Granulosa cells

颗粒细胞
  • 文章类型: Dataset
    颗粒细胞(GCs)在卵母细胞成熟中起着至关重要的作用。通过间隙连接和细胞外囊泡,它们介导微小RNA和信使RNA等分子的交换。不同的卵巢细胞类型表现出独特的基因表达谱,反映他们的专业功能和阶段。通过结合来自形成毛囊的各种细胞类型的RNA-seq数据,我们的目标是捕捉广泛的表达模式,提供对跨GC转录组调控的功能多样性和复杂性的见解。在这里,我们对公共数据库中存在的RNA测序数据集进行了综合生物信息学分析,具有独特和标准化的工作流程。,通过结合不同研究的数据,我们成功地提高了我们发现的稳健性和可靠性,并发现了新的基因,miRNA,以及与GCs功能和卵母细胞成熟相关的信号通路。此外,我们的研究结果为进一步的湿实验室研究GCs生物学及其对卵母细胞发育和能力的影响提供了宝贵的资源。
    Granulosa cells (GCs) play crucial roles in oocyte maturation. Through gap junctions and extracellular vesicles, they mediate the exchange of molecules such as microRNAs and messenger RNAs. Different ovarian cell types exhibit unique gene expression profiles, reflecting their specialized functions and stages. By combining RNA-seq data from various cell types forming the follicle, we aimed at capturing a wide range of expression patterns, offering insights into the functional diversity and complexity of the transcriptome regulation across GCs. Herein, we performed an integrated bioinformatics analysis of RNA sequencing datasets present in public databases, with a unique and standardized workflow., By combining the data from different studies, we successfully increased the robustness and reliability of our findings and discovered novel genes, miRNAs, and signaling pathways associated with GCs function and oocyte maturation. Moreover, our results provide a valuable resource for further wet-lab research on GCs biology and their impact on oocyte development and competence.
    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
    卵巢颗粒细胞(GC)对于卵泡发育至关重要。卵巢晚期糖基化终产物(AGEs)的积累与GCs功能障碍有关。α-硫辛酸(ALA)说明了不孕症相关疾病的治疗能力。因此,这项研究评估了ALA对AGEs诱导的GCs激素功能障碍的影响。
    该研究于2021年10月至2022年9月在医学遗传学系进行,设拉子医科大学。分离的GC(n=50)分为对照,人糖化白蛋白(HGA),HGA+ALA,和ALA治疗。通过qRT-PCR评估类固醇生成酶和AGE受体(RAGE)基因。使用ELISA和Western印迹评估类固醇激素和RAGE蛋白。使用GraphPadPrism软件(ver。9),P<0.05被认为是显著的。
    我们的发现表明,HGA治疗显着(P=0.0001)增加了RAGE(140.66%),明星(117.65%),3β-HSD(165.68%),和17β-HSD(122.15%)表达,而CYP19A1表达降低(68.37%)。RAGE蛋白水平(267.10%)在HGA处理的GC中也增加。还观察到雌二醇显着降低(59.66%),孕酮(30.40%)和总睾丸激素(158.24%)水平略有急剧升高。ALA治疗改善了HGA诱导的类固醇生成酶mRNA水平(P=0.001)和类固醇激素分泌(P=0.010)的变化。
    这项工作表明,ALA疗法可能会纠正由黄体化GC中的AGEs引起的激素功能障碍。这种效果可能是通过降低RAGE表达来实现的。临床研究需要了解AGEs和ALA如何在卵巢中相互作用,这可能导致更有针对性的卵巢功能障碍治疗。
    UNASSIGNED: Ovarian granulosa cells (GCs) are essential for follicular development. Ovarian advanced glycation end-products (AGEs) accumulation is related to GCs dysfunction. Alpha-lipoic acid (ALA) illustrates therapeutic capabilities for infertility-related disorders. Therefore, this study assessed the effects of ALA on AGEs-induced GCs hormonal dysfunction.
    UNASSIGNED: The study was conducted from October 2021 to September 2022 at the Department of Medical Genetics, Shiraz University of Medical Sciences. Isolated GCs (n=50) were divided into control, human glycated albumin (HGA), HGA+ALA, and ALA treatments. Steroidogenic enzymes and AGE receptor (RAGE) genes were assessed by qRT-PCR. Steroid hormones and RAGE protein were evaluated using ELISA and Western blotting. Data were analyzed using GraphPad Prism software (ver. 9), and P<0.05 was considered significant.
    UNASSIGNED: Our findings showed that HGA treatment significantly (P=0.0001) increased RAGE (by 140.66%), STAR (by 117.65%), 3β-HSD (by 165.68%), and 17β-HSD (by 122.15%) expression, while it decreased CYP19A1 (by 68.37%) expression. RAGE protein level (by 267.10%) was also increased in HGA-treated GCs. A significant decrease in estradiol (by 59.66%) and a slight and sharp elevation in progesterone (by 30.40%) and total testosterone (by 158.24%) levels was also observed. ALA treatment ameliorated the HGA-induced changes in steroidogenic enzyme mRNA levels (P=0.001) and steroid hormone secretion (P=0.010).
    UNASSIGNED: This work shows that ALA therapy likely corrects hormonal dysfunctions caused by AGEs in luteinized GCs. This effect is probably achieved by decreased RAGE expression. Clinical research is needed to understand how AGEs and ALA interact in the ovary, which might lead to a more targeted ovarian dysfunction therapy.
    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
    卵泡发育是指卵巢中的卵泡从初级阶段逐渐发育到成熟状态的过程,大多数初级卵泡不能正常发育,不形成致密的颗粒细胞层和细胞壁,被鉴定为闭锁性卵泡。颗粒细胞通过产生激素和提供支持来帮助卵泡发育,颗粒细胞与卵母细胞相互作用的干扰可能导致闭锁卵泡的形成。Ferroptosis,作为一种非凋亡的死亡形式,是由细胞积累致死水平的铁依赖性磷脂过氧化物引起的。将4至5mm的健康卵泡随机分为两组:对照组(DMSO)和治疗组(10uM的铁凋亡诱导因子erastin)。每组在三次重复培养24小时后进行测序。我们发现铁性凋亡与闭锁卵泡有关,并且用铁性凋亡诱导剂erastin对健康卵泡进行体外治疗产生了与闭锁卵泡相似的表型。总的来说,我们的研究阐明tRF-1:30-Gly-GCC-2参与了GCs的凋亡和铁凋亡。机械上,tRF-1:30-Gly-GCC-2通过抑制丝裂原活化蛋白激酶1(MAPK1)抑制颗粒细胞增殖并促进铁凋亡。tRF-1:30-Gly-GCC-2可能是改善卵巢功能障碍中闭锁卵泡发育的新分子靶标。总之,我们的研究为颗粒细胞功能障碍和卵泡闭锁的发病机制提供了新的视角。
    Follicle development refers to the process in which the follicles in the ovary gradually develop from the primary stage to a mature state, and most primary follicles fail to develop normally, without forming a dense granular cell layer and cell wall, which is identified as atretic follicles. Granulosa cells assist follicle development by producing hormones and providing support, and interference in the interaction between granulosa cells and oocytes may lead to the formation of atretic follicles. Ferroptosis, as a non-apoptotic form of death, is caused by cells accumulating lethal levels of iron-dependent phospholipid peroxides. Healthy follicles ranging from 4 to 5 mm were randomly divided into two groups: a control group (DMSO) and treatment group (10 uM of ferroptosis inducer erastin). Each group was sequenced after three repeated cultures for 24 h. We found that ferroptosis was associated with atretic follicles and that the in vitro treatment of healthy follicles with the ferroptosis inducer erastin produced a phenotype similar to that of atretic follicles. Overall, our study elucidates that tRF-1:30-Gly-GCC-2 is involved in the apoptosis and ferroptosis of GCs. Mechanistically, tRF-1:30-Gly-GCC-2 inhibits granulosa cell proliferation and promotes ferroptosis by inhibiting Mitogen-activated protein kinase 1 (MAPK1). tRF-1:30-Gly-GCC-2 may be a novel molecular target for improving the development of atretic follicles in ovarian dysfunction. In conclusion, our study provides a new perspective on the pathogenesis of granulosa cell dysfunction and follicular atresia.
    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
    背景:酪蛋白激酶1α(CK1α),在卵巢生殖细胞和体细胞中表达,参与小鼠卵母细胞的初始减数分裂和原始卵泡形成。本研究采用体外和体内实验,探讨CK1α在小鼠卵巢颗粒细胞雌激素合成中的作用及机制。
    方法:CK1α基因敲除(cKO)小鼠模型,特异性靶向卵巢颗粒细胞(GC),用于确定CK1α对体内雌激素合成的影响。通过免疫荧光分析和Westernblot测定体内和体外CK1α缺乏对GC的影响。转录组分析,差异表达基因和基因功能富集分析,和计算蛋白质-蛋白质对接,进一步用于评估CK1α途径。此外,用CK1α拮抗剂D4476治疗野生型雌性小鼠,以阐明CK1α在雌激素调节中的作用。
    结果:卵巢GCsCK1α缺乏损害雌性小鼠的生育力和超排卵;cKO雌性小鼠的平均产仔数和血清雌二醇(E2)水平分别降低了57.3%和87.4%。对照小鼠,分别。这种缺乏破坏了发情周期并增强了GCs的凋亡。我们观察到CK1α通过细胞色素P450亚家族19成员1(CYP19A1)介导小鼠卵巢GC中雌二醇的分泌。
    结论:这些发现提高了对女性生殖和雌激素合成调节机制的现有认识。
    背景:不适用。
    BACKGROUND: Casein kinase 1α (CK1α), expressed in both ovarian germ and somatic cells, is involved in the initial meiosis and primordial follicle formation of mouse oocytes. Using in vitro and in vivo experiments in this study, we explored the function and mechanism of CK1α in estrogen synthesis in mice ovarian granulosa cells.
    METHODS: A CK1α knockout (cKO) mouse model, targeted specifically to ovarian granulosa cells (GCs), was employed to establish the influence of CK1α on in vivo estrogen synthesis. The influence of CK1α deficiency on GCs was determined in vivo and in vitro by immunofluorescence analysis and Western blot assay. Transcriptome profiling, differentially expressed genes and gene functional enrichment analyses, and computation protein-protein docking, were further employed to assess the CK1α pathway. Furthermore, wild-type female mice were treated with the CK1α antagonist D4476 to elucidate the CK1α\'s role in estrogen regulation.
    RESULTS: Ovarian GCs CK1α deficiency impaired fertility and superovulation of female mice; also, the average litter size and the estradiol (E2) level in the serum of cKO female mice were decreased by 57.3% and 87.4% vs. control mice, respectively. This deficiency disrupted the estrous cycle and enhanced the apoptosis in the GCs. We observed that CK1α mediated the secretion of estradiol in mouse ovarian GCs via the cytochrome P450 subfamily 19 member 1 (CYP19A1).
    CONCLUSIONS: These findings improve the existing understanding of the regulation mechanism of female reproduction and estrogen synthesis.
    BACKGROUND: Not applicable.
    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
    关于南部白犀牛(SWR)的生殖生理以及在卵拾取(OPU)之前卵巢刺激的作用,目前尚不清楚。颗粒细胞(GC)提供了对卵泡生长和卵母细胞成熟状态的宝贵见解。这项研究的目的是从卵泡发育的三个阶段评估GC的转录组变化,并确定可能与卵巢刺激导致的卵泡生长和成熟相关的生物标志物。OPU后从SWR收集的GC根据卵泡大小分配阶段。分离总RNA,制备cDNA文库并在NovaSeq6000上测序。所有生物信息学分析均使用Galaxy网络平台进行。将读数与CerSimCot1.0对齐,并使用EquCab3.0进行手动管理。总的来说,在卵泡阶段鉴定了39,455个转录本(21,612个基因),和手动管理使基因鉴定比原始注释增加了61%。来自排卵前卵泡的颗粒细胞表达最高数量的独特转录本。基于聚类分析和表达模式确定了以下七个生物标志物:COL1A1,JMY,FBXW11,NRG1,TMPO,MACIR和COL4A1。这些数据可用于评估不同卵巢刺激方案对卵泡动力学的影响,改善OPU结果,并支持该物种的保护工作。
    Much remains unknown about the reproductive physiology of southern white rhinoceros (SWR) and the effect of ovarian stimulation prior to ovum pickup (OPU) have not been fully elucidated. Granulosa cells (GC) provide valuable insight into follicle growth and oocyte maturation status. The goals of this study were to evaluate transcriptomic changes in GC from three stages of follicle development and to identify biomarkers possibly associated with follicular growth and maturation as a result of ovarian stimulation. GC collected from SWRs following OPU were assigned stages based upon follicle size. Total RNA was isolated, and cDNA libraries were prepared and sequenced on a NovaSeq 6000. All bioinformatics analyses were performed utilizing the Galaxy web platform. Reads were aligned to CerSimCot1.0, and the manual curation was performed with EquCab3.0. Overall, 39,455 transcripts (21,612 genes) were identified across follicle stages, and manual curation yielded a 61% increase in gene identification from the original annotation. Granulosa cells from preovulatory follicles expressed the highest number of unique transcripts. The following seven biomarkers were determined based upon cluster analysis and patterns of expression: COL1A1, JMY, FBXW11, NRG1, TMPO, MACIR and COL4A1. These data can be used to potentially evaluate the effects of different ovarian stimulation protocols on follicle dynamics, improve OPU results, and support conservation efforts in this species.
    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
    盐诱导型激酶(SIKs),丝氨酸/苏氨酸激酶家族,被发现是女性生育能力的关键决定因素。SIK2沉默导致对促性腺激素的排卵反应增加。相比之下,SIK3基因敲除导致不孕症,促性腺激素不敏感,和卵巢缺乏窦和排卵前卵泡。这项研究假设SIK2和SIK3通过颗粒细胞(GC)的对比作用差异调节卵泡生长和生育力,卵泡的体细胞.因此,SIK2或SIK3GC特异性敲除(SIK2GCKD和SIK3GCKD,分别)通过将SIK培养的小鼠与Cyp19a1pII-Cre小鼠杂交来产生小鼠。生育力研究表明,SIK2GCKD小鼠在六个月内的幼崽积累和平均产仔数与对照组相似,尽管SIK3GCKD小鼠与对照组相比显着降低。与对照组相比,促性腺激素刺激青春期前SIK2GCKD小鼠导致血清雌二醇水平显著升高,而SIK3GCKD小鼠产生的雌二醇明显减少。在促性腺激素刺激的SIK2GCKD小鼠的GC中,Cyp11a1,Cyp19a1和StAR显着增加。然而,在SIK3GCKD小鼠中,Cyp11a1和StAR仍然显着低于对照组。有趣的是,与对照组相比,SIK3GCKD中的Cyp19a1刺激没有统计学差异。超数排卵导致SIK2GCKD小鼠排卵明显更多的卵母细胞,而SIK3GCKD小鼠排卵的卵母细胞明显少于对照组。值得注意的是,SIK3GCKD超排卵卵巢的腔前卵泡明显多于对照组。SIK3GCKD卵巢含有比对照明显更多的凋亡细胞和更少的增殖细胞。该数据指出了SIK2和SIK3对GC功能和卵泡发育的差异调节,并支持了靶向这些激酶治疗不育症或开发新避孕药的治疗潜力。
    Salt-inducible kinases (SIKs), a family of serine/threonine kinases, were found to be critical determinants of female fertility. SIK2 silencing results in increased ovulatory response to gonadotropins. In contrast, SIK3 knockout results in infertility, gonadotropin insensitivity, and ovaries devoid of antral and preovulatory follicles. This study hypothesizes that SIK2 and SIK3 differentially regulate follicle growth and fertility via contrasting actions in the granulosa cells (GCs), the somatic cells of the follicle. Therefore, SIK2 or SIK3 GC-specific knockdown (SIK2GCKD and SIK3GCKD, respectively) mice were generated by crossing SIK floxed mice with Cyp19a1pII-Cre mice. Fertility studies revealed that pup accumulation over 6 months and the average litter size of SIK2GCKD mice were similar to controls, although in SIK3GCKD mice were significantly lower compared to controls. Compared to controls, gonadotropin stimulation of prepubertal SIK2GCKD mice resulted in significantly higher serum estradiol levels, whereas SIK3GCKD mice produced significantly less estradiol. Cyp11a1, Cyp19a1, and StAR were significantly increased in the GCs of gonadotropin-stimulated SIK2GCKD mice. However, Cyp11a1 and StAR remained significantly lower than controls in SIK3GCKD mice. Interestingly, Cyp19a1 stimulation in SIK3GCKD was not statistically different compared to controls. Superovulation resulted in SIK2GCKD mice ovulating significantly more oocytes, whereas SIK3GCKD mice ovulated significantly fewer oocytes than controls. Remarkably, SIK3GCKD superovulated ovaries contained significantly more preantral follicles than controls. SIK3GCKD ovaries contained significantly more apoptotic cells and fewer proliferating cells than controls. These data point to the differential regulation of GC function and follicle development by SIK2 and SIK3 and supports the therapeutic potential of targeting these kinases for treating infertility or developing new contraceptives.
    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
    背景:多囊卵巢综合征(PCOS)对女性的生殖健康提出了重大挑战,以卵泡发育中断和排卵功能障碍为特征。PCOS发病机制的核心是颗粒细胞,其功能障碍有助于异常的类固醇激素产生和氧化应激。线粒体功能障碍成为关键角色,影响细胞能量学,氧化应激,和类固醇生成。这项研究调查了经血干细胞(MenSCs)及其外泌体在减轻PCOS颗粒细胞线粒体功能障碍和氧化应激方面的治疗潜力。
    方法:使用来曲唑诱导的PCOS大鼠模型,收获颗粒细胞并培养。MenSCs及其外泌体被用来评估它们对线粒体生物发生的影响。氧化应激,PCOS颗粒细胞产生雌激素。
    结果:结果显示PCOS颗粒细胞线粒体生物发生减少和氧化应激增加,同时减少雌激素的产生。用MenSCs及其外泌体治疗显示线粒体生物发生的显着改善,氧化应激水平,PCOS颗粒细胞产生雌激素。进一步分析显示,MenSCs优于外泌体,归因于它们持续分泌生物活性因子。机械上,与线粒体生物发生和抗氧化防御相关的MenSCs和外泌体激活途径,强调他们对PCOS的治疗潜力。
    结论:这项研究提供了颗粒细胞线粒体在PCOS发病机制中的作用的见解,并提出了MenSCs和外泌体作为减轻PCOS线粒体功能障碍和氧化应激的潜在策略。需要进一步的研究来了解潜在的机制并验证临床疗效。提出了解决PCOS复杂性的有希望的途径。
    BACKGROUND: Polycystic ovary syndrome (PCOS) presents a significant challenge in women\'s reproductive health, characterized by disrupted folliculogenesis and ovulatory dysfunction. Central to PCOS pathogenesis are granulosa cells, whose dysfunction contributes to aberrant steroid hormone production and oxidative stress. Mitochondrial dysfunction emerges as a key player, influencing cellular energetics, oxidative stress, and steroidogenesis. This study investigates the therapeutic potential of menstrual blood-derived stem cells (MenSCs) and their exosomes in mitigating mitochondrial dysfunction and oxidative stress in PCOS granulosa cells.
    METHODS: Using a rat model of PCOS induced by letrozole, granulosa cells were harvested and cultured. MenSCs and their exosomes were employed to assess their effects on mitochondrial biogenesis, oxidative stress, and estrogen production in PCOS granulosa cells.
    RESULTS: Results showed diminished mitochondrial biogenesis and increased oxidative stress in PCOS granulosa cells, alongside reduced estrogen production. Treatment with MenSCs and their exosomes demonstrated significant improvements in mitochondrial biogenesis, oxidative stress levels, and estrogen production in PCOS granulosa cells. Further analysis showed MenSCs\' superior efficacy over exosomes, attributed to their sustained secretion of bioactive factors. Mechanistically, MenSCs and exosomes activated pathways related to mitochondrial biogenesis and antioxidative defense, highlighting their therapeutic potential for PCOS.
    CONCLUSIONS: This study offers insights into granulosa cells mitochondria\'s role in PCOS pathogenesis and proposes MenSCs and exosomes as a potential strategy for mitigating mitochondrial dysfunction and oxidative stress in PCOS. Further research is needed to understand underlying mechanisms and validate clinical efficacy, presenting promising avenues for addressing PCOS complexity.
    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
    产蛋性能在家禽中具有重要的经济意义,但是潜在的遗传机制仍然难以捉摸。在这项工作中,我们对产蛋量不同的母鸡进行了多组学和多组织整合研究,检测hub候选基因并构建有助于产卵表型差异的hub分子网络。我们确定了三个枢纽候选基因作为产卵促进因子:TFPI2,其促进下丘脑神经元细胞中的GnRH分泌;CAMK2D,促进垂体细胞中FSHβ和LHβ的分泌;和OSTN,促进颗粒细胞增殖和性类固醇激素的合成。我们通过组织间串扰分析揭示了涉及产蛋的关键内分泌因素,并证明两者都是肝细胞因子,APOA4和脂肪因子,ANGPTL2可以通过与下丘脑-垂体-卵巢轴的组织间通讯来增加产卵量。一起,这些结果揭示了多组织协同调控鸡产蛋性能的分子机制,为禽类生殖调控提供了关键见解。
    Egg-laying performance is of great economic importance in poultry, but the underlying genetic mechanisms are still elusive. In this work, we conduct a multi-omics and multi-tissue integrative study in hens with distinct egg production, to detect the hub candidate genes and construct hub molecular networks contributing to egg-laying phenotypic differences. We identifiy three hub candidate genes as egg-laying facilitators: TFPI2, which promotes the GnRH secretion in hypothalamic neuron cells; CAMK2D, which promotes the FSHβ and LHβ secretion in pituitary cells; and OSTN, which promotes granulosa cell proliferation and the synthesis of sex steroid hormones. We reveal key endocrine factors involving egg production by inter-tissue crosstalk analysis, and demonstrate that both a hepatokine, APOA4, and an adipokine, ANGPTL2, could increase egg production by inter-tissue communication with hypothalamic-pituitary-ovarian axis. Together, These results reveal the molecular mechanisms of multi-tissue coordinative regulation of chicken egg-laying performance and provide key insights to avian reproductive regulation.
    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
    卵巢颗粒细胞对促性腺激素调节的雌激素产生至关重要,女性周期维持和生育能力。上皮Na+通道(ENaC)与女性生育能力有关;然而,它是否以及如何在卵巢细胞功能中发挥作用仍有待探索。这里,我们报道了人和小鼠卵巢颗粒细胞中ENaC表达和通道活性的膜片钳和Na+成像检测,由垂体促性腺激素促进,卵泡刺激素(FSH)或黄体生成素(LH)。小鼠中基于Cre重组酶和CRISPR-Cas9的颗粒特异性敲除ENaCα亚基(Scnn1a)导致早期发情时雌激素升高失败,黄体数量减少,异常延长的发情期,减少成年雌性小鼠的产仔数和低生育力。使用包括RNA测序和Ca2+成像在内的技术进行的进一步分析显示,基于shRNA的敲除或ENaC的敲除减少了自发或受刺激的Ca2振荡,降低了细胞内Ca2储存的能力,并损害了FSH/LH刺激的转录组变化,从而在小鼠和/或人颗粒细胞中产生雌激素。一起,这些结果揭示了ENaC在调节颗粒细胞中的促性腺激素信号以促进雌激素稳态和女性生育能力方面的作用。
    Ovarian granulosa cells are essential to gonadotrophin-regulated estrogen production, female cycle maintenance and fertility. The epithelial Na+ channel (ENaC) is associated with female fertility; however, whether and how it plays a role in ovarian cell function(s) remained unexplored. Here, we report patch-clamp and Na+ imaging detection of ENaC expression and channel activity in both human and mouse ovarian granulosa cells, which are promoted by pituitary gonadotrophins, follicle stimulating hormone (FSH) or luteinizing hormone (LH). Cre-recombinase- and CRISPR-Cas9-based granulosa-specific knockout of ENaC α subunit (Scnn1a) in mice resulted in failed estrogen elevation at early estrus, reduced number of corpus luteum, abnormally extended estrus phase, reduced litter size and subfertility in adult female mice. Further analysis using technologies including RNA sequencing and Ca2+ imaging revealed that pharmacological inhibition, shRNA-based knockdown or the knockout of ENaC diminished spontaneous or stimulated Ca2+ oscillations, lowered the capacity of intracellular Ca2+ stores and impaired FSH/LH-stimulated transcriptome changes for estrogen production in mouse and/or human granulosa cells. Together, these results have revealed a previously undefined role of ENaC in modulating gonadotrophin signaling in granulosa cells for estrogen homeostasis and thus female fertility.
    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
    背景:分泌型卷曲相关蛋白(SFRP)包含WNT信号拮抗剂家族,其在卵巢中的作用知之甚少。以前发现Sfrp4-null小鼠由于颗粒细胞对促性腺激素的反应增强而肥沃,导致窦卵泡闭锁减少和排卵率提高。本研究旨在阐明SFRP4拮抗FSH作用的机制。
    方法:用FSH和/或SFRP4处理来自野生型小鼠的颗粒细胞的原代培养物,并且通过RT-qPCR和RNAseq评价处理对基因表达的影响。进行生物信息学分析以分析SFRP4对转录组的影响,并将它们与FSH或FOXO1的组成型活性突变体进行比较。来自野生型或Sfrp4-null小鼠的其他颗粒细胞培养物,一些用特定信号效应物的药物抑制剂预处理,用于检查FSH和/或SFRP4对信号通路的影响,蛋白质印迹和TUNEL的自噬和凋亡。
    结果:发现用重组SFRP4处理培养的颗粒细胞可降低FSH靶基因的基础和FSH刺激的mRNA水平。出乎意料的是,发现这种效应既不通过规范(CTNNB1依赖性)也不通过规范的WNT信号机制发生,但被发现是GSK3β依赖性的。相反,发现SFRP4通过涉及AMPK的机制使AKT活性前变性。这导致FOXO1的低磷酸化和FSH和FOXO1转录组的一部分的表达减少。相反,FSH刺激的AMPK,发现相对于野生型对照,Sfrp4缺失小鼠的颗粒细胞中的AKT和FOXO1磷酸化水平增加。SFRP4处理颗粒细胞还通过经由AKT-mTORC1-ULK1的信号传导诱导自噬以及细胞凋亡。
    结论:本研究确定了一种新的GSK3β-AMPK-AKT信号机制,SFPR4通过该机制拮抗FSH作用,并进一步鉴定SFRP4为颗粒细胞自噬的新型调节因子。这些发现为先前在Sfrp4-null小鼠中观察到的表型变化提供了机制基础,并拓宽了我们对卵巢中WNT信号传导过程的生理作用的理解。
    BACKGROUND: Secreted frizzled-related proteins (SFRPs) comprise a family of WNT signaling antagonists whose roles in the ovary are poorly understood. Sfrp4-null mice were previously found to be hyperfertile due to an enhanced granulosa cell response to gonadotropins, leading to decreased antral follicle atresia and enhanced ovulation rates. The present study aimed to elucidate the mechanisms whereby SFRP4 antagonizes FSH action.
    METHODS: Primary cultures of granulosa cells from wild-type mice were treated with FSH and/or SFRP4, and effects of treatment on gene expression were evaluated by RT-qPCR and RNAseq. Bioinformatic analyses were conducted to analyse the effects of SFRP4 on the transcriptome, and compare them to those of FSH or a constitutively active mutant of FOXO1. Additional granulosa cell cultures from wild-type or Sfrp4-null mice, some pretreated with pharmacologic inhibitors of specific signaling effectors, were used to examine the effects of FSH and/or SFRP4 on signaling pathways, autophagy and apoptosis by western blotting and TUNEL.
    RESULTS: Treatment of cultured granulosa cells with recombinant SFRP4 was found to decrease basal and FSH-stimulated mRNA levels of FSH target genes. Unexpectedly, this effect was found to occur neither via a canonical (CTNNB1-dependent) nor non-canonical WNT signaling mechanism, but was found to be GSK3β-dependent. Rather, SFRP4 was found to antognize AKT activity via a mechanism involving AMPK. This lead to the hypophosphorylation of FOXO1 and a decrease in the expression of a portion of the FSH and FOXO1 transcriptomes. Conversely, FSH-stimulated AMPK, AKT and FOXO1 phosphorylation levels were found to be increased in the granulosa cells of Sfrp4-null mice relative to wild-type controls. SFRP4 treatement of granulosa cells also induced autophagy by signaling via AKT-mTORC1-ULK1, as well as apoptosis.
    CONCLUSIONS: This study identifies a novel GSK3β-AMPK-AKT signaling mechanism through which SFPR4 antagonizes FSH action, and further identifies SFRP4 as a novel regulator of granulosa cell autophagy. These findings provide a mechanistic basis for the phenotypic changes previously observed in Sfrp4-null mice, and broaden our understanding of the physiological roles of WNT signaling processes in the ovary.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号