaca mulatta/crescimento & desenvolvimento , Macaca mulatta/metabolismo , Células de Sertoli/fisiologia , Animais , AMP Cíclico/genética , AMP Cíclico/metabolismo , Hormônio Foliculoestimulante/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Masculino , Ligação Proteica , Maturidade Sexual , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa