1
|
Zhao X, Yu Z, Wang X, Li X, Liu Y, Wang L. The administration of human amniotic epithelial cells in premature ovarian insufficiency: From preclinical to clinical. Gynecol Endocrinol 2024; 40:2382818. [PMID: 39039858 DOI: 10.1080/09513590.2024.2382818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
Premature ovarian insufficiency (POI) or premature ovarian failure (POF) is a multifactorial disorder occurring in reproductive-age women, characterized by elevated levels of follicle-stimulating hormone (FSH) and irregular or absent menstrual cycles, often accompanied by perimenopausal symptoms and infertility. While assisted reproductive technology can address the reproductive aspirations of some POI-affected women, it is hindered by issues such as exorbitant expenses, substantial risks, and poor rates of conception. Encouragingly, extensive research is exploring novel approaches to enhance fertility, particularly in the realm of stem cell therapy, showcasing both feasibility and significant potential. Human amniotic epithelial cells (hAECs) from discarded placental tissues are crucial in regenerative medicine for their pluripotency, low immunogenicity, non-tumorigenicity, accessibility, and minimal ethical concerns. Preclinical studies highlight the underlying mechanisms and therapeutic effects of hAECs in POI treatment, and current research is focusing on innovative interventions to augment hAECs' efficacy. However, despite these strides, overcoming application challenges is essential for successful clinical translation. This paper conducted a comprehensive analysis of the aforementioned issues, examining the prospects and challenges of hAECs in POI, with the aim of providing some insights for future research and clinical practice.
Collapse
Affiliation(s)
- Xiaojing Zhao
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhongna Yu
- Department of Gynecology, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaojing Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yang Liu
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liang Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Notghi P, Mehranjani MS, Shariatzadeh SMA. Atorvastatin improves ovarian function and follicular reserve in rats with premature ovarian insufficiency. Reprod Biomed Online 2024; 49:104324. [PMID: 39197401 DOI: 10.1016/j.rbmo.2024.104324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 09/01/2024]
Abstract
RESEARCH QUESTION Can atorvastatin, with its antioxidant, anti-inflammatory and anti-apoptotic properties, improve ovarian function and follicular reserve in rats with cyclophosphamide-induced premature ovarian insufficiency (POI)? DESIGN In this experimental study, 24 adult female Wistar rats were divided into four groups: control; POI; POI + atorvastatin; and atorvastatin. After treatment with atorvastatin, serum concentrations of total antioxidant capacity, glutathione, malondialdehyde, FSH, oestradiol, anti-Müllerian hormone, tumour necrosis factor-alpha and interleukin-6 were evaluated. Additionally, mRNA and protein expression of Bax, Bcl-2 and VEGF-A; number of follicles; and total volume of the ovary, and volumes of the cortex and medulla were examined. RESULTS The results showed that serum concentrations of total antioxidant capacity (P < 0.001), glutathione, oestradiol and anti-Müllerian hormone (P < 0.05); mRNA and protein expression of Bcl-2 and VEGF-A (P < 0.05); number of primordial and primary follicles (P < 0.001), and preantral and antral follicles (P < 0.01); and total volume of the ovary, and volume of the cortex (P < 0.05) increased significantly in the POI + atorvastatin group compared with the POI group. Serum concentrations of malondialdehyde, FSH, tumour necrosis factor-alpha and interleukin-6; and mRNA and protein expression of Bax decreased significantly in the POI + atorvastatin group compared with the POI group (P < 0.05). CONCLUSIONS Atorvastatin reduces the detrimental effects of cyclophosphamide in the POI model significantly by reducing oxidative stress and pro-inflammatory cytokines; regulating the expression of Bax, Bcl-2 and VEGF-A; and improving ovarian function and follicular reserve.
Collapse
Affiliation(s)
- Parmis Notghi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | | | | |
Collapse
|
3
|
Guo Y, Xue L, Tang W, Xiong J, Chen D, Dai Y, Wu C, Wei S, Dai J, Wu M, Wang S. Ovarian microenvironment: challenges and opportunities in protecting against chemotherapy-associated ovarian damage. Hum Reprod Update 2024; 30:614-647. [PMID: 38942605 PMCID: PMC11369228 DOI: 10.1093/humupd/dmae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/27/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Chemotherapy-associated ovarian damage (CAOD) is one of the most feared short- and long-term side effects of anticancer treatment in premenopausal women. Accumulating detailed data show that different chemotherapy regimens can lead to disturbance of ovarian hormone levels, reduced or lost fertility, and an increased risk of early menopause. Previous studies have often focused on the direct effects of chemotherapeutic drugs on ovarian follicles, such as direct DNA damage-mediated apoptotic death and primordial follicle burnout. Emerging evidence has revealed an imbalance in the ovarian microenvironment during chemotherapy. The ovarian microenvironment provides nutritional support and transportation of signals that stimulate the growth and development of follicles, ovulation, and corpus luteum formation. The close interaction between the ovarian microenvironment and follicles can determine ovarian function. Therefore, designing novel and precise strategies to manipulate the ovarian microenvironment may be a new strategy to protect ovarian function during chemotherapy. OBJECTIVE AND RATIONALE This review details the changes that occur in the ovarian microenvironment during chemotherapy and emphasizes the importance of developing new therapeutics that protect ovarian function by targeting the ovarian microenvironment during chemotherapy. SEARCH METHODS A comprehensive review of the literature was performed by searching PubMed up to April 2024. Search terms included 'ovarian microenvironment' (ovarian extracellular matrix, ovarian stromal cells, ovarian interstitial, ovarian blood vessels, ovarian lymphatic vessels, ovarian macrophages, ovarian lymphocytes, ovarian immune cytokines, ovarian oxidative stress, ovarian reactive oxygen species, ovarian senescence cells, ovarian senescence-associated secretory phenotypes, ovarian oogonial stem cells, ovarian stem cells), terms related to ovarian function (reproductive health, fertility, infertility, fecundity, ovarian reserve, ovarian function, menopause, decreased ovarian reserve, premature ovarian insufficiency/failure), and terms related to chemotherapy (cyclophosphamide, lfosfamide, chlormethine, chlorambucil, busulfan, melphalan, procarbazine, cisplatin, doxorubicin, carboplatin, taxane, paclitaxel, docetaxel, 5-fluorouraci, vincristine, methotrexate, dactinomycin, bleomycin, mercaptopurine). OUTCOMES The ovarian microenvironment shows great changes during chemotherapy, inducing extracellular matrix deposition and stromal fibrosis, angiogenesis disorders, immune microenvironment disturbance, oxidative stress imbalances, ovarian stem cell exhaustion, and cell senescence, thereby lowering the quantity and quality of ovarian follicles. Several methods targeting the ovarian microenvironment have been adopted to prevent and treat CAOD, such as stem cell therapy and the use of free radical scavengers, senolytherapies, immunomodulators, and proangiogenic factors. WIDER IMPLICATIONS Ovarian function is determined by its 'seeds' (follicles) and 'soil' (ovarian microenvironment). The ovarian microenvironment has been reported to play a vital role in CAOD and targeting the ovarian microenvironment may present potential therapeutic approaches for CAOD. However, the relation between the ovarian microenvironment, its regulatory networks, and CAOD needs to be further studied. A better understanding of these issues could be helpful in explaining the pathogenesis of CAOD and creating innovative strategies for counteracting the effects exerted on ovarian function. Our aim is that this narrative review of CAOD will stimulate more research in this important field. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| |
Collapse
|
4
|
Cui X, Jing X. Stem cell-based therapeutic potential in female ovarian aging and infertility. J Ovarian Res 2024; 17:171. [PMID: 39182123 PMCID: PMC11344413 DOI: 10.1186/s13048-024-01492-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/11/2024] [Indexed: 08/27/2024] Open
Abstract
Premature ovarian insufficiency (POI) is defined as onset of menopause characterized by amenorrhea, hypergonadotropism, and hypoestrogenism, before the age of 40 years. The POI is increasing, which seriously affects the quality of patients' life. Due to its diversity of pathogenic factors, complex pathogenesis and limited treatment methods, the search for finding effective treatment of POI has become a hotspot. Stem cells are characterized by the ability of self-renewal and differentiation and play an important role in the regeneration of injured tissues, which is therapy is expected to be used in the treatment of POI. The aim of this review is to summarize the pathogenic mechanisms and the research progress of POI treatment with stem cells from different sources.
Collapse
Affiliation(s)
- Xiangrong Cui
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Children's Hospital of Shanxi, Shanxi Maternal and Child Health Hospital, Taiyuan, 030001, China
| | - Xuan Jing
- Clinical Laboratory, Shanxi Provincial People's Hospital, Taiyuan, 030001, China.
| |
Collapse
|
5
|
Ye X, Lin Y, Ying Y, Shen X, Ni F, Wang F, Chen J, Zhao W, Yu X, Zhang D, Liu Y. Human Amniotic Epithelial Stem Cells Alleviate Autoimmune Premature Ovarian Insufficiency in Mice by Targeting Granulosa Cells via AKT/ERK Pathways. Stem Cell Rev Rep 2024; 20:1618-1635. [PMID: 38831179 PMCID: PMC11319531 DOI: 10.1007/s12015-024-10745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Autoimmune factors play an important role in premature ovarian insufficiency (POI). Human amniotic epithelial stem cells (hAESCs) have recently shown promising treatment effects on chemotherapy-induced POI. However, the therapeutic efficacy and underlying mechanisms of hAESCs in autoimmune POI remain to be investigated. In this study, we showed for the first time that intravenous transplantation of hAESCs could reside in the ovary of zona pellucida 3 peptide (pZP3) induced autoimmune POI mice model for at least 4 weeks. hAESCs could improve ovarian function and fertility, alleviate inflammation and reduce apoptosis of granulosa cells (GCs) in autoimmune POI mice. The transcriptome analysis of mice ovaries and in vitro co-cultivation experiments suggest that activation of the AKT and ERK pathways may be the key mechanism in the therapeutic effect of hAESCs. Our work provides the theoretical and experimental foundation for optimizing the administration of hAESCs, as well as the clinical application of hAESCs in autoimmune POI patients.
Collapse
Affiliation(s)
- Xiaohang Ye
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Yifeng Lin
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Yanyun Ying
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Xuezhi Shen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Feida Ni
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Feixia Wang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Jianpeng Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Wei Zhao
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Xiaoming Yu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, 310006, China.
| | - Yifeng Liu
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang, 310006, China.
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, 310006, China.
| |
Collapse
|
6
|
Yang Y, Li L, Yan T, Hua J, Li S, Liu Y, Yu S, Zhang H, Tang S, Xue Z, Zhang X, Zheng C. Evaluation of Safety and Efficacy of Amniotic Mesenchymal Stem Cells for POI in Animals. Reprod Sci 2024; 31:1159-1169. [PMID: 38097900 DOI: 10.1007/s43032-023-01417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/27/2023] [Indexed: 03/24/2024]
Abstract
The efficacy of human amniotic mesenchymal stem cell (hAMSC) ovarian injection in improving ovarian function in primary ovarian insufficiency (POI) patients has been shown in some reports. However, the safety and efficacy of hAMSC vein injection remains unclear. In this study, we evaluated the safety and efficacy of hAMSC intravenous injection in cynomolgus macaques and SD rats and provided evidence for clinical trials. The hAMSCs were transplanted three times in SD rats at low, medium, and high doses. The animal behavior and biochemical and biophysical parameters were routinely monitored on a 2-month period posttransplantation, and histopathologic examinations were also performed. Experiments on the acute toxicity, allergy test, and hemolysis test showed that hAMSCs possess good biocompatibility. Our results showed that the maximum tolerated dose of hAMSCs in SD rats was 4.0 × 107 cells/kg. The maximum safe dose with three injections of hAMSCs in SD rats was 5.0 × 106 cells/kg. In addition, the results demonstrated that hAMSCs may restore POI rat ovarian function after two injections of 2.5 × 106 cells/kg or 5.0 × 106 cells/kg, which improved the disturbed estrous cycle, hormone levels, and ovarian lesions induced by pZP3. In conclusion, the preclinical results suggested that the transplantation of hAMSCs may be safe and efficacious for SD rats at doses of 5.0 × 106 cells/kg and lower.
Collapse
Affiliation(s)
- Yuan Yang
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | - Li Li
- Changsha Health Vocational College, Changsha, 410100, Hunan, China
| | - Tenglong Yan
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | - Jiangzhou Hua
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | - Shiping Li
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | - Yun Liu
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | - Sijie Yu
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
| | | | - Shihuan Tang
- Loudi Central Hospital, Loudi, 417000, Hunan, China
| | - Zhigang Xue
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China
- Department of Regenerative Medicine, Translational Center for Stem Cell Research, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | | | - Chunbing Zheng
- Hunan Yuanpin Cell Technology Co. Ltd. (Yuanpin Biotech), Changsha, 410100, Hunan, China.
- Changsha Institute of Industrial Technology for Stem Cell and Regenerative Medicine, Changsha, 410100, China.
| |
Collapse
|
7
|
Hu L, Tan R, He Y, Wang H, Pu D, Wu J. Stem cell therapy for premature ovarian insufficiency: a systematic review and meta-analysis of animal and clinical studies. Arch Gynecol Obstet 2024; 309:457-467. [PMID: 37264272 DOI: 10.1007/s00404-023-07062-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023]
Abstract
PURPOSE The aim of this systematic review and meta-analysis is to evaluate the efficacy of stem cell therapy in mouse models of POI and patients with POI. METHODS The PubMed, Web of Science, and Embase databases were searched from inception to February 2022 for relevant animal and clinical studies. The reference lists of the included reviews were manually searched to identify additional eligible studies. Data were independently extracted by two investigators, and disagreements were resolved by discussion. SYRCLE's risk of bias tool and the MINORS tool were used to assess the quality of animal and clinical studies by two independent investigators. All statistical analyses were conducted using Review Manager 5.3 software. RESULTS A total of twenty animal studies and six clinical studies were included in this meta-analysis. In animal studies, the results showed that stem cells could improve hormone levels, follicle count, estrous cycle and pregnancy outcome. For hormone levels, stem cells increased serum E2 and AMH levels and decreased serum FSH and LH levels compared with the control group (serum E2 level: SMD: 5.05, 95% CI 4.21-5.90, P < 0.00001; serum AMH level: SMD: 4.42, 95% CI 3.06-5.79, P < 0.00001; serum FSH level: SMD: - 3.79, 95% CI - 4.87 to - 2.70, P < 0.00001; serum LH level: SMD: - 1.31, 95% CI - 1.65 to - 0.96, P < 0.00001). All follicle counts, except for the antral follicle count, were significantly changed compared with the control group. (primordial follicle count: SMD: 4.61, 95% CI 3.65-5.56, P < 0.00001; primary follicle count: SMD: 3.35, 95% CI 1.08-5.63, P = 0.004; secondary follicle count: SMD: 3.23, 95% CI 1.92-4.55, P < 0.00001; total follicle count: SMD: 4.84, 95% CI 2.86-6.83, P < 0.00001; oocyte count: SMD: 7.56, 95% CI 5.92-9.20, P < 0.00001; atretic follicle count: SMD: - 1.79, 95% CI - 2.59 to - 1.00, P < 0.00001). For the estrous cycle, stem cell therapy increased the number of estrous cycles (WMD: 2.72, 95% CI 2.07-3.37, P < 0.00001) and decreased the duration of the estrous cycle (WMD: - 1.26, 95% CI - 1.84 to - 0.69, P < 0.0001) compared with the control group. For pregnancy outcomes, stem cell therapy increased the fertility rate (RR: 3.00, 95% CI 1.74-5.17, P < 0.0001) and litter size (WMD: 3.82, 95% CI 0.36-7.28, P = 0.03) compared with the control group. In animal studies, the asymmetric funnel plot of serum E2 and FSH levels indicated the possibility of publication bias. Unpublished and negative studies may be the source of publication bias. In clinical studies, the results showed that stem cell therapy could decrease serum FSH level (MD: - 30.32, 95% CI - 59.03 to - 1.01, P = 0.04) and increase AFC (MD: 1.07, 95% CI 0.70-1.43, P < 0.00001), pregnancy rate (RD: 0.19, 95% CI 0.04-0.34, P = 0.01) and live birth rate (RD: 0.19, 95% CI 0.07-0.31, P = 0.001) in POI patients. In addition, there was no significant difference in menstrual function regained (RD: 0.22, 95% CI - 0.03-0.46, P = 0.09), oocytes retrieved (MD: 1.00, 95% CI - 0.64-2.64, P = 0.23) and embryos (MD: 0.80, 95% CI - 0.15-1.76, P = 0.10) between different groups. CONCLUSION This meta-analysis suggested that stem cell therapy might be effective in POI mouse models and patients and could be considered a potential treatment to restore fertility capability in POI patients.
Collapse
Affiliation(s)
- Luanqian Hu
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rongrong Tan
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuheng He
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huiyuan Wang
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Danhua Pu
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jie Wu
- Department of Obstetrics and Gynecology, Jiangsu Province Hospital, Jiangsu Women and Children Health Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Moustaki M, Kontogeorgi A, Tsangkalova G, Tzoupis H, Makrigiannakis A, Vryonidou A, Kalantaridou SN. Biological therapies for premature ovarian insufficiency: what is the evidence? FRONTIERS IN REPRODUCTIVE HEALTH 2023; 5:1194575. [PMID: 37744287 PMCID: PMC10512839 DOI: 10.3389/frph.2023.1194575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Premature Ovarian Insufficiency (POI) is a multi-factorial disorder that affects women of reproductive age. The condition is characterized by the loss of ovarian function before the age of 40 years and several factors have been identified to be implicated in its pathogenesis. Remarkably though, at least 50% of women have remaining follicles in their ovaries after the development of ovarian insufficiency. Population data show that approximately up to 3.7% of women worldwide suffer from POI and subsequent infertility. Currently, the treatment of POI-related infertility involves oocyte donation. However, many women with POI desire to conceive with their own ova. Therefore, experimental biological therapies, such as Platelet-Rich Plasma (PRP), Exosomes (exos) therapy, In vitro Activation (IVA), Stem Cell therapy, MicroRNAs and Mitochondrial Targeting Therapies are experimental treatment strategies that focus on activating oogenesis and folliculogenesis, by upregulating natural biochemical pathways (neo-folliculogenesis) and improving ovarian microenvironment. This mini-review aims at identifying the main advantages of these approaches and exploring whether they can underpin existing assisted reproductive technologies.
Collapse
Affiliation(s)
- Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | | | | | | | - Antonis Makrigiannakis
- Department of Obstetrics and Gynecology, University of Crete Medical School, Heraklion, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Sophia N. Kalantaridou
- Serum IVF Fertility Center, Athens, Greece
- 3rd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| |
Collapse
|
9
|
Rezayat F, Esmaeil N, Rezaei A. Potential Therapeutic Effects of Human Amniotic Epithelial Cells on Gynecological Disorders Leading to Infertility or Abortion. Stem Cell Rev Rep 2023; 19:368-381. [PMID: 36331801 DOI: 10.1007/s12015-022-10464-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
The induction of feto-maternal tolerance, fetal non-immunogenicity, and the regulation of mother's immune system are essential variables in a successful pregnancy. Fetal membranes have been used as a source of stem cells and biological components in recent decades. Human amniotic epithelial cells (hAEC) have stem/progenitor characteristics like those found in the amniotic membrane. Based on their immunomodulatory capabilities, recent studies have focused on the experimental and therapeutic applications of hAECs in allograft transplantation, autoimmune disorders, and gynecological problems such as recurrent spontaneous abortion (RSA), recurrent implantation failure (RIF), and premature ovarian failure (POF). This review discusses some of the immunomodulatory features and therapeutic potential of hAECs in preventing infertility, miscarriage, and implantation failure by controlling the maternal immune system.
Collapse
Affiliation(s)
- Fatemeh Rezayat
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran. .,Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran. .,Department of Immunology, School of Medicine, Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, 81744-176, Isfahan, Iran.
| | - Abbas Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Amniotic stem cells as a source of regenerative medicine to treat female infertility. Hum Cell 2023; 36:15-25. [PMID: 36251241 PMCID: PMC9813167 DOI: 10.1007/s13577-022-00795-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/13/2022] [Indexed: 01/09/2023]
Abstract
Impaired reproductive health is a worldwide problem that affects the psychological well-being of a society. Despite the technological developments to treat infertility, the global infertility rate is increasing significantly. Many infertility conditions are currently treated using various advanced clinical approaches such as intrauterine semination (IUI), in vitro fertilization (IVF), and intracytoplasmic injection (ICSI). Nonetheless, clinical management of some conditions such as dysfunctional endometrium, premature ovarian failure, and ovarian physiological aging still pose significant challenges. Stem cells based therapeutic strategies have a long-standing history to treat many infertility conditions, but ethical restrictions do not allow the broad-scale utilization of adult mesenchymal stromal/stem cells (MSCs). Easily accessible, placental derived or amniotic stem cells present an invaluable alternative source of non-immunogenic and non-tumorigenic stem cells that possess multilineage potential. Given these characteristics, placental or amniotic stem cells (ASCs) have been investigated for therapeutic purposes to address infertility in the last decade. This study aims to summarize the current standing and progress of human amniotic epithelial stem cells (hAECs), amniotic mesenchymal stem cells (hAMSCs), and amniotic fluid stem cells (hAFSCs) in the field of reproductive medicine. The therapeutic potential of these cells to restore or enhance normal ovarian function and pregnancy outcomes are highlighted in this study.
Collapse
|
11
|
Huang Y, Zhu M, Liu Z, Hu R, Li F, Song Y, Geng Y, Ma W, Song K, Zhang M. Bone marrow mesenchymal stem cells in premature ovarian failure: Mechanisms and prospects. Front Immunol 2022; 13:997808. [PMID: 36389844 PMCID: PMC9646528 DOI: 10.3389/fimmu.2022.997808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022] Open
Abstract
Premature ovarian failure (POF) is a common female reproductive disorder and characterized by menopause, increased gonadotropin levels and estrogen deficiency before the age of 40 years old. The etiologies and pathogenesis of POF are not fully clear. At present, hormone replacement therapy (HRT) is the main treatment options for POF. It helps to ameliorate perimenopausal symptoms and related health risks, but can't restore ovarian function and fertility fundamentally. With the development of regenerative medicine, bone marrow mesenchymal stem cells (BMSCs) have shown great potential for the recovery of ovarian function and fertility based on the advantages of abundant sources, high capacity for self-renewal and differentiation, low immunogenicity and less ethical considerations. This systematic review aims to summarize the possible therapeutic mechanisms of BMSCs for POF. A detailed search strategy of preclinical studies and clinical trials on BMSCs and POF was performed on PubMed, MEDLINE, Web of Science and Embase database. A total of 21 studies were included in this review. Although the standardization of BMSCs need more explorations, there is no doubt that BMSCs transplantation may represent a prospective therapy for POF. It is hope to provide a theoretical basis for further research and treatment for POF.
Collapse
Affiliation(s)
- Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengdi Zhu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Mingmin Zhang, ; Kunkun Song,
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Mingmin Zhang, ; Kunkun Song,
| |
Collapse
|
12
|
Fathi I, Miki T. Human Amniotic Epithelial Cells Secretome: Components, Bioactivity, and Challenges. Front Med (Lausanne) 2022; 8:763141. [PMID: 35083233 PMCID: PMC8784524 DOI: 10.3389/fmed.2021.763141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) derived from placental tissue have received significant attention as a promising tool in regenerative medicine. Several studies demonstrated their anti-inflammatory, anti-fibrotic, and tissue repair potentials. These effects were further shown to be retained in the conditioned medium of hAECs, suggesting their paracrine nature. The concept of utilizing the hAEC-secretome has thus evolved as a therapeutic cell-free option. In this article, we review the different components and constituents of hAEC-secretome and their influence as demonstrated through experimental studies in the current literature. Studies examining the effects of conditioned medium, exosomes, and micro-RNA (miRNA) derived from hAECs are included in this review. The challenges facing the application of this cell-free approach will also be discussed based on the current evidence.
Collapse
Affiliation(s)
- Ibrahim Fathi
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Lange-Consiglio A, Capra E, Herrera V, Lang-Olip I, Ponsaerts P, Cremonesi F. Application of Perinatal Derivatives in Ovarian Diseases. Front Bioeng Biotechnol 2022; 10:811875. [PMID: 35141212 PMCID: PMC8818994 DOI: 10.3389/fbioe.2022.811875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/05/2022] [Indexed: 12/18/2022] Open
Abstract
Reproductive diseases could lead to infertility and have implications for overall health, most importantly due to psychological, medical and socio-economic consequences for individuals and society. Furthermore, economical losses also occur in animal husbandry. In both human and veterinary medicine, hormonal and surgical treatments, as well as assisted reproductive technologies are used to cure reproductive disorders, however they do not improve fertility. With ovarian disorders being the main reproductive pathology in human and bovine, over the past 2 decades research has approached regenerative medicine in animal model to restore normal function. Ovarian pathologies are characterized by granulosa cell and oocyte apoptosis, follicular atresia, decrease in oocyte quality and embryonic development potential, oxidative stress and mitochondrial abnormalities, ultimately leading to a decrease in fertility. At current, application of mesenchymal stromal cells or derivatives thereof represents a valid strategy for regenerative purposes. Considering their paracrine/autocrine mode of actions that are able to regenerate injured tissues, trophic support, preventing apoptosis and fibrosis, promoting angiogenesis, stimulating the function and differentiation of endogenous stem cells and even reducing the immune response, are all important players in their future therapeutic success. Nevertheless, obtaining mesenchymal stromal cells (MSC) from adult tissues requires invasive procedures and implicates decreased cell proliferation and a reduced differentiation capacity with age. Alternatively, the use of embryonic stem cells as source of cellular therapeutic encountered several ethical concerns, as well as the risk of teratoma formation. Therefore, several studies have recently focussed on perinatal derivatives (PnD) that can be collected non-invasively and, most importantly, display similar characteristics in terms of regenerating-inducing properties, immune-modulating properties and hypo-immunogenicity. This review will provide an overview of the current knowledge and future perspectives of PnD application in the treatment of ovarian hypofunction.
Collapse
Affiliation(s)
- Anna Lange-Consiglio
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Lodi, Italy
- Centro Clinico-Veterinario e Zootecnico-Sperimentale di Ateneo, Università Degli Studi di Milano, Lodi, Italy
- *Correspondence: Anna Lange-Consiglio,
| | - Emanuele Capra
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale Delle Ricerche IBBA CNR, Lodi, Italy
| | - Valentina Herrera
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Lodi, Italy
| | - Ingrid Lang-Olip
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Fausto Cremonesi
- Dipartimento di Medicina Veterinaria, Università Degli Studi di Milano, Lodi, Italy
- Centro Clinico-Veterinario e Zootecnico-Sperimentale di Ateneo, Università Degli Studi di Milano, Lodi, Italy
| |
Collapse
|
14
|
Xu H, Xia Y, Qin J, Xu J, Li C, Wang Y. Effects of low intensity pulsed ultrasound on expression of B-cell lymphoma-2 and BCL2-Associated X in premature ovarian failure mice induced by 4-vinylcyclohexene diepoxide. Reprod Biol Endocrinol 2021; 19:113. [PMID: 34284777 PMCID: PMC8290625 DOI: 10.1186/s12958-021-00799-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 07/09/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Premature ovarian failure (POF) is a common disease in the field of Gynecology. Low intensity pulsed ultrasound (LIPUS) can promote tissue repair and improve function. This study was performed to determine the effects of LIPUS on granulosa cells (GCs) apoptosis and protein expression of B-cell lymphoma-2 (Bcl-2) and BCL2-Associated X (Bax) in 4-vinylcyclohexene diepoxide (VCD)-induced POF mice and investigate the mechanisms of LIPUS on ovarian function and reserve capacity. METHODS The current POF mice model was administrated with VCD (160 mg/kg) by intraperitoneal injection for 15 consecutive days. The mice were divided into the POF group, LIPUS group and control group. In the LIPUS group, the right ovary of mice was treated by LIPUS (acoustic intensity was 200 mW/cm2, frequency was 0.3 MHz, and duty cycle was 20%) for 20 min, 15 consecutive days from day 16. The mice of the POF group and control group were treated without ultrasonic output. The basic observation and body weight were recorded. Hematoxylin and eosin staining (H&E staining) and enzyme-linked immunosorbent assay (ELISA) were applied to detect ovarian follicle development, ovarian morphology and sex hormone secretion. Ovarian GCs apoptosis was detected by TUNEL assay and immunohistochemistry. RESULTS The results showed that VCD can induce estrus cycle disorder, follicular atresia, sex hormone secretion decreased and GCs apoptosis in mice to establish POF model successfully. LIPUS significantly promoted follicular development, increased sex hormone secretion, inhibited excessive follicular atresia and GCs apoptosis. The mechanism might be achieved by increasing the protein expression of Bcl-2 and decreasing the expression of Bax in ovaries. CONCLUSIONS LIPUS can improve the POF induced by VCD. These findings have the potential to provide novel methodological foundation for the future research, which help treat POF patients in the clinic.
Collapse
Affiliation(s)
- Haopeng Xu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yi Xia
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Juan Qin
- Department of Gynaecology, Guiyang Maternal and Child Health Hospital, Guizhou, 550003, China
| | - Jie Xu
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Chongyan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, 400016, Chongqing, China
| | - Yan Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
15
|
Huang Y, Ma Z, Kuang X, Zhang Q, Li H, Lai D. Sodium alginate-bioglass-encapsulated hAECs restore ovarian function in premature ovarian failure by stimulating angiogenic factor secretion. Stem Cell Res Ther 2021; 12:223. [PMID: 33794993 PMCID: PMC8015041 DOI: 10.1186/s13287-021-02280-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/10/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Human amniotic epithelial cells (hAECs) exhibit a strong capability to restore ovarian function in chemotherapy-induced premature ovarian failure (POF). However, the therapeutic efficacy of hAECs is usually affected by the limited number and proliferative ability of grafted hAECs in target organs. The transplantation of stem cells encapsulated in sodium alginate-bioglass (SA-BG) composite hydrogel has recently been shown to be an effective strategy for tissue regeneration. The current study aims to investigate the therapeutic potential of hAECs or hAEC-derived conditioned medium (CM) encapsulated in SA-BG in mice with chemotherapy-induced POF. METHODS C57BL/6 mice were intraperitoneally injected with chemotherapy drugs to induce POF. hAECs or CM were harvested and encapsulated in SA-BG composite hydrogel, which were transplanted onto the injured ovaries of mice with POF. Follicle development, granulosa cell function, and ovarian angiogenesis were evaluated by morphological methods. To further elucidate the effect of SA-BG-encapsulated hAECs/CM on vascularization, the tube formation of human umbilical vein epithelial cells (hUVECs) was conducted in vitro. Cytokine array and ELISA were used to analyze and quantify the effects of bioactive components released by SA-BG on the secretion of angiogenic factors by hAECs. RESULTS The transplantation of SA-BG-encapsulated hAECs/CM restored follicle development, repaired granulosa cell function, and enhanced ovarian angiogenesis in POF mice. The further study showed that SA-BG significantly promoted the tube formation of hUVECs in vitro. Moreover, encapsulating hAECs could facilitate the effect of SA-BG on inducing the formation of the capillary tube in a paracrine manner. In addition, we found that SA-BG extracts significantly enhanced the viability of hAECs and stimulated the secretion of pro-angiogenic factors of hAECs. Notably, compared with SA-BG/CM, SA-BG/hAECs achieve better therapeutic effects, possibly because stimulation of BG enhanced the viability and paracrine capacity of hAECs. CONCLUSIONS The present study initially demonstrates that SA-BG-encapsulated hAECs or CM can exert a therapeutic effect on chemotherapy-induced POF mainly by protecting granulosa cell function and enhancing ovarian vascularization, which might provide a novel strategy for the delivery of hAECs for treating POF.
Collapse
Affiliation(s)
- Yating Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 1961 Hua-Shan Road, Shanghai, 200030, People's Republic of China
| | - Zhijie Ma
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua-Shan Road, Shanghai, 200030, People's Republic of China
| | - Xiaojun Kuang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 1961 Hua-Shan Road, Shanghai, 200030, People's Republic of China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 1961 Hua-Shan Road, Shanghai, 200030, People's Republic of China. .,Shanghai Key Laboratory of Embryo Original Diseases, 145 Guang-Yuan Road, Shanghai, 200030, People's Republic of China.
| | - Haiyan Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua-Shan Road, Shanghai, 200030, People's Republic of China. .,Chemical and Environmental Engineering, School of Engineering, RMIT University, 124 La Trobe St, Melbourne, VIC, 3000, Australia.
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 1961 Hua-Shan Road, Shanghai, 200030, People's Republic of China. .,Shanghai Key Laboratory of Embryo Original Diseases, 145 Guang-Yuan Road, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
16
|
Ahmadian S, Mahdipour M, Pazhang M, Sheshpari S, Mobarak H, Bedate AM, Rahbarghazi R, Nouri M. Effectiveness of Stem Cell Therapy in the Treatment of Ovarian Disorders and Female Infertility: A Systematic Review. Curr Stem Cell Res Ther 2020; 15:173-186. [PMID: 31746298 DOI: 10.2174/1574888x14666191119122159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/22/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Infertility is a major problem worldwide. Various strategies are being used to develop better treatments for infertility and The most trending strategy is the stem cell therapy. In this study, the literature on stem cell therapy for ovarian disorders is summarized with analysis of current developments. OBJECTIVE Different published studies on stem cell-based therapy for the treatment of various types of ovarian insufficiency and disorders such as Premature Ovarian Insufficiency (POI) in the affected female population in animal or human clinical studies are systematically reviewed. METHODS We monitored five databases, including PubMed, Cochrane, Embase, Scopus, and ProQuest. A comprehensive online search was done using the criteria targeting the application of stem cells in animal models for menopause. Two independent reviewers carefully evaluated titles and abstracts of studies. The stem cell type, source, dosage, route of administration were highlighted in various POI animals models. Non-relevant and review articles were excluded. OUTCOMES 648 published studies were identified during the initial comprehensive search process from which 41 were selected according to designed criteria. Based on our analysis, stem cells could accelerate ovarian tissues rejuvenation, regulate systemic sex-related hormones levels and eventually increase fertility rate. CONCLUSION The evidence suggests that stem cell-based therapies could be considered as an alternative modality to deal with women undergoing POI.
Collapse
Affiliation(s)
- Shahin Ahmadian
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Pazhang
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Sepideh Sheshpari
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Alberto Miranda Bedate
- Laboratory for Translational Immunology (LTI), Universitair Medisch Centrum Utrecht, (UMCU), Utrecht, Netherlands
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Zhang Q, Lai D. Application of human amniotic epithelial cells in regenerative medicine: a systematic review. Stem Cell Res Ther 2020; 11:439. [PMID: 33059766 PMCID: PMC7559178 DOI: 10.1186/s13287-020-01951-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) derived from placental tissues have gained considerable attention in the field of regenerative medicine. hAECs possess embryonic stem cell-like proliferation and differentiation capabilities, and adult stem cell-like immunomodulatory properties. Compared with other types of stem cell, hAECs have special advantages, including easy isolation, plentiful numbers, the obviation of ethical debates, and non-immunogenic and non-tumorigenic properties. During the past two decades, the therapeutic potential of hAECs for treatment of various diseases has been extensively investigated. Accumulating evidence has demonstrated that hAEC transplantation helps to repair and rebuild the function of damaged tissues and organs by different molecular mechanisms. This systematic review focused on summarizing the biological characteristics of hAECs, therapeutic applications, and recent advances in treating various tissue injuries and disorders. Relevant studies published in English from 2000 to 2020 describing the role of hAECs in diseases and phenotypes were comprehensively sought out using PubMed, MEDLINE, and Google Scholar. According to the research content, we described the major hAEC characteristics, including induced differentiation plasticity, homing and differentiation, paracrine function, and immunomodulatory properties. We also summarized the current status of clinical research and discussed the prospects of hAEC-based transplantation therapies. In this review, we provide a comprehensive understanding of the therapeutic potential of hAECs, including their use for cell replacement therapy as well as secreted cytokine and exosome biotherapy. Moreover, we showed that the powerful immune-regulatory function of hAECs reveals even more possibilities for their application in the treatment of immune-related diseases. In the future, establishing the optimal culture procedure, achieving precise and accurate treatment, and enhancing the therapeutic potential by utilizing appropriate preconditioning and/or biomaterials would be new challenges for further investigation.
Collapse
Affiliation(s)
- Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory of Embryo Original Diseases; Shanghai Municipal Key Clinical Speciality, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University; Shanghai Key Laboratory of Embryo Original Diseases; Shanghai Municipal Key Clinical Speciality, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
18
|
Cui L, Bao H, Liu Z, Man X, Liu H, Hou Y, Luo Q, Wang S, Fu Q, Zhang H. hUMSCs regulate the differentiation of ovarian stromal cells via TGF-β 1/Smad3 signaling pathway to inhibit ovarian fibrosis to repair ovarian function in POI rats. Stem Cell Res Ther 2020; 11:386. [PMID: 32894203 PMCID: PMC7487655 DOI: 10.1186/s13287-020-01904-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 01/01/2023] Open
Abstract
Objective The basic pathological changes of primary ovarian insufficiency (POI) include ovarian tissue fibrosis and follicular development disorders. The human umbilical cord mesenchymal stem cell (hUMSC) transplantation has been shown an effective method to improve the ovarian function in POI rat model; however, the exact mechanisms are still unclear. The purpose of this study is to investigate whether the recovery of ovarian function in POI rats is related to the inhibition of tissue fibrosis following hUMSC transplantation. Furthermore, the transforming growth factor-β1 (TGF-β1) signaling pathway is explored to determine the mechanisms of ovarian function recovery through its inhibition of tissue fibrosis. Methods The primary ovarian insufficiency (POI) rat model was established by intraperitoneal injection of chemotherapy drug cisplatin (CDDP) for 7 days. The levels of serum sex hormones were measured using enzyme-linked immunosorbent assay (ELISA). The tissue fibrosis in the ovary was examined using Masson staining and Sirius red staining. The collagen fibers in the ovarian tissues were detected by Western blot analysis. To investigate the mechanisms of ovarian function recovery following hUMSC transplantation, ovarian stromal cells were isolated from the ovarian cortex of immature rats. The expression of Cytochrome P450 17A1 (Cyp17a1) and fibrosis marker of alpha smooth muscle actin (α-SMA) in ovarian stromal cells was examined using immunofluorescence analysis. Also, the protein levels of Cyp17a1 and α-SMA in ovarian stromal cells were examined by Western blot analysis. The expression of TGF-β1 and Smad3 signals was measured by Western blot and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis. Results The results show that the function of the ovary in POI rats was significantly improved after hUMSC transplantation. The expression of fibrosis markers (α-SMA) and production of Collagen Type I (Collagen I) and Collagen Type III (Collagen III) in POI rats were significantly inhibited in POI rats following hUMSC transplantation. In the cultured ovarian stromal cells, the decrease of TGF-β1 and p-Smad3 protein expression was observed in hUMSC-treated POI rats. The treatment with TGF-β1 inhibitor of SB431542 further confirmed this signal pathway was involved in the process. Conclusion Our study demonstrated that the TGF-β1/Smad3 signaling pathway was involved in the inhibition of ovarian tissue fibrosis, which contributed to the restoration of ovarian function in POI rats following hUMSC transplantation.
Collapse
Affiliation(s)
- Linlu Cui
- College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China.,College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Hongchu Bao
- Department of Clinical Medicine, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Zhongfeng Liu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Xuejing Man
- Department of Clinical Medicine, Yantai Yuhuangding Hospital, Yantai, 264000, Shandong, China
| | - Hongyuan Liu
- Clinical Medical School, Binzhou Medical University, Yantai, Shandong, China
| | - Yun Hou
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Qianqian Luo
- College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Siyuan Wang
- Clinical Medical School, Binzhou Medical University, Yantai, Shandong, China
| | - Qiang Fu
- School of pharmacy, Binzhou Medical University, Yantai, Shandong, China.
| | - Hongqin Zhang
- College of Basic Medicine & Institute of Reproductive Diseases, Binzhou Medical University, Yantai, 264003, Shandong, China. .,College of Basic Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.
| |
Collapse
|
19
|
Bahrehbar K, Rezazadeh Valojerdi M, Esfandiari F, Fathi R, Hassani SN, Baharvand H. Human embryonic stem cell-derived mesenchymal stem cells improved premature ovarian failure. World J Stem Cells 2020; 12:857-878. [PMID: 32952863 PMCID: PMC7477659 DOI: 10.4252/wjsc.v12.i8.857] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/01/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Premature ovarian failure (POF) affects many adult women less than 40 years of age and leads to infertility. According to previous reports, various tissue-specific stem cells can restore ovarian function and folliculogenesis in mice with chemotherapy-induced POF. Human embryonic stem cells (ES) provide an alternative source for mesenchymal stem cells (MSCs) because of their similarities in phenotype and immunomodulatory and anti-inflammatory characteristics. Embryonic stem cell-derived mesenchymal stem cells (ES-MSCs) are attractive candidates for regenerative medicine because of their high proliferation and lack of barriers for harvesting tissue-specific MSCs. However, possible therapeutic effects and underlying mechanisms of transplanted ES-MSCs on cyclophosphamide and busulfan-induced mouse ovarian damage have not been evaluated. AIM To evaluate ES-MSCs vs bone marrow-derived mesenchymal stem cells (BM-MSCs) in restoring ovarian function in a mouse model of chemotherapy-induced premature ovarian failure. METHODS Female mice received intraperitoneal injections of different doses of cyclophosphamide and busulfan to induce POF. Either human ES-MSCs or BM-MSCs were transplanted into these mice. Ten days after the mice were injected with cyclophosphamide and busulfan and 4 wk after transplantation of the ES-MSCs and/or BM-MSCs, we evaluated body weight, estrous cyclicity, follicle-stimulating hormone and estradiol hormone concentrations and follicle count were used to evaluate the POF model and cell transplantation. Moreover, terminal deoxynucleotidyl transferase mediated 2-deoxyuridine 5-triphosphate nick end labeling, real-time PCR, Western blot analysis and immunohistochemistry and mating was used to evaluate cell transplantation. Enzyme-linked immunosorbent assay was used to analyze vascular endothelial growth factor, insulin-like growth factor 2 and hepatocyte growth factor levels in ES-MSC condition medium in order to investigate the mechanisms that underlie their function. RESULTS The human ES-MSCs significantly restored hormone secretion, survival rate and reproductive function in POF mice, which was similar to the results obtained with BM-MSCs. Gene expression analysis and the terminal deoxynucleotidyl transferase mediated 2-deoxyuridine 5-triphosphate nick end labeling assay results indicated that the ES-MSCs and/or BM-MSCs reduced apoptosis in the follicles. Notably, the transplanted mice generated new offspring. The results of different analyses showed increases in antiapoptotic and trophic proteins and genes. CONCLUSION These results suggested that transplantation of human ES-MSCs were similar to BM-MSCs in that they could restore the structure of the injured ovarian tissue and its function in chemotherapy-induced damaged POF mice and rescue fertility. The possible mechanisms of human ES-MSC were related to promotion of follicular development, ovarian secretion, fertility via a paracrine effect and ovarian cell survival.
Collapse
Affiliation(s)
- Khadijeh Bahrehbar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 1665659911, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Tehran 1665659911, Iran
- Department of Anatomy, Faculty of Medical Science, Tarbiat Modares University, Tehran 1665659911, Iran
| | - Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 1665659911, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, Tehran 1665659911, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 1665659911, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 1665659911, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 1665659911, Iran.
| |
Collapse
|
20
|
Ahmadian S, Sheshpari S, Pazhang M, Bedate AM, Beheshti R, Abbasi MM, Nouri M, Rahbarghazi R, Mahdipour M. Intra-ovarian injection of platelet-rich plasma into ovarian tissue promoted rejuvenation in the rat model of premature ovarian insufficiency and restored ovulation rate via angiogenesis modulation. Reprod Biol Endocrinol 2020; 18:78. [PMID: 32758249 PMCID: PMC7405361 DOI: 10.1186/s12958-020-00638-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Premature Ovarian Insufficiency (POI) is viewed as a type of infertility in which the menopausal status occurs before the physiological age. Several therapeutic strategies have been introduced in clinic for POI treatment, although the outputs are not fully convincing. Platelet-rich plasma (PRP) is a unique blood product widely applied in regenerative medicine, which is based on the releasing of the growth factors present in platelets α-granules. In the current investigation, we examined the effectiveness of PRP as a therapeutic alternative for POI animals. POI in Wistar albino rats was induced by daily intraperitoneal (IP) administration of gonadotoxic chemical agent, 4-vinylcyclohexene dioxide (VCD) (160 mg/ kg) for 15 consecutive days. After POI induction, the PRP solution was directly injected intra-ovarian in two concentrations via a surgical intervention. Every two weeks post-injection, pathological changes were monitored in the ovaries using Hematoxylin-Eosin staining method, until eight weeks. Follicle Stimulating Hormone (FSH) content in serum was measured, together with the expression of the angiogenic-related transcripts ANGPT2 and KDR by real-time qPCR. Furthermore the fertility status of the treated rats was evaluated by mating trials. Histopathological examination revealed successful POI induction via the depletion of morphologically normal follicles in rats following VCD treatment compared to the control rats. The injection of PRP at two concentrations reduced the number and extent of the follicular atresia and inflammatory responses (p < 0.05). The expression of both ANGPT2 and KDR transcripts were significantly increased in POI rats due to enhanced inflammation, while these values were modulated after PRP administration (p < 0.05) compared to POI rats. FSH showed a decreased trend in concentration eight weeks after PRP treatment, but not statistically significant (p > 0.05). Nevertheless, a clear improvement in litter counts was found in POI rats receiving PRP compared to the non-treated POI group, being able to consider PRP as a facile, quick, accessible, safe and relatively cheap alternative therapeutic strategy to revert POI-related pathologies.
Collapse
Affiliation(s)
- Shahin Ahmadian
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, 5138663134, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 537517169, Iran
| | - Sepideh Sheshpari
- Department of Midwifery, Faculty of Nursing and Midwifery, Tabriz University of Medical Sciences, Tabriz, 5138947977, Iran
| | - Mohammad Pazhang
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 537517169, Iran
| | - Alberto Miranda Bedate
- Laboratory for Translational Immunology (LTI), Universitair Medisch Centrum Utrecht, (UMCU), Utrecht, Heidelberglaan 100, 3584, CX, The Netherlands
| | - Rahim Beheshti
- Department of Veterinary Science, Islamic Azad University Shabestar Branch, Shabestar, 5381637181, Iran
| | | | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665811, Iran.
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 5166653431, Iran.
| |
Collapse
|
21
|
Zhang Y, Ouyang X, You S, Zou H, Shao X, Zhang G, Zhang C, Hu L. Effect of human amniotic epithelial cells on ovarian function, fertility and ovarian reserve in primary ovarian insufficiency rats and analysis of underlying mechanisms by mRNA sequencing. Am J Transl Res 2020; 12:3234-3254. [PMID: 32774697 PMCID: PMC7407690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 05/30/2020] [Indexed: 06/11/2023]
Abstract
Human amniotic epithelial cells (hAECs) show similar features to stem cells and have low immunogenicity. This study aims to investigate the therapeutic effect of hAEC transplantation on cyclophosphamide-induced primary ovarian insufficiency (POI) rats and evaluate the underlying mechanisms by mRNA sequencing of ovarian samples. Notably, hAECs mainly located in the interstitial area of the ovaries rather than follicles. hAEC transplantation led to a slight increase in body and ovary weight, normalized irregular estrous cycles, decreased serum follicle stimulating hormone (FSH) and increased anti-Mullerian hormone (AMH) level and restored follicle pools in POI rats. Ovarian expression of AMH, follicle stimulating hormone receptor (FSHR) and klotho in POI rats was also significantly upregulated following hAEC transplantation. Fetus number was higher in the hAEC transplantation group than the POI group. The mRNA sequencing results showed that hAEC transplantation led to the upregulation of several angiogenesis and inflammation molecules including interferon regulatory factor 7 (IRF7), Mx dynamin-like GTPase 1 (Mx1), vascular endothelial growth factor receptor (VEGFR)1 and VEGFR2. Moreover, hAEC therapy had an effect on ribosomes, protein digestion, protein absorption, neuroactive ligand-receptor interaction, cAMP signaling pathway and steroid biosynthesis pathways. The expression of several steroid biosynthesis proteins was significantly upregulated as measured by quantitative real-time polymerase chain reaction (RT-qPCR), immunohistochemical staining and Western blot analysis. In summary, hAECs can significantly restore ovarian function, and improve both ovarian reserve and fertility. This may be due to the paracrine effect of hAECs in regulating steroid biosynthesis, modulating follicle development from initiation to ovulation, promoting angiogenesis and reducing inflammation.
Collapse
Affiliation(s)
- Yulin Zhang
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
| | - Xiaolan Ouyang
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
| | - Shuang You
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
| | - Heng Zou
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
- Reproduction and Stem Cell Therapy Research Center of ChongqingChina
- Joint International Research Lab for Reproduction and Development, Ministry of EducationChina
| | - Xiaoyan Shao
- Shanghai iCELL Biotechnology Co., Ltd.Shanghai 200333, China
| | - Guanghui Zhang
- Chongqing Engineering Technology Research Center of Stem Cell and Neural RegenerationChongqing, China
- Chongqing Guolian Stem Cell Technology Co., Ltd.Chongqing, China
| | - Chanyu Zhang
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
- Reproduction and Stem Cell Therapy Research Center of ChongqingChina
- Joint International Research Lab for Reproduction and Development, Ministry of EducationChina
| | - Lina Hu
- The Center for Reproductive Medicine, Obstetrics and Gynecology Department, The Second Affiliated Hospital, Chongqing Medical UniversityChongqing, China
- Reproduction and Stem Cell Therapy Research Center of ChongqingChina
- Joint International Research Lab for Reproduction and Development, Ministry of EducationChina
| |
Collapse
|
22
|
Li B, Zhang Q, Sun J, Lai D. Human amniotic epithelial cells improve fertility in an intrauterine adhesion mouse model. Stem Cell Res Ther 2019; 10:257. [PMID: 31412924 PMCID: PMC6694540 DOI: 10.1186/s13287-019-1368-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/23/2019] [Accepted: 08/01/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Intrauterine adhesion (IUA) is an adhesion of the uterine cavity or cervical canal resulting from damage to the basal layer of the endometrium; this condition is usually accompanied by fibrosis of the endometrium. Previous studies have demonstrated that human amniotic epithelial cells (hAECs) have stem cell characteristics; however, it is unclear whether hAECs have the therapeutic potential to restore fertility after IUA. METHODS A murine IUA model was established by mechanical injury to the uterus. Then, 106 hAECs were transplanted by intraperitoneal injection. The endometrium thickness, number of glands, and fibrosis area were measured by hematoxylin and eosin (H&E) staining and Masson staining. Molecules (including vWF, VEGF, PCNA, ER, PR, LC3, and p62) related to endometrial angiogenesis, cell proliferation, and autophagy were assayed by IHC staining. Pregnancy outcomes were also evaluated. Finally, hAECs were cocultured with human endometrial mesenchymal stem cells (hEnSCs) damaged by H2O2 to verify the paracrine effect on endometrial stromal cells in vitro. RESULTS The IUA uterine cavity presented with adhesion and even atresia, accompanied by a thinner endometrium, fewer glands, increased fibrosis area, and fewer microvessels. However, hAECs significantly improved the uterine structure after IUA. After hAEC treatment, the endometrium was thicker, the number of endometrial glands was increased, fibrosis was reduced, and more microvessels were generated. The expression levels of VEGF, PCNA, and ER were increased in the hAEC-treated endometrium, indicating improvements in angiogenesis and stromal cell proliferation. hAECs also increased pregnancy outcomes in IUA mice, and the pregnancy rate and fetus number increased. Furthermore, we observed altered autophagy in the IUA uterine model, and hAEC transplantation upregulated autophagy. An in vitro study showed that hAECs activated autophagy in (hEnSCs) treated with H2O2 in a paracrine manner. CONCLUSIONS Our results demonstrated that hAECs have the potential to repair the uterus after injury, providing a new strategy for the prevention and treatment of Asherman syndrome.
Collapse
Affiliation(s)
- Boning Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qiuwan Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Junyan Sun
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
- Shanghai Municipal Key Clinical Speciality, Shanghai, 20030, China.
| |
Collapse
|
23
|
Zhang Q, Huang Y, Sun J, Gu T, Shao X, Lai D. Immunomodulatory effect of human amniotic epithelial cells on restoration of ovarian function in mice with autoimmune ovarian disease. Acta Biochim Biophys Sin (Shanghai) 2019; 51:845-855. [PMID: 31287492 DOI: 10.1093/abbs/gmz065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/28/2019] [Indexed: 12/23/2022] Open
Abstract
Autoimmune ovarian disease (AOD) is considered to be a major cause of premature ovarian failure (POF). The immunomodulatory properties of human amniotic epithelial cells (hAECs) have been studied in many disease models. We previously reported that hAECs restored ovarian function in chemotherapy-induced POF mice, but the immunomodulatory mechanism of hAECs is still unclear. To investigate the effect of hAECs on recipient mice, especially on regulatory Treg cells, hAECs and hAEC-conditioned medium (hAEC-CM) were intravenously injected into AOD mice immunized with zona pellucida protein 3 peptides (pZP3). Ovarian function was evaluated through estrous cycle, hormone secretion, follicle development, and cell apoptosis analysis. Immune cells including CD3, CD4, CD8 and Treg cells in the spleens were tested by flow cytometry. To elucidate the effect of hAEC-CM on macrophage function, inflammation model in vitro was established in RAW264.7 cells induced by lipopolysaccharide (LPS). hAECs and hAEC-CM regulated estrous cycles, promoted follicle development, ameliorated cell apoptosis and fibrosis in ovaries of AOD mice. In addition, hAECs significantly reversed the decrease of pZP3-induced Treg cells in the spleens. In vitro, hAEC-CM significantly inhibited the inflammatory reaction induced by LPS in RAW264.7 cells via up-regulating the expression of M2 macrophage genes. Further study demonstrated that hAEC-secreted transforming growth factor-beta and macrophage inhibitory factor played important roles in the macrophage polarization and migration under inflammatory stimulation. Taken together, hAECs restored ovarian function by up-regulating Treg cells in the spleens and reduced the inflammatory reaction via modulating the activated macrophage function in a paracrine manner in the ovaries of AOD mice.
Collapse
Affiliation(s)
- Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Shanghai Municipal Key Clinical Speciality, Shanghai, China
| | - Yating Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junyan Sun
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Gu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Shao
- Shanghai iCELL Biotechnology Co., Ltd, Shanghai, China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Shanghai Municipal Key Clinical Speciality, Shanghai, China
| |
Collapse
|
24
|
Human Amniotic Epithelial Cell-Derived Exosomes Restore Ovarian Function by Transferring MicroRNAs against Apoptosis. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:407-418. [PMID: 31022607 PMCID: PMC6479666 DOI: 10.1016/j.omtn.2019.03.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/20/2019] [Accepted: 03/16/2019] [Indexed: 02/08/2023]
Abstract
Premature ovarian failure (POF) is one of the most common complications among female patients with tumors treated with chemotherapy and requires advanced treatment strategies. Human amniotic epithelial cell (hAEC)-based therapy mediates tissue regeneration in a variety of diseases, and increasing evidence suggests that the therapeutic efficacy of hAECs mainly depends on paracrine action. This study aimed to identify exosomes derived from hAECs and explored the therapeutic potential in ovaries damaged by chemotherapy and the underlying molecular mechanism. hAEC-derived exosomes exhibited a cup- or sphere-shaped morphology with a mean diameter of 100 nm and were positive for Alix, CD63, and CD9. hAEC exosomes increased the number of follicles and improved ovarian function in POF mice. During the early stage of transplantation, hAEC exosomes significantly inhibited granulosa cell apoptosis, protected the ovarian vasculature from damage, and were involved in maintaining the number of primordial follicles in the injured ovaries. Enriched microRNAs (miRNAs) existed in hAEC exosomes, and target genes were enriched in phosphatidylinositol signaling and apoptosis pathways. Studies in vitro demonstrated that hAEC exosomes inhibited chemotherapy-induced granulosa cell apoptosis via transferring functional miRNAs, such as miR-1246. Our findings demonstrate that hAEC-derived exosomes have the potential to restore ovarian function in chemotherapy-induced POF mice by transferring miRNAs.
Collapse
|
25
|
Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism. Stem Cell Res Ther 2019; 10:46. [PMID: 30683144 PMCID: PMC6347748 DOI: 10.1186/s13287-019-1136-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/03/2019] [Accepted: 01/06/2019] [Indexed: 12/21/2022] Open
Abstract
Background Chemotherapy can induce premature ovarian insufficiency (POI) and reduce fertility in young female patients. Currently, there is no effective therapy for POI. Human amnion-derived mesenchymal stem cells (hAD-MSCs) may be a promising seed cell for regenerative medicine. This study investigated the effects and mechanisms of hAD-MSC transplantation on chemotherapy-induced POI in rats. Methods Chemotherapy-induced POI rat models were established by intraperitoneal injection of cyclophosphamide. Seventy-two female SD rats were randomly divided into control, POI, and hAD-MSC-treated groups. hAD-MSCs were labeled with PKH26 and injected into the tail veins of POI rats. To examine the underlying mechanisms, the differentiation of transplanted hAD-MSCs in the POI ovaries was analyzed by immunofluorescent staining. The in vitro expression of growth factors secreted by hAD-MSCs in hAD-MSC-conditioned media (hAD-MSC-CM) was analyzed by ELISA. Sixty female SD rats were divided into control, POI, and hAD-MSC-CM-treated groups, and hAD-MSC-CM was injected into the bilateral ovaries of POI rats. After hAD-MSC transplantation or hAD-MSC-CM injection, serum sex hormone levels, estrous cycles, ovarian pathological changes, follicle counts, granulosa cell (GC) apoptosis, and Bcl-2, Bax, and VEGF expression in ovaries were examined. Results PKH26-labeled hAD-MSCs mainly homed to ovaries after transplantation. hAD-MSC transplantation reduced ovarian injury and improved ovarian function in rats with POI. Transplanted hAD-MSCs were only located in the interstitium of ovaries, rather than in follicles, and did not express the typical markers of oocytes and GCs, which are ZP3 and FSHR, respectively. hAD-MSCs secreted FGF2, IGF-1, HGF, and VEGF, and those growth factors were detected in the hAD-MSC-CM. hAD-MSC-CM injection improved the local microenvironment of POI ovaries, leading to a decrease in Bax expression and an increase in Bcl-2 and endogenous VEGF expression in ovarian cells, which inhibited chemotherapy-induced GC apoptosis, promoted angiogenesis and regulated follicular development, thus partly reducing ovarian injury and improving ovarian function in rats with POI. Conclusions hAD-MSC transplantation can improve ovarian function in rats with chemotherapy-induced POI at least partly through a paracrine mechanism. The presence of a paracrine mechanism accounting for hAD-MSC-mediated recovery of ovarian function might be attributed to the growth factors secreted by hAD-MSCs.
Collapse
|
26
|
Tan B, Yuan W, Li J, Yang P, Ge Z, Liu J, Qiu C, Zhu X, Qiu C, Lai D, Guo L, Wang L, Yu L. Therapeutic effect of human amniotic epithelial cells in murine models of Hashimoto's thyroiditis and Systemic lupus erythematosus. Cytotherapy 2018; 20:1247-1258. [PMID: 30174233 DOI: 10.1016/j.jcyt.2018.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/26/2018] [Accepted: 04/02/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS The chronic inflammation of autoimmune diseases develops repetitive localized destruction or systemic disorders, represented by Hashimoto's thyroiditis (HT) and Systemic lupus erythematosus (SLE) respectively. Currently, there are no efficient ways to treat these autoimmune diseases. Therefore, it is critically important to explore new therapeutic strategies. The aim of this study was to investigate the therapeutic efficacy of human amniotic epithelial cells (hAECs) in murine models of HT and SLE. METHODS Experimental autoimmune thyroiditis (EAT) was induced in female CBA/J mice by immunization with porcine thyroglobulin (pTg). hAECs were intravenously administered at different time points during the disease course. MRL-Faslpr mice, a strain with spontaneously occurring SLE, were intravenously administered hAECs when their sera were positive for both anti-nuclear antibodies (ANAs) and anti-double-stranded DNA (anti-dsDNA) antibodies. Two weeks after the last cell transplantation, blood and tissue samples were collected for histological examination and immune system analysis. RESULTS hAECs prevented lymphocytes infiltration into the thyroid and improved the damage of thyroid follicular in EAT mice. Correspondingly, hAECs administration reduced anti-thyroglobulin antibodies (TGAb), anti-thyroid peroxidase antibodies (TPOAb) and thyroid stimulating hormone (TSH) levels. SLE mice injected with hAECs appeared negative for ANAs and anti-dsDNA antibodies and showed reduced immunoglobulin profiles. Mechanically, hAECs modulated the immune cells balance in EAT and SLE mice, by downregulating the ratios of Th17/Treg cells in both EAT and SLE mice and upregulating the proportion of B10 cells in EAT mice. This was confirmed by in vitro assay, in which hAECs inhibited the activation of EAT mice-derived splenocytes. Moreover, hAECs improved the cytokine environment in both EAT and SLE mice, by suppressing the levels of IL-17A and IFN-γ and enhancing TGF-β. CONCLUSION These results demonstrated the immunoregulatory effect of hAECs for inflammation inhibition and injury recovery in HT and SLE murine models. The current study may provide a novel therapeutic strategy for these autoimmune diseases in clinic.
Collapse
Affiliation(s)
- Bing Tan
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Weixin Yuan
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Jinying Li
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Pengjie Yang
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Zhen Ge
- Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, China
| | - Jia Liu
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Chen Qiu
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Xiaolong Zhu
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Cong Qiu
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lihe Guo
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai iCELL Biotechnology Co Ltd, Shanghai 200333, China
| | - Liang Wang
- Center for Stem Cell and Regenerative Medicine, Hangzhou, China; The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Luyang Yu
- Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, China; College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, China; Center for Stem Cell and Regenerative Medicine, Hangzhou, China.
| |
Collapse
|
27
|
Li J, Qiu C, Zhang Z, Yuan W, Ge Z, Tan B, Yang P, Liu J, Zhu X, Qiu C, Lai D, Guo L, Yu L. Subretinal Transplantation of Human Amniotic Epithelial Cells in the Treatment of Autoimmune Uveitis in Rats. Cell Transplant 2018; 27:1504-1514. [PMID: 30168350 PMCID: PMC6180726 DOI: 10.1177/0963689718796196] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
As a featured ocular inflammatory disease, autoimmune uveitis is the major cause of blindness in the clinic. Although current immunosuppressive regimens can alleviate the progression of autoimmune uveitis, they have serious side effects. Therefore, an alternative therapeutic strategy is urgently required. The present study investigated the therapeutic efficacy of human amniotic epithelial cells (hAECs) on autoimmune uveitis in a rat model. Herein, experimental autoimmune uveitis (EAU) was induced in rats via a subcutaneous injection of interphotoreceptor retinoid-binding protein. EAU rats were treated with hAECs or the vehicle solution via a subretinal injection on day 0 and day 6 after immunization, and rats were sacrificed on day 12 and day 18 for further analysis. The pathological development of EAU was evaluated by slit lamp microscopy. Immune cell infiltration and retinal structure damage were examined by histological examination of hematoxylin and eosin (H&E) and immunofluorescence staining. T-cell subsets were detected by flow cytometry, and the levels of inflammatory cytokines were quantified by enzyme-linked immunosorbent assay (ELISA). hAEC treatment ameliorated the pathological progression of EAU and preserved the retinal structure organization and thickness, especially in the preventive group that received a subretinal injection on day 0. Moreover, hAECs inhibited the retinal infiltration of macrophages and T-cells. Mechanistically, hAECs modulated the balance of T-cell subsets by downregulating T helper (Th)17 cells and upregulating T regulatory (Treg) cells, as confirmed by decreased interleukin (IL)-17 and increased IL-10 levels in the spleens and lymph nodes of EAU rats. Furthermore, hAECs improved the local cytokine environment in EAU rats by suppressing the monocyte chemoattractant protein (MCP)-1, IL-17 and interferon (IFN)-γ levels and enhancing the IL-10 in the aqueous humor. Therefore, subretinal transplantation of hAECs in EAU rats ameliorated ocular inflammation, preserved the retinal structure and coordinated the immune balance. The current study provides a novel therapeutic strategy for autoimmune uveitis and related ocular inflammatory diseases in the clinic.
Collapse
Affiliation(s)
- Jinying Li
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Chen Qiu
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Zheng Zhang
- 4 The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weixin Yuan
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Zhen Ge
- 5 Institute of Materia Medica, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang, China
| | - Bing Tan
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Pengjie Yang
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Jia Liu
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Xiaolong Zhu
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Cong Qiu
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| | - Dongmei Lai
- 6 International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lihe Guo
- 7 Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,8 Shanghai iCELL Biotechnology Co Ltd, Shanghai, China
| | - Luyang Yu
- 1 Institute of Genetics and Regenerative Biology, College of Life Sciences, Hangzhou, Zhejiang, China.,2 College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, Hangzhou, Zhejiang, China.,3 Center for Stem Cell and Regenerative Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Biological characterization of human amniotic epithelial cells in a serum-free system and their safety evaluation. Acta Pharmacol Sin 2018; 39:1305-1316. [PMID: 29565036 DOI: 10.1038/aps.2018.22] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/01/2018] [Indexed: 12/16/2022]
Abstract
Human amniotic epithelial cells (hAECs), derived from the innermost layer of the term placenta closest to the fetus, have been shown to be potential seed cells for allogeneic cell therapy. Previous studies have shown a certain therapeutic effect of hAECs. However, no appropriate isolation and culture system for hAECs has been developed for clinical applications. In the present study, we established a serum-free protocol for hAEC isolation and cultivation, in which better cell growth was observed compared with that in a traditional culture system with serum. In addition to specific expression of cell surface markers (CD29, CD166 and CD90), characterization of the biological features of hAECs revealed expression of the pluripotent markers SSEA4, OCT4 and NANOG, which was greater than that in human mesenchymal stem cells, whereas very low levels of HLA-DR and HLA-DQ were detected, suggesting the weak immunogenicity of hAECs. Intriguingly, CD90+ hAECs were identified as a unique population with a powerful immunoregulatory capacity. In a systemic safety evaluation, intravenous administration of hAEC did not result in hemolytic, allergy, toxicity issues or, more importantly, tumorigenicity. Finally, the therapeutic effect of hAECs was demonstrated in mice with radiation-induced damage. The results revealed a novel function of hAECs in systemic injury recovery. Therefore, the current study provides an applicable and safe strategy for hAEC cell therapy administration in the clinical setting.
Collapse
|
29
|
Li J, Yu Q, Huang H, Deng W, Cao X, Adu-Frimpong M, Yu J, Xu X. Human chorionic plate-derived mesenchymal stem cells transplantation restores ovarian function in a chemotherapy-induced mouse model of premature ovarian failure. Stem Cell Res Ther 2018; 9:81. [PMID: 29615109 PMCID: PMC5883538 DOI: 10.1186/s13287-018-0819-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/07/2018] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background Previous studies have reported that transplantation of mesenchymal stem cells (MSCs) from many human tissues could ameliorate ovarian dysfunction. However, no study has revealed the therapeutic efficiency of MSCs derived from the chorionic plate (CP-MSCs) for premature ovarian failure (POF). Methods We investigated the restorative effects of CP-MSCs on cyclophosphamide (CTX)-induced POF. The POF mouse models were established via intraperitoneal injection of 50 mg/kg CTX into female mice for 15 consecutive days. After that, CP-MSCs were intravenously transplanted into the mice once a week for 4 weeks. The serum estradiol (E2) and follicle-stimulating hormone (FSH) levels in the mouse models were detected using enzyme-linked immunosorbent assay (ELISA) before and after treatment. Ovarian function was evaluated through counting the follicles, estrous cycles, and oocytes. Results CP-MSC transplantation restored the serum hormone level and ovarian function of the mice in the mouse model of POF induced by CTX. The levels of serum E2 and FSH in the POF model group was 232.33 ± 17.16 pg/mL and 4.48 ± 0.29 mIU/mL, respectively, after 6 weeks of treatment, which were similar to the values in the wild-type (WT) group. The superovulation demonstrated that ovarian function was significantly improved compared with nontreated POF model mice. The CP-MSC transplantation could restore CTX-induced ovarian dysfunction. Conclusions Our results offer a potential application for human CP-MSCs in POF treatment.
Collapse
Affiliation(s)
- Jun Li
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China.,Sichuan Huahao Biotechnology Co. Ltd., Chengdu, 610041, People's Republic of China
| | - Qingtong Yu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China
| | - Haisen Huang
- Sichuan Huahao Biotechnology Co. Ltd., Chengdu, 610041, People's Republic of China
| | - Wenwen Deng
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China
| | - Xia Cao
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China
| | - Ximing Xu
- Department of Pharmaceutics and Tissue Engineering, School of Pharmacy, Jiangsu University, Zhenjiang, Xuefu Rd, 212013, People's Republic of China.
| |
Collapse
|
30
|
Ling L, Feng X, Wei T, Wang Y, Wang Y, Zhang W, He L, Wang Z, Zeng Q, Xiong Z. Effects of low-intensity pulsed ultrasound (LIPUS)-pretreated human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation on primary ovarian insufficiency in rats. Stem Cell Res Ther 2017; 8:283. [PMID: 29258619 PMCID: PMC5735876 DOI: 10.1186/s13287-017-0739-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/26/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Human amnion-derived mesenchymal stem cells (hAD-MSCs) have the features of mesenchymal stem cells (MSCs). Low-intensity pulsed ultrasound (LIPUS) can promote the expression of various growth factors and anti-inflammatory molecules that are necessary to keep the follicle growing and to reduce granulosa cell (GC) apoptosis in the ovary. This study aims to explore the effects of LIPUS-pretreated hAD-MSC transplantation on chemotherapy-induced primary ovarian insufficiency (POI) in rats. METHODS The animals were divided into control, POI, hAD-MSC treatment, and LIPUS-pretreated hAD-MSC treatment groups. POI rat models were established by intraperitoneal injection of cyclophosphamide (CTX). The hAD-MSCs isolated from the amnion were exposed to LIPUS or sham irradiation for 5 consecutive days and injected into the tail vein of POI rats. Expression and secretion of growth factors promoted by LIPUS in hAD-MSCs were detected by real-time quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) in vitro. Estrous cycle, serum sex hormone levels, follicle counts, ovarian pathological changes, GC apoptosis, Bcl2 and Bax expression, and pro-inflammatory cytokine levels in ovaries were examined. RESULTS Primary hAD-MSCs were successfully isolated from the amnion. LIPUS promoted the expression and secretion of growth factors in hAD-MSCs in vitro. Both hAD-MSC and LIPUS-pretreated hAD-MSC transplantation increased the body and reproductive organ weights, improved ovarian function, and reduced reproductive organ injuries in POI rats. Transplantation of hAD-MSCs increased the Bcl-2/Bax ratio and reduced GC apoptosis and ovarian inflammation induced by chemotherapy in ovaries. These effects could be improved by pretreatment with LIPUS on hAD-MSCs. CONCLUSION Both hAD-MSC transplantation and LIPUS-pretreated hAD-MSC transplantation can repair ovarian injury and improve ovarian function in rats with chemotherapy-induced POI. LIPUS-pretreated hAD-MSC transplantation is more advantageous for reducing inflammation, improving the local microenvironment, and inhibiting GC apoptosis induced by chemotherapy in ovarian tissue of POI rats.
Collapse
Affiliation(s)
- Li Ling
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Xiushan Feng
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Tianqin Wei
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Yan Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400010 China
| | - Yaping Wang
- Department of Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, 400010 China
| | - Wenqian Zhang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Lianli He
- Department of Obstetrics and Gynecology, the Third Affiliated Hospital, Zunyi Medical College, Zunyi, 563000 Guizhou China
| | - Ziling Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Qianru Zeng
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| | - Zhengai Xiong
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Chongqing Medical University, No. 76, Linjiang Road, Chongqing, 400010 China
| |
Collapse
|
31
|
Khanmohammadi N, Sameni HR, Mohammadi M, Pakdel A, Mirmohammadkhani M, Parsaie H, Zarbakhsh S. Effect of Transplantation of Bone Marrow Stromal Cell- Conditioned Medium on Ovarian Function, Morphology and Cell Death in Cyclophosphamide-Treated Rats. CELL JOURNAL 2017; 20:10-18. [PMID: 29308613 PMCID: PMC5759671 DOI: 10.22074/cellj.2018.4919] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/12/2017] [Indexed: 12/13/2022]
Abstract
Objective Although stem cell transplantation has beneficial effects on tissue regeneration, but there are still problems
such as high cost and safety issues. Since stem cell therapy is largely dependent on paracrine activity, in this study,
utilization of transplantation of bone marrow stromal cells (BMSCs)-secretome instead of the cells, into damaged
ovaries was evaluated to overcome the limitations of stem cell transplantation.
Materials and Methods In this experimental study, BMSCs were cultured and 25-fold concentrated conditioned
medium (CM) from BMSCs was prepared. Female rats were injected intraperitoneally with cyclophosphamide (CTX)
for 14 days. Then, BMSCs and CM were individually transplanted into bilateral ovaries, and the ovaries were excised
after four weeks of treatment. The follicle count was performed using hematoxylin and eosin (H&E) staining and the
apoptotic cells were counted using TUNEL assay. Ovarian function was evaluated by monitoring the ability of ovulation
and the levels of serum estradiol (E2) and follicle-stimulating hormone (FSH).
Results Evaluation of the ovarian function and structure showed that results of secretome transplantation were almost
similar to those of BMSCs transplantation and there was no significant differences between them.
Conclusion BMSCs-secretome is likely responsible for the therapeutic paracrine effect of BMSCs. Stem cell-
secretome is expected to overcome the limitations of stem cell transplantation and become the basis of a novel therapy
for ovarian damage.
Collapse
Affiliation(s)
- Nasrin Khanmohammadi
- Research Center of Nervous System Stem Cells, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamid Reza Sameni
- Research Center of Nervous System Stem Cells, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Moslem Mohammadi
- Molecular and Cell Biology Research Center, Department of Physiology and Pharmacology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbas Pakdel
- Research Center of Nervous System Stem Cells, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Mirmohammadkhani
- Research Center for Social Determinants of Health Community Medicine Department, Semnan University of Medical Sciences, Semnan, Iran
| | - Houman Parsaie
- Research Center of Nervous System Stem Cells, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sam Zarbakhsh
- Research Center of Nervous System Stem Cells, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
32
|
Zhang Q, Bu S, Sun J, Xu M, Yao X, He K, Lai D. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res Ther 2017; 8:270. [PMID: 29179771 PMCID: PMC5704397 DOI: 10.1186/s13287-017-0721-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/27/2017] [Accepted: 11/07/2017] [Indexed: 12/27/2022] Open
Abstract
Background Human amniotic epithelial cells (hAECs) are attractive candidates for regenerative medical therapy, with the potential to replace deficient cells and improve functional recovery after injury. Previous studies have demonstrated that transplantation of hAECs effectively alleviate chemotherapy-induced ovarian damage via inhibiting granulose cells apoptosis in animal models of premature ovarian failure/insufficiency (POF/POI). However, the underlying molecular mechanism accounting for hAECs-mediated ovarian function recovery is not fully understood. Methods To investigate whether hAECs-secreting cytokines act as molecular basis to attenuate chemotherapy-induced ovarian injury, hAECs or hAEC-conditioned medium (hAEC-CM) was injected into the unilateral ovary of POF/POI mouse. Follicle development was evaluated by H&E staining at 1, 2 months after hAECs or hAEC-CM treatment. In addition, we performed a cytokine array containing 507 human cytokines on hAECs-derived serum-free conditioned medium. Finally, we further investigated whether hAECs could affect chemotherapy-induced apoptosis in primary human granulosa-lutein (hGL) cells and the tube formation of human umbilical vein endothelial cells (hUVECs) via a co-culture system in vitro. Results We observed the existence of healthy and mature follicles in ovaries treated with hAECs or hAEC-CM, whereas seriously fibrosis and many atretic follicles were found in the contralateral untreated ovaries of the same mouse. To distinguish cytokines involved in the process of hAECs-restored ovarian function, hAEC-CM was analyzed with a human cytokines array. Results revealed that 109 cytokines in hAEC-CM might participate in a variety of biological processes including apoptosis, angiogenesis, cell cycle and immune response. In vitro experiments, hAECs significantly inhibited chemotherapy-induced apoptosis and activated TGF-β/Smad signaling pathway within primary granulosa-lutein cells in paracrine manner. Furthermore, hAEC-CM was shown to promote angiogenesis in the injured ovaries and enhance the tube formation of human umbilical vein endothelial cells (hUVECs) in co-culture system. Conclusions These findings demonstrated that paracrine might be a key pathway in the process of hAECs-mediating ovarian function recovery in animal models of premature ovarian failure/insufficiency (POF/POI). Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0721-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiuwan Zhang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China.,Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, People's Republic of China
| | - Shixia Bu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Junyan Sun
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Minhua Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Xiaofen Yao
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China
| | - Kunyan He
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 145, Guang-Yuan Road, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
33
|
Wang G, Zhao F, Yang D, Wang J, Qiu L, Pang X. Human amniotic epithelial cells regulate osteoblast differentiation through the secretion of TGFβ1 and microRNA-34a-5p. Int J Mol Med 2017; 41:791-799. [PMID: 29207015 PMCID: PMC5752186 DOI: 10.3892/ijmm.2017.3261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/20/2017] [Indexed: 01/08/2023] Open
Abstract
Since the beginning of the use of stem cells in tissue regenerative medicine, there has been a search for optimal sources of stem cells. Human amniotic epithelial cells (hAECs) are derived from human amnions, which are typically discarded as medical waste, but were recently found to include cells with trilineage differentiation potential in vitro. Previous study has focused on the osteogenic differentiation ability of hAECs as seed cells in bone regeneration; however, their paracrine effects on osteoblasts (OBs) are yet to be elucidated. In the present study, conditioned medium (CM) derived from hAECs was used to determine their paracrine effects on the human fetal OB cell line (hFOB1.19), and the potential bioactive factors involved in this process were investigated. The results suggested that hAEC-CM markedly promoted the proliferation, migration and osteogenic differentiation of hFOB1.19 cells. Expression of transforming growth factor β1 (TGFβ1) and microRNA 34a-5p (miR-34a-5p) were detected in hAECs. Furthermore, it was demonstrated that TGFβ1 and miR-34a-5p stimulated the differentiation of hFOB1.19 cells, and that TGFβ1 promoted cell migration. Moreover, the effects of hAEC-CM were downregulated following the depletion of either TGFβ1 or miR-34a-5p. These results demonstrated that hAECs promote OB differentiation through the secretion of TGFβ1 and miR-34a-5p, and that hAECs may be an optimal cell source in bone regenerative medicine.
Collapse
Affiliation(s)
- Guiling Wang
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning 110013, P.R. China
| | - Di Yang
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Jing Wang
- Department of Anal and Intestinal Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lihong Qiu
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, Liaoning 110002, P.R. China
| | - Xining Pang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang, Liaoning 110013, P.R. China
| |
Collapse
|
34
|
Bu S, Zhang Q, Wang Q, Lai D. Human amniotic epithelial cells inhibit growth of epithelial ovarian cancer cells via TGF‑β1-mediated cell cycle arrest. Int J Oncol 2017; 51:1405-1414. [PMID: 29048644 PMCID: PMC5642391 DOI: 10.3892/ijo.2017.4123] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/24/2017] [Indexed: 12/20/2022] Open
Abstract
It is reported that human amniotic epithelial cells (hAECs) endow intrinsic antitumor effects on certain kinds of cancer. This research was designed to evaluate whether hAECs endowed potential anticancer properties on epithelial ovarian cancer (EOC) cells in vivo and in vitro, which has not been reported before. In this study, we established a xenografted BALB/c nude mouse model by subcutaneously co-injecting ovarian cancer cell line, SK-OV-3, and hAECs for 28 days. In ex vivo experiments, CCK‑8 cell viability assay, real-time PCR, cell counting assay, cell cycle analysis and immunohistochemistry (IHC) assay were used to detect the effects of hAEC‑secreted factors on the proliferation and cell cycle progression of EOC cells. A cytokine array was conducted to detect anticancer-related cytokines released from hAECs. Human recombinant TGF‑β1 and TGF‑β1 antibody were used to treat EOC cells and analyzed whether TGF‑β1 contributed to the cell cycle arrest. Results from in vivo and ex vivo experiments showed that hAEC-secreted factors and rhTGF‑β1 decreased proliferation of EOC cells and induced G0/G1 cell cycle arrest in cancer cells, which could be partially reversed by excess TGF‑β1 antibody. These data indicate that hAECs endow potential anticancer properties on epithelial ovarian cancer in vivo and in vitro which is partially mediated by hAEC‑secreted TGF‑β1-induced cell cycle arrest. This study suggests a potential application of hAEC‑based therapy against epithelial ovarian cancer.
Collapse
Affiliation(s)
- Shixia Bu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Qian Wang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| | - Dongmei Lai
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200030, P.R. China
| |
Collapse
|
35
|
Sun L, Li D, Song K, Wei J, Yao S, Li Z, Su X, Ju X, Chao L, Deng X, Kong B, Li L. Exosomes derived from human umbilical cord mesenchymal stem cells protect against cisplatin-induced ovarian granulosa cell stress and apoptosis in vitro. Sci Rep 2017; 7:2552. [PMID: 28566720 PMCID: PMC5451424 DOI: 10.1038/s41598-017-02786-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/20/2017] [Indexed: 01/04/2023] Open
Abstract
Human umbilical cord mesenchymal stem cells (huMSCs) can treat primary ovarian insufficiency (POI) related to ovarian granulosa cell (OGC) apoptosis caused by cisplatin chemotherapy. Exosomes are a class of membranous vesicles with diameters of 30–200 nm that are constitutively released by eukaryotic cells. Exosomes mediate local cell-to-cell communication by transferring microRNAs and proteins. In the present study, we demonstrated the effects of exosomes derived from huMSCs (huMSC-EXOs) on a cisplatin-induced OGC model in vitro and discussed the preliminary mechanisms involved in these effects. We successfully extracted huMSC-EXOs from huMSC culture supernatant and observed the effective uptake of exosomes by cells with fluorescent staining. Using flow cytometry (with annexin-V/PI labelling), we found that huMSC-EXOs increased the number of living cells. Western blotting showed that the expression of Bcl-2 and caspase-3 were upregulated, whilst the expression of Bax, cleaved caspase-3 and cleaved PARP were downregulated to protect OGCs. These results suggest that huMSC-EXOs can be used to prevent and treat chemotherapy-induced OGC apoptosis in vitro. Therefore, this work provides insight and further evidence of stem cell function and indicates that huMSC-EXOs protect OGCs from cisplatin-induced injury in vitro.
Collapse
Affiliation(s)
- Liping Sun
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Kun Song
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China.
| | - Jianlu Wei
- Department of Orthopedics, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Shu Yao
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Zhao Li
- Department of obstetrics and gynecology, Qianfoshan Hospital, Shandong University, 16766 Jingshi Road, Jinan, 250014, Shandong Province, P.R. China
| | - Xuantao Su
- Institute of Biomedical Engineering, School of Control Science and Engineering, Shandong University, 17923 Jingshi Road, Jinan, Shandong, 250061, P.R. China
| | - Xiuli Ju
- Cryomedicine Laboratory, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Lan Chao
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China.,Reproduction Medicine Center, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Xiaohui Deng
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China.,Reproduction Medicine Center, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Beihua Kong
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China
| | - Li Li
- Department of obstetrics and gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China. .,Reproduction Medicine Center, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong Province, P.R. China.
| |
Collapse
|