Leydig cell

leydig 细胞
  • 文章类型: Journal Article
    男性衰老总是伴随着生育能力下降。据报道,叉头O(FOXO)转录因子FOXO4在衰老细胞中高度表达。激活后,它结合细胞核中的p53,防止衰老细胞凋亡和维持衰老细胞原位。睾丸间质细胞在辅助精子发生中起关键作用。睾丸间质细胞衰老导致睾丸微环境恶化并损害精子发生。在这项研究中,我们观察到FOXO4-DRI,一种特定的FOXO4-p53结合阻断剂,诱导衰老的睾丸间质细胞凋亡,减少某些衰老相关分泌表型的分泌,并改善共培养的GC-1SPG细胞的增殖。在自然衰老的小鼠中,FOXO4-DRI治疗的老年小鼠表现出提高的精子质量和改善的精子发生。
    Male ageing is always accompanied by decreased fertility. The forkhead O (FOXO) transcription factor FOXO4 is reported to be highly expressed in senescent cells. Upon activation, it binds p53 in the nucleus, preventing senescent cell apoptosis and maintaining senescent cells in situ. Leydig cells play key roles in assisting spermatogenesis. Leydig cell senescence leads to deterioration of the microenvironment of the testes and impairs spermatogenesis. In this study, we observed that FOXO4-DRI, a specific FOXO4- p53 binding blocker, induced apoptosis in senescent Leydig cells, reduced the secretion of certain Senescence-Associated Secretory Phenotype and improved the proliferation of cocultured GC-1 SPG cells. In naturally aged mice, FOXO4-DRI-treated aged mice exhibited increased sperm quality and improved spermatogenesis.
    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
    男性生育能力取决于正常的青春期发育。邻苯二甲酸二(2-乙基己基)酯(DEHP)是一种有效的抗雄激素化学物质,青春期暴露于DEHP会损害发育中的男性生殖系统,尤其是睾丸。然而,受DEHP影响的特定细胞靶标和分化过程,导致睾丸毒性,仍然定义不清。在这里,我们介绍了DEHP暴露后青春期小鼠睾丸的第一个单细胞转录组学图谱。为了进行实验,两组(每组8只)3周龄雄性小鼠,分别从出生后第21天至第48天每天口服0.5%羧甲基纤维素钠盐或100mg/kg体重DEHP。使用单细胞RNA测序,总共鉴定了31个不同的细胞群,特别是,Sertoli和Leydig细胞成为DEHP的重要靶标。DEHP暴露显着降低了表达成熟Sertoli标记(Sox9和Ar)的Sertoli细胞簇的比例,选择性降低胎儿睾丸间质细胞中睾酮合成基因的表达。通过细胞-细胞相互作用分析,我们观察到支持细胞1(SCs1)中相互作用的数量变化,睾丸间质细胞1(LCs1)和间质巨噬细胞(ITM),我们还确定了这些簇中的细胞特异性配体基因表达,比如Inha,Fyn,Vcam1和Apoe.补充体外试验证实,DEHP直接降低了与支持细胞粘附和细胞间通讯相关的基因的表达。总之,青春期周围的DEHP暴露会减少成熟的睾丸支持细胞的数量,并可能通过影响胎儿睾丸间质细胞而不是成年睾丸间质细胞中的睾丸激素合成基因来破坏睾丸类固醇生成。
    Male fertility depends on normal pubertal development. Di-(2-ethylhexyl) phthalate (DEHP) is a potent antiandrogen chemical, and exposure to DEHP during peripuberty can damage the developing male reproductive system, especially the testis. However, the specific cellular targets and differentiation processes affected by DEHP, which lead to testicular toxicity, remain poorly defined. Herein, we presented the first single-cell transcriptomic profile of the pubertal mouse testis following DEHP exposure. To carry out the experiment, 2 groups (n = 8 each) of 3-week-old male mice were orally administered 0.5% carboxymethylcellulose sodium salt or 100 mg/kg body weight DEHP daily from postnatal day 21-48, respectively. Using single-cell RNA sequencing, a total of 31 distinct cell populations were identified, notably, Sertoli and Leydig cells emerged as important targets of DEHP. DEHP exposure significantly decreased the proportions of Sertoli cell clusters expressing mature Sertoli markers (Sox9 and Ar), and selectively reduced the expression of testosterone synthesis genes in fetal Leydig cells. Through cell-cell interaction analyses, we observed changed numbers of interactions in Sertoli cells 1 (SCs1), Leydig cells 1 (LCs1), and interstitial macrophages, and we also identified cell-specific ligand gene expressions in these clusters, such as Inha, Fyn, Vcam1, and Apoe. Complementary in vitro assays confirmed that DEHP directly reduced the expression of genes related to Sertoli cell adhesion and intercellular communication. In conclusion, peripubertal DEHP exposure reduced the number of mature Sertoli cells and may disrupt testicular steroidogenesis by affecting the testosterone synthesis genes in fetal Leydig cells rather than adult Leydig cells.
    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
    睾丸间质细胞是睾丸间质组织的重要组成部分,是男性雄激素的主要来源。Leydig细胞的功能缺陷通常会导致严重的生殖障碍;然而,这些细胞的命运决定和类固醇生成背后的转录程序尚未完全定义。在这项研究中,我们报道,同源结构域转录因子PBX1是小鼠Leydig细胞分化和睾酮产生的主要调节因子。PBX1在成年睾丸间质细胞和肾小管周围肌样细胞中高表达。Leydig细胞中Pbx1的条件缺失导致生精缺陷和完全不育。组织学检查显示,Pbx1缺失损害了睾丸结构,并导致生精小管解体。单细胞RNA-seq分析显示,Pbx1功能的丧失影响了祖细胞Leydig细胞的命运决定,并改变了成年睾丸中与睾丸激素合成相关的基因的转录。Pbx1直接调节在类固醇生成中起重要作用的基因的转录(Prlr,Nr2f2和Nedd4)。进一步的分析表明,Pbx1的缺失导致睾酮水平显着降低,伴随着孕烯醇酮的增加,雄烯二酮和促黄体激素。总的来说,我们的数据显示,PBX1对于维持Leydig细胞功能是必不可少的。这些发现提供了有关睾丸发育不全和睾丸间质细胞激素分泌调节的见解。
    Leydig cells are essential components of testicular interstitial tissue and serve as a primary source of androgen in males. A functional deficiency in Leydig cells often causes severe reproductive disorders; however, the transcriptional programs underlying the fate decisions and steroidogenesis of these cells have not been fully defined. In this study, we report that the homeodomain transcription factor PBX1 is a master regulator of Leydig cell differentiation and testosterone production in mice. PBX1 was highly expressed in Leydig cells and peritubular myoid cells in the adult testis. Conditional deletion of Pbx1 in Leydig cells caused spermatogenic defects and complete sterility. Histological examinations revealed that Pbx1 deletion impaired testicular structure and led to disorganization of the seminiferous tubules. Single-cell RNA-seq analysis revealed that loss of Pbx1 function affected the fate decisions of progenitor Leydig cells and altered the transcription of genes associated with testosterone synthesis in the adult testis. Pbx1 directly regulates the transcription of genes that play important roles in steroidogenesis (Prlr, Nr2f2 and Nedd4). Further analysis demonstrated that deletion of Pbx1 leads to a significant decrease in testosterone levels, accompanied by increases in pregnenolone, androstenedione and luteinizing hormone. Collectively, our data revealed that PBX1 is indispensable for maintaining Leydig cell function. These findings provide insights into testicular dysgenesis and the regulation of hormone secretion in Leydig cells.
    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
    镉(Cd)是环境中常见的重金属污染物,含Cd化合物的产品在工业中的广泛使用导致了环境中的含量超标,通过食物链进入动物体内,从而严重影响动物的生殖发育。相关研究报道Cd严重影响动物精原细胞发育和精子发生。相比之下,Cd对雄性的生殖毒性及其作用机制尚未阐明。因此,本文综述了Cd对生殖细胞的毒性作用,雄性动物的精原细胞和下丘脑-垂体-性腺轴(HPG轴)及其氧化应激的毒性作用机制,细胞凋亡和自噬从细胞学的角度来看,遗传学和神经内分泌学。还分析了Cd胁迫对雄性动物生殖发育表观遗传修饰的影响。希望为深入研究Cd对雄性动物繁殖的毒性提供参考。
    Cadmium (Cd) is a common heavy metal pollutant in the environment, and the widespread use of products containing Cd compounds in industry has led to excessive levels in the environment, which enter the animal body through the food chain, thus seriously affecting the reproductive development of animals. Related studies have reported that Cd severely affects spermatogonia development and spermatogenesis in animals. In contrast, the reproductive toxicity of Cd in males and its mechanism of action have not been clarified. Therefore, this paper reviewed the toxic effects of Cd on germ cells, spermatogonia somatic cells and hypothalamic-pituitary-gonadal axis (HPG axis) of male animals and its toxic action mechanisms of oxidative stress, apoptosis and autophagy from the perspectives of cytology, genetics and neuroendocrinology. The effects of Cd stress on epigenetic modification of reproductive development in male animals were also analyzed. We hope to provide a reference for the in-depth study of the toxicity of Cd on male animal reproduction.
    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
    邻苯二甲酸二(2-乙基己基)酯(DEHP),被确定为一种内分泌干扰化学物质,与生殖毒性有关。这种关联在代谢功能不完全发育的新生儿中尤其值得注意,因为暴露于DEHP会对生殖系统造成持久的损害,可能影响成人生殖健康。在这项研究中,我们对出生后第5天(PD5)小鼠连续给药40μg/kg和80μg/kgDEHP10天,以模拟婴儿期的低剂量和高剂量DEHP暴露.利用单细胞RNA测序(scRNA-seq),我们的分析显示,婴儿期不同浓度的DEHP暴露在睾丸未分化精原细胞(UndiffSPG)中诱导了不同的DNA损伤反应特征.具体来说,DNA损伤引发线粒体功能障碍,导致乙酰辅酶A含量改变。随后,这种破坏导致组蛋白乙酰化模式异常,最终导致UndiffSPG在40μg/kgDEHP组中的凋亡和80μg/kgDEHP组中的自噬。此外,我们发现DEHP暴露通过局灶性粘附和PPAR信号通路影响Sertoli和Leydig细胞的发育和功能,分别。我们还发现,在DEHP暴露后,Leydig细胞通过Ptn-Sdc4和Mdk-Sdc4调节UndiffSPG的代谢环境。最后,我们的研究提供了开创性的证据,证明DEHP暴露在婴儿期引起的睾丸稳态破坏一直持续到成年期.总之,本研究阐明了婴儿期DEHP暴露影响睾丸细胞群发育的分子机制.
    Di (2-ethylhexyl) phthalate (DEHP), identified as an endocrine-disrupting chemical, is associated with reproductive toxicity. This association is particularly noteworthy in newborns with incompletely developed metabolic functions, as exposure to DEHP can induce enduring damage to the reproductive system, potentially influencing adult reproductive health. In this study, we continuously administered 40 μg/kg and 80 μg/kg DEHP to postnatal day 5 (PD5) mice for ten days to simulate low and high doses of DEHP exposure during infancy. Utilizing single-cell RNA sequencing (scRNA-seq), our analysis revealed that varying concentrations of DEHP exposure during infancy induced distinct DNA damage response characteristics in testicular Undifferentiated spermatogonia (Undiff SPG). Specifically, DNA damage triggered mitochondrial dysfunction, leading to acetyl-CoA content alterations. Subsequently, this disruption caused aberrations in histone acetylation patterns, ultimately resulting in apoptosis of Undiff SPG in the 40 μg/kg DEHP group and autophagy in the 80 μg/kg DEHP group. Furthermore, we found that DEHP exposure impacts the development and functionality of Sertoli and Leydig cells through the focal adhesion and PPAR signaling pathways, respectively. We also revealed that Leydig cells regulate the metabolic environment of Undiff SPG via Ptn-Sdc4 and Mdk-Sdc4 after DEHP exposure. Finally, our study provided pioneering evidence that disruptions in testicular homeostasis induced by DEHP exposure during infancy endure into adulthood. In summary, this study elucidates the molecular mechanisms through which DEHP exposure during infancy influences the development of testicular cell populations.
    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
    近年来观察到的预期寿命增加导致老年男性迟发性性腺功能减退症(LOH)的患病率更高。LOH的特征是睾丸激素水平下降,并可能对身心健康产生重大影响。虽然LOH的根本原因还没有完全理解,人们对探索炎症在其发展中的作用越来越感兴趣。炎症是一个描述慢性疾病的概念,低档,由于衰老而发生的全身性炎症。这种炎症状态与各种年龄相关疾病的发展有关。几种细胞和分子机制已被确定为炎症的贡献者,包括免疫衰老,细胞衰老,自噬缺陷,和线粒体功能障碍。尽管对炎症进行了广泛的研究,其与LOH的关系尚未在文献中得到全面回顾。为了解决这个差距,我们旨在回顾与炎症相关的最新发现及其对LOH发展的影响。此外,我们将探讨针对炎症的干预措施作为LOH的潜在治疗方法.
    The increasing life expectancy observed in recent years has resulted in a higher prevalence of late-onset hypogonadism (LOH) in older men. LOH is characterized by the decline in testosterone levels and can have significant impacts on physical and mental health. While the underlying causes of LOH are not fully understood, there is a growing interest in exploring the role of inflammaging in its development. Inflammaging is a concept that describes the chronic, low-grade, systemic inflammation that occurs as a result of aging. This inflammatory state has been implicated in the development of various age-related diseases. Several cellular and molecular mechanisms have been identified as contributors to inflammaging, including immune senescence, cellular senescence, autophagy defects, and mitochondrial dysfunction. Despite the extensive research on inflammaging, its relationship with LOH has not yet been thoroughly reviewed in the literature. To address this gap, we aim to review the latest findings related to inflammaging and its impact on the development of LOH. Additionally, we will explore interventions that target inflammaging as potential treatments for LOH.
    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
    草甘膦(GLY),一种广泛使用的除草剂,可以通过抑制睾酮合成对男性生殖健康产生不利影响。铁凋亡是铁依赖性氧化细胞死亡的一种形式,有助于抑制睾酮分泌。然而,目前尚不清楚铁凋亡是否参与GLY抑制的睾酮合成。特此,建立了1mMGLY暴露的睾丸Leydig(TM3)细胞的体外模型来阐明这一问题。数据首先表明GLY通过铁凋亡引起细胞毒性和睾酮合成抑制,而由于细胞内亚铁离子(Fe2)过载和谷胱甘肽消耗而导致的脂质过氧化物的积累被证实是铁死亡的决定因素。通过螯合Fe2+或抑制脂质过氧化来阻断铁凋亡可显著减轻GLY诱导的睾酮合成抑制。此外,在GLY处理的TM3细胞中发现自噬激活,核受体共激活因子4(NCOA4)介导的铁细胞通过释放过量的Fe2参与铁细胞凋亡。GLY诱导的细胞毒性和睾酮合成抑制被NCOA4敲低显著缓解,证明NCOA4介导的铁素吞噬在GLY抑制的睾酮合成中的关键作用。总之,这项研究提供了确凿的证据,证明NCOA4介导的铁细胞吞噬促进铁细胞凋亡以抑制睾酮合成,强调靶向NCOA4可能是GLY诱导的男性生殖毒性的潜在治疗方法。
    Glyphosate (GLY), a widely used herbicide, can adversely affect the male reproductive health by inhibiting testosterone synthesis. Ferroptosis is a form of iron-dependent oxidative cell death that contributes to inhibition of testosterone secretion. However, it still remains unclear whether ferroptosis is involved in GLY-inhibited testosterone synthesis. Hereby, an in vitro model of 1 mM GLY-exposed testicular Leydig (TM3) cells was established to elucidate this issue. Data firstly showed that GLY causes cytotoxicity and testosterone synthesis inhibition via ferroptosis, while accumulation of lipid peroxides due to intracellular ferrous ion (Fe2+) overload and glutathione depletion is confirmed as a determinant of ferroptosis. Blockage of ferroptosis via chelation of Fe2+ or inhibition of lipid peroxidation can markedly mitigate GLY-induced testosterone synthesis inhibition. Also, autophagy activation is revealed in GLY-treated TM3 cells and nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy is involved in ferroptosis through the release of excess Fe2+. GLY-induced cytotoxicity and testosterone synthesis inhibition are significantly alleviated by NCOA4 knockdown, demonstrating the crucial role of NCOA4-mediated ferritinophagy in GLY-inhibited testosterone synthesis. In summary, this study provides solid evidence that NCOA4-mediated ferritinophagy promotes ferroptosis to inhibit testosterone synthesis, highlighting that targeting NCOA4 may be a potential therapeutic approach in GLY-induced male reproductive toxicity.
    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
    四氯双酚A(TCBPA),卤化阻燃剂和内分泌干扰物,已在人体尿液和血清中检测到。虽然以前的研究表明它对生殖系统的影响,对其青春期机制的调查仍然有限。本研究旨在探讨TCBPA对青春期小鼠睾丸间质细胞的影响及其潜在机制。28日龄的雄性C57小鼠用50、100和200mg/kg/天的剂量给药28天。TCBPA不会改变体重和睾丸重量,但会降低100和200mg/kg的睾丸激素水平,并降低200mg/kg的附睾精子数量。TCBPA降低了200mg/kg的Leydig细胞数量,同时下调了关键的Leydig细胞基因(Lhcgr,Scarb1,Cyp11a1,Cyp17a1,Hsd3b6,Hsd17b3和Insl3)低至50mg/kg。进一步研究表明,TCBPA可诱导活性氧并引起内质网应激。在TM3小鼠Leydig细胞中进行的体外研究表明,TCBPA确实在75μM时诱导了活性氧并引起内质网应激,并且在此浓度下睾丸激素抑制了生产,添加抗氧化剂生育酚可以逆转它。这些发现为更深入地了解TCBPA对青春期Leydig细胞的毒性机制提供了新的见解和参考。
    Tetrachlorobisphenol A (TCBPA), a halogenated flame retardant and endocrine disruptor, has been detected in human urine and serum. While previous research has shown its impact on the reproductive system, investigations into its mechanisms during puberty remain limited. This study aims to explore the effects of TCBPA on Leydig cells in adolescent mice and potential underlying mechanisms. Male C57 mice of age 28 days were gavaged with 50, 100, and 200 mg/kg/day for 28 days. TCBPA did not alter body weight and testis weight but lowered testosterone levels at 100 and 200 mg/kg and reduced sperm count in the epididymis at 200 mg/kg. TCBPA lowered Leydig cell number at 200 mg/kg while it downregulated key Leydig cell gene (Lhcgr, Scarb1, Cyp11a1, Cyp17a1, Hsd3b6, Hsd17b3 and Insl3) as low as 50 mg/kg. Further study indicated that TCBPA induced reactive oxygen species and caused endoplasmic reticulum stress. In vitro study in TM3 mouse Leydig cells showed that TCBPA indeed induced reactive oxygen species and caused endoplasmic reticulum stress at 75 μM and inhibited testosterone production at this concentration and addition of antioxidant tocopherol can reverse it. These discoveries provide new insights and references for a deeper understanding of the toxic mechanisms of TCBPA on Leydig cells during puberty.
    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
    Klinefelter综合征(KS)曾经被认为是由于先天性染色体异常导致的不育,但是局灶性精子的存在改变了这一点。预测和促进精子发生的关键是寻找调控局灶性精子发生的靶点。
    探讨不同年龄段KS患者的生育力变化趋势,并确定潜在的治疗目标。
    使用文献计量分析从1992年至2022年的WebofScienceCoreCollection(WoSCC)收集了有关KS的临床研究数据。对2017年至2022年在现实世界中接受显微睾丸精子提取(mTESE)的75名KS患者进行了横断面研究。生殖激素,睾丸组织病理学,雄激素受体,分析胰岛素样因子3(INSL3)受体和精子恢复率(SRR)。
    男性不育,发育不良,支持细胞,Leydig细胞,睾酮和精子发生是与KS相关的研究热点。促黄体生成素(LH),睾丸激素,和INSL3是Leydig细胞功能的评价指标,随年龄波动。睾酮和LH在13-19岁和30-45岁达到峰值,而INSL3仅在13-19岁达到峰值。27名患者(27/75)通过mTESE恢复了精子,并在20、28、34和37岁时经历了SRR高峰。纤维化患者的SRR为46.15%,脂肪变性为7.14%,黑变病为40.00%。INSL3和雄激素受体在局灶性精子发生中高表达且大致平衡。
    睾丸间质细胞代谢异常导致INSL3和雄激素受体表达失衡,这可能是KS精子发生的潜在靶标。
    Klinefelter\'s syndrome (KS) was once considered infertile due to congenital chromosomal abnormalities, but the presence of focal spermatozoa changed this. The key to predict and promote spermatogenesis is to find targets that regulate focal spermatogenesis.
    To explore the trend of fertility changes in KS patients at different ages and identify potential therapeutic targets.
    Bibliometric analysis was used to collect clinical research data on KS from the Web of Science Core Collection (WoSCC) from 1992 to 2022. A cross-sectional study was conducted on 75 KS patients who underwent microscopic testicular sperm extraction (mTESE) from 2017 to 2022 in the real world. The reproductive hormones, testicular histopathology, androgen receptors, insulin-like factor 3 (INSL3) receptors and sperm recovery rate (SRR) were analyzed.
    Male infertility, dysplasia, Sertoli cells, Leydig cells, testosterone and spermatogenesis were the research focuses related to KS. Luteinizing hormone (LH), testosterone, and INSL3 were evaluation indicators of Leydig cell function that fluctuate with age. Testosterone and LH peaked at ages 13-19 and 30-45, while INSL3 only peaked at ages 13-19. 27 patients (27/75) recovered sperm through mTESE and experienced SRR peaks at the ages of 20, 28, 34, and 37. The SRR of fibrosis patients was 46.15%, fatty degeneration was 7.14%, and melanosis was 40.00%. The INSL3 and androgen receptors were highly expressed and roughly balanced in focal spermatogenesis.
    Abnormal metabolism of Leydig cells led to imbalanced expression of INSL3 and androgen receptors, which might be a potential target for spermatogenesis in KS.
    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:睾丸间质细胞周围的老化炎症微环境与男性睾丸激素水平降低有关。肿瘤坏死因子α诱导蛋白3(TNFAIP3)是多种衰老相关疾病中的重要抗炎因子。本研究旨在探讨TNFAIP3对衰老炎症微环境下睾丸间质细胞睾酮产生的保护作用。
    方法:生物信息学分析检查了衰老大鼠睾丸中TNFAIP3表达的差异,并验证了衰老小鼠睾丸中的发现。使用两种Leydig细胞系建立了炎症的体外模型,肿瘤坏死因子α(TNF-α)作为炎症因子。利用慢病毒转导来操纵这些细胞系中的TNFAIP3表达。转录组测序鉴定了TNFAIP3过表达细胞中差异表达的基因。
    结果:生物信息学分析和验证实验显示,衰老大鼠和小鼠睾丸中炎症信号传导增加和TNFAIP3表达升高。TNFAIP3敲低恶化睾酮合成抑制和细胞凋亡,而TNFAIP3过表达逆转了这些效应。转录组分析鉴定了TNFAIP3过表达后P38MAPK途径的改变。TNFAIP3敲低增强TNF诱导的P38MAPK信号,而其过度表达减弱了这种作用。发现TNFAIP3通过上调CEBPB表达来调节睾酮合成。
    结论:TNFAIP3对睾丸间质细胞的凋亡具有抑制作用,并促进睾丸激素的产生。TNFAIP3对炎症微环境中的Leydig细胞的保护性影响可能是通过抑制P38MAPK途径和上调CEBPB表达来介导的。
    The aging inflammatory microenvironment surrounding Leydig cells is linked to reduced testosterone levels in males. Tumor necrosis factor alpha-induced protein 3 (TNFAIP3) acts as a critical anti-inflammatory factor in various aging-related diseases. This study aims to investigate the protective effect of TNFAIP3 on testosterone production in Leydig cells under an aging inflammatory microenvironment.
    Bioinformatics analysis examined TNFAIP3 expression differences in aging rat testes and validated the findings in aging mouse testes. In vitro models of inflammation were established using two Leydig cell lines, with tumor necrosis factor alpha (TNF-α) as the inflammatory factor. Lentiviral transduction was utilized to manipulate TNFAIP3 expression in these cell lines. Transcriptomic sequencing identified differentially expressed genes in TNFAIP3-overexpressing cells.
    Bioinformatics analysis and validation experiments revealed increased inflammatory signaling and elevated TNFAIP3 expression in aging rat and mouse testes. TNFAIP3 knockdown worsened testosterone synthesis inhibition and apoptosis in cells, while TNFAIP3 overexpression reversed these effects. Transcriptome analysis identified alterations in the P38MAPK pathway following TNFAIP3 overexpression. TNFAIP3 knockdown enhanced TNF-induced P38MAPK signaling, whereas its overexpression attenuated this effect. TNFAIP3 was found to regulate testosterone synthesis by upregulating CEBPB expression.
    TNFAIP3 exhibits inhibitory effects on apoptosis and promotes testosterone production in Leydig cells. The protective influence of TNFAIP3 on Leydig cells within an inflammatory microenvironment is likely mediated through by inhibiting the P38MAPK pathway and upregulating CEBPB expression.
    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