Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 689
Filtrar
1.
Environ Sci Pollut Res Int ; 29(35): 52665-52674, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35267162

RESUMO

Biological effect of an individual nonylphenol (NP) isomer extremely relies upon the side chain structure. This research was designed to evaluate the impact of NP isomer, 4-[1-ethyl-1-methylhexy]-phenol (NP65), on Sertoli cells in vitro. Sertoli TM4 cells were exposed to various concentration (0, 0.1, 1, 10, or 20 µM) of NP65 for 24 h, and the outcomes indicated that treatment of NP65 induced reactive oxygen species (ROS) generation, oxidative stress, and apoptosis for Sertoli TM4 cells. In addition, it was found that NP65 exposure affected homeostasis of Ca2+ in Sertoli TM4 cells by increasing cytoplasm [Ca2+]i, inhibiting Ca2+-ATPase activity and decreasing cyclic adenosine monophosphate (cAMP) concentration. Pretreatment with ROS scavenger, N-acetylcysteine (NAC), attenuated NP65-induced oxidative stress as well as apoptosis for TM4 cells. Furthermore, NAC blocked NP65-induced disorders of Ca2+ homeostasis by attenuating the growth of intracellular [Ca2+]i and the inhibition of Ca2+-ATPase and cAMP activities. Thus, we have demonstrated that NP65 induced apoptosis as well as acted as a potent inhibitor of Ca2+-ATPase activity and resulted in disorder of Ca2+ homeostasis in Sertoli TM4 cells; ROS participated in the process. Our results supported the view that oxidative stress acted an essential role within the development of apoptosis and Ca2+ overload in TM4 cells as a consequence of NP65 stimulation.


Assuntos
Apoptose , Homeostase , Fenóis , Células de Sertoli , Acetilcisteína/farmacologia , Adenosina Trifosfatases/metabolismo , Humanos , Masculino , Fenóis/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Células de Sertoli/citologia , Células de Sertoli/efeitos dos fármacos
2.
FEBS J ; 289(10): 2809-2827, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34919331

RESUMO

Phagocytic clearance of apoptotic germ cells (GCs), as well as residual bodies released from developing spermatids, is critical for Sertoli cells (SCs) to maintain inner environment homeostasis within the testis. However, the molecular mechanisms controlling the phagocytosis are ill defined. Here, we identify a new role for alpha-enolase (ENO1), a key enzyme during glycolysis, as a molecule that facilitates testicular phagocytosis via transactivation of the engulfment and cell motility 1 (Elmo1) gene. Using immunohistochemistry and double-labeling immunofluorescence, ENO1 was observed to be expressed exclusively in the nuclei of SCs and its expression correlated with the completion of SC differentiation. By incubating TM4 cells with different pharmacological inhibitors and establishing TM4Tnfr1-/- cells, we demonstrated that SC-specific expression of ENO1 was under a delicate paracrine control from apoptotic GCs. In turn, persistent blockade of ENO1 expression by a validated small interfering RNA protocol resulted in the disturbance of spermatogenesis and impairment of male fertility. Furthermore, using ChIP, electrophoretic mobility shift and luciferase reporter assays, we showed that, in the presence of apoptotic GCs, ENO1 binds to the distal region of the Elmo1 promoter and facilitates transactivation of the Elmo1 gene. In agreement, overexpression of ELMO1 ameliorated ENO1 deficiency-induced impairment of phagocytosis in TM4 cells. These data reveal a novel role for SC-specific expression of ENO1 in regulating phagocytosis in testis, identify tumor necrosis factor-α and ELMO1 as critical upstream and downstream factors in mediating ENO1 action, and have important implications for our understanding of paracrine control of SC function by adjacent GCs.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Fosfopiruvato Hidratase , Células de Sertoli , Testículo , Fator de Necrose Tumoral alfa , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Movimento Celular , Expressão Gênica , Masculino , Camundongos , Fagocitose/genética , Fosfopiruvato Hidratase/metabolismo , Células de Sertoli/citologia , Testículo/citologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
J Clin Invest ; 131(23)2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34850745

RESUMO

Large-cell calcifying Sertoli cell tumors (LCCSCTs) are among the most frequent lesions occurring in male Carney complex (CNC) patients. Although they constitute a key diagnostic criterion for this rare multiple neoplasia syndrome resulting from inactivating mutations of the tumor suppressor PRKAR1A, leading to unrepressed PKA activity, LCCSCT pathogenesis and origin remain elusive. Mouse models targeting Prkar1a inactivation in all somatic populations or separately in each cell type were generated to decipher the molecular and paracrine networks involved in the induction of CNC testis lesions. We demonstrate that the Prkar1a mutation was required in both stromal and Sertoli cells for the occurrence of LCCSCTs. Integrative analyses comparing transcriptomic, immunohistological data and phenotype of mutant mouse combinations led to the understanding of human LCCSCT pathogenesis and demonstrated PKA-induced paracrine molecular circuits in which the aberrant WNT4 signal production is a limiting step in shaping intratubular lesions and tumor expansion both in a mouse model and in human CNC testes.


Assuntos
Complexo de Carney/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células de Sertoli/citologia , Neoplasias Testiculares/metabolismo , Proteína Wnt4/metabolismo , Animais , Apoptose , Complexo de Carney/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Genes Supressores de Tumor , Humanos , Masculino , Camundongos , Camundongos Knockout , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Comunicação Parácrina , Fenótipo , Pigmentação , Túbulos Seminíferos/metabolismo , Testículo/metabolismo , Transcriptoma
4.
Stem Cell Reports ; 16(11): 2798-2812, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34653405

RESUMO

We propose a new concept that human somatic cells can be converted to become male germline stem cells by the defined factors. Here, we demonstrated that the overexpression of DAZL, DAZ2, and BOULE could directly reprogram human Sertoli cells into cells with the characteristics of human spermatogonial stem cells (SSCs), as shown by their similar transcriptomes and proteomics with human SSCs. Significantly, human SSCs derived from human Sertoli cells colonized and proliferated in vivo, and they could differentiate into spermatocytes and haploid spermatids in vitro. Human Sertoli cell-derived SSCs excluded Y chromosome microdeletions and assumed normal chromosomes. Collectively, human somatic cells could be converted directly to human SSCs with the self-renewal and differentiation potentials and high safety. This study is of unusual significance, because it provides an effective approach for reprogramming human somatic cells into male germ cells and offers invaluable male gametes for treating male infertility.


Assuntos
Diferenciação Celular/genética , Autorrenovação Celular/genética , Reprogramação Celular/genética , Proteínas de Ligação a RNA/genética , Células de Sertoli/metabolismo , Espermatogônias/metabolismo , Animais , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Haploidia , Humanos , Masculino , Camundongos Nus , Proteômica/métodos , Proteínas de Ligação a RNA/metabolismo , Células de Sertoli/citologia , Espermátides/citologia , Espermátides/metabolismo , Espermatogônias/citologia , Transplante de Células-Tronco/métodos , Transplante Heterólogo
5.
Theriogenology ; 175: 7-22, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34481229

RESUMO

Metformin is a commonly used for treating type 2 diabetes and it acts on a variety of organs including the male reproductive system. 17ß-estradiol plays an important role in Sertoli cell (SC) proliferation which determines the germ cell development and spermatogenesis. The aim of this study is to investigate the effect of metformin on immature chicken SC proliferation and the potential mechanisms by which 17ß-estradiol regulate this process. Results showed that metformin significantly inhibited SC proliferation, whereas 17ß-estradiol weakened the inhibitory effects of metformin on SC viability, cell growth, and cell cycle progression. SC proliferation-inhibiting effect of metformin exposure was regulated by decreasing adenosine triphosphate level and respiratory enzyme activity in the mitochondria; this process was possibly mediated by the adenosine monophosphate-activated protein kinase (AMPK)/tuberous sclerosis complex 2 (TSC2)/mammalian target of rapamycin (mTOR) signaling pathway, which was regulated by the down-expressed miR-1764 and by the decreased antioxidant enzyme activity and excessive reactive oxygen species generation. In addition, SCs transfected with the miR-1764 agomir led to an improvement of proliferation capacity through down-regulating AMPKα2 level, which further decreased TSC2 expression and induced mTOR activation. However, the anti-proliferative effect of miR-1764 antagomir can be alleviated by 17ß-estradiol treatment via the up-expression of miR-1764 in transfected SCs. Our findings suggest appropriate dose of exogenous 17ß-estradiol treatment can ameliorate the inhibitory effect of metformin on SC proliferation via the regulation of AMPK/TSC2/mTOR signaling pathway, this might reduce the risk of poor male fertility caused by the abuse of anti-diabetic agents.


Assuntos
Estradiol , Metformina , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proliferação de Células , Galinhas , Estradiol/farmacologia , Masculino , Metformina/farmacologia , Células de Sertoli/citologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa
6.
PLoS Genet ; 17(8): e1009688, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34351902

RESUMO

Autophagy degrades unnecessary proteins or damaged organelles to maintain cellular function. Therefore, autophagy has a preventive role against various diseases including hepatic disorders, neurodegenerative diseases, and cancer. Although autophagy in germ cells or Sertoli cells is known to be required for spermatogenesis and male fertility, it remains poorly understood how autophagy participates in spermatogenesis. We found that systemic knockout mice of Rubicon, a negative regulator of autophagy, exhibited a substantial reduction in testicular weight, spermatogenesis, and male fertility, associated with upregulation of autophagy. Rubicon-null mice also had lower levels of mRNAs of Sertoli cell-related genes in testis. Importantly, Rubicon knockout in Sertoli cells, but not in germ cells, caused a defect in spermatogenesis and germline stem cell maintenance in mice, indicating a critical role of Rubicon in Sertoli cells. In mechanistic terms, genetic loss of Rubicon promoted autophagic degradation of GATA4, a transcription factor that is essential for Sertoli cell function. Furthermore, androgen antagonists caused a significant decrease in the levels of Rubicon and GATA4 in testis, accompanied by elevated autophagy. Collectively, we propose that Rubicon promotes Sertoli cell function by preventing autophagic degradation of GATA4, and that this mechanism could be regulated by androgens.


Assuntos
Fator de Transcrição GATA4/metabolismo , Técnicas de Inativação de Genes/métodos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Células de Sertoli/fisiologia , Animais , Autofagia , Linhagem Celular , Fertilidade , Humanos , Masculino , Camundongos , Proteólise , Células de Sertoli/citologia , Análise de Célula Única , Espermatogênese , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
7.
Front Endocrinol (Lausanne) ; 12: 648141, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34367061

RESUMO

Sustaining and maintaining the intricate process of spermatogenesis is liable upon hormones and growth factors acting through endocrine and paracrine pathways. The Sertoli cells (SCs) are the major somatic cells present in the seminiferous tubules and are considered to be the main regulators of spermatogenesis. As each Sertoli cell supports a specific number of germ cells, thus, the final number of Sertoli cells determines the sperm production capacity. Similarly, sex hormones are also major regulators of spermatogenesis and they can determine the proliferation of Sertoli cells. In the present review, we have critically and comprehensively discussed the role of sex hormones and some other factors that are involved in Sertoli cell proliferation, differentiation and maturation. Furthermore, we have also presented a model of Sertoli cell development based upon the recent advancement in the field of reproduction. Hence, our review article provides a general overview regarding the sex hormonal pathways governing Sertoli cell proliferation and development.


Assuntos
Hormônios Esteroides Gonadais/sangue , Células de Sertoli/citologia , Espermatogênese/fisiologia , Testículo/metabolismo , Ativinas/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Citocinas/metabolismo , Estrogênios/metabolismo , Fertilidade , Hormônio Foliculoestimulante/metabolismo , Humanos , Inibinas/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Progesterona/metabolismo , Prolactina/metabolismo , Ratos , Receptor de Insulina/metabolismo , Transdução de Sinais , Testosterona/metabolismo , Hormônios Tireóideos/metabolismo , Tretinoína/metabolismo , Proteínas Wnt/metabolismo
8.
Cells ; 10(6)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34207717

RESUMO

Timely and efficient elimination of apoptotic substrates, continuously produced during one's lifespan, is a vital need for all tissues of the body. This task is achieved by cells endowed with phagocytic activity. In blood-separated tissues such as the retina, the testis and the ovaries, the resident cells of epithelial origin as retinal pigmented epithelial cells (RPE), testis Sertoli cells and ovarian granulosa cells (GC) provide phagocytic cleaning of apoptotic cells and cell membranes. Disruption of this process leads to functional ablation as blindness in the retina and compromised fertility in males and females. To ensure the efficient elimination of apoptotic substrates, RPE, Sertoli cells and GC combine various mechanisms allowing maintenance of tissue homeostasis and avoiding acute inflammation, tissue disorganization and functional ablation. In tight cooperation with other phagocytosis receptors, MERTK-a member of the TAM family of receptor tyrosine kinases (RTK)-plays a pivotal role in apoptotic substrate cleaning from the retina, the testis and the ovaries through unconventional autophagy-assisted phagocytosis process LAP (LC3-associated phagocytosis). In this review, we focus on the interplay between TAM RTKs, autophagy-related proteins, LAP, and Toll-like receptors (TLR), as well as the regulatory mechanisms allowing these components to sustain tissue homeostasis and prevent functional ablation of the retina, the testis and the ovaries.


Assuntos
Células da Granulosa , Retina , Células de Sertoli , c-Mer Tirosina Quinase/metabolismo , Animais , Autofagia , Feminino , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Células da Granulosa/patologia , Humanos , Masculino , Fagocitose , Retina/citologia , Retina/metabolismo , Retina/patologia , Células de Sertoli/citologia , Células de Sertoli/metabolismo
9.
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
10.
Front Endocrinol (Lausanne) ; 12: 694796, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093450

RESUMO

The incidence of cancer in pre-pubertal boys has significantly increased and, it has been recognized that the gonado-toxic effect of the cancer treatments may lead to infertility. Here, we have evaluated the effects on porcine neonatal Sertoli cells (SCs) of three commonly used chemotherapy drugs; cisplatin, 4-Hydroperoxycyclophosphamide and doxorubicin. All three drugs induced a statistical reduction of 5-hydroxymethylcytosine in comparison with the control group, performed by Immunofluorescence Analysis. The gene and protein expression levels of GDNF, were significantly down-regulated after treatment to all three chemotherapy drugs comparison with the control group. Specifically, differences in the mRNA levels of GDNF were: 0,8200 ± 0,0440, 0,6400 ± 0,0140, 0,4400 ± 0,0130 fold change at 0.33, 1.66, and 3.33µM cisplatin concentrations, respectively (**p < 0.01 at 0.33 and 1.66 µM vs SCs and ***p < 0.001 at 3.33µM vs SCs); 0,6000 ± 0,0340, 0,4200 ± 0,0130 fold change at 50 and 100 µM of 4-Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7000 ± 0,0340, 0,6200 ± 0,0240, 0,4000 ± 0,0230 fold change at 0.1, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 µM vs SCs and ***p < 0.001 at 1 µM vs SCs). Differences in the protein expression levels of GDNF were: 0,7400 ± 0,0340, 0,2000 ± 0,0240, 0,0400 ± 0,0230 A.U. at 0.33, 1.66, and 3.33µM cisplatin concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,7300 ± 0,0340, 0,4000 ± 0,0130 A.U. at 50 and 100 µM of 4- Hydroperoxycyclophosphamide concentrations, respectively (**p < 0.01 at both these concentrations vs SCs); 0,6200 ± 0,0340, 0,4000 ± 0,0240, 0,3800 ± 0,0230 A.U. at 0.l, 0.2 and 1 µM doxorubicin concentrations, respectively (**p < 0.01 at 0.1 and 0.2 µM vs SCs and ***p < 0.001 at 1 µM vs SCs). Furthermore, we have demonstrated the protective effect of eicosapentaenoic acid on SCs only at the highest concentration of cisplatin, resulting in an increase in both gene and protein expression levels of GDNF (1,3400 ± 0,0280 fold change; **p < 0.01 vs SCs); and of AMH and inhibin B that were significantly recovered with values comparable to the control group. Results from this study, offers the opportunity to develop future therapeutic strategies for male fertility management, especially in pre-pubertal boys.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Ácido Eicosapentaenoico/farmacologia , Preservação da Fertilidade/métodos , Células de Sertoli/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Sobreviventes de Câncer , Células Cultivadas , Criança , Cisplatino/efeitos adversos , Ácido Eicosapentaenoico/uso terapêutico , Fertilidade/efeitos dos fármacos , Gônadas/efeitos dos fármacos , Gônadas/patologia , Humanos , Masculino , Células de Sertoli/citologia , Células de Sertoli/fisiologia , Suínos
11.
J Assist Reprod Genet ; 38(9): 2481-2490, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34050447

RESUMO

BACKGROUND: Mammalian spermatogenesis is responsible for male fertility and is supported by the self-renewal and differentiation of spermatogonial stem cells (SSCs). Sertoli cells provide a supportive microenvironment for SSCs, in part by the production of stem cell factor (SCF), which is a potent regulator of spermatogonia proliferation and survival. METHODS: We investigated the novel role of ß-estradiol in modulating the proliferation and apoptosis of fetal SSCs via the regulation of SCF secretion in Sertoli cells isolated from human fetal testes. The proliferation of SSCs in the co-culture system was determined by colony formation and BrdU incorporation assays. TUNEL assay was used to measure SSC apoptosis in co-culture in response to treatment with control, ß-estradiol, or the combination of ß-estradiol and the estrogen receptor inhibitor ICI 182780. RESULTS: In the system with purified human fetal Sertoli cells (MIS+/c-Kit-/AP-), ß-estradiol upregulated the production of SCF in a dose- and time-dependent manner. In the co-culture system of primary human fetal SSCs (c-Kit+/SSEA-4+/Oct-4+/AP+) and Sertoli cells (MIS+), ß-estradiol markedly increased the proliferation of SSCs. Moreover, SSC apoptosis was significantly inhibited by ß-estradiol and was completely reversed by the combination of ß-estradiol and ICI 182780. CONCLUSION: Here we report, for the first time, that ß-estradiol can induce the increase of SCF expression in human fetal Sertoli cells and regulates the growth and survival of human fetal SSCs. These novel findings provide new perspectives on the current understanding of the role of estrogen in human spermatogenesis.


Assuntos
Diferenciação Celular , Estradiol/farmacologia , Feto/citologia , Células de Sertoli/citologia , Espermatogônias/citologia , Fator de Células-Tronco/metabolismo , Células-Tronco/citologia , Técnicas de Cocultura , Estrogênios/farmacologia , Feto/efeitos dos fármacos , Feto/metabolismo , Idade Gestacional , Humanos , Masculino , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Espermatogênese , Espermatogônias/efeitos dos fármacos , Espermatogônias/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
12.
Reprod Biol Endocrinol ; 19(1): 64, 2021 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-33902598

RESUMO

BACKGROUND: Fluorochloridone (FLC), a selective pyrrolidone herbicide, has been recognized as a potential endocrine disruptor and reported to induce male reproductive toxicity, but the underlying mechanism is unclear. The aim of this study was to investigate the mechanism of FLC-induced reproductive toxicity on male mice with particular emphasis on the role of autophagy in mice' TM4 Sertoli cells. METHODS: Adult C57BL/6 mice were divided into one control group (0.5% sodium carboxymethyl cellulose), and four FLC-treated groups (3,15,75,375 mg/kg). The animals (ten mice per group) received gavage for 28 days. After treatment, histological analysis, sperm parameters, the microstructure of autophagy and the expression of autophagy-associated proteins in testis were evaluated. Furthermore, to explore the autophagy mechanism, TM4 Sertoli cells were treated with FLC (0,40,80,160 µM) in vitro for 24 h. Cell activity and cytoskeletal changes were measured by MTT assay and F-actin immunofluorescence staining. The formation of autophagosome, accumulation of reactive oxygen species (ROS), expression of autophagy marker proteins (LC3, Beclin-1 and P62) and AKT-related pathway proteins (AKT, mTOR) were observed. The ROS scavenger N-acetylcysteine (NAC) and AKT agonist (SC79) were used to treat TM4 cells to observe the changes of AKT-mTOR pathway and autophagy. RESULTS: In vivo, it showed that FLC exposure caused testicular injuries, abnormality in epididymal sperm. Moreover, FLC increased the formation of autophagosomes, the accumulation of LC3II/LC3I, Beclin-1 and P62 protein, which is related to the degradation of autophagy. In vitro, FLC triggered TM4 cell autophagy by increasing the formation of autophagosomes and upregulating of LC3II/LC3I, Beclin-1 and P62 levels. In addition, FLC induced ROS production and inhibited the activities of AKT and mTOR kinases. The Inhibition of AKT/mTOR signaling pathways and the activation of autophagy induced by FLC could be efficiently reversed by pretreatment of NAC. Additionally, decreased autophagy and increased cell viability were observed in TM4 cells treated with SC79 and FLC, compared with FLC alone, indicating that FLC-induced autophagy may be pro-death. CONCLUSION: Taken together, our study provided the evidence that FLC promoted autophagy in TM4 Sertoli cells and that this process may involve ROS-mediated AKT/mTOR signaling pathways.


Assuntos
Autofagia/efeitos dos fármacos , Disruptores Endócrinos/farmacologia , Herbicidas/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Pirrolidinonas/farmacologia , Células de Sertoli/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Acetatos/farmacologia , Acetilcisteína/farmacologia , Animais , Autofagia/fisiologia , Benzopiranos/farmacologia , Forma Celular , Herbicidas/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pirrolidinonas/toxicidade , Distribuição Aleatória , Espécies Reativas de Oxigênio , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Espermatozoides/efeitos dos fármacos , Espermatozoides/ultraestrutura
13.
J Cell Mol Med ; 25(8): 3950-3962, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33608983

RESUMO

Self-renewal and differentiation of spermatogonial stem cell (SSC) are critical for male fertility and reproduction, both of which are highly regulated by testicular microenvironment. Exosomal miRNAs have emerged as new components in intercellular communication. However, their roles in the differentiation of SSC remain unclear. Here, we observed miR-486-5p enriched in Sertoli cell and Sertoli cell-derived exosomes. The exosomes mediate the transfer of miR-486-5p from Sertoli cells to SSCs. Exosomes release miR-486-5p, thus up-regulate expression of Stra8 (stimulated by retinoic acid 8) and promote differentiation of SSC. And PTEN was identified as a target of miR-486-5p. Overexpression of miR-486-5p in SSCs down-regulates PTEN expression, which up-regulates the expression of STRA8 and SYCP3, promotes SSCs differentiation. In addition, blocking the exosome-mediated transfer of miR-486-5p inhibits differentiation of SSC. Our findings demonstrate that miR-486-5p acts as a communication molecule between Sertoli cells and SSCs in modulating differentiation of SSCs. This provides a new insight on molecular mechanisms that regulates SSC differentiation and a basis for the diagnosis, treatment, and prevention of male infertility.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular , Exossomos/metabolismo , MicroRNAs/genética , PTEN Fosfo-Hidrolase/metabolismo , Células de Sertoli/citologia , Testículo/citologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células Cultivadas , Exossomos/genética , Regulação da Expressão Gênica , Masculino , Camundongos , PTEN Fosfo-Hidrolase/genética , Células de Sertoli/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Testículo/metabolismo
14.
Anat Rec (Hoboken) ; 304(5): 1119-1135, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33022119

RESUMO

We studied the male reproductive tract of individuals of different body sizes of Caecilia thompsoni to describe morphological characteristics in comparison to other Gymnophiona. The reproductive tract consists of paired testes segmented into chains of primary and secondary lobes, sperm ducts that empty to Wolffian ducts, the cloaca that receives the Wolffian ducts and possesses a phallodeum. Müllerian ducts are present and develop into paired glands that empty into the cloacal urodeum. Testicular secondary lobes contain lobules with cysts of the entire germinal cell line, whereas primary lobes, in the terminal ends of the chains, only have spermagonia, Sertoli cells, and connective tissue. The smallest individual examined (21 cm body length) was immature and only possessed a few testicular primary lobes. Once the individuals reach sexual maturity, the morphological characteristics are quite consistent at macroscopic and histological level among males of very different body sizes. The histological features of the Wolffian and Müllerian glands suggest a complementary secretory role between the two ducts. In the cloaca we found the propulsor muscle, venous sinuses, and blind sacs in the phallodeum, which differentiate C. thompsoni from other species of the genus. Despite these slight differences, the general morphological characteristics, both macroscopic and microscopic, of the reproductive tracts of adult males of C. thompsoni follow the pattern known for the reproductively active males of Gymnophiona.


Assuntos
Anfíbios/anatomia & histologia , Ductos Paramesonéfricos/anatomia & histologia , Células de Sertoli/citologia , Espermatogônias/citologia , Testículo/anatomia & histologia , Ductos Mesonéfricos/anatomia & histologia , Animais , Masculino
15.
Mol Cell Endocrinol ; 523: 111099, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271218

RESUMO

Porcine Sertoli cell number including number present at puberty is increased if testicular estradiol synthesis is reduced during the neonatal interval. Evaluating the changes in gene expression during the crucial interval of suppressed estradiol that leads to the increased Sertoli cell population will increase our understanding of Sertoli cell biology but this evaluation first required a more precise determination of the critical interval for treatment and timing of a detectable response. Previously, reduced testicular estrogens from 1 week of age were accompanied by increased Sertoli cell number at 6.5 weeks of age but the age at which Sertoli cell numbers were initially increased was unknown, one of the current objectives. Additional experiments were designed to further delineate the essential timing of treatment for the Sertoli cell response. Finally, changes in gene expression induced by the reduced estradiol synthesis were evaluated to elucidate molecular mechanisms. Experimental design typically consisted of one member of littermate pairs of boars treated with the aromatase inhibitor, letrozole, beginning at 1 week of age and the remaining member treated with canola oil vehicle. Weekly treatments continued through 5 weeks of age or tissue collection, whichever came first. Increases in Sertoli cell numbers were not detectable prior to 6.5 weeks of age and persistent treatment through 5 weeks of age was required to induce the increase in Sertoli cell numbers. This increase resulted from prolonging the first interval of Sertoli cell proliferation in the treated animals. Few genes exhibited dramatically altered transcription and similarities in pathway analysis or principal modified genes were quite limited in 2, 3, and 5-week-old boars. The critical timing and prolonged treatment required and the sequential changes in gene expression suggest a complex mechanism is involved in this model of increased proliferation of Sertoli cells.


Assuntos
Estradiol/biossíntese , Regulação da Expressão Gênica , Células de Sertoli/citologia , Testículo/metabolismo , Animais , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Letrozol/farmacologia , Masculino , Proteínas de Membrana/metabolismo , Anotação de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células de Sertoli/efeitos dos fármacos , Células de Sertoli/metabolismo , Suínos , Testículo/citologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
16.
Molecules ; 25(22)2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33187275

RESUMO

More than 90% of diabetic patients suffer from sexual dysfunction, including diminished sperm count, sperm motility, and sperm viability, and low testosterone levels. The effects of Momordica charantia (MC) were studied by estimating the blood levels of insulin, glucose, glycosylated hemoglobin (HbA1c), testosterone (TST), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) in diabetic rats treated with 250 and 500 mg/kg b.w. of the total extract. Testicular antioxidants, epididymal sperm characteristics, testicular histopathology, and lesion scoring were also investigated. Testicular mRNA expression of apoptosis-related markers such as antiapoptotic B-cell lymphoma-2 (Bcl-2) and proapoptotic Bcl-2-associated X protein (Bax) were evaluated by real-time PCR. Furthermore, caspase-3 protein expression was evaluated by immunohistochemistry. MC administration resulted in a significant reduction in blood glucose and HbA1c and marked elevation of serum levels of insulin, TST, and gonadotropins in diabetic rats. It induced a significant recovery of testicular antioxidant enzymes, improved histopathological changes of the testes, and decreased spermatogenic and Sertoli cell apoptosis. MC effectively inhibited testicular apoptosis, as evidenced by upregulation of Bcl-2 and downregulation of Bax and caspase-3. Moreover, reduction in apoptotic potential in MC-treated groups was confirmed by reduction in the Bax/Bcl-2 mRNA expression ratio.


Assuntos
Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/metabolismo , Momordica charantia/química , Extratos Vegetais/farmacologia , Espermatogênese/efeitos dos fármacos , Animais , Apoptose , Caspase 3/metabolismo , Cromatografia Líquida , Diabetes Mellitus Experimental/metabolismo , Fertilidade , Hormônio Foliculoestimulante/metabolismo , Hemoglobinas Glicadas/análise , Imuno-Histoquímica , Hormônio Luteinizante/farmacologia , Masculino , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Espectrometria de Massas por Ionização por Electrospray , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Testículo/metabolismo , Testosterona/metabolismo , Proteína X Associada a bcl-2/metabolismo
17.
J Vis Exp ; (164)2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-33104058

RESUMO

Spermatogenesis is a unique differentiation process that ultimately gives rise to one of the most distinct cell types of the body, the sperm. Differentiation of germ cells takes place in the cytoplasmic pockets of somatic Sertoli cells that host 4 to 5 generations of germ cells simultaneously and coordinate and synchronize their development. Therefore, the composition of germ cell types within a cross-section is constant, and these cell associations are also known as stages (I-XII) of the seminiferous epithelial cycle. Importantly, stages can also be identified from intact seminiferous tubules based on their differential light absorption/scatter characteristics revealed by transillumination, and the fact that the stages follow each other along the tubule in a numerical order. This article describes a transillumination-assisted microdissection method for the isolation of seminiferous tubule segments representing specific stages of mouse seminiferous epithelial cycle. The light absorption pattern of seminiferous tubules is first inspected under a dissection microscope, and then tubule segments representing specific stages are cut and used for downstream applications. Here we describe immunostaining protocols for stage-specific squash preparations and for intact tubule segments. This method allows a researcher to focus on biological events taking place at specific phases of spermatogenesis, thus providing a unique tool for developmental, toxicological, and cytological studies of spermatogenesis and underlying molecular mechanisms.


Assuntos
Células Epiteliais/citologia , Túbulos Seminíferos/citologia , Coloração e Rotulagem , Transiluminação , Acrossomo/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Núcleo Celular/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Microdissecção , Células de Sertoli/citologia , Espermatogênese , Espermatozoides/citologia
18.
Anim Reprod Sci ; 222: 106608, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33039822

RESUMO

Spermatogenesis is important for male fertility, but has not been well-studied in Opsariichthys bidens, an economically important freshwater fish in China. In this study, there was investigation of the cytological features of spermatogenesis in O. bidens using light microscopy, transmission electron microscopy, and immunofluorescence detection of microtubules. O. bidens has tubular testis. Spermatogenesis in O. bidens is of the cystic type, in which the spermatogenic cells develop into spermatozoa in cysts. There was asynchronous development of primary spermatocytes within a single cyst. Spermiogenesis was classified as Type I, which develops into a Type I aquasperm with an oval nucleus, a small and simple midpiece, a flagellum and no acrosome. There was a nuage in spermatogonia, spermatocytes, and spermatids in different developmental stages of spermatids which may have important functions in fish spermatogenesis. Furthermore, microtubule dynamics may be involved in spermatid reshaping, material transport, and polar distribution of organelles during spermiogenesis.


Assuntos
Cyprinidae/fisiologia , Espermatogênese/fisiologia , Espermatozoides/citologia , Testículo/citologia , Animais , Aquicultura , China , Citoesqueleto/fisiologia , Água Doce , Masculino , Meiose , Microscopia Eletrônica , Microtúbulos/ultraestrutura , Células de Sertoli/citologia , Células de Sertoli/ultraestrutura , Espermátides/citologia , Espermátides/ultraestrutura , Espermatócitos/citologia , Espermatócitos/ultraestrutura , Espermatogônias/citologia , Espermatogônias/ultraestrutura , Espermatozoides/ultraestrutura , Testículo/ultraestrutura
19.
Stem Cell Reports ; 15(4): 968-982, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33053361

RESUMO

Sertoli cells are the major component of the spermatogonial stem cell (SSC) niche; however, regulatory mechanisms in Sertoli cells that dictate SSC fate decisions remain largely unknown. Here we revealed features of the N6-methyladenosine (m6A) mRNA modification in Sertoli cells and demonstrated the functions of WTAP, the key subunit of the m6A methyltransferase complex in spermatogenesis. m6A-sequencing analysis identified 21,909 m6A sites from 15,365 putative m6A-enriched transcripts within 6,122 genes, including many Sertoli cell-specific genes. Conditional deletion of Wtap in Sertoli cells resulted in sterility and the progressive loss of the SSC population. RNA sequencing and ribosome nascent-chain complex-bound mRNA sequencing analyses suggested that alternative splicing events of transcripts encoding SSC niche factors were sharply altered and translation of these transcripts were severely dysregulated by Wtap deletion. Collectively, this study uncovers a novel regulatory mechanism of the SSC niche and provide insights into molecular interactions between stem cells and their cognate niches in mammals.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fatores de Processamento de RNA/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Espermatogônias/citologia , Nicho de Células-Tronco , Adenosina/análogos & derivados , Adenosina/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/patologia , Masculino , Camundongos Knockout , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espermatogônias/metabolismo , Nicho de Células-Tronco/genética , Transcrição Gênica
20.
FASEB J ; 34(11): 15164-15179, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32918760

RESUMO

Sertoli cells play vital roles in normal spermatogenesis, and microRNAs (miRNAs) participate in regulating Sertoli cell development. However, the functions and mechanisms of action of most identified miRNAs in porcine Sertoli cells remain largely unknown. Herein, we primarily explored the regulatory roles of miR-130a in immature porcine Sertoli cells using EdU-based high-content screening assay. The results demonstrated that 27 miRNAs have potential roles in the promotion of immature porcine Sertoli cell proliferation, and miR-130a was identified as a promising candidate. miR-130a promoted cell cycle progression and cell proliferation, whereas it impeded cell apoptosis in immature porcine Sertoli cells. It also contributed to Sertoli cell proliferation and testis development in vivo. A TMT-based proteomics approach revealed that miR-130a regulated the expression of 91 proteins and multiple pathways, including the TGF-ß and PI3K/AKT signaling. miR-130a did not directly target the 3'-UTR of SMAD5; however, it increased SMAD5 phosphorylation. Moreover, miR-130a enhanced TGF-ß signaling by activating SMAD5 protein, and TGF-ß signaling further activated the PI3K/AKT signaling pathway to promote cell proliferation and inhibit cell apoptosis in porcine immature Sertoli cells. Collectively, miR-130a promoted immature porcine Sertoli cell growth by activating SMAD5 through the TGF-ß-PI3K/AKT signaling pathway. This study, therefore, provides novel insights into the effects of miR-130a on porcine spermatogenesis through the regulation of immature Sertoli cell proliferation and apoptosis.


Assuntos
MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células de Sertoli/citologia , Proteína Smad5/metabolismo , Espermatogênese , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células , Masculino , Camundongos Endogâmicos ICR , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Células de Sertoli/metabolismo , Proteína Smad5/genética , Suínos , Fator de Crescimento Transformador beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA