Endometriosis is a prevalent chronic inflammatory condition impacting 5-10% of reproductive-age women, commonly resulting in debilitating pelvic pain and infertility. Despite extensive research efforts, the precise underlying pathophysiology remains largely unclear. Emerging evidence increasingly suggests that cellular senescence, iron overload, and fibrosis collectively form a critical pathological axis that significantly contributes to the persistence and severity of the disease. However, the intricate mechanistic interplay between the immune system's failure to effectively clear senescent cells, the damaging effects of iron-induced oxidative stress, and the subsequent fibrotic remodelling is still poorly understood. This narrative review highlights the interconnected roles of impaired immune clearance of senescent cells, iron accumulation, and fibrosis development in driving endometriosis pathogenesis. The review aims to clarify how iron overload and cellular senescence contribute to the progression of endometriosis. It also evaluates novel therapeutic strategies that target iron dysregulation and senescence pathways. By exploring this detrimental triad, we seek to identify potential new avenues for transforming the management of endometriosis, offering hope for more effective treatments to alleviate the significant burden on affected women.
{"title":"The Nexus of Iron, Senescence, and Fibrosis in Endometriosis: A Narrative Review.","authors":"Richa Patel, Guruprasad Kalthur, Ratul Datta, Swar Shah, Rahul Dutta","doi":"10.1007/s43032-025-01999-0","DOIUrl":"10.1007/s43032-025-01999-0","url":null,"abstract":"<p><p>Endometriosis is a prevalent chronic inflammatory condition impacting 5-10% of reproductive-age women, commonly resulting in debilitating pelvic pain and infertility. Despite extensive research efforts, the precise underlying pathophysiology remains largely unclear. Emerging evidence increasingly suggests that cellular senescence, iron overload, and fibrosis collectively form a critical pathological axis that significantly contributes to the persistence and severity of the disease. However, the intricate mechanistic interplay between the immune system's failure to effectively clear senescent cells, the damaging effects of iron-induced oxidative stress, and the subsequent fibrotic remodelling is still poorly understood. This narrative review highlights the interconnected roles of impaired immune clearance of senescent cells, iron accumulation, and fibrosis development in driving endometriosis pathogenesis. The review aims to clarify how iron overload and cellular senescence contribute to the progression of endometriosis. It also evaluates novel therapeutic strategies that target iron dysregulation and senescence pathways. By exploring this detrimental triad, we seek to identify potential new avenues for transforming the management of endometriosis, offering hope for more effective treatments to alleviate the significant burden on affected women.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3783-3806"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12715047/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145378409","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
To investigate the pathogenicity of splice-site variants in DNAH17, and analyze their impact on sperm morphology and motility, we employed whole-exome sequencing (WES) and Sanger sequencing to identify and validate candidate variants. Computational predictions of splicing defects were performed using varSEAK and MobiDetails. Functional validation was conducted using minigene splicing assays in HEK293T cells. Structural modeling of mutant proteins was performed with AlphaFold3 and visualized by PyMOL. Two novel splice-site variants (DNAH17: c.11677 + 5G > T/c.11677 + 5G > A) were identified in a proband with asthenozoospermia and multiple morphological abnormalities of the sperm flagella (MMAF). Bioinformatics tools predicted disruption of the canonical donor splice site (MaxEntScan score reduction: 54.2% for G > A, 39.5% for G > T; SpliceAI donor loss scores > 0.8). Minigene assays confirmed exon 72 skipping, leading to a frameshift mutation (p.Asn3844Lysfs*13) that truncates the AAA6 domain. This study expands the mutational spectrum of DNAH17-related male infertility by demonstrating that splice-site variants disrupting the AAA6 domain represent a novel pathogenic mechanism underlying asthenozoospermia and MMAF. These findings underscore the necessity of integrating splice-site analysis into genetic diagnostics for male infertility.
为了研究DNAH17剪接位点变异的致病性,并分析其对精子形态和活力的影响,我们采用了全外显子组测序(WES)和Sanger测序来鉴定和验证候选变异。使用varSEAK和MobiDetails进行拼接缺陷的计算预测。在HEK293T细胞中使用minigene剪接实验进行功能验证。用AlphaFold3对突变蛋白进行结构建模,并用PyMOL进行可视化。两个新的剪接位点变异(DNAH17: c.11677 + 5G > T/c)。11677 + 5G > A)在一例弱精子症和精子鞭毛(MMAF)多重形态异常先证者中被鉴定出来。生物信息学工具预测了典型供体剪接位点的破坏(MaxEntScan评分降低:G > A 54.2%, G > T 39.5%; SpliceAI供体损失评分> 0.8)。miniigene实验证实了外显子72的跳跃,导致移码突变(p.a n3844lysfs *13)截断AAA6结构域。本研究通过证明破坏AAA6结构域的剪接位点变异是弱精子症和MMAF的一种新的致病机制,扩大了dnah17相关男性不育的突变谱。这些发现强调了将剪接位点分析整合到男性不育基因诊断中的必要性。
{"title":"Novel DNAH17 Splice-Site Mutations Truncating the AAA6 Domain Cause Asthenozoospermia with MMAF.","authors":"Leilei Feng, Feng Wan, Chenchen Cui, Ke Feng, Yanqing Xia, Xiaowei Qu, Bei Yang, Jinwei Wang, Longze Wang, Cuilian Zhang, Haibin Guo","doi":"10.1007/s43032-025-02002-6","DOIUrl":"10.1007/s43032-025-02002-6","url":null,"abstract":"<p><p>To investigate the pathogenicity of splice-site variants in DNAH17, and analyze their impact on sperm morphology and motility, we employed whole-exome sequencing (WES) and Sanger sequencing to identify and validate candidate variants. Computational predictions of splicing defects were performed using varSEAK and MobiDetails. Functional validation was conducted using minigene splicing assays in HEK293T cells. Structural modeling of mutant proteins was performed with AlphaFold3 and visualized by PyMOL. Two novel splice-site variants (DNAH17: c.11677 + 5G > T/c.11677 + 5G > A) were identified in a proband with asthenozoospermia and multiple morphological abnormalities of the sperm flagella (MMAF). Bioinformatics tools predicted disruption of the canonical donor splice site (MaxEntScan score reduction: 54.2% for G > A, 39.5% for G > T; SpliceAI donor loss scores > 0.8). Minigene assays confirmed exon 72 skipping, leading to a frameshift mutation (p.Asn3844Lysfs*13) that truncates the AAA6 domain. This study expands the mutational spectrum of DNAH17-related male infertility by demonstrating that splice-site variants disrupting the AAA6 domain represent a novel pathogenic mechanism underlying asthenozoospermia and MMAF. These findings underscore the necessity of integrating splice-site analysis into genetic diagnostics for male infertility.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3899-3905"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145550272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-13DOI: 10.1007/s43032-025-01998-1
Shoukat Hussain, Alia Javed, Dur EMaknoon Razia, Naosheen Ashiq, Muhammad Javid Iqbal, Muhammad Jahangeer, Uzma Shaheen, Muhammad Yameen, Asif Shahzad
Male infertility accounts for nearly half of all infertility cases globally, often resulting from oxidative stress and hormonal dysregulation. Conventional treatments offer limited efficacy, prompting interest in multi-target botanical therapies. This study evaluates a standardized polyherbal formulation (PHP), comprising Mucuna pruriens, Anacyclus pyrethrum, Asparagus racemosus, and Tribulus terrestris, for its protective effects against carbon tetrachloride (CCl4)-induced reproductive toxicity. In vitro, assays confirmed the antioxidant capacity of PHP, with dose-dependent radical scavenging in DPPH and FRAP assays and inhibition of GC-1/GC-2 spermatogonia cell proliferation. In vivo, PHP administration (100, 300, 500 mg/kg) in carbon tetrachloride (CCl4)-exposed rats significantly reduced oxidative stress, including a 52% reduction in thiobarbituric acid reactive substances (TBARS), 47% reduction in hydrogen perocide (H2O2), and normalization of enzymatic antioxidants glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and peroxidase (POD); P < 0.01 vs. CCl4. Hepatic injury biomarkedly improved, with alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin levels reduced by 40-55% compared to CCl4 group (P < 0.01). PHP also restored reproductive hormones testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH), and Prolactin levels elevated by 1.8 to 2.2-fold relative to intoxicated controls (p < 0.05). Gene expression analysis revealed significant recovery, with sex-hormone-binding globulin (SHBG), protein kinase B (AKT1), and androgen receptor (AR) transcripts upregulated 2.0-2.5-fold in PHP-treated animals (p < 0.05 vs. CCl4), approaching control values. Histopathological analysis confirmed structural restoration of testicular architecture, with reduced seminiferous tubule degeneration and active spermatogenesis. In silico docking identified luteolin and acacetin as key phytochemicals. Luteolin exhibits strong binding to the androgen receptor (AR: -9.6 kcal/mol), AKT1 (-9.9 kcal/mol), and SHBG (-9.5 kcal/mol). It complied with Lipinski's rule of five and displayed favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. Collectively, these findings suggest that PHP exerts multi-target protective effects against CCl4-induced male infertility by mitigating oxidative stress, restoring hormonal balance, and modulating key reproductive signaling pathways. Further mechanistic and clinical investigations are warranted to validate its therapeutic potential.
{"title":"Polyherbal Therapeutics Mitigate CCl<sub>4</sub>-Induced Testicular Toxicity through Modulation of SHBG, AKT1, and AR Pathways: An Integrated In Vitro, In Vivo, and In Silico Approach.","authors":"Shoukat Hussain, Alia Javed, Dur EMaknoon Razia, Naosheen Ashiq, Muhammad Javid Iqbal, Muhammad Jahangeer, Uzma Shaheen, Muhammad Yameen, Asif Shahzad","doi":"10.1007/s43032-025-01998-1","DOIUrl":"10.1007/s43032-025-01998-1","url":null,"abstract":"<p><p>Male infertility accounts for nearly half of all infertility cases globally, often resulting from oxidative stress and hormonal dysregulation. Conventional treatments offer limited efficacy, prompting interest in multi-target botanical therapies. This study evaluates a standardized polyherbal formulation (PHP), comprising Mucuna pruriens, Anacyclus pyrethrum, Asparagus racemosus, and Tribulus terrestris, for its protective effects against carbon tetrachloride (CCl<sub>4</sub>)-induced reproductive toxicity. In vitro, assays confirmed the antioxidant capacity of PHP, with dose-dependent radical scavenging in DPPH and FRAP assays and inhibition of GC-1/GC-2 spermatogonia cell proliferation. In vivo, PHP administration (100, 300, 500 mg/kg) in carbon tetrachloride (CCl<sub>4</sub>)-exposed rats significantly reduced oxidative stress, including a 52% reduction in thiobarbituric acid reactive substances (TBARS), 47% reduction in hydrogen perocide (H<sub>2</sub>O<sub>2</sub>), and normalization of enzymatic antioxidants glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and peroxidase (POD); P < 0.01 vs. CCl<sub>4</sub>. Hepatic injury biomarkedly improved, with alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin levels reduced by 40-55% compared to CCl<sub>4</sub> group (P < 0.01). PHP also restored reproductive hormones testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH), and Prolactin levels elevated by 1.8 to 2.2-fold relative to intoxicated controls (p < 0.05). Gene expression analysis revealed significant recovery, with sex-hormone-binding globulin (SHBG), protein kinase B (AKT1), and androgen receptor (AR) transcripts upregulated 2.0-2.5-fold in PHP-treated animals (p < 0.05 vs. CCl<sub>4</sub>), approaching control values. Histopathological analysis confirmed structural restoration of testicular architecture, with reduced seminiferous tubule degeneration and active spermatogenesis. In silico docking identified luteolin and acacetin as key phytochemicals. Luteolin exhibits strong binding to the androgen receptor (AR: -9.6 kcal/mol), AKT1 (-9.9 kcal/mol), and SHBG (-9.5 kcal/mol). It complied with Lipinski's rule of five and displayed favorable absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties. Collectively, these findings suggest that PHP exerts multi-target protective effects against CCl<sub>4</sub>-induced male infertility by mitigating oxidative stress, restoring hormonal balance, and modulating key reproductive signaling pathways. Further mechanistic and clinical investigations are warranted to validate its therapeutic potential.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3976-4002"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506592","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-13DOI: 10.1007/s43032-025-02005-3
Haruo Usuda, Hideyuki Ikeda, Shimpei Watanabe, Erin L Johnson, Sean W D Carter, Yusaku Kumagai, Yuya Saito, Michelle Seah, Binny P Sesurajan, Tsukasa Takahashi, Noriyoshi Mochi, Kantarou Sahara, Hannah Watson, Shinichi Kawamura, Masatoshi Saito, Matthew W Kemp
Fetal hypoxia remains a major unresolved clinical challenge. Using artificial placenta (AP) technology, we established a standardized model of progressive hypoxia that induces brain injury in fetal sheep. To support the development of new antenatal monitoring strategies, we further profiled gestation-specific physiological, biochemical, protein, and cfRNA responses during progressive hypoxia. Seven fetuses at 123 days gestational age (dGA) and eight fetuses at 98 dGA (term = 150 dGA) were supported on the AP. Following a 12-h stabilization period, during which FiO₂ and gas flow to the AP oxygenator were adjusted to maintain fetal SO₂ at 65-75% and pCO₂ at 35-45 mmHg, both parameters were reduced by 5% every 30 min until termination criteria were met (arterial pH < 7.0 or base excess < -12). This protocol produced 240-300 min of progressive hypoxia. Physiological parameters, blood gases, biochemical markers, and cardiac and vascular ultrasound were assessed hourly. At the end of the experiment, brain histology and plasma cfRNA profiling were performed. Progressive hypoxia induced significant increases in pCO₂ and lactate and decreases in SO₂, CtO₂, pH, and BE in both groups (p < 0.05). Gestation-specific differences were observed in pO₂, glucose, and vital signs (heart rate, mean arterial pressure, and circuit flow). AST, ALT, and NSE rose significantly only at 123 dGA (p < 0.05), whereas S100B and NT-proBNP increased in both groups. Histology confirmed hypoxic brain injury at both gestational ages. In 123 dGA fetuses, iliac artery pulsatility index and heart rate predicted dysregulation of pH, BE, SO₂, and lactate (R2 = 0.538-0.745, p < 0.05). Plasma cfRNA analysis demonstrated distinct gestation-specific gene expression patterns consistent with hypoxia-induced injury. We developed a standardized, regulable model of fetal hypoxia and demonstrated brain injury accompanied by gestation-specific physiological, biochemical, and transcriptomic responses. Plasma cfRNA profiling highlighted its potential as an antenatal biomarker of hypoxic insult. This platform may facilitate the development of new diagnostic tools and guide evaluation of postnatal interventions.
胎儿缺氧仍然是一个主要的未解决的临床挑战。采用人工胎盘(AP)技术,建立了羊胎进行性缺氧诱发脑损伤的标准化模型。为了支持新的产前监测策略的发展,我们进一步分析了进行性缺氧期间妊娠特异性生理、生化、蛋白质和cfRNA反应。7个胎龄为123天(dGA)的胎儿和8个胎龄为98天(足月= 150 dGA)的胎儿被支持在AP上。经过12小时的稳定期,在此期间调整FiO₂和AP氧合器的气体流量以维持胎儿SO₂在65-75%和pCO₂在35-45 mmHg,这两个参数每30分钟降低5%,直到满足终止标准(动脉pH 2 = 0.538-0.745, p
{"title":"Identification of Gestation-Specific Patterns of Physiological, Protein and Cell-Free RNA Injury Markers in a Sheep Model of Regulable Preterm Fetal Hypoxia.","authors":"Haruo Usuda, Hideyuki Ikeda, Shimpei Watanabe, Erin L Johnson, Sean W D Carter, Yusaku Kumagai, Yuya Saito, Michelle Seah, Binny P Sesurajan, Tsukasa Takahashi, Noriyoshi Mochi, Kantarou Sahara, Hannah Watson, Shinichi Kawamura, Masatoshi Saito, Matthew W Kemp","doi":"10.1007/s43032-025-02005-3","DOIUrl":"10.1007/s43032-025-02005-3","url":null,"abstract":"<p><p>Fetal hypoxia remains a major unresolved clinical challenge. Using artificial placenta (AP) technology, we established a standardized model of progressive hypoxia that induces brain injury in fetal sheep. To support the development of new antenatal monitoring strategies, we further profiled gestation-specific physiological, biochemical, protein, and cfRNA responses during progressive hypoxia. Seven fetuses at 123 days gestational age (dGA) and eight fetuses at 98 dGA (term = 150 dGA) were supported on the AP. Following a 12-h stabilization period, during which FiO₂ and gas flow to the AP oxygenator were adjusted to maintain fetal SO₂ at 65-75% and pCO₂ at 35-45 mmHg, both parameters were reduced by 5% every 30 min until termination criteria were met (arterial pH < 7.0 or base excess < -12). This protocol produced 240-300 min of progressive hypoxia. Physiological parameters, blood gases, biochemical markers, and cardiac and vascular ultrasound were assessed hourly. At the end of the experiment, brain histology and plasma cfRNA profiling were performed. Progressive hypoxia induced significant increases in pCO₂ and lactate and decreases in SO₂, CtO₂, pH, and BE in both groups (p < 0.05). Gestation-specific differences were observed in pO₂, glucose, and vital signs (heart rate, mean arterial pressure, and circuit flow). AST, ALT, and NSE rose significantly only at 123 dGA (p < 0.05), whereas S100B and NT-proBNP increased in both groups. Histology confirmed hypoxic brain injury at both gestational ages. In 123 dGA fetuses, iliac artery pulsatility index and heart rate predicted dysregulation of pH, BE, SO₂, and lactate (R<sup>2</sup> = 0.538-0.745, p < 0.05). Plasma cfRNA analysis demonstrated distinct gestation-specific gene expression patterns consistent with hypoxia-induced injury. We developed a standardized, regulable model of fetal hypoxia and demonstrated brain injury accompanied by gestation-specific physiological, biochemical, and transcriptomic responses. Plasma cfRNA profiling highlighted its potential as an antenatal biomarker of hypoxic insult. This platform may facilitate the development of new diagnostic tools and guide evaluation of postnatal interventions.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3906-3927"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506653","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abnormal levels of Inhibin B (INH-B), a major regulator of ovarian activity, are closely linked to the development and prognosis of several ovarian disorders. Understanding the molecular mechanisms governing its regulation in granulosa cells is essential for both diagnosis and therapy. Our earlier work demonstrated the precise localization of lncPrep + 96 kb in granulosa cells and its central influence on the estrogen biosynthetic pathway. In this study, the impact of lncPrep + 96 kb on INH-B expression was investigated further. We created knockout mice lacking the long non-coding RNA lncPrep + 96 kb, which is specifically expressed in granulosa cells of the ovary. RNA sequencing revealed that the inhibin subunit βB (INHBB) was significantly elevated in knockout mice. ELISA was utilized to quantify INH-B levels in serum and granulosa cell supernatants, revealing a significant increase in knockout mice compared to wild-type controls. Overexpression of lncPrep + 96 kb fragments (2.2 kb and 2.8 kb) reduced INH-B expression. Endothelial differentiation-related factor 1 (EDF1), a key intracellular transcription factor, was found to be upregulated by lncPrep + 96 kb, resulting in decreased INH-B expression. In summary, lncPrep + 96 kb regulates INH-B secretion in granulosa cells by modulating of EDF1, providing new insights into the mechanism of INH-B expression and offering new research directions for diagnostic and therapeutic studies of abnormal ovarian functions.
{"title":"LncPrep + 96 kb Inhibits the Secretion of Inhibin B in Ovarian Granulosa Cells Through Regulating Endothelial Differentiation-Associated Factor 1.","authors":"Hongdan Zhang, Jianwei Liu, Canglang Mou, Yingqi Tang, Yangfan Lv","doi":"10.1007/s43032-025-02007-1","DOIUrl":"10.1007/s43032-025-02007-1","url":null,"abstract":"<p><p>Abnormal levels of Inhibin B (INH-B), a major regulator of ovarian activity, are closely linked to the development and prognosis of several ovarian disorders. Understanding the molecular mechanisms governing its regulation in granulosa cells is essential for both diagnosis and therapy. Our earlier work demonstrated the precise localization of lncPrep + 96 kb in granulosa cells and its central influence on the estrogen biosynthetic pathway. In this study, the impact of lncPrep + 96 kb on INH-B expression was investigated further. We created knockout mice lacking the long non-coding RNA lncPrep + 96 kb, which is specifically expressed in granulosa cells of the ovary. RNA sequencing revealed that the inhibin subunit βB (INHBB) was significantly elevated in knockout mice. ELISA was utilized to quantify INH-B levels in serum and granulosa cell supernatants, revealing a significant increase in knockout mice compared to wild-type controls. Overexpression of lncPrep + 96 kb fragments (2.2 kb and 2.8 kb) reduced INH-B expression. Endothelial differentiation-related factor 1 (EDF1), a key intracellular transcription factor, was found to be upregulated by lncPrep + 96 kb, resulting in decreased INH-B expression. In summary, lncPrep + 96 kb regulates INH-B secretion in granulosa cells by modulating of EDF1, providing new insights into the mechanism of INH-B expression and offering new research directions for diagnostic and therapeutic studies of abnormal ovarian functions.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"4003-4014"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145445778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Polycystic ovary syndrome (PCOS) is a common endocrinopathy affecting 5-20% of reproductive-age women. Besides genetic factors, environmental triggers are considered major underlying causes that play a role through epigenetic alterations. Since epigenetic studies can be a possible link to explain the complexity of multifactorial disorders, it is worthwhile to investigate differentially methylated regions (DMRs) associated with PCOS. Therefore, a systematic review was conducted to identify epigenetic modifications associated with PCOS and analyse their functional role in the pathogenesis of the syndrome. After following inclusion and exclusion criteria, various databases (PubMed, Google Scholar, Central Cochrane Library, and Science Direct) were searched with predetermined keywords up to 30th June 2025, and quality of the selected articles assessed through the Newcastle Ottawa scale (NOS). In-silico analysis was performed by STRING, and Shiny GO. A total of 46 eligible studies were included, were further sub-categorized based on the sample type. A total of one hundred eleven genes were identified with altered DMRs, out of which thirty seven genes were reported from the ovarian tissues alone. Their GO analysis shows a significant interaction with other PCOS-susceptible genes and their functions in female sex differentiation and gonadal development. However, in-silico analysis of thirty six genes identified in blood showed their functions were significantly involved in the insulin-like growth factor binding activity. Gene enrichment analysis of fifteen genes with altered methylation identified in adipose tissue shows the significant involvement of female sex differentiation and insulin-activated receptor functions. Further, alterations in methylation of which genes (INSR, AMHR2, YAP1, CYP19A1, LHCGR, CDKN1A, LINE-1, AMH and TOX3) have been reported by at least two separate studies, irrespective of sample type. Their in-silico analysis shows significant involvement of these genes in the female sex characters, and gonadal development The current study highlights tissue-specific epigenetic modifications in PCOS, summarizes DMRs of genes which are involved in reproductive and insulin-related pathways. DMRs of identified genes may have a role in the pathogenesis of PCOS, insight into which may provide novel therapeutic targets in the future.
{"title":"Differential Methylation Signatures Associated with PCOS- A Systematic Review and In-Silico Analysis.","authors":"Priya Sharma, Ahelee Ghosal, Aiswarya Vs, Shweta Daryani, Tulsi Brahma, Preeti Khetarpal","doi":"10.1007/s43032-025-01976-7","DOIUrl":"10.1007/s43032-025-01976-7","url":null,"abstract":"<p><p>Polycystic ovary syndrome (PCOS) is a common endocrinopathy affecting 5-20% of reproductive-age women. Besides genetic factors, environmental triggers are considered major underlying causes that play a role through epigenetic alterations. Since epigenetic studies can be a possible link to explain the complexity of multifactorial disorders, it is worthwhile to investigate differentially methylated regions (DMRs) associated with PCOS. Therefore, a systematic review was conducted to identify epigenetic modifications associated with PCOS and analyse their functional role in the pathogenesis of the syndrome. After following inclusion and exclusion criteria, various databases (PubMed, Google Scholar, Central Cochrane Library, and Science Direct) were searched with predetermined keywords up to 30th June 2025, and quality of the selected articles assessed through the Newcastle Ottawa scale (NOS). In-silico analysis was performed by STRING, and Shiny GO. A total of 46 eligible studies were included, were further sub-categorized based on the sample type. A total of one hundred eleven genes were identified with altered DMRs, out of which thirty seven genes were reported from the ovarian tissues alone. Their GO analysis shows a significant interaction with other PCOS-susceptible genes and their functions in female sex differentiation and gonadal development. However, in-silico analysis of thirty six genes identified in blood showed their functions were significantly involved in the insulin-like growth factor binding activity. Gene enrichment analysis of fifteen genes with altered methylation identified in adipose tissue shows the significant involvement of female sex differentiation and insulin-activated receptor functions. Further, alterations in methylation of which genes (INSR, AMHR2, YAP1, CYP19A1, LHCGR, CDKN1A, LINE-1, AMH and TOX3) have been reported by at least two separate studies, irrespective of sample type. Their in-silico analysis shows significant involvement of these genes in the female sex characters, and gonadal development The current study highlights tissue-specific epigenetic modifications in PCOS, summarizes DMRs of genes which are involved in reproductive and insulin-related pathways. DMRs of identified genes may have a role in the pathogenesis of PCOS, insight into which may provide novel therapeutic targets in the future.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3833-3847"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145233273","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-12DOI: 10.1007/s43032-025-02006-2
Hui Feng, Yuhong Lin, Xiaoqi Zhao, Caiyun Ge, Wen Zhong, Xiaomeng Zha, Man Fang, Hui Wang, Ming Zhang
The "double-edged sword" effect of dexamethasone on fetal development has attracted significant attention. Currently, the "one-size-fits-all" prenatal corticosteroid regimen fails to confer uniform benefits to all pregnant women and fetuses. Simultaneously, there exist controversies regarding the wide time window for administration and the insufficient consideration of individual factors in dosage selection. Therefore, this study aimed to establish a precise prenatal dexamethasone exposure (PDE) mouse model that closely mimics clinical use, to evaluate its effects on placental morphology, development, differentiation, vascular formation, and nutrient transporter function. Dexamethasone was injected subcutaneously at diverse gestational stages, doses, and courses. The late-stage, high-dose, single-course PDE exhibited the most pronounced effects, including reduced placental weight, a decreased labyrinth-to-junctional zone (LZ/JZ) ratio, impaired trophoblast proliferation and differentiation, increased apoptosis, and decreased vascular endothelial growth factor (VEGF) expression. Amino acid and cholesterol transporter levels increased in both sexes, whereas glucose transporters showed sex-specific alterations-elevated in males but reduced in females. Further investigation revealed that PDE suppressed the "glucocorticoid (GC)-insulin-like growth factor 1 (IGF1) axis" programming, which was highly correlated with placental development and function indicators. In conclusion, PDE induced alterations in placental morphology, development, and nutrient transport function, which were influenced by stages, doses, courses, and sex differences. These changes may be associated with the "GC-IGF1 axis" programming. This study provides experimental and theoretical evidence to more precisely guide the clinical application of prenatal dexamethasone.
{"title":"The Effect of Prenatal Dexamethasone Exposure on Placental Morphology and Function at Different Stages, Doses, and Courses In Mice.","authors":"Hui Feng, Yuhong Lin, Xiaoqi Zhao, Caiyun Ge, Wen Zhong, Xiaomeng Zha, Man Fang, Hui Wang, Ming Zhang","doi":"10.1007/s43032-025-02006-2","DOIUrl":"10.1007/s43032-025-02006-2","url":null,"abstract":"<p><p>The \"double-edged sword\" effect of dexamethasone on fetal development has attracted significant attention. Currently, the \"one-size-fits-all\" prenatal corticosteroid regimen fails to confer uniform benefits to all pregnant women and fetuses. Simultaneously, there exist controversies regarding the wide time window for administration and the insufficient consideration of individual factors in dosage selection. Therefore, this study aimed to establish a precise prenatal dexamethasone exposure (PDE) mouse model that closely mimics clinical use, to evaluate its effects on placental morphology, development, differentiation, vascular formation, and nutrient transporter function. Dexamethasone was injected subcutaneously at diverse gestational stages, doses, and courses. The late-stage, high-dose, single-course PDE exhibited the most pronounced effects, including reduced placental weight, a decreased labyrinth-to-junctional zone (LZ/JZ) ratio, impaired trophoblast proliferation and differentiation, increased apoptosis, and decreased vascular endothelial growth factor (VEGF) expression. Amino acid and cholesterol transporter levels increased in both sexes, whereas glucose transporters showed sex-specific alterations-elevated in males but reduced in females. Further investigation revealed that PDE suppressed the \"glucocorticoid (GC)-insulin-like growth factor 1 (IGF1) axis\" programming, which was highly correlated with placental development and function indicators. In conclusion, PDE induced alterations in placental morphology, development, and nutrient transport function, which were influenced by stages, doses, courses, and sex differences. These changes may be associated with the \"GC-IGF1 axis\" programming. This study provides experimental and theoretical evidence to more precisely guide the clinical application of prenatal dexamethasone.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3943-3959"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145506627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-14DOI: 10.1007/s43032-025-01993-6
Habibollah Khodabandeh, Mohammad Amin Edalatmanesh, Mohsen Forouzanfar
Uteroplacental insufficiency (UPI) disrupts fetal brain development and induces oxidative damage. This study evaluates the neuroprotective effects of ferulic acid (FA) on oxidative stress biomarkers, neuroinflammation, the Akt/GSK-3β signaling pathway, and neuronal density in the medial prefrontal cortex (mPFC) following UPI in rats. Twenty pregnant Wistar rats were randomly assigned to four groups: Control, Sham Surgery, UPI + Vehicle (UPI + normal saline), and UPI + FA (UPI + FA at 100 mg/kg). UPI was induced via permanent ligation of the uterine arteries on embryonic day (ED) 18. FA or normal saline was administered orally from ED14 to ED21. On ED21, fetal brain tissue was analyzed for oxidative stress biomarkers (8-hydroxy-2'-deoxyguanosine, protein carbonyl, 4-hydroxy-2-nonenal, and malondialdehyde), inflammatory cytokines (interleukin-6 [IL-6], IL-1β, tumor necrosis factor-alpha [TNF-α], and IL-10), Akt/GSK-3β gene expression, and neuronal density in the mPFC. FA treatment significantly reduced oxidative stress biomarkers and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) while decreasing GSK-3β expression and increasing Akt expression. Additionally, FA enhanced neuronal density in the mPFC and elevated IL-10 levels compared to the UPI + Vehicle group (p < 0.05). Pre-treatment with FA prior to UPI induction mitigated oxidative stress, modulated the Akt/GSK-3β signaling pathway, suppressed neuroinflammation, and preserved cortical integrity in the fetal brain.
{"title":"The Effect of Ferulic Acid on the Akt-GSK3β Signaling Pathway, Neuroinflammation, Oxidative Stress, and Cortical Damage in the Fetal Brain with Uteroplacental Insufficiency.","authors":"Habibollah Khodabandeh, Mohammad Amin Edalatmanesh, Mohsen Forouzanfar","doi":"10.1007/s43032-025-01993-6","DOIUrl":"10.1007/s43032-025-01993-6","url":null,"abstract":"<p><p>Uteroplacental insufficiency (UPI) disrupts fetal brain development and induces oxidative damage. This study evaluates the neuroprotective effects of ferulic acid (FA) on oxidative stress biomarkers, neuroinflammation, the Akt/GSK-3β signaling pathway, and neuronal density in the medial prefrontal cortex (mPFC) following UPI in rats. Twenty pregnant Wistar rats were randomly assigned to four groups: Control, Sham Surgery, UPI + Vehicle (UPI + normal saline), and UPI + FA (UPI + FA at 100 mg/kg). UPI was induced via permanent ligation of the uterine arteries on embryonic day (ED) 18. FA or normal saline was administered orally from ED14 to ED21. On ED21, fetal brain tissue was analyzed for oxidative stress biomarkers (8-hydroxy-2'-deoxyguanosine, protein carbonyl, 4-hydroxy-2-nonenal, and malondialdehyde), inflammatory cytokines (interleukin-6 [IL-6], IL-1β, tumor necrosis factor-alpha [TNF-α], and IL-10), Akt/GSK-3β gene expression, and neuronal density in the mPFC. FA treatment significantly reduced oxidative stress biomarkers and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) while decreasing GSK-3β expression and increasing Akt expression. Additionally, FA enhanced neuronal density in the mPFC and elevated IL-10 levels compared to the UPI + Vehicle group (p < 0.05). Pre-treatment with FA prior to UPI induction mitigated oxidative stress, modulated the Akt/GSK-3β signaling pathway, suppressed neuroinflammation, and preserved cortical integrity in the fetal brain.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3960-3975"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145523769","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Recent advances in microbiome research have illuminated the complex bidirectional interactions between gut health and reproductive well-being. Understanding the gut microbiome's influence on the reproductive system and vice versa reveals how both of them can affect hormone production, immune function, and ultimately overall reproductive health. Dysbiosis, an imbalance in the gut microbial community, has been linked with a range of reproductive issues, including decreased sperm count and motility, erectile dysfunction, polycystic ovary syndrome (PCOS), endometriosis, infertility, and adverse pregnancy outcomes. This review critically evaluates emerging therapeutic interventions aimed at restoring microbial balance and enhancing reproductive health, such as use of prebiotics, probiotics, bacteriophage therapy, and fecal microbiota transplantation (FMT). By exploring the intricate interplay between gut microbiota and reproductive health, this review also emphasizes the need for integrated approaches in research and clinical practice to develop effective microbiome-based therapies for better reproductive health outcomes.
{"title":"From Gut to Reproductive Health: Exploring Microbiome Interactions and Future Interventions.","authors":"Samradhi Singh, Mona Kriti, Poonam Sharma, Namrata Pal, Devojit Kumar Sarma, Vinod Verma, Rajnarayan R Tiwari, Manoj Kumar","doi":"10.1007/s43032-025-02001-7","DOIUrl":"10.1007/s43032-025-02001-7","url":null,"abstract":"<p><p>Recent advances in microbiome research have illuminated the complex bidirectional interactions between gut health and reproductive well-being. Understanding the gut microbiome's influence on the reproductive system and vice versa reveals how both of them can affect hormone production, immune function, and ultimately overall reproductive health. Dysbiosis, an imbalance in the gut microbial community, has been linked with a range of reproductive issues, including decreased sperm count and motility, erectile dysfunction, polycystic ovary syndrome (PCOS), endometriosis, infertility, and adverse pregnancy outcomes. This review critically evaluates emerging therapeutic interventions aimed at restoring microbial balance and enhancing reproductive health, such as use of prebiotics, probiotics, bacteriophage therapy, and fecal microbiota transplantation (FMT). By exploring the intricate interplay between gut microbiota and reproductive health, this review also emphasizes the need for integrated approaches in research and clinical practice to develop effective microbiome-based therapies for better reproductive health outcomes.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3816-3832"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145422636","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-05-23DOI: 10.1007/s43032-025-01879-7
Mei Lv, Anni Feng, Di Cheng, Zejun Xu, Yuanjie Xie, Jian Tu
Polycystic ovary syndrome (PCOS) is a common and frequent disease and always leads to endocrine and metabolic disorders among women of reproductive age. In most PCOS patients, the serum level of anti-mullerian hormone (AMH) is significantly higher than that of normal women of childbearing age. AMH is an important auxiliary diagnostic method for PCOS. AMH may play an important role in the occurrence and development of PCOS and potentially affect the success of in vitro fertilization (IVF) treatments, although the exact mechanisms remain unclear. This review focuses on the relationships and mechanisms of dysregulated AMH and follicular development in PCOS based on recent research. It is mainly manifested in the regulation of related signal pathways, the negative feedback regulatory loop of several gonadal hormones such as follicle-stimulating hormone, luteinizing hormone, estradiol, and testosterone, as well as the influence of AMH on IVF in PCOS patients. Additionally, it puts forward some suggestions, which will provide some helpful ideas or directions for future further research on PCOS.
{"title":"The Effect of AMH on Folliculogenesis.","authors":"Mei Lv, Anni Feng, Di Cheng, Zejun Xu, Yuanjie Xie, Jian Tu","doi":"10.1007/s43032-025-01879-7","DOIUrl":"10.1007/s43032-025-01879-7","url":null,"abstract":"<p><p>Polycystic ovary syndrome (PCOS) is a common and frequent disease and always leads to endocrine and metabolic disorders among women of reproductive age. In most PCOS patients, the serum level of anti-mullerian hormone (AMH) is significantly higher than that of normal women of childbearing age. AMH is an important auxiliary diagnostic method for PCOS. AMH may play an important role in the occurrence and development of PCOS and potentially affect the success of in vitro fertilization (IVF) treatments, although the exact mechanisms remain unclear. This review focuses on the relationships and mechanisms of dysregulated AMH and follicular development in PCOS based on recent research. It is mainly manifested in the regulation of related signal pathways, the negative feedback regulatory loop of several gonadal hormones such as follicle-stimulating hormone, luteinizing hormone, estradiol, and testosterone, as well as the influence of AMH on IVF in PCOS patients. Additionally, it puts forward some suggestions, which will provide some helpful ideas or directions for future further research on PCOS.</p>","PeriodicalId":20920,"journal":{"name":"Reproductive Sciences","volume":" ","pages":"3848-3860"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144128549","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}