切换至 "中华医学电子期刊资源库"

中华细胞与干细胞杂志(电子版) ›› 2023, Vol. 13 ›› Issue (02) : 108 -114. doi: 10.3877/cma.j.issn.2095-1221.2023.02.007

综述

干细胞治疗多囊卵巢综合征的研究进展
符莞孟, 王晓黎, 刘玉, 张潍, 张菊()   
  1. 571437 琼海,海南博鳌未来医院研发中心
    570312 海口,海南省妇女儿童医学中心
    710038 西安,空军军医大学西京医院妇产科
    571437 琼海,海南博鳌未来医院研发中心;710038 西安,空军军医大学西京医院妇产科
  • 收稿日期:2022-09-19 出版日期:2023-04-01
  • 通信作者: 张菊
  • 基金资助:
    陕西省重点研发计划(2017ZDXM-SF-037); 全军医学科技青年培育项目(16QNP112)

Research progress of stem cell therapy in polycystic ovary syndrome

Wanmeng Fu, Xiaoli Wang, Yu Liu, Wei Zhang, Ju Zhang()   

  1. Hainan BoaoWorldlight Hospital R&D Center, Qionghai 571437, China
    Hainan Province Women and Children Medical Center, Haikou 570312, China
    Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an 710038, China
    Hainan BoaoWorldlight Hospital R&D Center, Qionghai 571437, China; Department of Gynecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an 710038, China
  • Received:2022-09-19 Published:2023-04-01
  • Corresponding author: Ju Zhang
  • About author:

    Fu Wanmeng and Wang Xiaoli are the first authors who contributed equally to the article

引用本文:

符莞孟, 王晓黎, 刘玉, 张潍, 张菊. 干细胞治疗多囊卵巢综合征的研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(02): 108-114.

Wanmeng Fu, Xiaoli Wang, Yu Liu, Wei Zhang, Ju Zhang. Research progress of stem cell therapy in polycystic ovary syndrome[J]. Chinese Journal of Cell and Stem Cell(Electronic Edition), 2023, 13(02): 108-114.

干细胞治疗在医学领域发展迅速且应用日益广泛。在基础研究和临床试验中,干细胞治疗多种生殖系统疾病均显示良好效果。近年来不同来源的间充质干细胞(MSCs)在多囊卵巢综合征(PCOS)治疗中取得一系列研究进展。本文总结了MSCs及其外泌体治疗PCOS的机制,包括调节激素和糖、脂代谢,抑制炎症反应和调节免疫功能,归巢效应以及促血管生成,改善排卵与生殖潜能等。回顾了诱导多能干细胞(iPSCs)和胚胎干细胞(ESCs)在PCOS实验研究进展,为干细胞在PCOS的临床研究与应用提供探索思路。

Stem cell based therapy is rapidly developing and widely applied in medical field. Several reproductive diseases have been treated using various types of stem cells in experimental studies and clinical trials, which have produced favorable results. In recent years, some experimental research progress of mesenchymal stem cells (MSCs) derived from different sources in the treatment of polycystic ovary syndrome (PCOS) has been reported. This article reviewed the mechanism of MSCs and their exosomes in the treatment of PCOS, including regulating hormones, sugar and lipid metabolism, inhibiting inflammatory response, regulating immune function, homing effect, promoting angiogenesis, and enhancing ovulation and reproductive potential function. This review also summarized the progress of experimental research on induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) in PCOS and provided some exploration ideas for the clinical research and application of stem cells in PCOS.

图1 干细胞与PCOS发病和治疗作用机制注:NF-κB为核因子κB亚基1;AKT为蛋白激酶B;P-IκB为磷酸化核因子κB抑制剂;p-p65为磷酸化核因子κB p65亚基;CTGF为结缔组织生长因子;INF-γ为干扰素γ;TNF-α为肿瘤坏死因子α;IL-6/10/16为白细胞介素6/10/16;IL1-β为白细胞介素1β;CYP11A1为细胞色素P450家族11亚家族A成员1;DENND1A为DENN结构域包含1A;AR为雄激素受体;p-IRS1为磷酸化胰岛素受体底物1;p-AKT为磷酸化蛋白激酶B;BTG2为BTG抗增殖因子2;PGC1α为过氧化物酶体增生物激活受体γ共激活剂1α;CIDEA为细胞死亡诱导DFFA样效应因子A;UCP1为解偶联蛋白1;AMH为抗缪勒管激素;AMHR为抗缪勒管激素受体;FSHR为卵泡刺激素受体;LH为黄体生成素;FSH为卵泡刺激素;LHR为黄体生成素受体;Erβ为雌激素β受体;BAX为BCL2相关X;VEGF为血管内皮生长因子;VEGFA为血管生长因子A;PDCD4为程序性细胞死亡因子4;BCL-2为B细胞淋巴瘤2;CD31为内皮细胞黏附分子1;VCAM-1为血管细胞黏附分子-1;CXCR4为C-X-C趋化因子受体4;α4β1又称极晚期抗原4 (VLA-4);SDF-1为基质细胞衍生因子1;FOXL2为叉头框转录因子L2基因;CREB为环腺苷酸响应元件结合蛋白;GPI为葡萄糖-6-磷酸异构酶;FBP1为果糖双磷酸酶1;PYGL为糖原磷酸化酶L;UGP2为UDP葡萄糖焦磷酸化酶2;NR0B2为核受体亚家族0,B组成员2
1
Ali AT, Guidozzi F. Midlife women's health consequences associated with polycystic ovary syndrome[J]. Climacteric, 2020, 23(2):116-122.
2
Yalameha B, Nejabati HR, Nouri M. Circulating microparticles as indicators of cardiometabolic risk in PCOS[J]. Clin Chim Acta, 2022, 533:63-70.
3
Ding H, Zhang J, Zhang F, et al. Resistance to the insulin and elevated level of androgen: a major cause of polycystic ovary syndrome[J]. Front Endocrinol (Lausanne), 2021, 12:741764. doi: 10.3389/fendo.2021.741764.
4
Glueck CJ, Goldenberg N. Characteristics of obesity in polycystic ovary syndrome: etiology, treatment, and genetics[J]. Metabolism, 2019, 92:108-120.
5
Shaaban Z, Khoradmehr A, Jafarzadeh Shirazi MR, et al. Pathophysiological mechanisms of gonadotropins- and steroid hormones-related genes in etiology of polycystic ovary syndrome[J]. Iran J Basic Med Sci, 2019, 22(1):3-16.
6
Dewailly D, Robin G, Peigne M, et al. Interactions between androgens, FSH, anti-Mullerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary[J]. Hum Reprod Update, 2016, 22(6):709-724.
7
Di Pietro M, Pascuali N, Parborell F, et al. Ovarian angiogenesis in polycystic ovary syndrome[J]. Reproduction, 2018, 155(5):R199-R209.
8
Rudnicka E, Suchta K, Grymowicz M, et al. Chronic low grade inflammation in pathogenesis of PCOS[J]. Int J Mol Sci, 2021, 22(7): 3789. doi: 10.3390/ijms22073789.
9
Motta AB. Dehydroepiandrosterone to induce murine models for the study of polycystic ovary syndrome[J]. J Steroid Biochem Mol Biol, 2010, 119(3-5):105-111.
10
Wang MY, Wang YX, Li-Ling J, et al. Adult stem cell therapy for premature ovarian failure: from bench to bedside[J]. Tissue Eng Part B Rev, 2022, 28(1):63-78.
11
Zhao YX, Chen SR, Su PP, et al. Using mesenchymal stem cells to treat female infertility: an update on female reproductive diseases[J]. Stem Cells Int, 2019, 2019:9071720. doi: 10.1155/2019/9071720.
12
Kim HJ, Kim G, Lee J, et al. Secretome of stem cells: roles of extracellular vesicles in diseases, stemness, differentiation, and reprogramming[J]. Tissue Eng Regen Med, 2022, 19(1):19-33.
13
Huang QY, Chen SR, Chen JM, et al. Therapeutic options for premature ovarian insufficiency: an updated review[J]. Reprod Biol Endocrinol, 2022, 20(1):28. doi: 10.1186/s12958-022-00892-8.
14
Mazini L, Rochette L, Amine M, et al. Regenerative capacity of adipose derived stem cells (ADSCs), comparison with mesenchymal stem cells (MSCs)[J]. Int J Mol Sci, 2019, 20(10):2523. doi:10.3390/ijms20102523.
15
Fu YX, Ji J, Shan F, et al. Human mesenchymal stem cell treatment of premature ovarian failure: new challenges and opportunities[J]. Stem Cell Res Ther, 2021, 12(1):161. doi:10.1186/s13287-021-02212-0.
16
He Y, Chen D, Yang L, et al. The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure[J]. Stem Cell Res Ther, 2018, 9(1):263.doi: 10.1186/s13287-018-1008-9.
17
Park HS, Chugh RM, Pergande MR, et al. Non-cytokine protein profile of the mesenchymal stem cell secretome that regulates the androgen production pathway[J]. Int J Mol Sci, 2022, 23(9):4633. doi: 10.3390/ijms23094633.
18
Chugh RM, Park HS, El Andaloussi A, et al. Mesenchymal stem cell therapy ameliorates metabolic dysfunction and restores fertility in a PCOS mouse model through interleukin-10[J]. Stem Cell Res Ther, 2021, 12(1):388. doi: 10.1186/s13287-021-02472-w.
19
Chugh RM, Park HS, Esfandyari S, et al. Mesenchymal stem cell-conditioned media regulate steroidogenesis and inhibit androgen secretion in a PCOS cell model via BMP-2[J]. Int J Mol Sci, 2021, 22(17):9184. doi:10.3390/ijms22179184.
20
Kalhori Z, Azadbakht M, Soleimani Mehranjani M, et al. Improvement of the folliculogenesis by transplantation of bone marrow mesenchymal stromal cells in mice with induced polycystic ovary syndrome[J]. Cytotherapy, 2018, 20(12):1445-1458.
21
郑雅静,代芳芳,杨冬咏,等. 棕色脂肪组织在多囊卵巢综合征中的研究进展[J]. 中国计划生育和妇产科, 2021, 13(12):9-11.
22
李子涵,李修阳,张宁,等. 棕色脂肪组织及其与多囊卵巢综合征相关性研究新进展[J]. 国际妇产科学杂志, 2018, 45(2):188-191.
23
Sanchez-Garrido MA, Tena-Sempere M. Metabolic dysfunction in polycystic ovary syndrome: pathogenic role of androgen excess and potential therapeutic strategies[J]. Mol Metab, 2020, 35:100937. doi: 10.1016/j.molmet.2020.01.001.
24
相迪,张真真,万贵平.白色脂肪组织褐变及其在多囊卵巢综合征治疗中的研究进展[J]. 航空航天医学杂志, 2020, 31(6):723-726.
25
Zhang Q, Ye R, Zhang YY, et al. Brown adipose tissue and novel management strategies for polycystic ovary syndrome therapy[J]. Front Endocrinol (Lausanne), 2022, 13:847249. doi:10.3389/fendo.2022.847249.
26
Zhao Y, Pan S, Wu X. Human umbilical cord mesenchymal stem cell-derived exosomes inhibit ovarian granulosa cells inflammatory response through inhibition of NF-kappaB signaling in polycystic ovary syndrome[J]. J Reprod Immunol, 2022, 152:103638. doi:10.1016/j.jri.2022.103638.
27
李玮. 人经血间充质干细胞对多囊卵巢综合征大鼠卵巢CYP19A1的影响[D]. 青岛:青岛大学, 2020.
28
刘荣华,何文容. MenSCs移植对大鼠多囊卵巢综合征的影响及其机制[J]. 天津医药, 2019, 47(7):709-712.
29
Cao M, Zhao Y, Chen T, et al. Adipose mesenchymal stem cell-derived exosomal microRNAs ameliorate polycystic ovary syndrome by protecting against metabolic disturbances[J]. Biomaterials, 2022, 288:121739. doi: 10.1016/j.biomaterials.2022.121739.
30
Park H, Ashour D, Elsharoud A, et al. Towards stem cell therapy of polycystic ovary syndrome (PCOS): therapeutic effect of human mesenchymal stem cells transplantation in pcos mouse model by regulating ovarian vascularization[J]. Cytotherapy, 2019, 21(5):259.
31
Xie Q, Xiong X, Xiao N, et al. Mesenchymal stem cells alleviate DHEA-induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice[J]. Stem Cells Int, 2019, 2019:9782373. doi: 10.1155/2019/9782373.
32
吴晨婷,伯乐,伊娜,等. 脐带间充质干细胞介导AKT/NF-κB信号通路改善PCOS小鼠卵巢损伤的作用机制研究[J]. 解剖科学进展, 2021, 27(5):619-623.
33
Wang J, Guan E, Roderiquez G, et al. Role of tyrosine phosphorylation in ligand-independent sequestration of CXCR4 in human primary monocytes-macrophages[J]. J Biol Chem, 2001, 276(52):49236-49243.
34
De Becker A, Riet IV. Homing and migration of mesenchymal stromal cells: How to improve the efficacy of cell therapy?[J]. World J Stem Cells, 2016, 8(3):73-87.
35
Ullah M, Liu DD, Thakor AS. Mesenchymal stromal cell homing: mechanisms and strategies for improvement[J]. iScience, 2019, 15:421-438.
36
Benor A, Gay S, DeCherney A. An update on stem cell therapy for Asherman syndrome[J]. J Assist Reprod Genet, 2020, 37(7):1511-1529.
37
Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy[J]. Front Physiol, 2012, 3:359. doi:10.3389/fphys.2012.00359.
38
Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the details[J]. Cell Stem Cell, 2009, 4(3):206-216.
39
曾辉锋,王玉霞,杨莹. 脂肪干细胞经两种方法移植后在多囊卵巢综合征大鼠体内分布的比较[J]. 暨南大学学报(自然科学与医学版), 2017, 38(5):427-432.
40
Ling L, Hou J, Liu D, et al. Important role of the SDF-1/CXCR4 axis in the homing of systemically transplanted human amnion-derived mesenchymal stem cells (hAD-MSCs) to ovaries in rats with chemotherapy-induced premature ovarian insufficiency (POI)[J]. Stem Cell Res Ther, 2022, 13(1):79. doi: 10.1186/s13287-022-02759-6.
41
Jafarzadeh H, Nazarian H, Ghaffari Novin M, et al. Improvement of oocyte in vitro maturation from mice with polycystic ovary syndrome by human mesenchymal stromal cell-conditioned media[J]. J Cell Biochem, 2018, 119(12):10365-10375.
42
Zhao Y, Tao M, Wei M, et al. Mesenchymal stem cells derived exosomal miR-323-3p promotes proliferation and inhibits apoptosis of cumulus cells in polycystic ovary syndrome (PCOS)[J]. Artif Cells Nanomed Biotechnol, 2019, 47(1):3804-3813.
43
Zhang Y, Zhang YS, Xue FX. Characterization of embryonic stem cell model of polycystic ovary syndrome[J]. In Vitro Cell Dev Biol Anim, 2016, 52(5):507-511.
44
Wang F, Liu WW, Chen XM, et al. Differential genes in adipocytes induced from polycystic and non-polycystic ovary syndrome-derived human embryonic stem cells[J]. Syst Biol Reprod Med, 2014, 60(3):136-142.
45
Li J, Wang F, Kong HJ, et al. Differentiation of polycystic ovary syndrome-derived human embryonic stem cells into adipocytes and their glucose consumption[J]. Gynecol Endocrinol, 2012, 28(11):871-873.
46
Yang S, Ding S, Jiang X, et al. Establishment and adipocyte differentiation of polycystic ovary syndrome-derived induced pluripotent stem cells[J]. Cell Prolif, 2016, 49(3):352-361.
47
Min Z, Zhao Y, Hang J, et al. Neuroendocrine characteristics of induced pluripotent stem cells from polycystic ovary syndrome women[J]. Protein Cell, 2019, 10(7):526-532.
48
Min Z, Zhao Y, Hang J, et al. Correction to: neuroendocrine characteristics of induced pluripotent stem cells from polycystic ovary syndrome women[J]. Protein Cell, 2020, 11(3):227-229.
49
Huang CC, Chen MJ, Lan CW, et al. Hyperactive CREB signaling pathway involved in the pathogenesis of polycystic ovarian syndrome revealed by patient-specific induced pluripotent stem cell modeling[J]. Fertil Steril, 2019, 112(3):594-607.e12.
50
Jiang X, Hu R, Li C, et al. Generation of an induced pluripotent stem cell line AMUFAHi002-A from polycystic ovary syndrome patient[J]. Stem Cell Res, 2022, 63:102875. doi: 10.1016/j.scr.2022.102875.
51
Prodia Stem Cell Indonesia PT. Stem cells and secretomes for infertility therapy in polycystic ovary syndrome (PCOS) patients with insulin resistance. ClinicalTrials [OL].

URL    
52
Li Z, Zhang M, Zheng J, et al. Human umbilical cord mesenchymal stem cell-derived exosomes improve ovarian function and proliferation of premature ovarian insufficiency by regulating the hippo signaling pathway[J]. Front Endocrinol (Lausanne), 2021, 12:711902. doi: 10.3389/fendo.2021.711902.
53
Cui L, Bao H, Liu Z, et al. hUMSCs regulate the differentiation of ovarian stromal cells via TGF-beta(1)/Smad3 signaling pathway to inhibit ovarian fibrosis to repair ovarian function in POI rats[J]. Stem Cell Res Ther, 2020, 11(1):386. doi: 10.1186/s13287-020-01904-3.
[1] 王岩, 马剑雄, 郎爽, 董本超, 田爱现, 李岩, 孙磊, 靳洪震, 卢斌, 王颖, 柏豪豪, 马信龙. 外泌体在骨质疏松症诊疗中应用的研究进展[J]. 中华关节外科杂志(电子版), 2023, 17(05): 673-678.
[2] 贺敬龙, 孙炜, 高明宏, 谢伟, 姜骆永, 何琦非, 岳家吉. 外泌体非编码RNA在骨关节炎发病机制中的研究进展[J]. 中华关节外科杂志(电子版), 2023, 17(04): 520-527.
[3] 代雯荣, 赵丽娟, 李智慧. 细胞外囊泡对胚胎着床影响的研究进展[J]. 中华妇幼临床医学杂志(电子版), 2023, 19(05): 616-620.
[4] 周东杰, 蒋敏, 范海瑞, 高玲玲, 孔祥, 卢丹, 王丽萍. 非编码RNA在卵泡发育成熟中作用及其机制的研究现状[J]. 中华妇幼临床医学杂志(电子版), 2023, 19(04): 387-393.
[5] 高雷, 李芳, 巴雅力嘎, 李全, 巴特. 干细胞源性外泌体在创伤修复中免疫作用的研究进展[J]. 中华损伤与修复杂志(电子版), 2023, 18(04): 364-367.
[6] 李晓晖, 上官昌盛, 向英, 裴芝皆, 车俊志, 谢飞. 3D腹腔镜袖状胃切除术后机体能量代谢与多囊卵巢综合征患者性激素水平关系[J]. 中华普外科手术学杂志(电子版), 2023, 17(05): 538-541.
[7] 唐英俊, 李华娟, 王赛妮, 徐旺, 刘峰, 李羲, 郝新宝, 黄华萍. 人脐带间充质干细胞治疗COPD小鼠及机制分析[J]. 中华肺部疾病杂志(电子版), 2023, 16(04): 476-480.
[8] 李晔, 何洁, 胡锦秀, 王金祥, 田川, 潘杭, 陈梦蝶, 赵晓娟, 叶丽, 张敏, 潘兴华. 高活性间充质干细胞干预猕猴卵巢衰老的研究[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 210-219.
[9] 龙慧玲, 林蜜, 邵婷. 三维球体间充质干细胞培养技术的研究进展及其应用[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 229-234.
[10] 刘文慧, 吴涛, 张曦. 间充质干细胞联合血小板生成素受体激动剂在异基因造血干细胞移植后血小板恢复中的研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 242-246.
[11] 王红敏, 谢云波, 王彦虎, 王福生. 间充质干细胞治疗新冠病毒感染的临床研究进展[J]. 中华细胞与干细胞杂志(电子版), 2023, 13(04): 247-256.
[12] 王楚风, 蒋安. 原发性肝癌的分子诊断[J]. 中华肝脏外科手术学电子杂志, 2023, 12(05): 499-503.
[13] 杨蕴钊, 周诚, 石美涵, 赵静, 白雪源. 人羊水间充质干细胞对膜性肾病大鼠的治疗作用[J]. 中华肾病研究电子杂志, 2023, 12(04): 181-186.
[14] 宋艳琪, 任雪景, 王文娟, 韩秋霞, 续玥, 庄凯婷, 肖拓, 蔡广研. 间充质干细胞对顺铂诱导的小鼠急性肾损伤中细胞铁死亡的作用[J]. 中华肾病研究电子杂志, 2023, 12(04): 187-193.
[15] 梁宇同, 丁旭, 马国慧, 黄艳红. 间充质干细胞在宫腔粘连治疗中的研究进展[J]. 中华临床医师杂志(电子版), 2023, 17(05): 596-599.
阅读次数
全文


摘要