1
|
Moradi K, Moghaddami R, Ghaffari-Nasab A, Khordadmehr M, Pagheh AS, Mosajakhah H, Rezaei S, Gharepapagh E, Ahmadi M, Montazeri M, Pazoki H, Ahmadpour E. Toxoplasma gondii modulates immune responses and mitigates type 1 diabetes progression in a streptozotocin-induced rat model. Cell Commun Signal 2025; 23:172. [PMID: 40200271 PMCID: PMC11980074 DOI: 10.1186/s12964-025-02168-1] [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: 01/26/2025] [Accepted: 03/22/2025] [Indexed: 04/10/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune disease characterized by the destruction of insulin-producing β-cells in the pancreas. Emerging evidence suggests that infections, including Toxoplasma gondii (T. gondii), may modulate immune responses and influence disease outcomes. This study aimed to investigate the effects of T. gondii infection on the development of T1DM in a Streptozotocin (STZ)-induced rat model, with an emphasis on immune modulation, cytokine profiles, and organ inflammation. In rats experimentally infected with pathogenic and non-pathogenic Toxoplasma strains, diabetes was induced via STZ injection and compared to a control group. Blood glucose levels and the expression of IL-10, IL-1β, and TNF-α at both gene and protein levels were assessed. Histopathological examinations of the pancreas and kidneys were conducted, alongside small-animal PET scans to evaluate metabolic activity in these organs. The T. gondii-infected diabetic groups showed reduced blood glucose levels, increased IL-10, and decreased TNF-α and IL-1β levels compared to the STZ group. Histopathological and PET imaging analyses revealed improved pancreatic and renal tissues and reduced metabolic activity, indicating improvement effects associated with decreased inflammation and immune modulation. T. gondii infection seems to influence immune responses and slow the progression of T1DM in a rat model. These results suggest a potential therapeutic role for parasitic infections in autoimmune diseases, offering valuable insights into the complex relationship between infections, immune regulation, and metabolic health.
Collapse
Affiliation(s)
- Kimia Moradi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Moghaddami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arshad Ghaffari-Nasab
- School of Nursing and Allied Medical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Monir Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Abdol Sattar Pagheh
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Mosajakhah
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Rezaei
- Department of Nuclear Medicine, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Gharepapagh
- Department of Nuclear Medicine, Medical School, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Ahmadi
- School of Medical Sciences, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Mahbobeh Montazeri
- Toxoplasmosis Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Pazoki
- Faculty of Medicine, Department of Medical Microbiology, Infectious Diseases Research Center, Gonabad University of Medical Science, Gonabad, Iran
| | - Ehsan Ahmadpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Wang C, Feng Y, Chen Y, Lin X, Li X. Proximity extension assay revealed novel inflammatory biomarkers for follicular development and ovarian function: a prospective controlled study combining serum and follicular fluid. Front Endocrinol (Lausanne) 2025; 16:1525392. [PMID: 39996063 PMCID: PMC11847672 DOI: 10.3389/fendo.2025.1525392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
Background Many components in follicular fluid (FF), such as peptide hormones, cytokines, and steroids, undergo dynamic changes during folliculogenesis and have important roles in follicular development. Because systemic inflammation has also been found to contribute to diminished ovarian reserve (DOR) in previous studies, do certain serum/FF inflammatory biomarkers affect both follicular development and ovarian function? Methods Serum samples from the menstruation phase (n=26), serum samples from the ovulation phase (n=26), FF samples of mature oocytes (n=26), and FF samples of immature oocytes (n=10) were collected. Olink proteomic proximity extension assay (PEA) technology was used to compare the differentially expressed proteins (DEPs), and patients were divided into two subgroups-the normal ovarian reserve (NOR) group and the DOR group-for further bioinformatics analysis and verification by enzyme-linked immunosorbent assay (ELISA). Results In total, 16 DEPs were detected between the mature group and the immature group (FF), and 11 DEPs were detected between the ovulation group and the menstruation group (serum). Further subdivision of the ovarian reserve subgroups revealed 22 DEPs in FF and 3 DEPs in serum. Among all four comparisons, only the expression of oncostatin M (OSM) significantly differed. The OSM signaling pathway, the IL-10 anti-inflammatory signaling pathway, and the PI3K-Akt signaling pathway are three notable pathways involved in affecting ovarian reserve capacity according to bioinformatics analysis. In addition, the concentration of estradiol on the hCG day was slightly but positively correlated with OSM (r=0.457, P=0.029). A significantly greater level of OSM (5.41 ± 2.65 vs. 3.94 ± 1.23 pg/mL, P=0.007) was detected in the serum of NOR patients via ELISA verification, and the sensitivity and specificity of ovarian reserve division were 50.00% and 83.33%, respectively. Conclusion This study proposed that immunological changes assessed by PEA technology affect ovarian function in humans and that OSM may serve as a potential inflammatory biomarker for ovarian function in serum, thus revealing alterations in FF.
Collapse
Affiliation(s)
- Chong Wang
- Department of Reproductive Medicine, Hangzhou Women’s Hospital, Hangzhou, China
| | - Ying Feng
- Department of Reproductive Medicine, Hangzhou Women’s Hospital, Hangzhou, China
| | - Yu Chen
- Department of Clinical Laboratory, Hangzhou Women’s Hospital, Hangzhou, China
| | - Xianhua Lin
- Department of Reproductive Medicine, Hangzhou Women’s Hospital, Hangzhou, China
| | - Xiangjuan Li
- Department of Obstetrics and Gynaecology, Hangzhou Women’s Hospital, Hangzhou, China
| |
Collapse
|
3
|
Ahmadian S, Jafari N, Tamadon A, Ghaffarzadeh A, Rahbarghazi R, Mahdipour M. Different storage and freezing protocols for extracellular vesicles: a systematic review. Stem Cell Res Ther 2024; 15:453. [PMID: 39593194 PMCID: PMC11600612 DOI: 10.1186/s13287-024-04005-7] [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: 08/27/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) have been considered promising tools in regenerative medicine. However, the nanoscale properties of EVs make them sensitive to environmental conditions. Optimal storage protocols are crucial for maintaining EV structural, molecular, and functional integrity. This systematic review aimed to gather evidence on the effects of various storage protocols on EV characteristics and integrity. STRATEGY A comprehensive search was conducted for original studies investigating the impacts of storage temperature, freezing techniques, freeze-thaw cycles, and stabilizing strategies on EV concentration, size distribution, morphology, cargo content, and bioactivity. Results from 50 included studies were analyzed. RESULTS Data indicated that rapid freezing procedures and constant subzero temperatures (optimally - 80 °C) resulted in appropriate EV quantity and cargo preservation. Subjecting EVs to multiple freeze-thaw cycles decreased particle concentrations, RNA content, impaired bioactivity, and increased EV size and aggregation. Electron microscopy revealed vesicle enlargement, and fusion, along with membrane deformation after being exposed to substandard storage protocols. The addition of stabilizers like trehalose helped EVs to maintain integrity. Of note, storage in native biofluids offered improved stability over purified EVs in buffers. CONCLUSION Data emphasize the critical need for precise storage protocols for EVs to ensure reproducible research outcomes and clinical applications. Further studies using reliable methods are necessary to create specific guidelines for improving the stability of EVs in various applications.
Collapse
Affiliation(s)
- Shahin Ahmadian
- 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
| | - Negin Jafari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Tamadon
- Department of Research and Development, PerciaVista R&D Co, Shiraz, Iran
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | | | - 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
| | - Mahdi Mahdipour
- 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.
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Firouzabadi SR, Mohammadi I, Ghafourian K, Mofidi SA, Firouzabadi SR, Hashemi SM, Tehrani FR, Jafarabady K. Mesenchymal stem cell-derived extracellular vesicles therapy for primary ovarian insufficiency: a systematic review and meta-analysis of pre-clinical studies. J Ovarian Res 2024; 17:200. [PMID: 39402602 PMCID: PMC11472498 DOI: 10.1186/s13048-024-01513-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/10/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) manifests with hormonal imbalances, menstrual irregularities, follicle loss, and infertility. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are emerging as a promising treatment for POI. This systematic review aims to assess the effects of MSC-EVs on follicle number, hormonal profile, and fertility in POI animal models. METHODS A systematic search of PubMed, Scopus, and Web of Science databases up to December 14th, 2023 was conducted. Two reviewers independently conducted screening, risk of bias assessment, and data extraction. Meta-analysis was performed to analyze treatment versus control outcomes using a random effects model. Publication bias was assessed using Egger's regression test and sensitivity analysis was assessed using the leave-one-out method. Subgroup analyses and meta-regressions were conducted based on EV source, induction model, type of animal, study quality, administration route, administration frequency and route, and dose. RESULTS a total of 29 studies were included. MSC-EVs treatment significantly increased total follicle count (SMD, (95CI), p-value; 3.56, (0.91, 6.21), < 0.001), including primordial (SMD, (95CI), p-value; 2.86, (1.60, 4.12), < 0.001), primary (SMD, (95CI), p-value; 3.17, (2.28, 4.06), < 0.001), mature (SMD, (95CI), p-value; 2.26, (1.02, 3.50), < 0.001), and antral follicles (SMD, (95CI), p-value; 2.44, (1.21, 3.67), < 0.001). Administration frequency and route did not affect this outcome, but EV source affected primordial, primary, secondary and antral follicle count. Additionally, MSC-EVs treatment elevated anti-müllerian hormone (SMD, (95CI); 3.36, (2.14, 4.58)) and estradiol (SMD, (95CI); 3.19, (2.20, 4.17)) levels while reducing follicle stimulating hormone levels (SMD, (95CI); -2.68, (-4.42, -0.94)). Unlike EV source, which had a significant impact on all three hormones, administration frequency, route, and EV dose did not affect this outcome. Moreover, treatment increased offspring number (SMD, (95CI); 3.70, (2.17, 5.23)) and pregnancy odds (OR, (95CI); 10.25, (4.29, 24.46)) compared to controls. Publication bias and a high level of heterogeneity was evident in all analyses, except for the analysis of the pregnancy odds. However, sensitivity analysis indicated that all of the analyses were stable. CONCLUSION MSC-EVs therapy shows promise for POI treatment, potentially facilitating clinical translation. However, Further research is warranted to optimize methodology and assess side effects.
Collapse
Affiliation(s)
| | - Ida Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiana Ghafourian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mofidi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kyana Jafarabady
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
5
|
Le DC, Ngo MHT, Kuo YC, Chen SH, Lin CY, Ling TY, Pham QTT, Au HK, Myung J, Huang YH. Secretome from estrogen-responding human placenta-derived mesenchymal stem cells rescues ovarian function and circadian rhythm in mice with cyclophosphamide-induced primary ovarian insufficiency. J Biomed Sci 2024; 31:95. [PMID: 39390588 PMCID: PMC11468397 DOI: 10.1186/s12929-024-01085-8] [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: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is an early decline in ovarian function that leads to ovarian failure. Conventional treatments for POI are inadequate, and treatments based on mesenchymal stem cells (MSCs) have emerged as an option. However, the lack of consideration of the estrogen niche in ovarian tissue significantly reduces the therapeutic efficacy, with an unclear mechanism in the MSCs in POI treatment. Furthermore, the disruption of circadian rhythm associated with POI has not been previously addressed. METHODS Conditioned medium (CM) and estradiol-conditioned medium (E2-CM) were generated from estrogen receptor positive MSCs (ER+pcMSCs). Chemotherapy-induced POI models were established using C57BL/6 mice (in vivo) and KGN cells (in vitro) treated with cyclophosphamide (CTX) or 4-hydroperoxycyclophosphamide (4-OOH-CP). Gene/protein expressions were detected using RT-qPCR, Western blotting, and immunohistochemistry assays. Locomotor activity was monitored for behavioral circadian rhythmicity. Cytokine arrays and miRNA analysis were conducted to analyze potential factors within CM/E2-CM. RESULTS The secretome of ER+pcMSCs (CM and E2-CM) significantly reduced the CTX-induced defects in ovarian folliculogenesis and circadian rhythm. CM/E2-CM also reduced granulosa cell apoptosis and rescued angiogenesis in POI ovarian tissues. E2-CM had a more favorable effect than the CM. Notably, ER+pcMSC secretome restored CTX-induced circadian rhythm defects, including the gene expressions associated with the ovarian circadian clock (e.g., Rora, E4bp4, Rev-erbα, Per2 and Dbp) and locomotor activity. Additionally, the cytokine array analysis revealed a significant increase in cytokines and growth factors associated with immunomodulation and angiogenesis, including angiogenin. Neutralizing the angiogenin in CM/E2-CM significantly reduced its ability to promote HUVEC tube formation in vitro. Exosomal miRNA analysis revealed the miRNAs involved in targeting the genes associated with POI rescue (PTEN and PDCD4), apoptosis (caspase-3, BIM), estrogen synthesis (CYP19A1), ovarian clock regulation (E4BP4, REV-ERBα) and fibrosis (COL1A1). CONCLUSION This study is the first to demonstrate that, in considering the estrogen niche in ovarian tissue, an estrogen-priming ER+pcMSC secretome achieved ovarian regeneration and restored the circadian rhythm in a CTX-induced POI mouse model. The potential factors involved include angiogenin and exosomal miRNAs in the ER+pcMSC secretome. These findings offer insights into potential stem cell therapies for chemotherapy-induced POI and circadian rhythm disruption.
Collapse
Affiliation(s)
- Duy-Cuong Le
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Laboratory, Vinmec International Hospital, Minh Khai Street, Hai Ba Trung, Hanoi, Vietnam
| | - Mai-Huong T Ngo
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Yung-Che Kuo
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Fishery Sciences, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, 10617, Taiwan
| | - Thai-Yen Ling
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Quoc Thao Trang Pham
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Heng-Kien Au
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, 11042, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
| | - Jihwan Myung
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre (BCRC), TMU-Shuang Ho Hospital, New Taipei City, 23561, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11042, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Wuxing Street, Taipei, 11031, Taiwan.
| |
Collapse
|
6
|
Ha YH, Kim JH, Ryu CS, Kim JW, Ko EJ, Lee JY, Shin JE, Kim YR, Ahn EH, Kim NK. Association between TGF-β/BMP signaling pathway polymorphisms and the risk of primary ovarian insufficiency in Korean women. Genes Genomics 2024; 46:1201-1208. [PMID: 39259486 DOI: 10.1007/s13258-024-01564-0] [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: 07/05/2024] [Accepted: 08/24/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Primary ovarian insufficiency (POI) is one of the leading female infertility diseases in which ovarian function stops before the age of 40. Reports that POI is associated with transforming growth factor (TGF)-β/bone morphogenetic protein (BMP) signaling pathway-associated genes (e.g., TGF-β, and BMP15) have been continuous since publication that the TGF-β superfamily acts as important regulators for ovary and placenta function in humans. Mechanistically, the secretion of follicle-stimulating hormone, progesterone, and estrogen is affected by the TGF-β superfamily in granulosa cells, which are involved in the development of theca cells, oocytes, and granulosa cells. OBJECTIVE This study aimed to identify the association between genes related to the TGF-β/BMP signaling pathway and the risk of POI pathogenesis. METHODS Possible associations between six gene polymorphisms and POI susceptibility were examined in 139 patients with POI and 345 control subjects. RESULTS Allele combination of TGFBR1 rs334348 G > A and TGFBR3 rs1805110G > A exhibited association with decreased POI risk (adjusted odds ratio [AOR] = 0.165; 95% confidence interval [CI] 0.032-0.847; P = 0.031). Also, TGFBR1 rs1590 G > T and rs334348 G > A and TGFBR3 rs1805110 G > A allele combination exhibited association with decreased POI risk (OR = 0.553; 95% CI 0.374-0.816; P = 0.003). CONCLUSION This study suggests that polymorphisms in the TGF-β signaling pathway genes can be useful biomarkers for POI diagnosis and treatment.
Collapse
Affiliation(s)
- Yong Hyun Ha
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Chang Soo Ryu
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Ji Won Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul, South Korea
| | - Eun Ju Ko
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jeong Yong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Ji Eun Shin
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Young Ran Kim
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Eun Hee Ahn
- Department of Obstetrics and Gynecology, CHA Bundang Medical Center, CHA University, Seongnam, South Korea.
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea.
| |
Collapse
|
7
|
Elahi N, Astaneh ME, Ai J, Makoolati Z. Histological assessment for investigation of dose-dependent ovarian toxicity of cyclophosphamide in the rat. Heliyon 2024; 10:e36767. [PMID: 39281529 PMCID: PMC11400909 DOI: 10.1016/j.heliyon.2024.e36767] [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: 12/17/2023] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024] Open
Abstract
Background Cyclophosphamide (CPA) have significant effects on ovarian follicles which lead to ovarian toxicity and impair the normal female reproductive function. This study aimed to evaluate the dose-dependent effects of CPA on rat follicle numbers. Methods The experimental groups consisted of rats administered a single intraperitoneal injection of CPA at doses of either 50, 75,150, or 200 mg/kg followed by daily doses of 8 mg/kg for 14 days and control group given no treatment. After the treatment period, the histological evaluation was done. Results Primordial and primary follicles were affected by all doses of CPA, but differential follicle counts revealed that graaf and preantral follicles were most sensitive to CPA, followed by primary and primordial follicles. The greatest reduction in all type of studied follicles caused by CPA doses of 50 mg/kg. Conclusion Differential follicle counts revealed that CPA-induced ovarian toxicity is exhibited in structural feature of the ovary, particularly in destruction of graaf and preantral follicles in a dose-dependent manner so that the highest decrease in all type of studied follicles caused by 50 mg/kg of CPA and is suggested as the best concentration for ovotoxicity induction. These findings give insight into ovarian response to structural disruption of folliculogenesis.
Collapse
Affiliation(s)
- Narges Elahi
- Students Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Ebrahim Astaneh
- Department of Anatomical Sciences, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohreh Makoolati
- Department of Anatomical Sciences, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
8
|
Luo Y, Chen J, Ning J, Sun Y, Chai Y, Xiao F, Huang B, Li G, Tian F, Hao J, Zhang Q, Zhao J, Li Y, Li H. Stem cell-derived extracellular vesicles in premature ovarian failure: an up-to-date meta-analysis of animal studies. J Ovarian Res 2024; 17:182. [PMID: 39252114 PMCID: PMC11382489 DOI: 10.1186/s13048-024-01489-y] [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: 05/14/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND There has been a significant surge in animal studies of stem cell-derived extracellular vesicles (EVs) therapy for the treatment of premature ovarian failure (POF) but its efficacy remains unknown and a comprehensive and up-to-date meta-analysis is lacking. Before clinical translation, it is crucial to thoroughly understand the overall impact of stem cell-derived EVs on POF. METHODS PubMed, EMBASE, Cochrane Library, Web of Science were searched up to February 18, 2024. The risk of bias was evaluated according to Cochrane Handbook criteria, while quality of evidence was assessed using the SYRCLE system. The PRISMA guidance was followed. Trial sequential analysis was conducted to assess outcomes, and sensitivity analysis and publication bias analysis were performed using Stata 14. RESULTS Data from 25 studies involving 339 animals were extracted and analyzed. The analysis revealed significant findings: stem cell-derived EVs increase ovary weight (SMD = 3.88; 95% CI: 2.50 ~ 5.25; P < 0.00001; I2 = 70%), pregnancy rate (RR = 3.88; 95% CI: 1.94 ~ 7.79; P = 0.0001; I2 = 0%), count of births (SMD = 2.17; 95% CI: 1.31 ~ 3.04; P < 0.00001; I2 = 69%) and counts of different types of follicles. In addition, it elevates the level of AMH (SMD = 4.15; 95% CI: 2.75 ~ 5.54; P < 0.00001; I2 = 88%) and E2 (SMD = 2.88; 95% CI: 2.02 ~ 3.73; P < 0.00001; I2 = 80%) expression, while reducing FSH expression (SMD = -5.05; 95% CI: -6.60 ~ -3.50; P < 0.00001; I2 = 90%). Subgroup analysis indicates that the source of EVs, animal species, modeling method, administration route, and test timepoint affected efficacy. Trial sequential analysis showed that there was sufficient evidence to confirm the effects of stem cell-derived EVs on birth counts, ovarian weights, and follicle counts. However, the impact of stem cell-derived EVs on pregnancy rates needs to be further demonstrated through more animal experimental evidence. CONCLUSIONS Stem cell-derived EVs demonstrate safety and efficacy in treating POF animal models, with potential improvements in fertility outcomes. TRIAL REGISTRATION PROSPERO registration number: CRD42024509699.
Collapse
Affiliation(s)
- Yan Luo
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Jingjing Chen
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jinyao Ning
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Sun
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Yitong Chai
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Medicine Eight-Year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Fen Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bixia Huang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Ge Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Fen Tian
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jie Hao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Qiong Zhang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Jing Zhao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Yanping Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China
| | - Hui Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province, 410008, China.
- Clinical Research Center for Women's, Reproductive Health in Hunan Province, Hunan Province, Changsha, 410008, China.
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Yamchi NN, Ahmadian S, Mobarak H, Amjadi F, Beheshti R, Tamadon A, Rahbarghazi R, Mahdipour M. Correction: Amniotic fluid-derived exosomes attenuated fibrotic changes in POI rats through modulation of the TGF-β/Smads signaling pathway. J Ovarian Res 2024; 17:168. [PMID: 39160613 PMCID: PMC11331637 DOI: 10.1186/s13048-024-01488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Affiliation(s)
| | - Shahin Ahmadian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Amjadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahim Beheshti
- Faculty of Veterinary Medicine, Shabestar Islamic Azad University, Shabestar, Iran
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz, Iran
- Department for Scientific Work, Marat Ospanov Medical University, West, Aktobe, Kazakhstan
| | - Reza Rahbarghazi
- Drug Applied 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
| | - 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.
| |
Collapse
|
10
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
11
|
Zhylkybekova A, Koshmaganbetova GK, Rysmakhanov MS, Abenova NA, Mussin NM, Kaliyev AA, Mahdipour M, Tamadon A. Engineering of ovarian tissue for ovarian dysfunctions: A review. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2024; 13:3-11. [DOI: 10.4103/apjr.apjr_81_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/20/2023] [Indexed: 04/30/2025] Open
Abstract
This review explores tissue engineering as a potential solution for reproductive health issues in women caused by genetic or acquired diseases, such as premature ovarian failure or oophorectomy. The loss of ovarian function can lead to infertility, osteoporosis, and cardiovascular disease. Hormone replacement therapy is a common treatment, but it has limitations and risks. The review focuses on two main approaches in tissue engineering: scaffold-based (3D printing, electrospinning, decellularization) and scaffold-free (stem cell transplantation, organoid cultivation). Both approaches show promise in preclinical studies for creating functional ovarian tissue. Challenges include vascularization, innervation, long-term function, and safety. Despite these challenges, tissue engineering offers a potential avenue for restoring fertility and hormone balance in women with ovarian dysfunction.
Collapse
Affiliation(s)
- Aliya Zhylkybekova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Gulbakit K. Koshmaganbetova
- Department of Evidence-Based Medicine and Scientific Management, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Myltykbay S. Rysmakhanov
- Department of Surgery and Urology No.2, West Kazakhstan Medical University, Aktobe, Kazakhstan
- Department of Surgery and Transplantation, Aktobe Medical Center, Aktobe, Kazakhstan
| | | | | | | | - 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
| | - Amin Tamadon
- Department for Scientific Work, West-Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
12
|
Zhao X, Peng X, Wang Z, Zheng X, Wang X, Wang Y, Chen J, Yuan D, Liu Y, Du J. MicroRNAs in Small Extracellular Vesicles from Amniotic Fluid and Maternal Plasma Associated with Fetal Palate Development in Mice. Int J Mol Sci 2023; 24:17173. [PMID: 38139002 PMCID: PMC10743272 DOI: 10.3390/ijms242417173] [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: 10/14/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Cleft palate (CP) is a common congenital birth defect. Cellular and morphological processes change dynamically during palatogenesis, and any disturbance in this process could result in CP. However, the molecular mechanisms steering this fundamental phase remain unclear. One study suggesting a role for miRNAs in palate development via maternal small extracellular vesicles (SEVs) drew our attention to their potential involvement in palatogenesis. In this study, we used an in vitro model to determine how SEVs derived from amniotic fluid (ASVs) and maternal plasma (MSVs) influence the biological behaviors of mouse embryonic palatal mesenchyme (MEPM) cells and medial edge epithelial (MEE) cells; we also compared time-dependent differential expression (DE) miRNAs in ASVs and MSVs with the DE mRNAs in palate tissue from E13.5 to E15.5 to study the dynamic co-regulation of miRNAs and mRNAs during palatogenesis in vivo. Our results demonstrate that some pivotal biological activities, such as MEPM proliferation, migration, osteogenesis, and MEE apoptosis, might be directed, in part, by stage-specific MSVs and ASVs. We further identified interconnected networks and key miRNAs such as miR-744-5p, miR-323-5p, and miR-3102-5p, offering a roadmap for mechanistic investigations and the identification of early CP biomarkers.
Collapse
Affiliation(s)
- Xige Zhao
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xia Peng
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Zhiwei Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xiaoyu Zheng
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Xiaotong Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Yijia Wang
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Jing Chen
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Dong Yuan
- Department of Geriatric Dentistry, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China;
| | - Ying Liu
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
| | - Juan Du
- Laboratory of Orofacial Development, Laboratory of Molecular Signaling and Stem Cells Therapy, Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China; (X.Z.); (X.P.); (Z.W.); (X.Z.); (X.W.); (Y.W.); (J.C.); (Y.L.)
- Department of Geriatric Dentistry, Capital Medical University School of Stomatology, Tiantan Xili No. 4, Beijing 100050, China;
| |
Collapse
|
13
|
Elahi N, Ai J, Makoolati Z. A Review on Treatment of Premature Ovarian Insufficiency: Characteristics, Limitations, and Challenges of Stem Cell versus ExosomeTherapy. Vet Med Int 2023; 2023:5760011. [PMID: 38023426 PMCID: PMC10673665 DOI: 10.1155/2023/5760011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a complex disorder that can result in varying degrees of infertility. Recently, mesenchymal stem cell (MSC) therapy and its derivatives, such as exosomes, have been introduced as novel strategies for the treatment of POI. This review discusses the features, limitations, and challenges of MSC and exosome therapy in the treatment of POI and provides readers with new insights for comparing and selecting chemical agents, optimizing doses, and other factors involved in study design and treatment strategies. MSC therapy has been shown to improve ovarian function in some animals with POI, but it can also have side effects such as high cost, time-consuming processes, limited lifespan and cell sources, loss of original characteristics during in vitro proliferation, dependence on specific culture environments, potential immune reactions, unknown therapeutic mechanisms, etc. However, exosome therapy is a newer therapy that has not been studied as extensively as MSC therapy, but that it has shown some promise in animal studies. The evidence for the effectiveness of MSC and exosome therapy is still limited, and more research is needed to determine whether these therapies are effective and safe for women with POI. This study presents a new perspective for researchers to advance their research in the fields of cell-based and cell-free therapies.
Collapse
Affiliation(s)
- Narges Elahi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zohreh Makoolati
- Department of Anatomical Sciences, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
14
|
Zhang Z, Shi C, Wang Z. The physiological functions and therapeutic potential of exosomes during the development and treatment of polycystic ovary syndrome. Front Physiol 2023; 14:1279469. [PMID: 38028777 PMCID: PMC10657906 DOI: 10.3389/fphys.2023.1279469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Polycystic ovary syndrome is a very common disease of gynecological endocrine, accompanied by irregular menstruation, hyperandrogenism, metabolic abnormalities, reproductive disorders and other clinical symptoms, which seriously endangers women's physical and mental health, but its etiology and pathogenesis are not completely clear. Recently, the contribution of exosomes to the diagnosis and treatment of various diseases in the biomedical field has attracted much attention, including PCOS. Exosomes are extracellular vesicles secreted by cells, containing various biologically active molecules such as cell-specific proteins, lipids, and nucleic acids. They are important signaling regulators in vivo and widely participate in various physiopathological processes. They are new targets for disease diagnosis and treatment. Considering the important role of non-coding RNAs during the development and treatment of PCOS, this article takes exosomal miRNAs as the breakthrough point for elucidating the physiological functions and therapeutic potential of exosomes during the development and treatment of PCOS through analyzing the effects of exosomal miRNAs on ovarian follicle development, hormone secretion, oxidative stress, inflammatory response and insulin resistance, thus providing new research directions and theoretical basis for PCOS pathogenesis, clinical diagnosis and prognosis improvement.
Collapse
Affiliation(s)
| | | | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
15
|
Ahmadpour E, Spotin A, Moghimi A, Shahrivar F, Jadidi-Niaragh F, Hajizadeh F, Mehrani S, Mazhab-Jafari K. Tumor suppressor p73 induces apoptosis of murine peritoneal cell after exposure to hydatid cyst antigens; a possibly survival mechanism of cystic echinococcosis in vivo mice model. PLoS One 2023; 18:e0292434. [PMID: 37796859 PMCID: PMC10553360 DOI: 10.1371/journal.pone.0292434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023] Open
Abstract
Cystic echinococcosis (CE) is a life-threatening helminthic disease caused by the Echinococcus granulosus sensulato complex. Previous evidence indicates that the host's innate immune responses against CE can combat and regulate the growth rate and mortality of hydatid cyst in the host's internal organs. However, the survival mechanisms of CE are not yet fully elucidated in the human body. In the present study, the apoptotic effects of fertile and infertile hydatid fluid (HF) were tested on murine peritoneal cells in vivo mice model. Mice were divided into five groups including; control group, fertile HF-treated peritoneal cells, infertile HF-treated peritoneal cells, protoscolices (PSCs)-treated peritoneal cells and HF+PSCs-treated peritoneal cells group. Mice groups were intraperitoneally inoculated with PBS, HF, and/or PSCs. Afterwards, peritoneal cells were isolated and mRNA expression of STAT3, caspase-3, p73 and Smac genes were evaluated by quantitative Real-time PCR. After 48 hours of exposure, the protein levels of Smac and STAT3 was determined by western blotting technique. After 6 hours of exposure, Caspase-3 activity was also measured by fluorometric assay. The intracellular reactive oxygen species (ROS) production was examined in all groups. The mRNA expression levels of p73, caspase-3 and also Caspase-3 activity in HF+PSCs-treated peritoneal cells were higher than in the test and control groups (Pv<0.05), while the mRNA expression level of anti-apoptotic STAT3 and Smac genes in HF+PSC-treated peritoneal cells were lower than in the other groups (Pv<0.05). As well, the level of intracellular ROS in the fertile HCF-treated peritoneal cells, infertile HCF-treated peritoneal cells, PSC-treated peritoneal cells and HF+PSC-treated peritoneal cells groups were significantly higher than in the control group (Pv<0.05).Current findings indicates that oxidative stress and p73 can trigger the apoptosis of murine peritoneal cells through modulator of HF-treated PSCs that is likely one of the hydatid cyst survival mechanisms in vivo mice model.
Collapse
Affiliation(s)
- Ehsan Ahmadpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Spotin
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Moghimi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Firooz Shahrivar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farnaz Hajizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sirous Mehrani
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Komeil Mazhab-Jafari
- Department of Laboratory Sciences, Abadan University of Medical Sciences, Abadan, Iran
- Department of Parasitology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
16
|
Dai W, Yang H, Xu B, He T, Liu L, Ma X, Ma J, Yang G, Si R, Pei X, Du X, Fu X. Human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) alleviate excessive autophagy of ovarian granular cells through VEGFA/PI3K/AKT/mTOR pathway in premature ovarian failure rat model. J Ovarian Res 2023; 16:198. [PMID: 37777781 PMCID: PMC10542694 DOI: 10.1186/s13048-023-01278-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] [Received: 06/11/2023] [Accepted: 09/12/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Premature ovarian failure (POF) is one of the leading causes of female infertility and is accompanied by abnormal endocrine, seriously affecting female quality of life. Previous studies have demonstrated that mesenchymal stem cells (MSCs) transplantation is a promising therapeutic strategy for POF. However, the mechanism remains obscure. This study aims to investigate the therapeutic effect of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) on ovarian function in the POF rat model and explore the underlying mechanisms. METHODS The ovarian function was evaluated by ovarian morphology, histology, estrous cycle, hormone levels (AMH, E2, FSH, and LH), and fertility ability to investigate the effect of hUC-MSCs on the POF rats model. The cytokines levels were assayed in serum using protein array to explore the mechanisms of hUC-MSCs therapy for POF. The excessive autophagy levels were evaluated using a co-culture system of 3D MSCs spheroids with human ovarian granulosa cell line (KGN) or primary ovarian granulosa cells (GCs) to understand the paracrine effect of hUC-MSCs on GCs. The related proteins expression of autophagy and PI3K/AKT/mTOR pathway was detected using Western Blotting and/or in various inhibitors supplement to further demonstrate that vascular endothelial growth factor A (VEGFA) secreted by hUC-MSCs can alleviate excessive autophagy of ovarian GCs via PI3K/AKT/mTOR signaling pathway. The ovarian culture model in vitro was applied to confirm the mechanism. RESULTS The ovarian function of POF and the excessive autophagy of ovarian GCs were restored after hUC-MSCs transplantation. The protein array result demonstrated that VEGF and PI3K/AKT might improve ovarian function. in vitro experiments demonstrated that VEGFA secreted by hUC-MSCs could decrease oxidative stress and inhibit excessive autophagy of ovarian GCs via PI3K/AKT/mTOR pathway. The ovarian culture model results confirmed this mechanism in vitro. CONCLUSION The hUC-MSCs can alleviate excessive autophagy of ovarian GCs via paracrine VEGFA and regulate the PI3K/AKT/mTOR signaling pathway, thereby improving the ovarian function of POF.
Collapse
Affiliation(s)
- Wenjie Dai
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Hong Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Bo Xu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Tiantian He
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Ling Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaoqian Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiaxue Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Guoqin Yang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Rui Si
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xing Du
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| | - Xufeng Fu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
17
|
Qi J, Zhang X, Zhang S, Wu S, Lu Y, Li S, Li P, Tan J. P65 mediated UBR4 in exosomes derived from menstrual blood stromal cells to reduce endometrial fibrosis by regulating YAP Ubiquitination. J Nanobiotechnology 2023; 21:305. [PMID: 37644565 PMCID: PMC10463480 DOI: 10.1186/s12951-023-02070-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Intrauterine adhesion (IUA) is a recurrent and refractory reproductive dysfunction disorder for which menstrual blood-derived stromal cells (MenSCs) might be a promising intervention. We reported that administration of MenSCs-derived exosomes (MenSCs-EXO) could achieve similar therapeutic effects to MenSCs transplantation, including alleviating endometrial fibrosis and improving fertility in IUA rats. The mass spectrometry sequencing result suggested that UBR4, a member of the proteasome family, was abundantly enriched in MenSCs-EXO. This study aimed to investigate the key role of UBR4 in MenSCs-EXO for the treatment of IUA and the specific molecular mechanism. RESULTS UBR4 was lowly expressed in the endometrial stromal cells (EndoSCs) of IUA patients. MenSCs-EXO treatment could restore the morphology of IUA endometrium, reduce the extent of fibrosis, and promote endometrial and vascular proliferation. Knockdown of UBR4 in MenSCs did not affect the characteristics of exosomes but attenuated the therapeutic effect of exosomes. UBR4 in MenSCs-EXO could alleviate endometrial fibrosis by boosting YAP ubiquitination degradation and promoting YAP nuclear-cytoplasmic translocation. Moreover, P65 could bind to the UBR4 promoter region to transcriptionally promote the expression level of UBR4 in MenSCs. CONCLUSION Our study clarified that MenSCs-EXO ameliorated endometrial fibrosis in IUA primarily by affecting YAP activity mediated through UBR4, while inflammatory signaling P65 may affect UBR4 expression in MenSCs to enhance MenSCs-EXO therapeutic effects. This revealed a novel mechanism for the treatment of IUA with MenSCs-EXO, proposing a potential option for the clinical treatment of endometrial injury.
Collapse
Affiliation(s)
- Jiarui Qi
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Xudong Zhang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Siwen Zhang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Shanshan Wu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Yimeng Lu
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Shuyu Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Pingping Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China
| | - Jichun Tan
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China.
- Key Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling of Liaoning Province, No. 39 Huaxiang Road, Tiexi District, Shenyang, 110022, China.
- Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, Shenyang, China.
| |
Collapse
|