慢性炎症及代谢与多囊卵巢综合征及其相关治疗的研究进展
Research Progress on Chronic Inflammation and Polycystic Ovary Syndrome and Related Treatments
DOI: 10.12677/acm.2024.1441190, PDF, HTML, XML, 下载: 29  浏览: 43 
作者: 王明伟:济宁医学院临床医学院,山东 济宁;姚红梅*:济宁医学院附属医院,山东 济宁
关键词: 多囊卵巢综合征炎症治疗Polycystic Ovary Syndrome Inflammation Therapy
摘要: 多囊卵巢综合征(Polycystic Ovary Syndrome, PCOS)是育龄期女性最常见的内分泌代谢紊乱疾病,以排卵障碍、高雄激素血症及卵巢多囊样改变为特征,主要临床表现为月经不规律、不孕、多毛和/或痤疮等。虽然PCOS的发病机制尚不清楚,但是其与低度慢性炎症密切相关。本研究对外周血炎症、卵巢炎症、子宫内膜炎症与PCOS的发病机制的关系,以及炎症因子、肥胖、胰岛素抵抗和高雄激素血症之间的相互作用和其相关的治疗方法进行总结。
Abstract: Polycystic ovary syndrome (PCOS) is the most common endocrine and metabolic disorder in women of childbearing age, characterized by ovulation disorders, hyperandrogenism, and polycystic ovary like changes. The main clinical manifestations include irregular menstruation, infertility, hirsutism, and/or acne. Although the pathogenesis of PCOS is still unclear, it is closely related to low-grade chronic inflammation. This study summarizes the relationship between peripheral blood inflammation, ovarian inflammation, endometritis, and the pathogenesis of PCOS, as well as the interaction and related treatment methods between inflammatory factors, obesity, insulin resistance, and hyperandrogenism.
文章引用:王明伟, 姚红梅. 慢性炎症及代谢与多囊卵巢综合征及其相关治疗的研究进展[J]. 临床医学进展, 2024, 14(4): 1547-1554. https://doi.org/10.12677/acm.2024.1441190

1. 引言

PCOS是全球妇科最常见的内分泌代谢疾病之一,占育龄期女性的6%~13% [1] 。我国女性PCOS患病率为5.6%,且发病率呈现逐年上升的趋势 [2] 。PCOS常伴有多种并发症,如不孕、肥胖、胰岛素抵抗(insulin resistance, IR)、心血管疾病及其他健康问题等,这些并发症会严重降低患者的生活质量,甚至影响预期寿命 [3] 。随着对PCOS的深入研究,越来越多的学者认为氧化应激(oxidative stress, OS)和慢性炎症是PCOS致病的主要因素,并可通过相关途径引起PCOS患者高雄激素血症、排卵功能障碍和IR等症状 [4] 。

炎症是一种由宿主的防御系统与致病微生物之间相互识别和相互作用引发的先天免疫反应,是机体发挥免疫功能的重要屏障之一 [5] 。炎症可分为急性和慢性,如果炎症因子不能在短期内清除或在机体内持续存在,导致炎症过程迁延不愈,急性炎症就会转化为慢性炎症 [6] 。全身慢性炎症是由细胞因子和炎性趋化因子的产生而引起的,包括白细胞介素(interleukin, IL)-1、IL-6、IL-10、IL-12、一氧化氮(Nitric Oxide, NO)和肿瘤坏死因子α (tumor necrosis factor-α, TNF-α)等,它们会影响卵巢功能,阻碍卵泡的生长和发育,影响PCOS的疾病进程 [7] 。由于许多研究认为PCOS是一种代谢综合征,本文将对相关炎症因子在PCOS发病机制中的影响及其相关治疗归纳总结如下。

2. 肥胖、胰岛素抵抗、高雄激素血症与炎症

包括遗传、内分泌、新陈代谢和环境等在内的许多因素都与PCOS疾病发生机制有关。肥胖、IR、高雄激素血症和慢性炎症等可以单独或相互作用,对PCOS女性产生一定的影响。先前的报道中提到大多数的PCOS女性患有糖耐量受损和2型糖尿病,此外肥胖患者发病率更高 [8] ,尤其是体内内脏脂肪堆积,其可引起低度慢性炎症,导致高胰岛素血症、胰岛素抵抗和高雄激素症等。然而,非肥胖的多囊卵巢综合征患者也可能出现内脏脂肪堆积从而导致代谢异常 [9] 。患有PCOS的肥胖女性可能存在葡萄糖和脂质分解代谢异常,比非肥胖PCOS女性更容易发生IR;此外,胰岛素的增加不仅刺激卵巢卵泡生长、激素分泌和卵巢类固醇生成,增加雄烯二酮和睾酮的产生,还会增加血液中游离睾酮水平和卵泡中雄激素水平,加速颗粒细胞凋亡,抑制卵泡生成,此外,还通过影响脑垂体和下丘脑,增加黄体生成素和促性腺激素释放激素的产生,从而影响卵泡生长,导致排卵功能障碍 [10] 。65%~70%的PCOS患者中伴有IR,特别是肥胖PCOS女性,但是正常体重健康女性中也可能会伴有IR [11] 。Khichar等 [12] 研究了PCOS患者中炎症因素与IR的相关性,其研究发现PCOS组存在着高胰岛素血症和IR,其血清中的超敏C反应蛋白(hs-CRP)和C3也较对照组升高,并且高胰岛素血症和IR与hs-CRP和C3呈正相关。一项分析报告表明,炎症因子产生的主要部位是脂肪组织,与正常体脂非肥胖者相比,正常体重肥胖者(体重正常但体脂百分比高)具有较高水平的CRP和IL-6 [13] ,会因肥胖和高胰岛素血症而加重。Stokkeland等 [14] 研究发现,妊娠期PCOS患者的血清炎症因子受到胎儿性别的显著影响,在孕早期和孕中期,孕育男性胎儿的PCOS女性的血清中IL-1Ra、IL-2、IL-7、MCP-1和碱性成纤维细胞生长因子(Basic fibroblast growth factor, FGF-b)水平均高于孕育女性胎儿的妇女。这一发现揭露了炎症水平与高雄激素水平的相关关系。这些研究结果表明PCOS女性中肥胖、胰岛素抵抗、高雄激素血症和炎症之间具有相关性,但其具体的作用机制及相关信号通路仍需进一步研究。

3. PCOS与炎症

3.1. 外周血炎症与PCOS

子宫多囊卵巢综合征的发病机制复杂,受多种遗传、环境和激素因素的影响,研究表明慢性低度炎症可能是PCOS发病机制和疾病发展过程中的一个重要因素 [15] 。

He等 [16] 研究发现,PCOS患者血液中的白细胞(white blood cell, WBC)计数、中性粒细胞计数和淋巴细胞计数明显增加,且总T淋巴细胞计数、CD4+T细胞和自然杀伤细胞(natural killer cell, NK)的百分比显著增加。上述文献表明PCOS的发病机制与炎症因子及免疫细胞有密切的关系。但Nadiah A Alhabardi等 [17] 研究中认为PCOS患者的WBC、中性粒细胞计数和淋巴细胞计数在内的血液学参数水平与健康对照组没有显著差异。所以,慢性炎症在PCOS女性血清中的作用机制仍需进一步研究。

PCOS患者中存在着慢性低度炎症,肥胖患者中也存在慢性低度炎症,因此肥胖的PCOS女性的慢性炎症状态无法确定于来自PCOS本身还是肥胖,此问题存在争议。Gonzalez等 [18] 首次证实了PCOS与血清中TNF-α之间的关系,研究结果表明正常体重PCOS女性的血清TNF-α平均浓度显著高于相应对照组,且无论是否患有PCOS,肥胖患者的TNF-α水平都要高。这项研究表明,PCOS患者血清中TNF-α的升高与肥胖无关。为了研究PCOS患者的WBC计数和CRP浓度,Rudnicka [19] 等将200名18~40岁PCOS女性与105名年龄分布、身体质量指数(body mass index, BMI)相匹配的健康女性进行了比较,结果表明PCOS组血清中WBC和CRP水平显著升高,并且PCOS组中BMI与WBC和CRP水平之间存在正相关关系,推断肥胖PCOS女性的炎症水平增加。因此,对于患有PCOS的肥胖女性来说,炎症的发生与PCOS和肥胖的关系仍不清楚。

3.2. 卵巢炎症与PCOS

在卵泡发育和排卵过程中,卵巢存在适度的炎症在生理上是正常的,但持续的慢性炎症可能会影响卵巢功能、卵母细胞质量和子宫内膜容受性,造成不良的妊娠结局 [20] 。Yu等 [21] 通过定量PCR对PCOS女性颗粒细胞中慢性炎症状态相关基因的表达水平进行测定,发现颗粒细胞中γ干扰素(IFN-γ)、TNF-α和IL-12水平相较于对照组显著升高,这提示卵巢颗粒细胞中存在炎症状态。C1QTNF6是一种与炎症相关的脂联素旁系,Yan等 [22] 研究发现,C1QTNF6在PCOS患者颗粒细胞中的表达升高,并表明C1QTNF6通过上调TNF-α、IL-6和CRP的水平,通过激活AKT/NF-κB信号通路影响PCOS患者的炎症反应。Lai等 [23] 发现PCOS组卵泡液中IL-6、IL-8和IL-18浓度显著升高,并且与脂肪酸水平呈正相关;此外,脂肪酸可以通过ERK1/2信号通路上调IL-6和IL-8的转录水平,也可抑制AMPK信号通路,诱导氧化应激和炎症反应。这表明卵泡液中存在着炎症反应,并且和脂质代谢密切相关。Liu等 [24] 研究发现PCOS患者卵泡液中IL-1b和IL-18的水平较对照组升高,这会影响颗粒细胞的增殖,减缓卵泡的发育与成熟。这提示卵泡液中存在炎症环境,这可能是PCOS疾病进展的原因之一。Liu等 [25] 评估了PCOS女性和PCOS小鼠模型卵巢组织中IL-15的表达水平,发现其卵巢组织中IL-15均明显增加,此外PCOS患者MPO、IL-1a、Kallikrin3、MCP-1和IL-8均显著升高,这表明PCOS女性和PCOS小鼠模型的卵巢组织中确实存在慢性炎症状态。有研究团队通过深度测序技术在PCOS小鼠模型的卵巢中发现436个基因差异性表达,其与T细胞介导的细胞毒性和同型半胱氨酸代谢过程有关,并且参与了调节PCOS小鼠模型炎症过程的几种信号通路,如NF-kB信号传导、酪氨酸代谢和苯丙氨酸代谢等 [26] 。这表明PCOS卵巢中基因差异性表达与慢性炎症有关性。

3.3. 子宫内膜炎症与PCOS

炎症因子随月经周期的激素变化而波动,在雌孕激素撤退导致的子宫内膜脱落期间,子宫内膜局部产生的炎症介质可能通过单独或协同作用,诱导血管舒张、改变细胞粘附分子表达并募集白细胞导致炎症反应,并且在排卵期间,子宫内膜分泌的巨噬细胞和中性粒细胞也会增加 [27] 。Aksak等 [28] 使用光学显微镜对32~39岁正常女性的子宫内膜组织进行观察,发现子宫内膜由柱状上皮、子宫内膜腺体和间质组成,这些细胞存在着细长的嗜碱性细胞核,并且间质中有淋巴细胞和巨噬细胞;通过免疫组化发现TNF-α和单核细胞趋化蛋白-2 (monocyte chemoattractant protein-2, MCP-2)在子宫内膜中表达显著增加,这表明正常子宫内膜也存在着炎症反应。Long等 [29] 为了探讨慢性低度炎症对PCOS女性子宫内膜的影响,通过宫腔镜手术获得了23名PCOS女性和20名健康女性子宫内膜的组织样本,研究发现PCOS组与对照组相比,其子宫内膜中IL-18显著增加,该研究还表明在超重的PCOS女性患者中,IL-18过表达更加明显。这些研究表明了PCOS患者的子宫内膜中也有炎症反应,并且与肥胖有显著相关性。

4. PCOS基于炎症的治疗

长期的慢性炎症会损害机体的多个器官,可能会使得患糖尿病、心脏病、动脉粥样硬化和高血压的风险增加。在心脑血管疾病等并发症发生之前,早期诊断和治疗管理至关重要。

4.1. 中西医药物治疗

针对性的抗炎药物,如中成药及靶向药物等已被证实对PCOS女性的治疗具有积极作用。Peng等 [30] 研究发现,在芹菜素的治疗后的经脱氢表雄酮处理的PCOS大鼠的体重较前减轻,其TNF-α和IL-6的水平降低,并且该大鼠紊乱的激素水平、脂质状况和抗氧化状态得到改善。在一项随机、双盲、安慰剂对照的临床试验中,Mousavi等 [31] 研究发现,给予PCOS患者补充镁和褪黑素对缓解多毛症、增加细胞总抗氧化能力和降低血清TNF-α水平有一定的帮助;并且褪黑素独立使用可降低血清TNF-α水平。螺内酯最显著的作用是降低雄激素水平,并改善其引起的多毛和痤疮等症状,Stephanie等 [32] 研究发现低剂量螺内酯可减轻来曲唑诱导的PCOS大鼠模型中的炎症状态和氧化应激,减少细胞凋亡及纠正内分泌代谢紊乱。

趋化因子CXC配体(CXCL) 13及其受体CXCR5在炎症相关的生殖系统疾病中高度表达,可能参与PCOS的发病,Ullah等 [33] 研究发现二甲双胍可以通过降低CXCL13和CXCR5的表达来缓解PCOS的症状。然而,在最近一项针对妊娠期PCOS女性的研究表明:二甲双胍并不能抑制炎性细胞因子,而是诱导全身免疫反应;这说明对于妊娠期PCOS患者来说,二甲双胍具有免疫调节功能而非抗炎作用 [34] 。这些研究表明,二甲双胍在纠正PCOS的炎症状态中的功能尚无统一结论,仍需要进一步探究。

白藜芦醇具有抗衰老、抗氧化和抗炎作用,Liang等 [35] 对35岁以下不孕妇女的卵巢颗粒细胞进行了研究,发现白藜芦醇可减缓PCOS患者的氧化应激并抑制卵巢颗粒细胞的凋亡,从而改善卵巢状态和增加辅助生殖技术的成功率。磷酸二酯酶-4抑制剂罗氟司特可以抑制PCOS小鼠的慢性炎症和脂质积聚,有利于改善卵巢功能并减少颗粒细胞凋亡 [36] 。Izadi等 [37] 研究发现PCOS小鼠间充质干细胞衍生的外泌体和细胞外囊泡通过抗炎和抑制细胞凋亡,减少卵巢损失,保护颗粒细胞,改善小鼠卵巢功能,并可调节类固醇生成及抑制雄激素的产生。石榴汁提取物通过其抗氧化、抗炎、抗纤维化、抗增殖和抗雄激素的作用使来曲唑诱导的PCOS大鼠的激素水平正常化,减少子宫内膜氧化应激、增殖、纤维化和雄激素受体过表达,对PCOS大鼠子宫内膜具有保护作用 [38] 。

4.2. 手术治疗

尽管手术治疗存在卵巢损伤和生育能力下降等风险,但腹腔镜卵巢打孔术和减肥手术等手术方式已被考虑用于治疗PCOS女性。比如,Nur等 [39] 研究发现腹腔镜卵巢打孔术可以降低子宫内膜NF-κB p65的表达,从而改善PCOS患者的慢性炎症。肥胖会加重PCOS的慢性炎症和代谢紊乱,Héctor F等 [40] 表明减肥可以改善生殖和代谢功能障碍,降低糖尿病和心血管疾病的发病风险,改善PCOS患者的月经异常、多毛症和高雄激素血症。Hu等 [41] 研究发现,与药物治疗相比,减肥手术治疗肥胖型PCOS患者具有更快、更好的效果,可以稳定可靠地减轻体重,恢复正常的月经周期,以及改善激素水平和代谢状况。与减肥手术相比,PCOS患者通过控制饮食和改变生活方式减轻体重效果相对较慢,因此减肥手术对于肥胖PCOS女性来说可能是一种可行的治疗方法。

4.3. 一般治疗

饮食干预是PCOS女性纠正炎症状态的方法之一。Wang等 [42] 表明高纤维饮食可以缓解PCOS患者的慢性炎症,调解肠道微生物菌群,其联合阿卡波糖可以更好地改善内分泌代谢紊乱、高胰岛素血症、IR及肠道微生物菌群。一项临床研究表明有氧运动可有效降低多囊卵巢妇女的IL-6、TNF-α和CRP表达水平;有氧运动是控制PCOS的有效方式,有利于缓解PCOS患者的慢性炎症 [43] 。Pandit等 [44] 研究表明PCOS患者在进行了20周的有氧运动后,BMI、HOMA-IR和hs-CRP表达水平较前有明显降低,这表明了有氧运动可以有效改善PCOS患者的炎症状态以及IR。

以上研究已证实炎症因子与PCOS关系密切,基于炎症的药物治疗、手术治疗及生活方式干预等可显著改善PCOS女性的症状,对未来研发针对纠正PCOS慢性低度炎症的新型药物具有一定的启示。

5. 总结

PCOS是一种多基因疾病,其发病机制受复杂多样的因素影响,目前研究对其尚无统一定论。迄今为止,炎症因子在PCOS患者外周血、颗粒细胞、卵泡液、卵巢组织及子宫内膜组织等中均有研究,大多数集中于对外周血炎症因子的探讨。炎症因子与肥胖、IR、高雄激素血症共同参与PCOS患者的疾病进程,但PCOS发病机制和信号通路尚需进一步研究。通过关注炎症因子与PCOS的相关关系,了解其作用机制,可以为此疾病的诊疗和并发症预防提供更广阔的思路。

NOTES

*通讯作者。

参考文献

[1] Shrivastava, S. and Conigliaro, R.L. (2023) Polycystic Ovarian Syndrome. The Medical Clinics of North America, 107, 227-234.
https://doi.org/10.1016/j.mcna.2022.10.004
[2] Li, R., Zhang, Q., Yang, D., et al. (2013) Prevalence of Polycystic Ovary Syndrome in Women in China: A Large Community-Based Study. Human Reproduction, 28, 2562-2569.
https://doi.org/10.1093/humrep/det262
[3] Patel, S. (2018) Polycystic Ovary Syndrome (PCOS), an Inflammatory, Systemic, Lifestyle Endocrinopathy. The Journal of Steroid Biochemistry and Molecular Biology, 182, 27-36.
https://doi.org/10.1016/j.jsbmb.2018.04.008
[4] Dabravolski, S.A., Nikiforov, N.G., Eid, A.H., et al. (2021) Mitochondrial Dysfunction and Chronic Inflammation in Polycystic Ovary Syndrome. International Journal of Molecular Sciences, 22, Article 3923.
https://doi.org/10.3390/ijms22083923
[5] Pang, Y., Wu, L., Tang, C., et al. (2022) Autophagy-Inflammation Interplay during Infection: Balancing Pathogen Clearance and Host Inflammation. Frontiers in Pharmacology, 13, Article 832750.
https://doi.org/10.3389/fphar.2022.832750
[6] Metsios, G.S., Moe, R.H. and Kitas, G.D. (2020) Exercise and Inflammation. Best Practice & Research. Clinical Rheumatology, 34, Article ID: 101504.
https://doi.org/10.1016/j.berh.2020.101504
[7] Shamsi, M., Ghazavi, A., Saeedifar, A.M., et al. (2022) The Immune System’s Role in PCOS. Molecular Biology Reports, 49, 10689-10702.
https://doi.org/10.1007/s11033-022-07695-5
[8] Kakoly, N.S., Khomami, M.B., Joham, A.E., et al. (2018) Ethnicity, Obesity and the Prevalence of Impaired Glucose Tolerance and Type 2 Diabetes in PCOS: A Systematic Review and Meta-Regression. Human Reproduction Update, 24, 455-467.
https://doi.org/10.1093/humupd/dmy007
[9] Sadeghi, H.M., Adeli, I., Calina, D., et al. (2022) Polycystic Ovary Syndrome: A Comprehensive Review of Pathogenesis, Management, and Drug Repurposing. International Journal of Molecular Sciences, 23, Article 583.
https://doi.org/10.3390/ijms23020583
[10] Xing, C., Li, C. and He, B. (2020) Insulin Sensitizers for Improving the Endocrine and Metabolic Profile in Overweight Women with PCOS. The Journal of Clinical Endocrinology and Metabolism, 105, 2950-2963.
https://doi.org/10.1210/clinem/dgaa337
[11] Garzia, E., Galiano, V., Marfia, G., et al. (2022) Hyperandrogenism and Menstrual Imbalance Are the Best Predictors of Metformin Response in PCOS Patients. Reproductive Biology and Endocrinology, 20, Article No. 6.
https://doi.org/10.1186/s12958-021-00876-0
[12] Khichar, A., Gupta, S., Mishra, S., et al. (2021) Assessment of Inflammatory Markers in Women with PCOS and Their Correlation with Insulin Resistance. Clinical Laboratory, 67, 2439-2446.
https://doi.org/10.7754/Clin.Lab.2021.210310
[13] Mohammadian Khonsari, N., Baygi, F., Tabatabaei-Malazy, O., et al (2023) Association of Normal Weight Obesity Phenotype with Inflammatory Markers: A Systematic Review and Meta-Analysis. Frontiers in Immunology, 14, Article 1044178.
https://doi.org/10.3389/fimmu.2023.1044178
[14] Stokkeland, L., Giskeødegård, G.F., Ryssdal, M., et al. (2022) Changes in Serum Cytokines throughout Pregnancy in Women with Polycystic Ovary Syndrome. The Journal of Clinical Endocrinology and Metabolism, 107, 39-52.
https://doi.org/10.1210/clinem/dgab684
[15] Abraham Gnanadass, S., Divakar Prabhu, Y. and Valsala Gopalakrishnan, A. (2021) Association of Metabolic and Inflammatory Markers with Polycystic Ovarian Syndrome (PCOS): An Update. Archives of Gynecology and Obstetrics, 303, 631-643.
https://doi.org/10.1007/s00404-020-05951-2
[16] He, S., Mao, X., Lei, H., et al. (2020) Peripheral Blood Inflammatory-Immune Cells as a Predictor of Infertility in Women with Polycystic Ovary Syndrome. Journal of Inflammation Research, 13, 441-450.
https://doi.org/10.2147/JIR.S260770
[17] ALhabardi, N.A., Al-Wutayd, O., Eltayieb, K.M., et al. (2020) Peripheral Hematological Parameters in Women with Polycystic Ovary Syndrome. The Journal of International Medical Research, 48, 1-6.
https://doi.org/10.1177/0300060520952282
[18] Gonzalez, F., Thusu, K., Abdel-Rahman, E., et al. (1999) Elevated Serum Levels of Tumor Necrosis Factor α in Normal-Weight Women with Polycystic Ovary Syndrome. Metabolism: Clinical and Experimental, 48, 437-441.
https://doi.org/10.1016/S0026-0495(99)90100-2
[19] Rudnicka, E., Kunicki, M., Suchta, K., et al. (2020) Inflammatory Markers in Women with Polycystic Ovary Syndrome. BioMed Research International, 2020, Article ID: 4092470.
https://doi.org/10.1155/2020/4092470
[20] Velez, L., M., Seldin, M. and Motta, A.B. (2021) Inflammation and Reproductive Function in Women with Polycystic Ovary Syndrome. Biology of Reproduction, 104, 1205-1217.
https://doi.org/10.1093/biolre/ioab050
[21] Yu, Y., Li, G., He, X., et al. (2021) MicroRNA-21 Regulate the Cell Apoptosis and Cell Proliferation of Polycystic Ovary Syndrome (PCOS) Granulosa Cells through Target Toll Like Receptor TLR8. Bioengineered, 12, 5789-5796.
https://doi.org/10.1080/21655979.2021.1969193
[22] Yan, S., Ding, J., Zhang, Y., et al. (2021) C1QTNF6 Participates in the Pathogenesis of PCOS by Affecting the Inflammatory Response of Granulosa Cells. Biology of Reproduction, 105, 427-438.
https://doi.org/10.1093/biolre/ioab094
[23] Lai, Y., Ye, Z., Mu, L., et al. (2022) Elevated Levels of Follicular Fatty Acids Induce Ovarian Inflammation via ERK1/2 and Inflammasome Activation in PCOS. The Journal of Clinical Endocrinology and Metabolism, 107, 2307-2317.
https://doi.org/10.1210/clinem/dgac281
[24] Liu, Y., Liu, H., Li, Z., et al. (2021) The Release of Peripheral Immune Inflammatory Cytokines Promote an Inflammatory Cascade in PCOS Patients via Altering the Follicular Microenvironment. Frontiers in Immunology, 12, Article ID: 685724.
https://doi.org/10.3389/fimmu.2021.685724
[25] Liu, Y., Li, Z., Wang, Y., et al. (2022) IL-15 Participates in the Pathogenesis of Polycystic Ovary Syndrome by Affecting the Activity of Granulosa Cells. Frontiers in Endocrinology, 13, Article 787876.
https://doi.org/10.3389/fendo.2022.787876
[26] Ullah, A., Wang, M.J., Yang, J.P., et al. (2022) Ovarian Inflammatory MRNA Profiles of a Dehydroepiandrosterone plus High-Fat Diet-Induced Polycystic Ovary Syndrome Mouse Model. Reproductive Biomedicine Online, 44, 791-802.
https://doi.org/10.1016/j.rbmo.2021.10.024
[27] Critchley, H., Maybin, J.A., Armstrong, G.M., et al. (2020) Physiology of the Endometrium and Regulation of Menstruation. Physiological Reviews, 100, 1149-1179.
https://doi.org/10.1152/physrev.00031.2019
[28] Aksak, T., Gümürdülü, D., Çetin, M.T., et al. (2021) Expression of Monocyte Chemotactic Protein 2 and Tumor Necrosis Factor α in Human Normal Endometrium and Endometriotic Tissues. Journal of Gynecology Obstetrics and Human Reproduction, 50, Article ID: 101971.
https://doi.org/10.1016/j.jogoh.2020.101971
[29] Long, X., Li, R., Yang, Y., et al. (2017) Overexpression of IL-18 in the Proliferative Phase Endometrium of Patients with Polycystic Ovary Syndrome. Reproductive Sciences, 24, 252-257.
https://doi.org/10.1177/1933719116653681
[30] Peng, F., Hu, Y., Peng, S., et al. (2022) Apigenin Exerts Protective Effect and Restores Ovarian Function in Dehydroepiandrosterone Induced Polycystic Ovary Syndrome Rats: A Biochemical and Histological Analysis. Annals of Medicine, 54, 578-587.
https://doi.org/10.1080/07853890.2022.2034933
[31] Mousavi, R., Alizadeh, M., Asghari Jafarabadi, M., et al. (2022) Effects of Melatonin and/or Magnesium Supplementation on Biomarkers of Inflammation and Oxidative Stress in Women with Polycystic Ovary Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Biological Trace Element Research, 200, 1010-1019.
https://doi.org/10.1007/s12011-021-02725-y
[32] Areloegbe, S.E., Peter, M.U., Oyeleke, M.B., et al. (2022) Low-Dose Spironolactone Ameliorates Adipose Tissue Inflammation and Apoptosis in Letrozole-Induced PCOS Rat Model. BMC Endocrine Disorders, 22, Article No. 224.
https://doi.org/10.1186/s12902-022-01143-y
[33] Ullah, A., Pervaz, S., Adu-Gyamfi, E.A,, et al. (2022) CXCL13, and, CXCR5, Are, Upregulated, in, PCOS, Mice, Ovaries, But, Downregulated Following Metformin Administration. Molecular and Cellular Endocrinology, 556, Article ID: 111730.
https://doi.org/10.1016/j.mce.2022.111730
[34] Ryssdal, M., Vanky, E., Stokkeland, L., et al. (2023) Immunomodulatory Effects of Metformin Treatment in Pregnant Women with PCOS. The Journal of Clinical Endocrinology and Metabolism, 108, e743-e753.
https://doi.org/10.1210/clinem/dgad145
[35] Liang, Y., Xu, M.L., Gao, X., et al. (2023) Resveratrol Improves Ovarian State by Inhibiting Apoptosis of Granulosa Cells. Gynecological Endocrinology, 39, 2181652.
https://doi.org/10.1080/09513590.2023.2181652
[36] Ji, X., Ye, Y., Wang, L., et al. (2023) PDE4 Inhibitor Roflumilast Modulates Inflammation and Lipid Accumulation in PCOS Mice to Improve Ovarian Function and Reduce DHEA-Induced Granulosa Cell Apoptosis in Vitro. Drug Development Research, 84, 226-237.
https://doi.org/10.1002/ddr.22027
[37] Izadi, M., Rezvani, M.E., Aliabadi, A., et al. (2022) Mesenchymal Stem Cells-Derived Exosomes as a Promising New Approach for the Treatment of Infertility Caused by Polycystic Ovary Syndrome. Frontiers in Pharmacology, 13, Article 1021581.
https://doi.org/10.3389/fphar.2022.1021581
[38] Ibrahim, M., Sadek, M.T. and Sharaf Eldin, H. (2022) Role of Pomegranate Extract in Restoring Endometrial Androgen Receptor Expression, Proliferation, and Pinopodes in a Rat Model of Polycystic Ovary Syndrome. Morphologie, 106, 145-154.
https://doi.org/10.1016/j.morpho.2021.04.004
[39] Dokuzeylül, Güngör, N., Güngör, K., Yurci, A., et al. (2022) Ovarian Drilling Down-Regulates Endometrial Nuclear Factor-κB P65 Expression in Women with PCOS: A Prospective Case-Control Study. Turkish Journal of Obstetrics and Gynecology, 19, 45-50.
https://doi.org/10.4274/tjod.galenos.2022.44845
[40] Escobar-Morreale, H.F. (2012) Surgical Management of Metabolic Dysfunction in PCOS. Steroids, 77, 312-316.
https://doi.org/10.1016/j.steroids.2011.12.004
[41] Hu, L., Ma, L., Xia, X., et al. (2022) Efficacy of Bariatric Surgery in the Treatment of Women with Obesity and Polycystic Ovary Syndrome. The Journal of Clinical Endocrinology and Metabolism, 107, e3217-e3229.
https://doi.org/10.1210/clinem/dgac294
[42] Wang, X., Xu, T., Liu, R., et al. (2022) High-Fiber Diet Or Combined with Acarbose Alleviates Heterogeneous Phenotypes of Polycystic Ovary Syndrome by Regulating Gut Microbiota. Frontiers in Endocrinology, 12, Article 806331.
https://doi.org/10.3389/fendo.2021.806331
[43] Elbandrawy, A.M., Yousef, A.M., Morgan, E.N., et al. (2022) Effect of Aerobic Exercise on Inflammatory Markers in Polycystic Ovary Syndrome: A Randomized Controlled Trial. European Review for Medical and Pharmacological Sciences, 26, 3506-3513.
[44] Pandit, U., Singh, M., Ranjan, R., et al. (2022) The Effect of Exercise Training on Body Composition, Insulin Resistance and High Sensitivity C-Reactive Protein (Hs-CRP) in Women with Polycystic Ovary Syndrome: A Pilot Study from North India. Cureus, 14, e23994.
https://doi.org/10.7759/cureus.23994