RESUMO
OBJECTIVES: To observe the effect of penetration needling from "Zhibian" (BL54) to "Shuidao"(ST28) on silencing information regulator 1 (SIRT1) /peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) /nuclear factor E2 related factor 2 (Nrf2) signaling in rats with premature ovarian insufficiency (POI), so as to explore its mechanisms underlying improvement of POI. METHODS: A total of 48 female SD rats were equally and randomly allocated to blank control, POI model, shallow needling and penetration needling (from "Zhibian" ï¼»BL54ï¼½ to "Shuidao" ï¼»ST28ï¼½) groups. The POI model was established by intraperitoneal injection of cyclophosphamide (50 mg·kg-1·d-1 on the 1st day and 8 mg·kg-1·d-1 from the 2nd to 15th day, for a total of 15 days). After successful modeling, for rats of the shallow needling group, a filiform needle was inserted into BL54 to a depth about 5-8 mm, and then retained for 30 min. And for rats of the penetration needling group, a filiform needle was inserted into BL54 area and advanced to the unilateral ST28 to a depth about 12-15 mm, and then retained for 30 min (bilateral acupoints were used at the same time). The treatments were conducted once daily, 6 times a week for 4 weeks. After the interventions, the contents of serum follicle stimulating hormone (FSH), luteinizing hormone (LH), estradiol (E2) and anti-Müllerian hormone (AMH) were detected using ELISA, and the activity of superoxide dismutase (SOD), catalase (CAT) and content of malondialdehyde (MDA) in the ovarian tissue were detected using colorimetry. Histopathological changes of the ovarian tissue were observed after H.E. staining. The immunoactivities and expression levels of SIRT1, PGC-1α, and Nrf2 mRNA and protein in the ovarian tissues were detected using immunohistochemistry, quantitative real-time PCR and Western blot, respectively. RESULTS: After modeling, the rats' estrus cycles were disordered, contents of serum FSH and LH levels significantly increased, and the E2 level markedly decreased compared with those of the blank control group (P<0.01), indicating that the POI model was successfully established. Relevant to the blank control group, the model group had an increase in serum FSH and LH, ovarian MDA contents, and the number of atretic oocytes (P<0.01), and a decrease in serum E2 and AMH contents, ovarian SOD and CAT activities, number of growing oocytes, immunoactivities and expressions of ovarian SIRT1, PGC-1α and Nrf2 protein and mRNA (P<0.01, P<0.05). Following interventions, both the increased levels of serum FSH and LH and ovarian MDA contents, and the number of atretic oocytes, and the decreased levels of E2 and AMH contents, ovarian SOD and CAT activities, number of growing oocytes, immunoactivities and expressions of ovarian SIRT1, PGC-1α and Nrf2 protein and mRNA were reversed by penetration needling of BL54-ST28 (P<0.01, P<0.05), but not by shallow needling, except serum FSH, LH, E2 and AMH contents. The effects of penetration needling were obviously superior to those of shallow needling in up-regulating the levels of serum AMH, ovarian SOD and CAT, number of growing oocytes, and the expressions of ovarian SIRT1, PGC-1α and Nrf2 protein and mRNA (P<0.05, P<0.01), and in down-regulating the level of MDA and the number of atretic oocytes (P<0.05). CONCLUSIONS: Penetration needling stimulation of BL54 to ST28 can increase the number of ovarian growing oocytes and reduce the number of atretic oocytes, regulate the serum hormone levels and relieve the ovarian oxidative stress level in POI rats, which may be associated with its functions in activating ovarian SIRT1/PGC-1α/Nrf2 signaling pathway.
Assuntos
Terapia por Acupuntura , Fator 2 Relacionado a NF-E2 , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Insuficiência Ovariana Primária , Ratos Sprague-Dawley , Transdução de Sinais , Sirtuína 1 , Animais , Feminino , Ratos , Sirtuína 1/metabolismo , Sirtuína 1/genética , Terapia por Acupuntura/instrumentação , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Humanos , Pontos de AcupunturaRESUMO
BACKGROUND: Restoring the function of the ovary is important for chemotherapy-induced ovarian failure (COF) patients. Stem cell and extracellular vesicles (EVs) therapy show promise but need further improvement. METHODS: Human umbilical cord mesenchymal stem cells (hUC-MSCs) were primarily cultured and further three-dimensional (3D) cultured using an ultra-low attachment surface method. The expression levels of nutritional cytokines and immunomodulatory and stemness-related genes of 3D-cultured hUC-MSCs were analyzed. EVs were isolated by ultracentrifugation and characterized. Ovaries were decellularized with sodium dodecyl sulfate to obtain extracellular matrix (ECM). Lyophilized EVs from three-dimensional (2D) or 3D hUC-MSCs were mixed with ECM to prepare the 2D/3D-MSC-EVs-ECM gels. The therapeutic effect of the MSC-EVs-ECM gel on cyclophosphamide (CTX) -treated rats was analyzed through various tests. RNA sequencing was used to analyze the expression changes of genes before and after treatment. RESULTS: After culturing in ultra-low attachment dishes, hUC-MSCs aggregated into spheroids and significantly upregulated the expression levels of immunomodulatory and stemness-related genes. The total EVs yield was also upregulated (5.6-fold) after 3D culture. The cell viability of CTX-treated ovarian granulosa cells (OGCs) was significantly rescued by coculture with the 3D-MSC-EVs-ECM gel. Hormones indicative of ovarian function, AMH, E2, and FSH, were recovered in both the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group, while the apoptosis-related protein Bax was significantly downregulated. The 3D-MSC-EVs-ECM gel was more effective than the 2D-MSC-EVs-ECM gel. Significantly differentially expressed genes, such as Hbb-b1, Gpd1, and Sirpa, were detected by RNA sequencing. Hbb-b1 was increased in the ovaries of CTX-treated rats, and this increase was attenuated by injecting the 2D/3D-MSC-EVs-ECM gel. Gpd1 was increased after CTX treatment, and this increase was reversed by the 3D-MSC-EVs-ECM gel. Sirpa was decreased in the ovaries of CTX-treated rats, and this decrease was attenuated by injecting the 3D-MSC-EVs-ECM gel. CONCLUSIONS: Our study demonstrated that the 3D-MSC-EVs-ECM gel is an efficient strategy for the recovery of ovarian function in CTX-induced ovarian failure.
Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Ovário , Feminino , Animais , Ratos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Humanos , Ovário/efeitos dos fármacos , Ovário/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais/métodos , Ratos Sprague-Dawley , Ciclofosfamida/farmacologia , Géis , Células Cultivadas , Insuficiência Ovariana Primária/terapia , Técnicas de Cultura de Células em Três Dimensões/métodosRESUMO
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.
Assuntos
Ritmo Circadiano , Ciclofosfamida , Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Insuficiência Ovariana Primária , Feminino , Animais , Ciclofosfamida/efeitos adversos , Camundongos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/metabolismo , Células-Tronco Mesenquimais/metabolismo , Ritmo Circadiano/efeitos dos fármacos , Humanos , Gravidez , Secretoma/metabolismo , Placenta/metabolismo , Placenta/efeitos dos fármacos , Estrogênios/farmacologia , Estrogênios/metabolismo , Ovário/metabolismo , Ovário/efeitos dos fármacosRESUMO
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.
Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Insuficiência Ovariana Primária , Animais , Feminino , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/metabolismo , Folículo Ovariano , Insuficiência Ovariana Primária/terapia , Modelos Animais de DoençasRESUMO
Premature ovarian failure (POF) is a leading cause of infertility in women, causing significant psychological and physical distress. Current therapeutic options are limited, necessitating the exploration of new treatments. Mesenchymal stem cells (MSCs), known for their remarkable homing and regenerative properties, have emerged as a promising intervention for POF. However, their clinical efficacy has been inconsistent. This paper aims to address these challenges by examining the cellular heterogeneity within MSC populations, which is crucial for identifying and selecting specific functional subpopulations for clinical applications. Understanding this heterogeneity can enhance therapeutic efficacy and ensure treatment stability. Additionally, this review comprehensively examines the literature on the effectiveness, safety, and ethical considerations of MSCs for ovarian regeneration, with a focus on preclinical and clinical trials. We also discuss potential strategies involving genetically and tissue-engineered MSCs. By integrating insights from these studies, we propose new directions for the design of targeted MSC treatments for POF and related disorders, potentially improving outcomes, addressing safety concerns, and expanding therapeutic options while ensuring ethical compliance.
Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Insuficiência Ovariana Primária , Insuficiência Ovariana Primária/terapia , Humanos , Feminino , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Engenharia Tecidual/métodos , Ensaios Clínicos como AssuntoRESUMO
A discrete mathematical model based on ordinary differential equations and the associated continuous model formed by a partial differential equation, which simulate the generational and temporal evolution of a stem cell population, are proposed. The model parameters are the maximum proliferation potential and the rates of mitosis, death events and telomerase activity. The mean proliferation potential at each point in time is suggested as an indicator of population aging. The model is applied on hematopoietic stem cells (HSCs), with different telomerase activity rates, in a range of variation of maximum proliferation potential in healthy individuals, to study the temporal evolution of aging. HSCs express telomerase, however not at levels that are sufficient for maintaining constant telomere length with aging [1,2]. Women with primary ovarian insufficiency (POI) are known to have low telomerase activity in granulosa cells and peripheral blood mononuclear cells [3]. Extrapolating this to hematopoietic stem cells, the mathematical model shows the differences in proliferation potential of the cell populations when telomerase expression is activated using sexual steroids, though the endogenous promoter or with gene therapy using exogenous, stronger promoters within the adeno-associated virus. In the first case, proliferation potential of cells from POI condition increases, but when adeno-associated viruses are used, the proliferation potential reaches the levels of healthy cell populations.
Assuntos
Proliferação de Células , Insuficiência Ovariana Primária , Telomerase , Humanos , Feminino , Telomerase/metabolismo , Telomerase/genética , Insuficiência Ovariana Primária/terapia , Células-Tronco Hematopoéticas/citologia , Modelos Biológicos , Simulação por Computador , Terapia Genética/métodosRESUMO
PURPOSE: Is it safe and effective to perform controlled ovarian stimulation (COS) and oocyte retrieval (OR) in prepubertal and peripubertal patients? METHODS: In this retrospective cohort study, data of 20 pre-/peripubertal patients who underwent COS and OR for the purpose of oocyte cryopreservation (OC) between 2008 and 2023 were reviewed. Following COS, all OR procedures were performed transabdominally using a vaginal ultrasound probe. Ovarian reserve was assessed by serum FSH, LH, estradiol, AMH, and antral follicle counts (AFC) in all subjects. All mature oocytes were vitrified. RESULTS: Mean age of the patients was 15.05 ± 1.87, mean AMH was 0.84 ± 0.8 ng/ml, mean FSH was 6.39 ± 3.95 IU/L, mean estradiol was 61.6 ± 51.9 pg/ml, mean LH was 4.69 ± 3.46 IU/L, and mean AFC was 5.5 ± 5.82. Among the patients, 12 had regular menstrual cycle, 5 had irregular menstrual cycle, whereas 3 patients still did not have their menarche yet. The indications for OC were as follows: primary ovarian insufficiency (n = 7), ovarian surgery for ovarian tumors (n = 5) or ovarian torsion (n = 1), mosaic Turner syndrome (n = 2), acute lymphoblastic leukemia (n = 1) anaplastic B-cell lymphoma (n = 1), Ewing's sarcoma (n = 1), Noonan syndrome (n = 1), and Thalassemia (n = 1). The mean number of oocytes retrieved, MII oocytes frozen, and maturation rate were 5.11 ± 5.0, 3.92 ± 4.48, and 75.1 ± 25.6%, respectively. Stepwise linear regression analysis demonstrated a positive correlation between AFC and number of total oocytes retrieved and number of MII oocytes. In the case diagnosed with Noonan syndrome, all 7 retrieved oocytes were MI and all frozen at MI phase. No patient had any complication related to COS or OR. CONCLUSION: Even though number of the enrolled subjects is limited and mean AMH is lower in our cohort, we demonstrated that performing COS and OR is safe in pre-/peripubertal patients. If required, transabdominal route can be performed in this age group for OR. AFC appears as a prognostic factor for stimulation outcome in this age group. Pediatric patients or young adolescents at risk for primary ovarian insufficiency should not be discouraged from utilizing OC.
Assuntos
Criopreservação , Recuperação de Oócitos , Oócitos , Indução da Ovulação , Humanos , Feminino , Indução da Ovulação/métodos , Recuperação de Oócitos/métodos , Criopreservação/métodos , Adolescente , Oócitos/crescimento & desenvolvimento , Estudos Retrospectivos , Reserva Ovariana/fisiologia , Preservação da Fertilidade/métodos , Gravidez , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/terapia , Folículo Ovariano/crescimento & desenvolvimento , Hormônio Foliculoestimulante/sangue , Criança , Estradiol/sangue , Puberdade , Fertilização in vitro/métodos , Taxa de Gravidez , Hormônio Antimülleriano/sangue , Hormônio Luteinizante/sangueRESUMO
Premature ovarian failure (POF), a condition influenced by genetic and immune factors, remains incurable despite years of intensive research and significant efforts. This persisting challenge underscores the urgency to address this escalating health concern. Fortunately, stem cell regenerative medicine has emerged as a promising avenue for developing therapeutic strategies and innovative treatments for POF. Bibliometric analysis, renowned for its objectivity, systematic approach, and comprehensive coverage of a given field, has yet to be applied to the study of stem cell research in POF. This study used CiteSpace software to assess contributions and co-occurrence relationships among various countries/regions, institutes, journals, and authors. This approach also allowed us to identify research hotspots and promising future trends within this field. Additionally, we generated visualizing maps utilizing the Web of Science Core Collection (WOSCC) and PubMed publications. By providing valuable information and references, we aim to enhance the understanding of the challenges involved in translating stem cell regeneration into clinical therapeutic potential for POF. Furthermore, our analysis and findings guide researchers and clinicians, facilitating future collaborative research and clinical intervention efforts.
Assuntos
Bibliometria , Insuficiência Ovariana Primária , Insuficiência Ovariana Primária/terapia , Feminino , Humanos , Células-Tronco , Pesquisa com Células-Tronco , Transplante de Células-Tronco/métodos , Medicina Regenerativa/métodosRESUMO
Intraovarian injection of platelet-rich plasma (PRP) has been recently proposed, with encouraging results to provide an alternative option to patients diagnosed with POR or POI. However, the broad spectrum of PRP effects on the reproductive function and the mechanisms of action in follicular activation, response to stimulation, and embryo quality have not yet been studied. In this study, we first induced poor ovarian reserve (POR) and premature ovarian insufficiency (POI) ovarian phenotypes in CD1 mice undergoing PRP or sham intraovarian injection. PRP administration reduced those alterations induced by chemotherapy in ovarian stroma and follicle morphology in both the POR and POI conditions. After ovarian stimulation, we found that PRP did not modify the MII-oocyte yield. Nevertheless, the amount of obtained 2-cell embryos and fertilization rate were increased, being especially relevant for the POI model. Further in vitro embryo culture led to improved blastocyst formation rates and higher numbers of good quality blastocysts in PRP vs. sham females in both the POR and POI conditions. These positive results of PRP injection were also validated in the C57Bl/6 stain. Altogether, our findings suggest a possible effect on oocyte and embryo quality. This effect is likely due to the increase of local paracrine signaling through the released growth factors in PRP-treated ovaries.
Assuntos
Desenvolvimento Embrionário , Oócitos , Reserva Ovariana , Plasma Rico em Plaquetas , Insuficiência Ovariana Primária , Animais , Feminino , Oócitos/efeitos dos fármacos , Reserva Ovariana/efeitos dos fármacos , Camundongos , Desenvolvimento Embrionário/efeitos dos fármacos , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/terapia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversosRESUMO
In-vitro maturation (IVM) is the process of cultivating early-stage follicles from the primordial to the antral stage and facilitating the maturation of oocytes outside the body within a supportive environment. This intricate procedure requires the careful coordination of various factors to replicate the natural ovarian conditions. Advanced techniques for IVM are designed to mimic the natural ovarian environment and enhance the development of follicles. Three-dimensional (3D) culture systems provide a more biologically relevant setting for follicle growth compared to traditional two-dimensional (2D) cultures. Traditional culture systems, often fail to support the complex process of follicle development effectively. However, modern engineered reproductive tissues and culture systems are making it possible to create increasingly physiological in-vitro models of folliculogenesis. These innovative methods are enabling researchers and clinicians to better replicate the dynamic and supportive environment of the ovary, thereby improving the outcomes of IVM offering new hope for fertility preservation and treatment. This paper focuses on the routine 3D culture, and innovative 3D culture of ovary and follicles, including a tissue engineering scaffolds, microfluidic (dynamic) culture system, organ-on-chip models, EVATAR system, from a clinical perspective to determine the most effective approach for achieving in-vitro maturation of follicles. These techniques provide critical support for ovarian function in various ovarian-associated disorders, including primary ovarian insufficiency (POI), premature ovarian failure (POF), ovarian cancer, and age-related infertility.
Assuntos
Preservação da Fertilidade , Folículo Ovariano , Engenharia Tecidual , Feminino , Humanos , Engenharia Tecidual/métodos , Preservação da Fertilidade/métodos , Ovário/fisiologia , Animais , Oócitos/fisiologia , Alicerces Teciduais , Técnicas de Cultura de Células em Três Dimensões/métodos , Insuficiência Ovariana Primária/terapiaRESUMO
<b>Background and Objective:</b> Platelet-Rich Plasma (PRP) is proposed to have a potential regenerative effect on the ovaries following premature ovarian insufficiency (POI), however, the main impact on the process of follicle development (folliculogenesis) remains unclear. Protein-protein interaction (PPI) or network interaction analysis enables us to predict the targets of PRP's biomolecules using biological data through public databases and platforms. Therefore, this method is more efficient in time and cost than traditional laboratory procedures. The purpose of this study was to predict the targets of PRP's biomolecules on folliculogenesis following POI using PPI analysis and to clarify if those predictive targets integrate into PI3K/Akt signaling pathway which is an important pathway in folliculogenesis. <b>Materials and Methods:</b> Mining data targets of POI, PRP and folliculogenesis was done by GeneCards. Only genes with "protein-coding" category were analyzed further. Network analysis was performed using Cytoscape and STRING. Finally, STRING, Enricher and ShinyGO platforms were conducted to analyze gene ontology, including biological processes, molecular function and cellular components, as well as pathways. <b>Results:</b> Network analysis with Cytoscape and STRING discovered 107 gene hubs for POI, PRP and folliculogenesis. Analysis of KEGG pathway using STRING, Enricher and ShinyGO identified 43 genes integrated into PI3K/Akt signaling pathway. From the KEGG pathway, PI3K and Akt were revealed as two main targets following PRP treatment for POI patients. <b>Conclusion:</b> Biomolecules in PRP may recover ovarian follicle development following POI through the PI3K/Akt signaling pathway.
Assuntos
Folículo Ovariano , Plasma Rico em Plaquetas , Insuficiência Ovariana Primária , Transdução de Sinais , Feminino , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/metabolismo , Humanos , Plasma Rico em Plaquetas/metabolismo , Folículo Ovariano/metabolismo , Folículo Ovariano/crescimento & desenvolvimento , Mapas de Interação de Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismoRESUMO
BACKGROUND: Emerging evidence has highlighted the therapeutic potential of human umbilical cord mesenchymal stem cells (UC-MSCs) in chemotherapy-induced premature ovarian failure (POF). This study was designed to investigate the appropriate timing and molecular mechanism of UC-MSCs treatment for chemotherapy-induced POF. METHODS: Ovarian structure and function of mice were assessed every 3 days after injections with cyclophosphamide (CTX) and busulfan (BUS). UC-MSCs and UC-MSCs-derived extracellular vesicles (EVs) were infused into mice via the tail vein, respectively. Ovarian function was analyzed by follicle counts, the serum levels of hormones and ovarian morphology. The apoptosis and proliferation of ovarian granulosa cells were analyzed in vitro and in vivo. Label-free quantitative proteomics was used to detect the differentially expressed proteins in UC-MSC-derived EVs. RESULTS: After CTX/BUS injection, we observed that the ovarian function of POF mice was significantly deteriorated on day 9 after CTX/BUS infusion. TUNEL assay indicated that the number of apoptotic cells in the ovaries of POF mice was significantly higher than that in normal mice on day 3 after CTX/BUS injection. Transplantation of UC-MSCs on day 6 after CTX/BUS injection significantly improved ovarian function, enhanced proliferation and inhibited apoptosis of ovarian granulosa cells, whereas the therapeutic effect of UC-MSCs transplantation decreased on day 9, or day 12 after CTX/BUS injection. Moreover, EVs derived from UC-MSCs exerted similar therapeutic effects on POF. UC-MSCs-derived EVs could activate the PI3K/AKT signaling pathway and reduce ovarian granulosa cell apoptosis. Quantitative proteomics analysis revealed that clusterin (CLU) was highly expressed in the EVs of UC-MSCs. The supplementation of CLU proteins prevented ovarian granulosa cells from chemotherapy-induced apoptosis. Further mechanistic analysis showed that CLU-knockdown blocked the PI3K/AKT signaling and reversed the protective effects of UC-MSCs-derived EVs. CONCLUSIONS: Administration of UC-MSCs and UC-MSCs-derived EVs on day 6 of CTX/BUS injection could effectively improve the ovarian function of POF mice. UC-MSCs-derived EVs carrying CLU promoted proliferation and inhibited apoptosis of ovarian granulosa cells through activating the PI3K/AKT pathway. This study identifies a previously unrecognized molecular mechanism of UC-MSCs-mediated protective effects on POF, which pave the way for the use of cell-free therapeutic approach for POF.
Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Fosfatidilinositol 3-Quinases , Insuficiência Ovariana Primária , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Cordão Umbilical , Feminino , Animais , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/induzido quimicamente , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Cordão Umbilical/citologia , Clusterina/metabolismo , Apoptose , Transplante de Células-Tronco Mesenquimais/métodos , Ovário/metabolismo , Células da Granulosa/metabolismo , Proliferação de Células , Bussulfano/farmacologiaRESUMO
Gonadotoxicity resulting from systemic and locoregional cancer treatments significantly threatens women's reproductive health, often culminating in premature ovarian insufficiency. These therapies, particularly alkylating agents and ionizing radiation, induce DNA damage and apoptosis in ovarian follicles, leading to infertility, amenorrhea, and estrogen deficiency, which exacerbate risks of osteoporosis and cardiovascular diseases. Existing fertility preservation methods do not prevent immediate ovarian damage, underscoring the need for innovative protective strategies. Menstrual blood-derived stem cells (MenSC) and their extracellular vesicles (EV) present promising regenerative potential due to their therapeutic cargo delivery and pathway modulation capabilities. Preclinical studies demonstrate that MenSC-derived EV ameliorate premature ovarian insufficiency by inhibiting granulosa cell apoptosis, promoting angiogenesis, and activating pivotal pathways such as SMAD3/AKT/MDM2/P53. However, comprehensive research is imperative to ensure the safety, efficacy, and long-term effects of MenSC-derived EV in clinical practice. In this review, we update the current knowledge and research regarding the use of MenSC-derived EV as a novel therapeutic weapon for ovarian regeneration in the context of gonadotoxicity induced by systemic anticancer treatment.
Assuntos
Antineoplásicos , Exossomos , Insuficiência Ovariana Primária , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/etiologia , Humanos , Feminino , Exossomos/metabolismo , Antineoplásicos/efeitos adversos , Animais , Menstruação , Células-Tronco/metabolismo , Neoplasias/terapia , Neoplasias/tratamento farmacológicoRESUMO
Premature ovarian insufficiency (POI), a major cause of female infertility, is defined as follicular atresia and a rapid loss of germ cells in women of reproductive age due to ovarian failure. Recently, findings from several studies have indicated that human umbilical cord mesenchymal stem cells (hUMSCs) can alleviate ovarian dysfunction resulting from POI. However, the mechanisms underlying this effect require further clarification. In this study, a mouse model of POI was established as achieved with an intraperitoneal injection of cyclophosphamide (CTX) into female C57BL/6J mice in vivo. These POI mice received a 1-week intervention of hUMACs. In addition, an in vitro POI model was also included. The cultured supernatants of hUMSCs and glycogen synthase kinase 3 beta (GSK3ß) inhibitor (SB216763) were used to treat theca cells (TCs) exposed to CTX. Hematoxylin and Eosin (H&E) staining and Enzyme-linked immunosorbent assay (ELISA) were used to assess ovarian structure and morphology, as well as endocrine function in these POI mice. Based on results from the ELISA and JC-1 labeling, CTX exerted significant detrimental effects on testosterone levels and the mitochondrial membrane potential in TCs. Subsequently, Western Blot, Immunofluorescence staining (IF), and Quantitative real-time polymerase chain reaction (qRT-PCR) were used to evaluate various indicators of testosterone synthesis function and mitochondrial dynamics in ovaries and TCs of POI mice. In vivo, dysfunctions in ovarian structure and function in the POI mouse model were effectively restored following hUMSCs treatment, and abnormalities in hormone synthesis were significantly reduced. Furthermore, when the stem cell supernatants of hUMSCs were applied to TCs in vitro we found that GSK3ß expression was reduced, the imbalance of mitochondrial dynamics was alleviated, and the ability of mitochondrial testosterone synthesis was increased. Taken together, our results indicate that hUMSCs treatment can restore the imbalance of mitochondrial dynamics and restart testosterone synthesis of TCs by suppressing GSK3ß expression, ultimately alleviating POI damage.
Assuntos
Glicogênio Sintase Quinase 3 beta , Células-Tronco Mesenquimais , Dinâmica Mitocondrial , Insuficiência Ovariana Primária , Células Tecais , Animais , Feminino , Glicogênio Sintase Quinase 3 beta/metabolismo , Camundongos , Células Tecais/metabolismo , Células Tecais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Humanos , Insuficiência Ovariana Primária/metabolismo , Insuficiência Ovariana Primária/terapia , Dinâmica Mitocondrial/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Ovário/metabolismo , Ovário/efeitos dos fármacos , Cordão Umbilical/citologia , Ciclofosfamida/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Testosterona , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Indóis , MaleimidasRESUMO
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.
Assuntos
Infertilidade Feminina , Insuficiência Ovariana Primária , Células-Tronco , Humanos , Feminino , Insuficiência Ovariana Primária/terapia , Infertilidade Feminina/terapia , Ovário , Envelhecimento , Transplante de Células-Tronco/métodos , AnimaisRESUMO
BACKGROUND: Drug-free in vitro activation (IVA) is a new protocol to activate residual dormant follicles for fertility restoration in patients with premature ovarian insufficiency (POI). However, several deficiencies have reduced its clinical efficacy rate. Our previous studies have confirmed that the combination of adipose-derived stem cells (ADSCs) and drug-free IVA can improve the effectiveness of drug-free IVA and restore ovarian function of rats with POI. Increasing evidence has demonstrated that mesenchymal stem cell-derived exosomes have similar therapeutic effects as their source cells. Here, we performed a preclinical study to evaluate the therapeutic effects of ADSCs-derived exosomes (ADSCs-Exos) combined with drug-free IVA in the POI rats and the mechanism in restoring ovarian function. RESULTS: In vivo, the effects of ADSCs-Exos were comparable to those of ADSCs, and the ADSCs-Exos combined with drug-free IVA was better than ADSCs-Exos alone therapy in promoting follicular development. Moreover, transplantation of ADSCs/ADSCs-Exos lead to up-regulation of BCL-2 expression and down-regulation of the expression of Bax and Cleaved Caspase-3, thus reducing the apoptosis of chemotherapy-induced follicle cells, and further promoting the development of the follicles and rescuing ovarian function in POI-damaged ovary. In vitro, ovarian fragmentation could activate follicular growth and development, and in combination with ADSCs-Exos could prevent the loss of follicles, promote follicular proliferation and inhibit apoptosis. CONCLUSIONS: ADSCs-Exos combined drug-free IVA had remarkable therapeutic effects in restoring ovarian function of POI rats, and markedly promoted follicular development and inhibited apoptosis of ovarian cells in vitro. Our study confirmed that the combination therapy might be a promising and effective treatment for POI.
Assuntos
Exossomos , Ovário , Insuficiência Ovariana Primária , Animais , Feminino , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/metabolismo , Exossomos/metabolismo , Exossomos/transplante , Ratos , Ovário/metabolismo , Apoptose , Células-Tronco Mesenquimais/metabolismo , Folículo Ovariano/metabolismo , Tecido Adiposo/citologia , Ratos Sprague-DawleyRESUMO
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.
Assuntos
Âmnio , Células Epiteliais , Insuficiência Ovariana Primária , Humanos , Insuficiência Ovariana Primária/terapia , Feminino , Células Epiteliais/transplante , Âmnio/citologia , Âmnio/transplanteRESUMO
In brief: This study reveals that orthotopic transplantation of 3D hUC-MSC spheroids is more effective than monolayer-cultured hUC-MSCs in improving POF and distinctly reducing oxidative stress through the paracrine effect, thereby preventing apoptosis and autophagy of GCs. Abstract: Premature ovarian failure (POF) is a common reproductive disease in women younger than 40 years old, and studies have demonstrated that the application of human umbilical cord mesenchymal stem cells (hUC-MSCs) is a promising therapy strategy for POF. Given the previously established therapeutic advantages of 3D MSC spheroids, and to evaluate their effectiveness, both 3D hUC-MSC spheroids and monolayer-cultured hUC-MSCs were employed to treat a cyclophosphamide-induced POF rat model through orthotopic transplantation. The effects of these two forms on POF were subsequently assessed by examining apoptosis, autophagy, and oxidative damage in ovarian granulosa cells (GCs). The results indicated that hUC-MSC spheroids exhibited superior treatment effects on resisting autophagy, apoptosis, and oxidative damage in GCs compared to monolayer-cultured hUC-MSCs. To further elucidate the impact of hUC-MSC spheroids in vitro, a H2O2-induced KGN cells model was established and co-cultured with both forms of hUC-MSCs. As expected, the hUC-MSC spheroids also exhibited superior effects in resisting apoptosis and autophagy caused by oxidative damage. Therefore, this study demonstrates that 3D hUC-MSC spheroids have potential advantages in POF therapy; however, the detailed mechanisms need to be further investigated. Furthermore, this study will provide a reference for the clinical treatment strategy of POF.
Assuntos
Apoptose , Autofagia , Modelos Animais de Doenças , Células da Granulosa , Células-Tronco Mesenquimais , Estresse Oxidativo , Insuficiência Ovariana Primária , Esferoides Celulares , Feminino , Animais , Ratos , Células da Granulosa/patologia , Células da Granulosa/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/induzido quimicamente , Humanos , Transplante de Células-Tronco Mesenquimais , Ratos Sprague-Dawley , Cordão Umbilical/citologia , Células CultivadasRESUMO
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.
Assuntos
Âmnio , Células Epiteliais , Células da Granulosa , Sistema de Sinalização das MAP Quinases , Insuficiência Ovariana Primária , Proteínas Proto-Oncogênicas c-akt , Células-Tronco , Feminino , Animais , Insuficiência Ovariana Primária/terapia , Insuficiência Ovariana Primária/patologia , Insuficiência Ovariana Primária/metabolismo , Humanos , Camundongos , Âmnio/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células da Granulosa/metabolismo , Células Epiteliais/metabolismo , Células-Tronco/metabolismo , Células-Tronco/citologia , Modelos Animais de Doenças , Transplante de Células-Tronco , Apoptose , Doenças Autoimunes/terapia , Doenças Autoimunes/patologia , Glicoproteínas da Zona Pelúcida/metabolismo , Glicoproteínas da Zona Pelúcida/genéticaRESUMO
BACKGROUND: The therapeutic potential of exosomes from human umbilical cord mesenchymal stem cells (HUMSCs-Exo) for delivering specific circular RNAs (circRNAs) in treating premature ovarian failure (POF) is not well understood. This study aimed to explore the efficacy of HUMSCs-Exo in delivering hsa_circ_0002021 for POF treatment, focusing on its effects on granulosa cell (GC) senescence and ovarian function. METHODS: Bioinformatic analysis was conducted on circRNA profiles using the GSE97193 dataset from GEO, targeting granulosa cells from varied age groups. To simulate granulosa cell senescence, KGN cells were treated with cyclophosphamide (CTX). HUMSCs were transfected with pcDNA 3.1 vectors to overexpress hsa_circ_0002021, and the HUMSCs-Exo secreted were isolated. These exosomes were characterized by transmission electron microscopy (TEM) and Western blotting to confirm exosomal markers CD9 and CD63. Co-culture of these exosomes with CTX-treated KGN cells was performed to assess ß-galactosidase activity, oxidative stress markers, ROS levels, and apoptosis via flow cytometry. Interaction between hsa_circ_0002021, microRNA-125a-5p (miR-125a-5p), and cyclin-dependent kinase 6 (CDK6) was investigated using dual-luciferase assays and RNA immunoprecipitation (RIP). A POF mouse model was induced with CTX, treated with HUMSCs-Exo, and analyzed histologically and via immunofluorescence staining. Gene expression was quantified using RT-qPCR and Western blot. RESULTS: hsa_circ_0002021 was under expressed in both in vivo and in vitro POF models and was effectively delivered by HUMSCs-Exo to KGN cells, showing a capability to reduce GC senescence. Overexpression of hsa_circ_0002021 in HUMSCs-Exo significantly enhanced these anti-senescence effects. This circRNA acts as a competitive adsorbent of miR-125a-5p, regulating CDK6 expression, which is crucial in modulating cell cycle and apoptosis. Enhanced expression of hsa_circ_0002021 in HUMSCs-Exo ameliorated GC senescence in vitro and improved ovarian function in POF models by modulating oxidative stress and cellular senescence markers. CONCLUSION: This study confirms that hsa_circ_0002021, when delivered through HUMSCs-Exo, can significantly mitigate GC senescence and restore ovarian function in POF models. These findings provide new insights into the molecular mechanisms of POF and highlight the therapeutic potential of circRNA-enriched exosomes in treating ovarian aging and dysfunction.