GnRH neurons

GnRH 神经元
  • 文章类型: Journal Article
    丙烯酰胺(AA)在环境和饮食中受到广泛污染。然而,AA和性激素的关系很少被调查,尤其是青少年,对性激素破坏特别敏感的时期。在这项研究中,在2013-2016年国家健康和营养调查(NHANES)共3268名受试者中,采用调查加权多元线性回归模型确定AAHb生物标志物[HbAA和缩水甘油胺(HbGA)]与性激素[总睾酮(TT)和雌二醇(E2)]之间的关联.此外,用AA治疗成年和青春期小鼠,以评估AA对性激素的影响并探索潜在的机制。在所有科目中,仅在年轻人(6-19岁)中发现了HbGA和性激素的显着阴性模式,男性TT的最低β为-0.53(95%CI:-0.80至-0.26),女性E2的最低β为-0.58(95%CI:-0.93至-0.23)。分层分析进一步显示,青少年中HbGA与性激素之间存在显着负相关,男性TT的最低β为-0.58(95%CI:-1.02至-0.14),女性E2的最低β为-0.54(95%CI:-1.03至-0.04),而儿童或晚期青少年之间没有显着差异。在老鼠身上,在AA处理的青春期小鼠中,TT和E2的水平显著降低,但在成年小鼠中没有降低.AA扰乱了下丘脑-垂体-性腺(HPG)轴中基因的表达,诱导下丘脑产生的促性腺激素释放激素(GnRH)神经元凋亡,并降低青春期小鼠的血清和下丘脑GnRH水平。我们的研究表明,在青春期,AA可以通过损伤GnRH神经元和破坏HPG轴降低TT和E2水平。表现为青少年严重的内分泌干扰。我们的发现加强了青春期是AA引起的性激素破坏的脆弱阶段的观点。
    Acrylamide (AA) is widely contaminated in environment and diet. However, the association of AA and sex hormones has rarely been investigated, especially in adolescents, a period of particular susceptibility to sex hormone disruption. In this study, survey-weighted multivariate linear regression models were conducted to determine the association between AA Hb biomarkers [HbAA and glycidamide (HbGA)] and sex hormones [total testosterone (TT) and estradiol (E2)] in a total of 3268 subjects from National Health and Nutrition Examination Survey (NHANES) 2013-2016 waves. Additionally, adult and pubertal mice were treated with AA to assess the effect of AA on sex hormones and to explore the potential mechanisms. Among all the subjects, significant negative patterns for HbGA and sex hormones were identified only in youths (6-19 years old), with the lowest β being - 0.53 (95% CI: -0.80 to -0.26) for TT in males and - 0.58 (95% CI: -0.93 to -0.23) for E2 in females. Stratified analysis further revealed significant negative associations between HbGA and sex hormones in adolescents, with the lowest β being - 0.58 (95% CI: -1.02 to -0.14) for TT in males and - 0.54 (95% CI: -1.03 to -0.04) for E2 in females, while there were no significant differences between children or late adolescents. In mice, the levels of TT and E2 were dramatically reduced in AA-treated pubertal mice but not in adult mice. AA disturbed the expression of genes in the hypothalamic-pituitary-gonadal (HPG) axis, induced apoptosis of hypothalamus-produced gonadotropin-releasing hormone (GnRH) neurons in the hypothalamus and reduced serum and hypothalamic GnRH levels in pubertal mice. Our study indicates AA could reduce TT and E2 levels by injuring GnRH neurons and disrupting the HPG axis in puberty, which manifested as severe endocrine disruption on adolescents. Our findings reinforce the idea that adolescence is a vulnerable stage in AA-induced sex hormone disruption.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    本研究旨在从下丘脑-垂体-睾丸轴的角度评估味精对小鼠睾丸精子发生的影响,以及这种破坏作用是否随时间减轻。
    将新生小鼠随机分为味精(MSG)组和对照组,出生后在肩胛骨间区域以下,用10微升味精递送4毫克/克(体重),或等量的0.9%盐水。涉及血液的样本,收集小鼠的大脑和睾丸,并在青春期60天和成年期90天进行测量。
    结果表明,GnRH神经纤维的荧光强度,卵泡刺激素(FSH)的水平,黄体生成素(LH),和生殖系统中的睾丸激素,睾丸切片中精母细胞和精子的数量,身体长度,体重,睾丸重量,味精组(MSG60组)和MSG90组60日龄小鼠睾丸指数均低于正常对照组(NC60组)60日龄小鼠睾丸指数(p<0.05),但睾丸切片凋亡细胞数高于NC60组(p<0.05)。味精组(MSG90组)90日龄小鼠与MSG60组比较,除了体重和睾丸重量增加(p<0.05),上述其他参数无显著差异(p>0.05)。
    味精通过损伤GnRH神经元对雄性小鼠产生生殖毒性,这种生殖毒性不能随时间自发缓解。观察到的组织学变化支持了这些发现。
    UNASSIGNED: The present study aims to evaluate the effect of monosodium glutamate on testicular spermatogenesis in mice from the perspective of the hypothalamic-pituitary-testicular axis and whether this destructive effect is alleviated with time.
    UNASSIGNED: Neonatal mice were randomly divided into a monosodium glutamate (MSG) group and a control group, just below the interscapular region after birth with 10 µL MSG to deliver 4 mg/g (body mass), or with equivalent volumes of 0.9% saline. Samples which involved blood, brains and testicles of mice were collected and measured at puberty at 60 days and adulthood at 90 days.
    UNASSIGNED: The results show that the fluorescence intensity of GnRH nerve fibers, the levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), and testosterone (T) hormones in the reproductive system, the number of spermatocytes and spermatozoa in testicular sections, the body length, body weight, testicular weight, and testicular index in the 60-day-old mice in monosodium glutamate group (MSG60 group) and the MSG90 group were lower than those in the 60-day-old mice in normal control group (NC60 group) (p < 0.05), but the number of apoptotic cells in the testicular section was higher than in the NC60 group (p < 0.05). When the 90-day-old mice in monosodium glutamate group (MSG90 group) was compared with the MSG60 group, except for body weight and testicular weight increase (p < 0.05), there is no significant difference in the other parameters mentioned above (p > 0.05).
    UNASSIGNED: Monosodium glutamate can cause reproductive toxicity to male mice by damaging GnRH neurons, and this reproductive toxicity cannot be relieved spontaneously over time. These findings are supported by observed histological changes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Microcystin-leucine arginine (MC-LR) is a kind of toxin produced by cyanobacterial, resulting in decrease of testosterone levels in serum and leading to impaired spermatogenesis. Gonadotropin-releasing hormone (GnRH) neurons play crucial roles in the regulation of testosterone release. Meanwhile, it has been demonstrated that MC-LR is capable of entering the GnRH neurons and inducing apoptosis. Nevertheless, the molecular mechanism of MC-LR induced apoptosis of GnRH neurons remains elusive. In present study, we found that MC-LR inhibited the cell viability of GT1-7 cells. In addition, we discovered apoptosis of GnRH neurons and GT1-7 cells treated with MC-LR. And increased intracellular ROS production and the release of intracellular Ca2+ were all observed following exposure to MC-LR. Furthermore, we also found the endoplasmic reticulum stress (ERs) and autophagy were activated by MC-LR. Additionally, pretreatment of the ERs inhibitor (4-Phenyl butyric acid) reduced the apoptotic rate of GT1-7 cells comparing with MC-LR exposure alone. Comparing with MC-LR treatment alone, apoptotic cell death was increased by pretreatment of GT1-7 cells with an autophagy inhibitor (3-methyladenine). Together, our data implicated that the treatment of MC-LR induced the apoptosis of GnRH neurons by activating the ERs resulting in a decrease of serum testosterone level in mice. Autophagy is a protective cellular process which was activated by ER stress and thus protected cells from apoptosis upon MC-LR exposure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Microcystin-leucine arginine (MC-LR) enters into gonadotropin-releasing hormone (GnRH) neurons and induces decline of serum GnRH levels resulting in male reproductive toxicity via hypothalamic-pituitary-testis axis. The organic anion transporting polypeptide 1a5 (Oatp1a5) is a critical transporter for the uptake of MC-LR by GnRH neurons. However, the underlying molecular mechanisms of the transport process are still elusive. In this study, we found that the transmembrane domains 2, 8, and 9 played important roles in transporting function of Oatp1a5. In addition, our data demonstrated that N-linked glycosylation was involved in the transport of MC-LR by Oatp1a5. Moreover, we showed that N-linked glycosylation sites Asn483 and Asn492 were vital for the transport function of Oatp1a5. In summary, the study furthered our understanding of mechanisms that the uptake of MC-LR by GnRH neurons and laid a theoretical foundation for preventing MC-LR from injuring male reproductive health.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    We previously reported Microcystin-LR (MC-LR) could enter the hypothalamus, reduce the expression of gonadotropin-releasing hormone (GnRH), and induce male reproductive barriers. However, the molecular mechanisms underlying in the hypothalamus have not been elucidated in detail. In this study, we further showed that MC-LR inhibited the synthesis of GnRH in GnRH neurons via activating protein kinase a (PKA), cAMP-response element binding protein (Creb), protein kinase c (PKC), nuclear factor kappa B (NF-κB), extracellular regulated protein kinases (Erk) and P38 protein, and thus resulted in the change of activity of transcriptional enhancers or suppressors such as Oct-1, Otx-2, Pbx1a, Dlx-2, c-Jun and c-Fos. Following exposure, MC-LR-treated mice exhibited decreased GnRH level. Our data demonstrated that MC-LR can stimulate intracellular Ca2+ and cAMP to activate PKC, PKA and MAPK signaling pathways in GnRH neurons, and then inhibit Pbx1a, Oct-1, Dlx-2, Otx-2 and upregulate c-Jun and c-Fos to initiate the transcription of GnRH, which provides novel insights to explore the mechanism associated with MC-LR-induced male reproductive barriers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号