1
|
Yin Q, Zheng J, Cao Y, Yan X, Zhang H. Proteomic analysis of human follicular fluid based on the 4D label free method to identify proteins that may affect oocyte quality in hyperandrogenic PCOS patients. Front Endocrinol (Lausanne) 2025; 16:1579469. [PMID: 40444232 PMCID: PMC12119282 DOI: 10.3389/fendo.2025.1579469] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Objective Proteomic analysis was conducted on human follicular fluid (FF) using the 4D label-free method to identify proteins potentially influencing oocyte quality in hyperandrogenic (HA) polycystic ovary syndrome (PCOS) patients. Methods FF was collected from 3 different groups: HA PCOS patients, non-hyperandrogenic (NHA) PCOS patients, and controls. Protein profiles of FF from HA PCOS patients (n = 10) were constructed utilizing 4D label-free proteomics technology. Differentially expressed proteins were identified by comparing these profiles with those from NHA PCOS (n = 10) and control patients (n = 10). In addition, FF was collected from 34 HA, 33 NHA, and 23 control patients for enzyme-linked immunosorbent assay (ELISA) validation of differentially expressed proteins. Associations between the levels of differentially expressed proteins in FF and various embryonic outcome indicators were evaluated. Results The HA group demonstrated significantly reduced normal cleavage rates, D3 available embryo rates, D3 high-quality embryo rates, available blastocyst rates, and high-quality blastocyst rates compared to the NHA and control groups (HA vs. NHA vs. Control, 88.3 vs. 93.6 vs. 94.23, 70.57 vs. 81.76 vs. 83.77, 42.49 vs. 56.39 vs. 61.83, 55.0 vs. 65.96 vs. 67.26, 27.62 vs. 45.19 vs. 44.75, respectively), with statistically significant differences (P < 0.05). 23 differentially expressed proteins were identified in FF profiles of the HA group relative to the control group, while 9 differentially expressed proteins were noted in comparison with the NHA group. Cross-comparison highlighted three potential target proteins: insulin-like growth factor binding protein 5 (IGFBP5), lysosomal-associated membrane protein 2 (LAMP2), and cadherin-5 (CDH5). Adjusting for age and body mass index (BMI), IGFBP5 levels in FF exhibited negative correlations with normal cleavage rate, D3 high-quality embryo rate, available blastocyst rate, and high-quality blastocyst rate (P ≤ 0.05). Similarly, LAMP2 levels were negatively correlated with normal cleavage rate, D3 available embryo rate, D3 high-quality embryo rate, and high-quality blastocyst rate (P < 0.05). CDH5 levels demonstrated positive correlations with D3 high-quality embryo rate and high-quality blastocyst rate (P < 0.05). Conclusion The proteins IGFBP5, LAMP2, and CDH5 may contribute to the mechanisms underlying the adverse effects of hyperandrogenism on oocyte quality in PCOS patients.
Collapse
Affiliation(s)
- Qianqian Yin
- Center for Reproductive Medicine, XuZhou Central Hospital, Xuzhou, Jiangsu, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jianhua Zheng
- Department of Obstetrics and Gynecology, XuZhou Central Hospital, Xuzhou, Jiangsu, China
| | - Yijuan Cao
- Center for Reproductive Medicine, XuZhou Central Hospital, Xuzhou, Jiangsu, China
| | - Xiaonan Yan
- Center for Reproductive Medicine, XuZhou Central Hospital, Xuzhou, Jiangsu, China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
2
|
Kalimuthu V, Chandran Manimegalai S, Venkatesan R, Krishnamoorthy SP, Dey N, Ramesh T, Balamuthu K. Exploring the Therapeutic Potential of Terminalia chebula Retz. in Alleviating the Complications of Letrozole-Induced PCOS in Rat Model. Reprod Sci 2025; 32:836-853. [PMID: 39939489 DOI: 10.1007/s43032-025-01813-x] [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: 06/28/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
Polycystic Ovarian Syndrome is a leading gynecological condition that is being known to affect women fertility irrespective of their reproductive age. Though its prevalency and adverse effects in causing the female infertility is know to be reported worldwide, it has the steroidal pills having remarkable side effects as their effective medication to treat and manage its symptoms. Inorder to find a novel curative plant besed therapy, this study investigates the therapeutic potential of Terminalia chebula Retz. to manage the complications of PCOS. In this present study, the bioactive compounds of Terminalia chebula Retz. fruit extract were identified by GC-MS and the experimental animals (female Wistar rats) were categorized into six groups including control, letrozole-induced PCOS group, metformine treated as standard control, along with the groups orally treated with T. chebula fruit extracts at various concentrations. As a result of PCOS induction, the level of LPx got increased evidencing the increased lipid metabolism where the other antioxidant levels were decreased. The serum hormonal profile revealed a considerable decrease in estrogen and progesterone levels while the levels of LH, FSH, testosterone, and insulin were increased. The mRNA and protein expressions of CYP17A1, was upregulated whereas the CYP19A1 and PPAR-γ found to have lower expression on concerning the control group. These entire physiological, and biochemical observed during the successful induction of PCOS got restored to normal after being treated with the fruit extract of T. chebula in the experimental animals and implied its potentiality in managing the complications of PCOS.
Collapse
Affiliation(s)
- Vignesh Kalimuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | | | - Ramya Venkatesan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | | | - Nigamananda Dey
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Kadalmani Balamuthu
- Department of Animal Science, Bharathidasan University, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|
3
|
Jin L, Ren C, Yang L, Zhu Y, Li G, Chang Y, Du J, Yang Z, Yuan Y. Efficacy and Safety of Human Umbilical Cord Mesenchymal Stem Cells in Improving Fertility in Polycystic Ovary Syndrome Mice. Curr Stem Cell Res Ther 2025; 20:279-290. [PMID: 40351081 DOI: 10.2174/011574888x287937240424074937] [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: 12/20/2023] [Revised: 02/20/2024] [Accepted: 03/02/2024] [Indexed: 05/14/2025]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most prevalent reproductive endocrine illness in women of reproductive age and is one of the most important causes of female infertility. The pathogenesis of PCOS is complex. Although mesenchymal stem cell therapy is anticipated to be a successful treatment for PCOS, its long-term safety, including tumorigenesis in patients, remains unknown. OBJECTIVE This study aimed to confirm the efficacy and safety of human umbilical cord mesenchymal stem cells in improving fertility in PCOS mice. METHODS In this study, dehydroepiandrosterone (DHEA) was used to construct a C56BL/6 mouse PCOS model, human umbilical cord mesenchymal stem cells (hUC-MSCs) were used as a treatment, and the reproductive phenotype was observed in parallel breeding experiments to confirm the efficacy of the treatment. A 4-month follow-up period, final blood tests, and organ histology were carried out to confirm the long-term safety of the treatment. RESULTS After hUC-MSCs treatment, the sex hormone disorder of mice was corrected, the morphology and function of the ovary were improved, the number of offspring was significantly increased compared to the control group, and no adverse reactions related to stem cell transplantation such as tumor formation were found within 4 months. CONCLUSION The treatment of hUC-MSCs is safe and effective in treating PCOS over the long term.
Collapse
Affiliation(s)
- Lukuo Jin
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Chenchen Ren
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Li Yang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Yuanhang Zhu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Genxia Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yun Chang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Junxiao Du
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Zhaoyuan Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yuchao Yuan
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
4
|
Hu R, Huang Y, Liu Z, Dong H, Ma W, Song K, Xu X, Wu X, Geng Y, Li F, Zhang M, Song Y. Characteristics of polycystic ovary syndrome rat models induced by letrozole, testosterone propionate and high-fat diets. Reprod Biomed Online 2025; 50:104296. [PMID: 39626468 DOI: 10.1016/j.rbmo.2024.104296] [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: 12/05/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 01/29/2025]
Abstract
RESEARCH QUESTION What are the long-term effects of different models of polycystic ovary syndrome (PCOS), and which model could be used in future research? DESIGN PCOS models induced by letrozole, letrozole plus high-fat diet (LE+HFD), testosterone propionate (TP) or testosterone propionate plus HFD (TP+HFD) were established in rats. Body weight, energy intake, blood glucose, sex hormone concentrations, lipid profiles and the oestrus cycle were observed. Histology of ovaries, large intestine and fat was displayed. Protein and mRNA levels relating to hormone synthesis, oocyte maturation, the gut barrier, lipid metabolism and inflammation were evaluated using western blotting, immunohistochemistry and PCR. The composition of the microbial community was measured using 16S RNA sequencing. RESULTS Letrozole treatment induced hyperandrogenaemia, polycystic ovarian morphology, a disrupted oestrus cycle and impaired ovarian function, which could be restored within 42 days. Concurrently, letrozole disturbed glucose, fat, and energy metabolism, affected the inflammatory state and compromised intestinal homeostasis. HFD could amplify the disturbances in the metabolism and intestinal microenvironment, and the pituitary-ovarian axis was more efficiently and consistently affected by testosterone propionate. Testosterone propionate and TP+HFD treatment also disturbed the intestinal microenvironment. Although the metabolic effects of testosterone propionate were not as profound as those of letrozole, they were enhanced by HFD. CONCLUSIONS Letrozole is useful for studies on metabolic disturbances in PCOS, and LE+HFD treatment is suitable for investigations on PCOS metabolic abnormalities and the gut-PCOS link. Testosterone propionate injection is appropriate for studying reproductive abnormalities in PCOS, while TP+HFD treatment is the most comprehensive for studying PCOS reproductive abnormalities, metabolic disturbances and the gut-PCOS link.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohu Xu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Wu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Mallya P, Kalthur G, Sravani AB, Lewis SA. Improving the Dehydroepiandrosterone Induced PCOS Rat Model: Interplay of Age, High Fat Diet, and Treatment Regimen on Reproductive and Metabolic Phenotypes. Reprod Sci 2025; 32:187-199. [PMID: 39567467 PMCID: PMC11729200 DOI: 10.1007/s43032-024-01742-1] [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: 05/06/2024] [Accepted: 10/26/2024] [Indexed: 11/22/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a ubiquitous reproductive condition with triggering hallmarks such as glucose intolerance, hyperandrogenism, and dyslipidemia. Despite the existence of various PCOS animal models, an ideal model which could encompass all PCOS-specific phenotype is of dire need. Dehydroepiandrosterone (DHEA) induced PCOS rats are frequently employed; though, determining the superior model among pubertal and prepubertal rats, incorporation of high fat diet (HFD), and their sustainability remains uncertain. This study aims to examine the age factor, impact of HFD, and DHEA regimen in model development. Prepubertal and pubertal Sprague-Dawley rats were subcutaneously injected with DHEA (6 mg/kg and 60 mg/kg/day, respectively) with and without HFD up to 21 days. Serum testosterone, glucose, lipid profile, ovary morphology, and estrous cycle were evaluated. Following 21 days of treatment with DHEA, pubertal PCOS rats exhibited better reproductive phenotype than prepubertal rats. However, there was no significant difference in the lipid profile. Accordingly, both the age-group rats were concomitantly treated with DHEA and HFD for additional 3 weeks on alternate day basis after model development. The persistence of reproductive and metabolic features on treatment withdrawal were also simultaneously investigated by alienating the rats into continuous and stop dosing groups. The DHEA + HFD and DHEA treated pubertal rats in continuous dosing group showed significant PCOS features (p < 0.05) compared to stop dosing, prepubertal, and control groups. To conclude, continual dosing with DHEA on alternate days for 3 weeks is necessary to sustain metabolic and reproductive phenotypes of PCOS.
Collapse
Affiliation(s)
- Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shaila A Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
6
|
Li Y, Song H, Xu J, Wang Y, Bai L, Wang H, Zhang J. TIGAR relieves PCOS by inhibiting granulosa cell apoptosis and oxidative stress through activating Nrf2. Mol Cell Endocrinol 2024; 594:112381. [PMID: 39341450 DOI: 10.1016/j.mce.2024.112381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
This study aimed to elucidate the role of TP53-induced glycolysis and apoptosis regulator (TIGAR) in polycystic ovary syndrome (PCOS). A rat model PCOS was constructed by subcutaneous injection with dehydroepiandrosterone (DHEA). Follicular atresia and reduced granular cells (GCs) in ovaries suggested successful modeling. The low expression of TIGAR was observed in ovarian tissue of PCOS rat. To explore the role of TIGAR in PCOS, lentivirus carrying the TIGAR were used to up-regulate TIGAR expression. TIGAR overexpression reduced the DHEA-induced increase of ovarian weight, the levels of estradiol (E2), and the ratio of luteinizing hormone/follicle-stimulating hormone (LH/FSH) in the serum, as well as improved the morphology of the follicle, especially increased the thickness of the GC layer, which attributed to the inhibition of apoptosis by TIGAR. In addition, high expression of TIGAR inhibited oxidative stress in ovaries of PCOS rat, as evidenced by decreased level of malondialdehyde (MDA), and reactive oxygen species (ROS), and enhanced activity of glutathione peroxidase (GPX) and superoxide dismutase (SOD). Mechanically, Nrf2/OH-1 signal pathway was activated by TIGAR. The effect of TIGAR on PCOS were verified in the primary rat GCs treated with dihydrotestosterone, but also the rescue experiment was performed. Downregulation of Nrf2 reversed the effects of TIGAR, indicating that TIGAR suppressed oxidative stress and GC apoptosis by activating Nrf2/OH-1 pathway in PCOS. Finally, non-targeted metabolomics revealed that TIGAR might affect the energy metabolic pathway, thereby altering the metabolic profile of primary rat GCs. This study provided new insights into the prevention and treatment of PCOS.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Hui Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Jia Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Yunping Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Lu Bai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Haixu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China.
| | - Jianfang Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
7
|
Fang YQ, Ding H, Li T, Zhao XJ, Luo D, Liu Y, Li Y. N-acetylcysteine supplementation improves endocrine-metabolism profiles and ovulation induction efficacy in polycystic ovary syndrome. J Ovarian Res 2024; 17:205. [PMID: 39415242 PMCID: PMC11484282 DOI: 10.1186/s13048-024-01528-8] [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: 05/27/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) affects 6-20% of women worldwide, with insulin resistance and hyperinsulinemia occurring in 50-70% of patients. Hyperinsulinemia exacerbates oxidative stress, contributing to PCOS pathogenesis. N-acetylcysteine (NAC) is an antioxidant and insulin sensitizer that shows promise as a therapeutic for PCOS. Our current study aimed to investigate the effects of NAC supplementation on endocrine-metabolic parameters in PCOS mice and its effect on ovulation induction (OI) efficacy in women with PCOS. METHODS Female C57BL/6 mice were orally administered letrozole (LE) to induce PCOS and then randomly divided into groups receiving daily oral administration of 160 mg/kg NAC (PCOS + NAC group), 200 mg/kg metformin (PCOS + Met group), or 0.5% carboxymethyl cellulose (drug solvent) (pure PCOS group) for 12 days. Healthy female mice served as pure controls. Estrous cycles were monitored during the intervention. Metabolic and hormone levels, ovarian phenotypes, antioxidant activity in ovarian tissues, and oxidative stress levels in oocytes were assessed post-intervention. Furthermore, a pragmatic, randomized, controlled clinical study was conducted with 230 PCOS women, randomly assigned to the NAC group (1.8 g/day oral NAC, n = 115) or the control group (n = 115). Patients in both groups underwent ≤ 3 cycles of OI with sequential LE and urinary follicle-stimulating hormone (uFSH). Cycle characteristics and pregnancy outcomes were compared between groups. RESULTS Similar to metformin, NAC supplementation significantly improved the estrous cycles and ovarian phenotypes of PCOS mice; reduced the LH concentration, LH/FSH ratio, and T level; and increased glucose clearance and insulin sensitivity. Notably, NAC significantly reduced oocyte ROS levels and increased the mitochondrial membrane potential in PCOS mice. Additionally, NAC significantly enhanced enzymatic and nonenzymatic antioxidant activities in PCOS mouse ovaries, whereas metformin had no such effect. In the clinical trial, compared to women in the control group, women receiving NAC had significantly lower average uFSH dosage and duration (p < 0.005) and significantly greater clinical pregnancy rates per OI cycle and cumulative clinical pregnancy rates per patient (p < 0.005). CONCLUSION NAC supplementation improved endocrine-metabolic parameters in PCOS mice and significantly enhanced OI efficacy with sequential LE and uFSH in women with PCOS. Therefore, NAC could be a valuable adjuvant in OI for women with PCOS.
Collapse
Affiliation(s)
- Yu-Qing Fang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hui Ding
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Tao Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Xiao-Jie Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Dan Luo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yanhui Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
8
|
Berkel C. Inducers and Inhibitors of Pyroptotic Death of Granulosa Cells in Models of Premature Ovarian Insufficiency and Polycystic Ovary Syndrome. Reprod Sci 2024; 31:2972-2992. [PMID: 39026050 PMCID: PMC11438836 DOI: 10.1007/s43032-024-01643-3] [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: 04/04/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
Granulosa cells (GCs), the largest cell population and primary source of steroid hormones in the ovary, are the important somatic ovarian components. They have critical roles in folliculogenesis by supporting oocyte, facilitating its growth, and providing a microenvironment suitable for follicular development and oocyte maturation, thus having essential functions in maintaining female fertility and in reproductive health in general. Pyroptotic death of GCs and associated inflammation have been implicated in the pathogenesis of several reproductive disorders in females including Premature Ovarian Insufficiency (POI) and Polycystic Ovary Syndrome (PCOS). Here, I reviewed factors, either intrinsic or extrinsic, that induce or inhibit pyroptosis in GCs in various models of these disorders, both in vitro and in vivo, and also covered associated molecular mechanisms. Most of these studied factors influence NLRP3 inflammasome- and GSDMD (Gasdermin D)-mediated pyroptosis in GCs, compared to other inflammasomes and gasdermins (GSDMs). I conclude that a more complete mechanistic understanding of these factors in terms of GC pyroptosis is required to be able to develop novel strategies targeting inflammatory cell death in the ovary.
Collapse
Affiliation(s)
- Caglar Berkel
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| |
Collapse
|
9
|
Corrie L, Singh H, Gulati M, Vishwas S, Chellappan DK, Gupta G, Paiva-Santos AC, Veiga F, Alotaibi F, Alam A, Eri RD, Prasher P, Adams J, Paudel KR, Dua K, Singh SK. Polysaccharide-fecal microbiota-based colon-targeted self-nanoemulsifying drug delivery system of curcumin for treating polycystic ovarian syndrome. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6721-6743. [PMID: 38507103 DOI: 10.1007/s00210-024-03029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024]
Abstract
The gut microbiome is involved in the pathogenesis of many diseases including polycystic ovarian syndrome (PCOS). Modulating the gut microbiome can lead to eubiosis and treatment of various metabolic conditions. However, there is no proper study assessing the delivery of microbial technology for the treatment of such conditions. The present study involves the development of guar gum-pectin-based solid self-nanoemulsifying drug delivery system (S-SNEDDS) containing curcumin (CCM) and fecal microbiota extract (FME) for the treatment of PCOS. The optimized S-SNEDDS containing FME and CCM was prepared by dissolving CCM (25 mg) in an isotropic mixture consisting of Labrafil M 1944 CS, Transcutol P, and Tween-80 and solidified using lactose monohydrate, aerosil-200, guar gum, and pectin (colon-targeted CCM solid self-nanoemulsifying drug delivery system [CCM-CT-S-SNEDDS]). Pharmacokinetic and pharmacodynamic evaluation was carried out on letrozole-induced female Wistar rats. The results of pharmacokinetic studies indicated about 13.11 and 23.48-fold increase in AUC of CCM-loaded colon-targeted S-SNEDDS without FME (CCM-CT-S-SNEDDS (WFME)) and CCM-loaded colon-targeted S-SNEDDS with FME [(CCM-CT-S-SNEDDS (FME)) as compared to unprocessed CCM. The pharmacodynamic study indicated excellent recovery/reversal in the rats treated with CCM-CT-S-SNEDDS low and high dose containing FME (group 13 and group 14) in a dose-dependent manner. The developed formulation showcasing its improved bioavailability, targeted action, and therapeutic activity in ameliorating PCOS can be utilized as an adjuvant therapy for developing a dosage form, scale-up, and technology transfer.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Hardeep Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Faisal Alotaibi
- Department of Pharmacology, College of Pharmacy (Al-Duwadimi Campus), Shaqra University, Shaqra, Saudi Arabia
| | - Aftab Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Rajaraman D Eri
- School of Science, STEM College, RMIT University, Melbourne, VIC, 3001, Australia
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India
| | - Jon Adams
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Keshav Raj Paudel
- Centre of Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
10
|
Danduga RCSR, Kurapati AS, Shaik RA, Kola PK, Konidala SK, Varada HB. Synergistic Amelioration of Letrozole-induced Polycystic Ovary Syndrome in Rats: A Therapeutic Approach with Apple Cider Vinegar and Metformin Combination. Reprod Sci 2024; 31:2861-2876. [PMID: 38777948 DOI: 10.1007/s43032-024-01545-4] [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: 10/14/2023] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
The present study was conducted to evaluate the combination effect of apple cider vinegar (ACV) and metformin against letrozole-induced polycystic ovary syndrome (PCOS). Female Wistar rats were administered letrozole (1 mg/kg/day, p.o) for 21 days, except for the control group of animals. On the 22nd day, PCOS-induced animals were segregated into 4 groups and administered with CMC, ACV, metformin, and a combination of ACV and metformin, respectively. The treatments were continued for 15 days, and on the 36th day, all the animals were sacrificed for biochemical (blood glucose, lipid profile), hormonal (sex hormones and adiponectin), and pro-inflammatory mediator estimations in blood samples. The ovarian tissue samples were used for oxidative stress parameters and histological alterations. The PCOS control animals showed a significant alteration in the estrous cycle. The administration of letrozole resulted in the alteration of hormonal balance and elevation of body weights, glycemic state, lipid profile, pro-inflammatory mediators in serum, and oxidative stress in ovarian samples. Individual treatment groups and combination treatment groups reversed the letrozole-induced alterations in PCOS animals, and more promising results were observed with combination therapy than with individual treatment groups. Further, the therapeutic potential of the combination treatment group was also confirmed by the histological observations in the ovarian samples. The study showed that the combination of ACV and metformin significantly alleviated letrozole-induced PCOS complications in rats. This might have been achieved by mitigating the hormonal imbalance, pro-inflammatory, hyperglycemic, and hyperlipidemic states in serum, and oxidative stress in the ovary samples.
Collapse
Affiliation(s)
- Ravi Chandra Sekhara Reddy Danduga
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| | - Aarathi Shalom Kurapati
- Department of Pharmacology, Acharya Nagarjuna University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Guntur, 522510, India
| | - Roohi Anju Shaik
- Department of Pharmacology, Acharya Nagarjuna University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Guntur, 522510, India
| | - Phani Kumar Kola
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Sathish Kumar Konidala
- Department of Pharmaceutical Sciences, School of Biotechnology and Pharmaceutical Sciences, Vignan's Foundation for Sciences, Technology, and Research, Vadlamudi, Guntur, 522213, India
| | - Hema Bharathi Varada
- Department of Pharmacology, Acharya Nagarjuna University College of Pharmaceutical Sciences, Acharya Nagarjuna University, Guntur, 522510, India
| |
Collapse
|
11
|
Chen Y, Wang G, Chen J, Wang C, Dong X, Chang HM, Yuan S, Zhao Y, Mu L. Genetic and Epigenetic Landscape for Drug Development in Polycystic Ovary Syndrome. Endocr Rev 2024; 45:437-459. [PMID: 38298137 DOI: 10.1210/endrev/bnae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/26/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
The treatment of polycystic ovary syndrome (PCOS) faces challenges as all known treatments are merely symptomatic. The US Food and Drug Administration has not approved any drug specifically for treating PCOS. As the significance of genetics and epigenetics rises in drug development, their pivotal insights have greatly enhanced the efficacy and success of drug target discovery and validation, offering promise for guiding the advancement of PCOS treatments. In this context, we outline the genetic and epigenetic advancement in PCOS, which provide novel insights into the pathogenesis of this complex disease. We also delve into the prospective method for harnessing genetic and epigenetic strategies to identify potential drug targets and ensure target safety. Additionally, we shed light on the preliminary evidence and distinctive challenges associated with gene and epigenetic therapies in the context of PCOS.
Collapse
Affiliation(s)
- Yi Chen
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Guiquan Wang
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Reproduction and Genetics, Xiamen University, Xiamen 361023, China
| | - Jingqiao Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Congying Wang
- The Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang 322000, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 40400, Taiwan
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm 171 65, Sweden
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing 100007, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University, Beijing 100191, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Liu Y, Jiang JJ, Du SY, Mu LS, Fan JJ, Hu JC, Ye Y, Ding M, Zhou WY, Yu QH, Xia YF, Xu HY, Shi YJ, Qian SW, Tang Y, Li W, Dang YJ, Dong X, Li XY, Xu CJ, Tang QQ. Artemisinins ameliorate polycystic ovarian syndrome by mediating LONP1-CYP11A1 interaction. Science 2024; 384:eadk5382. [PMID: 38870290 DOI: 10.1126/science.adk5382] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/19/2024] [Indexed: 06/15/2024]
Abstract
Polycystic ovary syndrome (PCOS), a prevalent reproductive disorder in women of reproductive age, features androgen excess, ovulatory dysfunction, and polycystic ovaries. Despite its high prevalence, specific pharmacologic intervention for PCOS is challenging. In this study, we identified artemisinins as anti-PCOS agents. Our finding demonstrated the efficacy of artemisinin derivatives in alleviating PCOS symptoms in both rodent models and human patients, curbing hyperandrogenemia through suppression of ovarian androgen synthesis. Artemisinins promoted cytochrome P450 family 11 subfamily A member 1 (CYP11A1) protein degradation to block androgen overproduction. Mechanistically, artemisinins directly targeted lon peptidase 1 (LONP1), enhanced LONP1-CYP11A1 interaction, and facilitated LONP1-catalyzed CYP11A1 degradation. Overexpression of LONP1 replicated the androgen-lowering effect of artemisinins. Our data suggest that artemisinin application is a promising approach for treating PCOS and highlight the crucial role of the LONP1-CYP11A1 interaction in controlling hyperandrogenism and PCOS occurrence.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing-Jing Jiang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shao-Yue Du
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200032, China
| | - Liang-Shan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian-Jun Fan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400010, China
| | - Jun-Chi Hu
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400010, China
| | - Yao Ye
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Meng Ding
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei-Yu Zhou
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qiu-Han Yu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yi-Fan Xia
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong-Yu Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Jie Shi
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yong-Jun Dang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400010, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Ying Li
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cong-Jian Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Disease, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
13
|
Chen W, Liao B, Yun C, Zhao M, Pang Y. Interlukin-22 improves ovarian function in polycystic ovary syndrome independent of metabolic regulation: a mouse-based experimental study. J Ovarian Res 2024; 17:100. [PMID: 38734641 PMCID: PMC11088773 DOI: 10.1186/s13048-024-01428-x] [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: 11/22/2023] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder with multiple metabolic abnormalities. Most PCOS patients have concomitant metabolic syndromes such as insulin resistance and obesity, which often lead to the development of type II diabetes and cardiovascular disease with serious consequences. Current treatment of PCOS with symptomatic treatments such as hormone replacement, which has many side effects. Research on its origin and pathogenesis is urgently needed. Although improving the metabolic status of the body can alleviate reproductive function in some patients, there is still a subset of patients with metabolically normal PCOS that lacks therapeutic tools to address ovarian etiology. METHODS The effect of IL-22 on PCOS ovarian function was verified in a non-metabolic PCOS mouse model induced by dehydroepiandrosterone (DHEA) and rosiglitazone, as well as granulosa cell -specific STAT3 knockout (Fshrcre+Stat3f/f) mice (10 groups totally and n = 5 per group). Mice were maintained under controlled temperature and lighting conditions with free access to food and water in a specific pathogen-free (SPF) facility. Secondary follicles separated from Fshrcre+Stat3f/f mice were cultured in vitro with DHEA to mimic the hyperandrogenic environment in PCOS ovaries (4 groups and n = 7 per group) and then were treated with IL-22 to investigate the specific role of IL-22 on ovarian function. RESULTS We developed a non-metabolic mice model with rosiglitazone superimposed on DHEA. This model has normal metabolic function as evidenced by normal glucose tolerance without insulin resistance and PCOS-like ovarian function as evidenced by irregular estrous cycle, polycystic ovarian morphology (PCOM), abnormalities in sex hormone level. Supplementation with IL-22 improved these ovarian functions in non-metabolic PCOS mice. Application of DHEA in an in vitro follicular culture system to simulate PCOS follicular developmental block and ovulation impairment. Follicles from Fshrcre+Stat3f/f did not show improvement in POCS follicle development with the addition of IL-22. In DHEA-induced PCOS mice, selective ablation of STAT3 in granulosa cells significantly reversed the ameliorative effect of IL-22 on ovarian function. CONCLUSION IL-22 can improve non-metabolic PCOS mice ovarian function. Granulosa cells deficient in STAT3 reverses the role of IL-22 in alleviating ovary dysfunction in non-metabolic PCOS mice.
Collapse
Affiliation(s)
- Weixuan Chen
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Baoying Liao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Min Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynaecology, (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yanli Pang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynaecology, (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
14
|
Yu S, Hou C, Zhang X, Wei Z. Mesencephalic astrocyte-derived neurotrophic factor ameliorates inflammatory response in polycystic ovary syndrome via inhibiting TLR4-NF-κB-NLRP3 pathway. Biochem Biophys Res Commun 2024; 707:149782. [PMID: 38493745 DOI: 10.1016/j.bbrc.2024.149782] [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: 02/05/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder in women of reproductive age, which often leads to female infertility. Chronic inflammation is a significant factor in the development of PCOS. Our study aimed to explore the impact of mesencephalic astrocyte-derived neurotrophic factor (MANF), a scientifically validated anti-inflammatory factor, on 99 diagnosed PCOS patients. We also investigated its effects on PCOS mice induced with dehydroepiandrosterone (DHEA) and KGN cells induced with dihydrotestosterone (DHT). Our findings revealed a decrease in serum MANF levels in PCOS patients, which were negatively associated with serum tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) levels. The administration of recombinant human MANF (rhMANF) in PCOS mice demonstrated a decrease in pro-inflammatory cytokines and monocytes/macrophages in both peripheral blood and ovarian tissues. Furthermore, the inclusion of rhMANF notably ameliorated DHEA-induced ovarian dysfunction and fibrosis by negatively regulating the toll-like receptor 4 (TLR4)-nuclear factor kappa B (NF-κB)-NLR family, pyrin domain containing protein 3 (NLRP3) pathway. Additionally, in vitro experiments showed that the up-regulation of MANF offset DHT-induced inhibition of viability and apoptosis in KGN cells. Collectively, this study highlights the anti-inflammatory properties of MANF in PCOS and suggests its potential as a therapeutic approach for the management of PCOS.
Collapse
Affiliation(s)
- Shujun Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Chao Hou
- School of Basic Medical Science, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xinru Zhang
- School of Basic Medical Science, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Zhaolian Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
15
|
Tanaka T, Urata Y, Harada M, Kunitomi C, Kusamoto A, Koike H, Xu Z, Sakaguchi N, Tsuchida C, Komura A, Teshima A, Takahashi N, Wada-Hiraike O, Hirota Y, Osuga Y. Cellular senescence of granulosa cells in the pathogenesis of polycystic ovary syndrome. Mol Hum Reprod 2024; 30:gaae015. [PMID: 38603629 PMCID: PMC11060870 DOI: 10.1093/molehr/gaae015] [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: 11/02/2023] [Revised: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders in women of reproductive age, but its pathology has not been fully characterized and the optimal treatment strategy remains unclear. Cellular senescence is a permanent state of cell-cycle arrest that can be induced by multiple stresses. Senescent cells contribute to the pathogenesis of various diseases, owing to an alteration in secretory profile, termed 'senescence-associated secretory phenotype' (SASP), including with respect to pro-inflammatory cytokines. Senolytics, a class of drugs that selectively eliminate senescent cells, are now being used clinically, and a combination of dasatinib and quercetin (DQ) has been extensively used as a senolytic. We aimed to investigate whether cellular senescence is involved in the pathology of PCOS and whether DQ treatment has beneficial effects in patients with PCOS. We obtained ovaries from patients with or without PCOS, and established a mouse model of PCOS by injecting dehydroepiandrosterone. The expression of the senescence markers p16INK4a, p21, p53, γH2AX, and senescence-associated β-galactosidase and the SASP-related factor interleukin-6 was significantly higher in the ovaries of patients with PCOS and PCOS mice than in controls. To evaluate the effects of hyperandrogenism and DQ on cellular senescence in vitro, we stimulated cultured human granulosa cells (GCs) with testosterone and treated them with DQ. The expression of markers of senescence and a SASP-related factor was increased by testosterone, and DQ reduced this increase. DQ reduced the expression of markers of senescence and a SASP-related factor in the ovaries of PCOS mice and improved their morphology. These results indicate that cellular senescence occurs in PCOS. Hyperandrogenism causes cellular senescence in GCs in PCOS, and senolytic treatment reduces the accumulation of senescent GCs and improves ovarian morphology under hyperandrogenism. Thus, DQ might represent a novel therapy for PCOS.
Collapse
Affiliation(s)
- Tsurugi Tanaka
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Urata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akari Kusamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Koike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Zixin Xu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nanoka Sakaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chihiro Tsuchida
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Airi Komura
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayaka Teshima
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Hussain MS, Moglad E, Bansal P, Kaur H, Deorari M, Almalki WH, Kazmi I, Alzarea SI, Singh M, Kukreti N. Exploring the oncogenic and tumor-suppressive roles of Circ-ADAM9 in cancer. Pathol Res Pract 2024; 256:155257. [PMID: 38537524 DOI: 10.1016/j.prp.2024.155257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/14/2024]
Abstract
Circular RNAs (circRNAs) constitute a recently identified category of closed continuous loop RNA transcripts, serving as a subset of competing endogenous RNAs (ceRNAs) with the capacity to modulate genes by acting as microRNA sponges. In the context of cancer growth, numerous investigations have explored the potential functions of circRNAs, revealing their diverse functions either as oncogenes, promoting cancer progression, or as tumor suppressors, mitigating disease development. Among these, circRNA ADAM9 (Circ-ADAM9) is now recognized as an important player in a variety of mechanisms, both physiological and pathological, especially in cancer. The aberrant expression of Circ-ADAM9 has been observed across multiple human malignancies, implying a significant involvement in tumorigenesis. This comprehensive review aims to synthesize recent findings elucidating the function of Circ-ADAM9 in many malignancies. Additionally, the review explores the possibility of Circ-ADAM9 as a valuable biomarker, offering insights into its prognostic, diagnostic, and therapeutic implications. By summarizing the latest discoveries in this field, the review contributes to our understanding of the multifaceted contribution of Circ-ADAM9 in tumor biology and its potential applications in clinical settings.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, Rajasthan 302017, India
| | - Ehssan Moglad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs, NMIMS University, Shirpur campus, Maharastra 425405, India
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| |
Collapse
|
17
|
Li R, Hu R, Huang Y, Li D, Ma X, Yang Y. Astragalus polysaccharide alleviates polycystic ovary syndrome by reducing insulin resistance and oxidative stress and increasing the diversity of gut microbiota. Endocrine 2024; 83:783-797. [PMID: 37824046 DOI: 10.1007/s12020-023-03553-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder, which is frequently accompanied by insulin resistance, oxidative stress (OS), and dyslipidemia. Astragalus polysaccharide (APS)-as a water-soluble heteropolysaccharide-can lower blood sugar and lipid and exert anti-aging effects and thus has been proven to be beneficial to various types of metabolic diseases. However, specific mechanisms of the action of APS on PCOS are yet to be studied. METHODS Herein, BALB/C female mice aged 3 weeks were randomly divided into three groups (10 mice/group): oil + PBS group, DHEA + PBS group, and DHEA + APS group. Changes in the estrous cycle, ovarian tissue sections, serum levels of the hormone, blood glucose, blood lipid, and OS were studied. The intestinal microbiome was sequenced and Spearman correlation analysis was used to analyze the correlation between serum metabolic indexes and microflora. RESULTS The results revealed that APS treatment ameliorated insulin resistance, OS, and dyslipidemia in PCOS mice. The results of 16S rDNA sequencing indicated that there were significant differences in the composition and diversity of intestinal microorganisms between DHEA and APS treatments. Firmicutes, Lachnospiraceae, Bacilli, Lactobacillaceae, and Lachnospiraceae_NK4A13_group were abundant in the oil + PBS group. Bacteroidota and Muribaculaceae were enriched in the DHEA + PBS group, while Rikenellaceae, Odoribacter, and Marinifilaceae were enriched in the DHEA + APS group. Furthermore, Spearman correlation analysis showed that there were close interactions and correlations between intestinal bacteria and indicators of blood glucose, blood lipids, steroid hormones, and OS in PCOS mice. CONCLUSIONS Overall, the study showed that APS improved PCOS in mice by correcting serum metabolic disorders and increasing microbiome diversity, which may provide insight into understanding the pathogenesis and be a beneficial intervention for PCOS.
Collapse
Affiliation(s)
- Ruiyun Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Rui Hu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yi Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dan Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaoling Ma
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuan Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China.
- The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
18
|
Cao P, Li H, Wang P, Zhang X, Guo Y, Zhao K, Guo J, Li X, Nashun B. DNA Hypomethylation-Mediated Transcription Dysregulation Participates in Pathogenesis of Polycystic Ovary Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00072-5. [PMID: 38403164 DOI: 10.1016/j.ajpath.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a highly heterogeneous and genetically complex endocrine disorder. Although the etiology remains mostly elusive, growing evidence suggested abnormal changes of DNA methylation correlate well with systemic and tissue-specific dysfunctions in PCOS. A dehydroepiandrosterone-induced PCOS-like mouse model was generated, which has a similar metabolic and reproductive phenotype as human patients with PCOS, and was used to experimentally validate the potential role of aberrant DNA methylation in PCOS in this study. Integrated DNA methylation and transcriptome analysis revealed the potential role of genomic DNA hypomethylation in transcription regulation of PCOS and identified several key candidate genes, including BMP4, Adcy7, Tnfaip3, and Fas, which were regulated by aberrant DNA hypomethylation. Moreover, i.p. injection of S-adenosylmethionine increased the overall DNA methylation level of PCOS-like mice and restored expression of the candidate genes to similar levels as the control, alleviating reproductive and metabolic abnormalities in PCOS-like mice. These findings provided direct evidence showing the importance of normal DNA methylation in epigenetic regulation of PCOS and potential targets for diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Pengbo Cao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China; Inner Mongolia Qilu Pharmaceutical Company, Hohhot, China
| | - Haoran Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Peijun Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xinna Zhang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yuxuan Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Keyu Zhao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jiaojiao Guo
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xihe Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China; Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot, China
| | - Buhe Nashun
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China.
| |
Collapse
|
19
|
Yan Y, Di F, Zhang R, Song L, Zhang R, Qiu J. Arachidonic acid inhibit granulosa cell function by affecting metabolic function of liver in brown adipose transplantation rats. J Ovarian Res 2024; 17:43. [PMID: 38374173 PMCID: PMC10875878 DOI: 10.1186/s13048-024-01374-8] [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: 08/10/2023] [Accepted: 02/13/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a gynecological endocrine disease and could be considered a metabolic disease because it is often accompanied by obesity and insulin resistance. Brown adipose tissue (BAT) transplantation has been shown to be effective in treating PCOS rats. RESULTS The study demonstrated that BAT successfully recovered the reproductive and metabolic phenotype of PCOS rats. The disorder estrous cycle, abnormal hyperglycemia and the expression of liver factors were improved. Differentially expressed metabolites were analyzed, among them, arachidonic acid may play a role in inhibiting cell proliferation, enhancing oxidative stress reaction, promoting estrogen expression, and reducing progesterone level in KGN cells. CONCLUSION Our findings suggest that BAT transplantation may be a therapeutic strategy for PCOS by changing the expression of some cytokines and metabolites. Differentially expressed metabolites might be crucially important for the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Yan Yan
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China
| | - Fangfang Di
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China
| | - Ruoxi Zhang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China
| | - Liwen Song
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China.
| | - Runjie Zhang
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China.
| | - Jin Qiu
- Obstetrics and Gynecology Department, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai, 200336, China.
| |
Collapse
|
20
|
Liu H, Tu M, Yin Z, Zhang D, Ma J, He F. Unraveling the complexity of polycystic ovary syndrome with animal models. J Genet Genomics 2024; 51:144-158. [PMID: 37777062 DOI: 10.1016/j.jgg.2023.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a highly familial and heritable endocrine disorder. Over half of the daughters born to women with PCOS may eventually develop their own PCOS-related symptoms. Progress in the treatment of PCOS is currently hindered by the complexity of its clinical manifestations and incomplete knowledge of its etiopathogenesis. Various animal models, including experimentally induced, naturally occurring, and spontaneously arising ones, have been established to emulate a wide range of phenotypical and pathological traits of human PCOS. These studies have led to a paradigm shift in understanding the genetic, developmental, and evolutionary origins of this disorder. Furthermore, emerging evidence suggests that animal models are useful in evaluating state-of-the-art drugs and treatments for PCOS. This review aims to provide a comprehensive summary of recent studies of PCOS in animal models, highlighting the power of these disease models in understanding the biology of PCOS and aiding high-throughput approaches.
Collapse
Affiliation(s)
- Huanju Liu
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhiyong Yin
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Clinical Research Center on Birth Defect Prevention and Intervention of Zhejiang Province, Hangzhou, Zhejiang 310006, China.
| | - Jun Ma
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| | - Feng He
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
21
|
Zhu H, Wu Y, Zhuang Z, Xu J, Chen F, Wang Q, Tang Q. Ampelopsis japonica aqueous extract improves ovulatory dysfunction in PCOS by modulating lipid metabolism. Biomed Pharmacother 2024; 170:116093. [PMID: 38159378 DOI: 10.1016/j.biopha.2023.116093] [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: 10/11/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a highly prevalent endocrine and metabolic disorder that is closely associated with the proliferation and apoptosis of ovarian granulosa cells (GCs). Ampelopsis japonica (AJ) is the dried tuberous root of Ampelopsis japonica (Thunb.) Makino (A. japonica), with anti-inflammatory, antioxidant, antibacterial, antiviral, wound-healing, and antitumor properties; however, it is unclear whether this herb has a therapeutic effect on PCOS. Therefore, this study aimed to investigate the pharmacological effect of AJ on PCOS and reveal its potential mechanism of action. A PCOS rat model was established using letrozole. After establishing the PCOS model, the rats received oral treatment of AJ and Diane-35 (Positive drug: ethinylestradiol + cyproterone tablets) for 2 weeks. Lipidomics was conducted using liquid-phase mass spectrometry and chromatography. AJ significantly regulated serum hormone levels and attenuated pathological variants in the ovaries of rats with PCOS. Furthermore, AJ significantly reduced the apoptotic rate of ovarian GCs. Lipidomic analysis revealed that AJ modulated glycerolipid and glycerophospholipid metabolic pathways mediated by lipoprotein lipase (Lpl), diacylglycerol choline phosphotransferase (Chpt1), and choline/ethanolamine phosphotransferase (Cept1). Therefore, we established that AJ may reduce ovarian GC apoptosis by modulating lipid metabolism, ultimately improving ovulatory dysfunction in PCOS. Therefore, AJ is a novel candidate for PCOS treatment.
Collapse
Affiliation(s)
- Huiqing Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Yuanyuan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Ziming Zhuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Jing Xu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Feilong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China
| | - Qirui Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China.
| | - Qingfa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Guangzhou 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, China.
| |
Collapse
|
22
|
Tezuka M, Yonekubo-Awaka S, Tamai Y, Tsuchioka K, Kobayashi K, Kuramochi Y, Tatemichi S, Nagasawa T, Kiguchi S. Suppression of hypothalamic-pituitary-gonadal function by linzagolix in benign prostatic hyperplasia and polycystic ovary syndrome animal models. Clin Exp Pharmacol Physiol 2023; 50:914-923. [PMID: 37622483 DOI: 10.1111/1440-1681.13817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/15/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
The hypothalamic-pituitary-gonadal (HPG) axis is an important regulatory mechanism involved primarily in the development and regulation of the reproductive systems. The suppression of the HPG axis by gonadotropin-releasing hormone (GnRH) analogues is expected to be effective for the treatment of sex hormone-dependent diseases, such as endometriosis, uterine fibroid, prostate cancer, benign prostatic hyperplasia (BPH) and polycystic ovary syndrome (PCOS). Despite the established involvement of GnRH signalling in these disorders, the therapeutic efficacy of small molecular GnRH antagonists for BPH and PCOS has not been adequately evaluated in non-clinical studies. Therefore, the purpose of the present study was to evaluate the potential of linzagolix, a small molecular GnRH antagonist, as a potential new treatment option for BPH and PCOS. Dogs and rats exhibiting normal prostates and dogs diagnosed with prostatic hyperplasia were used to evaluate the effects of linzagolix in BPH. The effects of linzagolix were also examined in a rat model of PCOS induced by repeated administration of letrozole, an aromatase inhibitor. Linzagolix reduced serum luteinizing hormone and testosterone levels in male rats and normal or BPH model dogs and suppressed prostate weight without testosterone depletion, suggesting the existence of an optimal therapeutic testosterone level for BPH treatment. In a PCOS rat model, linzagolix improved both insulin resistance and ovarian dysfunction. Treatment with linzagolix decreased follicle-stimulating hormone levels, but did not alter serum luteinizing hormone and testosterone levels. These results indicate that linzagolix may provide a new treatment option for GnRH-related disorders, such as BPH and PCOS.
Collapse
Affiliation(s)
- Motohiro Tezuka
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | | | - Yasuaki Tamai
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Kumi Tsuchioka
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Yu Kuramochi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Satoshi Tatemichi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| | - Tatsuya Nagasawa
- Safety Research Department, Kissei Pharmaceuticals Co., Ltd., Azumino, Japan
| | - Sumiyoshi Kiguchi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd., Azumino, Japan
| |
Collapse
|
23
|
Corrie L, Ajjarapu S, Banda S, Parvathaneni M, Bolla PK, Kommineni N. HPMCAS-Based Amorphous Solid Dispersions in Clinic: A Review on Manufacturing Techniques (Hot Melt Extrusion and Spray Drying), Marketed Products and Patents. MATERIALS (BASEL, SWITZERLAND) 2023; 16:6616. [PMID: 37895598 PMCID: PMC10608006 DOI: 10.3390/ma16206616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023]
Abstract
Today, therapeutic candidates with low solubility have become increasingly common in pharmaceutical research pipelines. Several techniques such as hot melt extrusion, spray drying, supercritical fluid technology, electrospinning, KinetiSol, etc., have been devised to improve either or both the solubility and dissolution to enhance the bioavailability of these active substances belonging to BCS Class II and IV. The principle involved in all these preparation techniques is similar, where the crystal lattice of the drug is disrupted by either the application of heat or dissolving it in a solvent and the movement of the fine drug particles is arrested with the help of a polymer by either cooling or drying to remove the solvent. The dispersed drug particles in the polymer matrix have higher entropy and enthalpy and, thereby, higher free energy in comparison to the crystalline drug. Povidone, polymethaacrylate derivatives, hydroxypropyl methyl cellulose (HPMC) and hydroxypropyl methylcellulose acetate succinate derivatives are commonly used as polymers in the preparation of ASDs. Specifically, hydroxypropylmethylcellulose acetate succinate (HPMCAS)-based ASDs have become well established in commercially available products and are widely explored to improve the solubility of poorly soluble drugs. This article provides an analysis of two widely used manufacturing techniques for HPMCAS ASDs, namely, hot melt extrusion and spray drying. Additionally, details of HPMCAS-based ASD marketed products and patents have been discussed to emphasize the commercial aspect.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | | | - Srikanth Banda
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA;
| | - Madhukiran Parvathaneni
- Department of Biotechnology, Harrisburg University of Science and Technology, Harrisburg, PA 17101, USA;
| | - Pradeep Kumar Bolla
- Department of Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, USA
| | | |
Collapse
|
24
|
Zuo W, Liu X, Chen J, Zuo W, Yin Y, Nie X, Tang P, Huang Y, Yu Q, Hu Q, Zhou J, Tan Y, Huang X, Ren Q. Single-cell sequencing provides insights into the landscape of ovary in PCOS and alterations induced by CUMS. Am J Physiol Endocrinol Metab 2023; 325:E346-E362. [PMID: 37584608 DOI: 10.1152/ajpendo.00165.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder related to psychological distress. However, the mechanism underlying increased prevalence of depression in PCOS remained unclear. This study aimed to explore the unique transcriptional landscape of ovary and offered a platform to explore the mechanism of PCOS, as well as the influences caused by depression. The PCOS rat model was established by letrozole whereas PCOS rat model with depression was established by letrozole combined with chronic unpredicted mild stress (CUMS). Then single-cell RNA sequencing (scRNA-Seq) was applied to analyze the transcriptional features of rat ovaries. Granulosa cells (GCs) and fibroblasts (Fibros) accounted for the top two clusters of total 12 cell types. There were nine clusters in GCs, related to inflammatory response, endoplasmic reticulum (ER) stress, and steroidogenesis. The expression of differentially expressed genes (DEG) Hes1 was higher in PCOS and PCOS + CUMS groups, exhibiting enhanced expression by pseudotime and positively related to inflammation. Pseudotemporal analysis revealed that inflammation contributed to the different GCs distributions. Moreover, analysis of DEGs and gene ontology (GO) function enrichment revealed CUMS aggravated inflammation in PCOS GCs possibly via interferon signaling pathway. In theca cells (TCs), nine clusters were observed and some of them were relevant to inflammation, ER stress, and lipid metabolism. DEGs Ass1, Insl3, and Ifi27 were positively related to Cyp17a1, and Ces1d might contribute to the different trajectory of TCs. Subsequent scRNA-seq revealed a signature profile of endothelial cells (ECs) and Fibros, which suggest that inflammation-induced damage of ECs and Fibro, further exacerbated by CUMS. Finally, analysis of T cells and mononuclear phagocytes (MPs) revealed the existence of immune dysfunction, among which interferon signaling played a critical role. These findings provided more knowledge for a better understanding PCOS from the view of inflammation and identified new biomarkers and targets for the treatment of PCOS with psychological diseases.NEW & NOTEWORTHY In this study, we mapped the landscape of polycystic ovary syndrome (PCOS) ovary with rat model induced by letrozole and provided a novel insight into the molecular mechanism of PCOS accompanied by chronic unpredicted mild stress (CUMS) at single-cell transcriptomic level. These observations highlight the importance of inflammation in the pathogenesis of PCOS, which might also be the bridge between PCOS and psychological diseases.
Collapse
Affiliation(s)
- Wenting Zuo
- Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Xiangfei Liu
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jianhuai Chen
- Department of Andrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Wenren Zuo
- Department of Urology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Yanyun Yin
- Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Xiaowei Nie
- Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Peipei Tang
- Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Yunke Huang
- Department of Gynaecology, Women's Hospital School of Zhejiang University, Hangzhou, People's Republic of China
| | - Qian Yu
- Department of Science and Technology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Qiaoyun Hu
- Singleron Biotechnologies, Nanjing, People's Republic of China
| | - Jie Zhou
- Department of Traditional Chinese Medicine, The First People's Hospital of Nantong, Nantong, People's Republic of China
| | - Yong Tan
- Department of Reproductive Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Xi Huang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Qingling Ren
- Department of Gynaecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
25
|
Zhou J, Mo H, Feng Q, Li L, La J. ApoC3 is expressed in oocytes and increased expression is associated with PCOS progression. J Ovarian Res 2023; 16:188. [PMID: 37689737 PMCID: PMC10493025 DOI: 10.1186/s13048-023-01263-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a lifelong metabolic disorder and the most common cause of anovulatory infertility affecting women in reproductive age. Our recent study reported that apolipoprotein C3 (ApoC3) could be a potential diagnostic serum marker for metabolism disturbance in PCOS patients, but whether it is present in the ovaries and what role it plays has not yet been described. OBJECTIVE Aimed to investigate ApoC3 expression in ovary of PCOS, and to discuss its potential role in PCOS progression. METHODS ApoC3 expression in ovarian tissue samples from 12 PCOS patients along with 12 healthy controls were measured via immunohistochemistry (IHC). Also, the level of ApoC3 in follicular fluid from 14 patients diagnosed with PCOS and 13 control subjects were detected by ELISA. The expression and location of ApoC3 in ovaries of PCOS mice were tested weekly for three consecutive weeks during PCOS formation using real time PCR, Western Blot, IHC and immunofluorescence. The relation of ApoC3 and sex hormones was analyzed in mouse plasma. Additionally, the dynamic changes of ApoC3 level in ovaries of healthy mice during postnatal development was also investigated. RESULTS ApoC3 levels in ovarian tissue and follicular fluid were significantly higher in PCOS patients than in controls (33.87 ± 4.11 vs. 27.71 ± 3.65, P < 0.01; 0.87 ± 0.09 vs. 0.51 ± 0.32 ng/mL, P < 0.05), respectively. In ovary, ApoC3 was found to be located in the cytoplasm of oocyte, and its expression gradually increased with PCOS progression (P < 0.05). Furthermore, correlation analysis showed that plasma ApoC3 level was closely associated with luteinizing hormone (r = 0.709, P = 0.001), testosterone (r = 0.627, P = 0.005) and anti-mullerian hormone (r = 0.680, P = 0.002) in PCOS mice. In addition, ApoC3 level in oocyte was physiologically increased and peaked on postnatal age 21 (P21), then decreased following P21 in healthy mice. CONCLUSIONS We identified ApoC3 expression in oocyte. It may be involved in PCOS progression and possibly participate in the regulation of oocyte development.
Collapse
Affiliation(s)
- Jiahe Zhou
- Guangdong Women and Children Hospital, Guangzhou, 511442, China
- Guangzhou Medical University, Guangzhou, 511436, China
| | - Hui Mo
- Faculty of Chinese Medicines, Macau University of Science and Technology, Macao, 000853, China
| | - Qian Feng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Li Li
- Guangdong Women and Children Hospital, Guangzhou, 511442, China.
- Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jiahui La
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| |
Collapse
|
26
|
Kommineni N, Chaudhari R, Conde J, Tamburaci S, Cecen B, Chandra P, Prasad R. Engineered Liposomes in Interventional Theranostics of Solid Tumors. ACS Biomater Sci Eng 2023; 9:4527-4557. [PMID: 37450683 DOI: 10.1021/acsbiomaterials.3c00510] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Engineered liposomal nanoparticles have unique characteristics as cargo carriers in cancer care and therapeutics. Liposomal theranostics have shown significant progress in preclinical and clinical cancer models in the past few years. Liposomal hybrid systems have not only been approved by the FDA but have also reached the market level. Nanosized liposomes are clinically proven systems for delivering multiple therapeutic as well as imaging agents to the target sites in (i) cancer theranostics of solid tumors, (ii) image-guided therapeutics, and (iii) combination therapeutic applications. The choice of diagnostics and therapeutics can intervene in the theranostics property of the engineered system. However, integrating imaging and therapeutics probes within lipid self-assembly "liposome" may compromise their overall theranostics performance. On the other hand, liposomal systems suffer from their fragile nature, site-selective tumor targeting, specific biodistribution and premature leakage of loaded cargo molecules before reaching the target site. Various engineering approaches, viz., grafting, conjugation, encapsulations, etc., have been investigated to overcome the aforementioned issues. It has been studied that surface-engineered liposomes demonstrate better tumor selectivity and improved therapeutic activity and retention in cells/or solid tumors. It should be noted that several other parameters like reproducibility, stability, smooth circulation, toxicity of vital organs, patient compliance, etc. must be addressed before using liposomal theranostics agents in solid tumors or clinical models. Herein, we have reviewed the importance and challenges of liposomal medicines in targeted cancer theranostics with their preclinical and clinical progress and a translational overview.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ruchita Chaudhari
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa; Lisboa 1169-056, Portugal
| | - Sedef Tamburaci
- Department of Chemical Engineering, Izmir Institute of Technology, Gulbahce Campus, Izmir 35430, Turkey
| | - Berivan Cecen
- Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
- Department of Mechanical Engineering, Rowan University, Glassboro, New Jersey 08028, United States
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rajendra Prasad
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
27
|
Das D, Shruthi NR, Banerjee A, Jothimani G, Duttaroy AK, Pathak S. Endothelial dysfunction, platelet hyperactivity, hypertension, and the metabolic syndrome: molecular insights and combating strategies. Front Nutr 2023; 10:1221438. [PMID: 37614749 PMCID: PMC10442661 DOI: 10.3389/fnut.2023.1221438] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023] Open
Abstract
Metabolic syndrome (MetS) is a multifaceted condition that increases the possibility of developing atherosclerotic cardiovascular disease. MetS includes obesity, hypertension, dyslipidemia, hyperglycemia, endothelial dysfunction, and platelet hyperactivity. There is a concerning rise in the occurrence and frequency of MetS globally. The rising incidence and severity of MetS need a proactive, multipronged strategy for identifying and treating those affected. For many MetS patients, achieving recommended goals for healthy fat intake, blood pressure control, and blood glucose management may require a combination of medicine therapy, lifestyles, nutraceuticals, and others. However, it is essential to note that lifestyle modification should be the first-line therapy for MetS. In addition, MetS requires pharmacological, nutraceutical, or other interventions. This review aimed to bring together the etiology, molecular mechanisms, and dietary strategies to combat hypertension, endothelial dysfunction, and platelet dysfunction in individuals with MetS.
Collapse
Affiliation(s)
- Diptimayee Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Nagainallur Ravichandran Shruthi
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Asim K. Duttaroy
- Faculty of Medicine, Department of Nutrition, Institute of Medical Sciences, University of Oslo, Oslo, Norway
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| |
Collapse
|
28
|
Lissaman AC, Girling JE, Cree LM, Campbell RE, Ponnampalam AP. Androgen signalling in the ovaries and endometrium. Mol Hum Reprod 2023; 29:gaad017. [PMID: 37171897 PMCID: PMC10663053 DOI: 10.1093/molehr/gaad017] [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: 11/22/2022] [Revised: 04/27/2023] [Indexed: 05/14/2023] Open
Abstract
Currently, our understanding of hormonal regulation within the female reproductive system is largely based on our knowledge of estrogen and progesterone signalling. However, while the important functions of androgens in male physiology are well known, it is also recognized that androgens play critical roles in the female reproductive system. Further, androgen signalling is altered in a variety of gynaecological conditions, including endometriosis and polycystic ovary syndrome, indicative of regulatory roles in endometrial and ovarian function. Co-regulatory mechanisms exist between different androgens, estrogens, and progesterone, resulting in a complex network of steroid hormone interactions. Evidence from animal knockout studies, in vitro experiments, and human data indicate that androgen receptor expression is cell-specific and menstrual cycle stage-dependent, with important regulatory roles in the menstrual cycle, endometrial biology, and follicular development in the ovaries. This review will discuss the expression and co-regulatory interactions of androgen receptors, highlighting the complexity of the androgen signalling pathway in the endometrium and ovaries, and the synthesis of androgens from additional alternative pathways previously disregarded as male-specific. Moreover, it will illustrate the challenges faced when studying androgens in female biology, and the need for a more in-depth, integrative view of androgen metabolism and signalling in the female reproductive system.
Collapse
Affiliation(s)
- Abbey C Lissaman
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jane E Girling
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Lynsey M Cree
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Rebecca E Campbell
- Department of Physiology and Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Anna P Ponnampalam
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Pūtahi Manawa-Healthy Hearts for Aotearoa New Zealand, Centre of Research Excellence, New Zealand
| |
Collapse
|
29
|
Almeida GHDR, Iglesia RP, Rinaldi JDC, Murai MK, Calomeno CVAQ, da Silva Junior LN, Horvath-Pereira BDO, Pinho LBM, Miglino MA, Carreira ACO. Current Trends on Bioengineering Approaches for Ovarian Microenvironment Reconstruction. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36355603 DOI: 10.1089/ten.teb.2022.0171] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian tissue has a unique microarchitecture and a complex cellular and molecular dynamics that are essential for follicular survival and development. Due to this great complexity, several factors may lead to ovarian insufficiency, and therefore to systemic metabolic disorders and female infertility. Techniques currently used in the reproductive clinic such as oocyte cryopreservation or even ovarian tissue transplant, although effective, have several limitations, which impair their wide application. In this scenario, mimetic ovarian tissue reconstruction comes as an innovative alternative to develop new methodologies for germ cells preservation and ovarian functions restoration. The ovarian extracellular matrix (ECM) is crucial for oocyte viability maintenance, once it acts actively in folliculogenesis. One of the key components of ovarian bioengineering is biomaterials application that mimics ECM and provides conditions for cell anchorage, proliferation, and differentiation. Therefore, this review aims at describing ovarian tissue engineering approaches and listing the main limitations of current methods for preservation and reestablishment of ovarian fertility. In addition, we describe the main elements that structure this study field, highlighting the main advances and the challenges to overcome to develop innovative methodologies to be applied in reproductive medicine. Impact Statement This review presents the main advances in the application of tissue bioengineering in the ovarian tissue reconstruction to develop innovative solutions for ovarian fertility reestablishment.
Collapse
Affiliation(s)
| | - Rebeca Piatniczka Iglesia
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mikaelly Kiemy Murai
- Department of Morphological Sciences, State University of Maringa, Maringá, Brazil
| | | | | | | | - Letícia Beatriz Mazo Pinho
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Center of Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
| |
Collapse
|
30
|
Haddad-Filho H, Tosatti JAG, Vale FM, Gomes KB, Reis FM. Updates in diagnosing polycystic ovary syndrome-related infertility. Expert Rev Mol Diagn 2023; 23:123-132. [PMID: 36856088 DOI: 10.1080/14737159.2023.2177536] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a condition that affects approximately 13% of reproductive age women and is characterized by androgen excess, menstrual irregularity and altered ovarian morphology. PCOS presents a complex etiology and pathophysiology, which still requires a detailed investigation of biochemical signatures to identify the molecules and mechanisms that govern it. AREAS COVERED This narrative review summarizes the main molecular alterations found in the ovarian follicular fluid, endometrium and placenta of women with PCOS, and the genotypes potentially associated with the outcome of infertility treatments in PCOS. EXPERT OPINION PCOS is associated with multiple alterations in growth factors, sex steroid hormones, reactive oxygen species, proinflammatory cytokines and adipokines, which contribute to follicle arrest/ anovulation or suboptimal corpus luteum function, and ultimately to menstrual irregularity and hyperandrogenic symptoms. A panel of PCOS biomarkers should include, besides ovarian products, markers of adipose tissue function, insulin resistance, vascular health, and low-grade chronic inflammation. The effects of ovarian stimulation drugs on infertile women with PCOS are likely to be modified by genetic factors, but the available evidence is heterogeneous; therefore, future studies should evaluate standard treatments and pre-specified outcomes of interest to provide more conclusive answers.
Collapse
Affiliation(s)
- Hélio Haddad-Filho
- Graduate Program in Surgery, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Department of Medicine, Universidade Federal de Lavras, Lavras, Brazil
| | - Jéssica A G Tosatti
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda M Vale
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karina B Gomes
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando M Reis
- Division of Human Reproduction, Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
31
|
Ataabadi MS, Bahmanpour S, Yousefinejad S, Alaee S. Blood volatile organic compounds as potential biomarkers for poly cystic ovarian syndrome (PCOS): An animal study in the PCOS rat model. J Steroid Biochem Mol Biol 2023; 226:106215. [PMID: 36332782 DOI: 10.1016/j.jsbmb.2022.106215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Polycystic ovary syndrome (PCOS) as a common metabolic and endocrinological disorder can affect the metabolic profile in biological fluids. We studied the profile of blood volatile organic compounds (VOCs) in rats with PCOS and controls to identify potential specific biomarkers of blood VOCs in PCOS rats. For this purpose, 30 female adult Wistar rats were assigned to two groups: control and PCOS groups. PCOS model was induced using letrozole gavage (1 mg/kg) for 21 days. The rats in the control group received water of the same volume for 21 days. During treatment, a collection of vaginal smears was done every day for estrus cycle determination and weight was measured weekly. On the day after the last administration of letrozole, the rats were killed and their blood and ovaries were collected. Testosterone levels and histologic changes in ovaries were examined. Also, headspace-solid phase microextraction-gas chromatography-mass spectrometry (HS-SPME-GC-MS) analyzed the VOCs in the blood of PCOS and control rats. Multivariate and univariate statistical analyses were used to find the potential biomarkers for a rat model of PCOS. Weight gain, ovarian and vaginal pathological alteration, as well as hyperandrogenemia, confirmed the successful induction of the PCOS in rats. The results of blood VOCs analysis showed that nine VOCs were significantly elevated and one VOC decreased in the PCOS group than the control group (P < 0/05). The partial least-squares discriminant analysis (PLS-DA) and principal component analysis (PCA) showed good separation of VOCs between the PCOS rats and the control group. The 4-ethylphenol and capric (decanoic) acid were selected as the potential biomarkers for PCOS diagnosis in the blood of the PCOS rats. The blood of PCOS rats had a specific profile of VOCs, which could be detected by GC-MS analysis. These findings can pave the way for further studies towards developing a new screening method for PCOS detection and studying their pathology, based on VOCs analysis.
Collapse
Affiliation(s)
- Mahmoud Sadeghi Ataabadi
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz Univcersity of Medical Sciences, Shiraz, Iran
| | - Soghra Bahmanpour
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Yousefinejad
- Research Center for Health Sciences, Institute of Health, Department of Occupational Health Engineering, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sanaz Alaee
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz Univcersity of Medical Sciences, Shiraz, Iran
| |
Collapse
|
32
|
Corrie L, Awasthi A, Kaur J, Vishwas S, Gulati M, Kaur IP, Gupta G, Kommineni N, Dua K, Singh SK. Interplay of Gut Microbiota in Polycystic Ovarian Syndrome: Role of Gut Microbiota, Mechanistic Pathways and Potential Treatment Strategies. Pharmaceuticals (Basel) 2023; 16:197. [PMID: 37259345 PMCID: PMC9967581 DOI: 10.3390/ph16020197] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 11/26/2023] Open
Abstract
Polycystic Ovarian Syndrome (PCOS) comprises a set of symptoms that pose significant risk factors for various diseases, including type 2 diabetes, cardiovascular disease, and cancer. Effective and safe methods to treat all the pathological symptoms of PCOS are not available. The gut microbiota has been shown to play an essential role in PCOS incidence and progression. Many dietary plants, prebiotics, and probiotics have been reported to ameliorate PCOS. Gut microbiota shows its effects in PCOS via a number of mechanistic pathways including maintenance of homeostasis, regulation of lipid and blood glucose levels. The effect of gut microbiota on PCOS has been widely reported in animal models but there are only a few reports of human studies. Increasing the diversity of gut microbiota, and up-regulating PCOS ameliorating gut microbiota are some of the ways through which prebiotics, probiotics, and polyphenols work. We present a comprehensive review on polyphenols from natural origin, probiotics, and fecal microbiota therapy that may be used to treat PCOS by modifying the gut microbiota.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Jaskiran Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jaipur 302017, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600007, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | | | - Kamal Dua
- ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
33
|
Ahmed ASF, Sharkawi SS, AbdelHameed SS, Bayoumi AM, Moussa RS, Alhakamy NA, Al Sadoun H, Mansouri RA, El-Moselhy MA, El-Daly M, Anter AF, Truhan TE. Ketogenic diet restores hormonal, apoptotic/proliferative balance and enhances the effect of metformin on a letrozole-induced polycystic ovary model in rats. Life Sci 2023; 313:121285. [PMID: 36526050 DOI: 10.1016/j.lfs.2022.121285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Polycystic ovaries (PCO) is a hormonal disorder that is a leading cause of infertility. The formation of multiple persistent cysts and hormonal imbalance are hallmarks of PCO. Recent clinical studies reported a beneficial effect of the ketogenic diet (KD; high-fat, low-carbohydrate) on PCO. The aim of this study was to investigate the effect of the KD alone and in combination with metformin on letrozole-induced PCO in female rats. METHODS Female rats were grouped into control and PCO (letrozole; 1 mg/kg for 21 days). The PCO group was subdivided into PCO (non-treated), PCO-metformin (300 mg/kg), PCO rats fed with KD only, and PCO rats treated with metformin and fed with KD. All groups continued to receive letrozole during the 21-day treatment period. At the end of the experiment, serum and ovaries were collected for further analysis. RESULTS The untreated-PCO rats showed increased testosterone, LH/FSH ratio, and ovary weights. Disturbed apoptosis and proliferation balance were evident as a low caspase-3 activation and proliferating cell nuclear antigen expression and increased TGF-β expression. The KD improved the letrozole-induced effects, which was comparable to the effect of metformin. Combining the KD with metformin treatment additively enhanced the metformin effect. CONCLUSION Our results indicate that the KD has a protective role against PCO in rats, especially when combined with metformin. This study reveals a potential therapeutic role of the KD in PCO, which could prompt valuable future clinical applications.
Collapse
Affiliation(s)
- Al-Shaimaa F Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Sara S Sharkawi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Sara S AbdelHameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Asmaa M Bayoumi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Rabab S Moussa
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Mohamed Saeed Tamer Chair for Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hadeel Al Sadoun
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Rasha A Mansouri
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed A El-Moselhy
- Clinical Pharmacy and Pharmacology Department, Ibn Sina National College for Medical Studies, Jeddah 21589, Saudi Arabia
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Aliaa F Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia, Egypt
| | | |
Collapse
|
34
|
Szukiewicz D, Trojanowski S, Kociszewska A, Szewczyk G. Modulation of the Inflammatory Response in Polycystic Ovary Syndrome (PCOS)-Searching for Epigenetic Factors. Int J Mol Sci 2022; 23:ijms232314663. [PMID: 36498989 PMCID: PMC9736994 DOI: 10.3390/ijms232314663] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in women of reproductive age. Despite its incidence, the syndrome is poorly understood and remains underdiagnosed, and female patients are diagnosed with a delay. The heterogenous nature of this complex disorder results from the combined occurrence of genetic, environmental, endocrine, and behavioral factors. Primary clinical manifestations of PCOS are derived from the excess of androgens (anovulation, polycystic ovary morphology, lack of or scanty, irregular menstrual periods, acne and hirsutism), whereas the secondary manifestations include multiple metabolic, cardiovascular, and psychological disorders. Dietary and lifestyle factors play important roles in the development and course of PCOS, which suggests strong epigenetic and environmental influences. Many studies have shown a strong association between PCOS and chronic, low-grade inflammation both in the ovarian tissue and throughout the body. In the vast majority of PCOS patients, elevated values of inflammatory markers or their gene markers have been reported. Development of the vicious cycle of the chronic inflammatory state in PCOS is additionally stimulated by hyperinsulinemia and obesity. Changes in DNA methylation, histone acetylation and noncoding RNA levels are presented in this review in the context of oxidative stress, reactive oxygen species, and inflammatory signaling in PCOS. Epigenetic modulation of androgenic activity in response to inflammatory signaling is also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
- Correspondence:
| | - Seweryn Trojanowski
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Anna Kociszewska
- Chair and Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, 03-242 Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
35
|
Corrie L, Kaur J, Awasthi A, Vishwas S, Gulati M, Saini S, Kumar B, Pandey NK, Gupta G, Dureja H, Chellapan DK, Dua K, Tewari D, Singh SK. Multivariate Data Analysis and Central Composite Design-Oriented Optimization of Solid Carriers for Formulation of Curcumin-Loaded Solid SNEDDS: Dissolution and Bioavailability Assessment. Pharmaceutics 2022; 14:2395. [PMID: 36365213 PMCID: PMC9697677 DOI: 10.3390/pharmaceutics14112395] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 10/29/2023] Open
Abstract
The study was initiated with two major purposes: investigating the role of isomalt (GIQ9) as a pharmaceutical carrier for solid self-nanoemulsifying drug delivery systems (S-SNEDDSs) and improving the oral bioavailability of lipophilic curcumin (CUN). GIQ9 has never been explored for solidification of liquid lipid-based nanoparticles such as a liquid isotropic mixture of a SNEDDS containing oil, surfactant and co-surfactant. The suitability of GIQ9 as a carrier was assessed by calculating the loading factor, flow and micromeritic properties. The S-SNEDDSs were prepared by surface adsorption technique. The formulation variables were optimized using central composite design (CCD). The optimized S-SNEDDS was evaluated for differential scanning calorimetry (DSC), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD), microscopy, dissolution and pharmacokinetic studies. The S-SNEDDS showed a particle size, zeta potential and PDI of 97 nm, -26.8 mV and 0.354, respectively. The results of DSC, XRD, FTIR and microscopic studies revealed that the isotropic mixture was adsorbed onto the solid carrier. The L-SNEDDS and S-SNEDDS showed no significant difference in drug release, indicating no change upon solidification. The optimized S-SNEDDS showed 5.1-fold and 61.7-fold enhancement in dissolution rate and oral bioavailability as compared to the naïve curcumin. The overall outcomes of the study indicated the suitability of GIQ9 as a solid carrier for SNEDDSs.
Collapse
Affiliation(s)
- Leander Corrie
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Jaskiran Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Ankit Awasthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Sumant Saini
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Bimlesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Narendra Kumar Pandey
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur 302017, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Dinesh Kumar Chellapan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Descipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
36
|
Abstract
Polycystic ovary syndrome (PCOS) is a complex disease affecting up to 15% of women of reproductive age. Women with PCOS suffer from reproductive dysfunctions with excessive androgen secretion and irregular ovulation, leading to reduced fertility and pregnancy complications. The syndrome is associated with a wide range of comorbidities including type 2 diabetes, obesity, and psychiatric disorders. Despite the high prevalence of PCOS, its etiology remains unclear. To understand the pathophysiology of PCOS, how it is inherited, and how to predict PCOS, and prevent and treat women with the syndrome, animal models provide an important approach to answering these fundamental questions. This minireview summarizes recent investigative efforts on PCOS-like rodent models aiming to define underlying mechanisms of the disease and provide guidance in model selection. The focus is on new genetic rodent models, on a naturally occurring rodent model, and provides an update on prenatal and peripubertal exposure models.
Collapse
|
37
|
Chávez-Genaro R, Toledo A, Hernández K, Anesetti G. Structural and functional changes in rat uterus induced by neonatal androgenization. J Mol Histol 2022; 53:903-914. [PMID: 36201133 DOI: 10.1007/s10735-022-10106-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Fetal or neonatal androgen exposure has a programming effect on ovarian function inducing a polycystic ovarian syndrome-like condition. Its effects on uterine structure and function are poorly studied. The aim of this work was to characterize the temporal course of changes in the rat uterine structure induced by neonatal exposure to aromatizable or not aromatizable androgens. Rats were daily treated with testosterone, dihydrotestosterone or vehicle during follicle assembly period (postnatal days 1 to 5). Uterine histoarchitecture, hormonal milieu, endometrial stromal collagen and capillary density were analyzed at prepubertal, pubertal and adult ages. Our data shows that neonatal androgen exposure induces early and long-lasting deleterious effects on uterine development, including altered adenogenesis and superficial epithelial alterations and suggest a role for altered serum estradiol levels in the maintenance and worsening of the situation. Our results suggest that alterations of the neonatal androgenic environment on the uterus could be responsible for alterations in the processes of implantation and maintenance of the embryo in women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- Rebeca Chávez-Genaro
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Agustina Toledo
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Karina Hernández
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Gabriel Anesetti
- Laboratorio de Biología de la Reproducción, Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
38
|
Poojary PS, Nayak G, Panchanan G, Rao A, Kundapur SD, Kalthur SG, Mutalik S, Adiga SK, Zhao Y, Bakkum-Gamez J, Chang AY, DeStephano C, Sherman M, Kannan N, Kalthur G. Distinctions in PCOS Induced by Letrozole Vs Dehydroepiandrosterone With High-fat Diet in Mouse Model. Endocrinology 2022; 163:6625847. [PMID: 35776497 DOI: 10.1210/endocr/bqac097] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Indexed: 11/19/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is a complex health condition associated with metabolic disturbances and infertility. Recent data suggest that the prevalence of PCOS is increasing among women globally, although the etiology of these trends is undefined. Consequently, preclinical models that better reflect the biology of PCOS are urgently needed to facilitate research that can lead to the discovery of prevention strategies or improved management. The existing animal models have several limitations as they do not reflect all the PCOS features metabolically and/or phenotypically. Therefore, there is no clear consensus on the use of appropriate animal model and selection of the most appropriate PCOS-inducing agent. To that end, we have established a Swiss albino mouse model of PCOS based on 3 weeks of daily treatment with letrozole (50 μg/day; intraperitoneal) and dehydroepiandrosterone (DHEA, 6 mg/100 g body weight; subcutaneous) in 5-week-old female mice fed on normal or high-fat diet (HFD). Mice were regularly assessed for body weight, blood glucose, and estrous cycle. Three weeks after drug administration, mice were sacrificed and assessed for blood-based metabolic parameters as well as ovarian function. Our results indicate that DHEA combined with HFD produces changes mimicking those of clinical PCOS, including elevated serum testosterone and luteinizing hormone, dyslipidemia, poor ovarian microenvironment, and development of multiple ovarian cysts, recapitulating cardinal features of PCOS. In comparison, normal diet and/or letrozole produced fewer features of PCOS. The data from the experimental models presented here can improve our understanding of PCOS, a growing concern in women's health.
Collapse
Affiliation(s)
- Pooja Suresh Poojary
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Guruprasad Nayak
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Gangotri Panchanan
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Arpitha Rao
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Sanjna Das Kundapur
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Sneha Guruprasad Kalthur
- Department of Anatomy, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Satish Kumar Adiga
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
| | - Yulian Zhao
- Department of Obstetrics and Gynecology and Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Jamie Bakkum-Gamez
- Division of Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Alice Y Chang
- Division of Gynecologic Oncology Surgery, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 55902, USA
| | - Christopher DeStephano
- Division of Endocrinology, Diabetes, Metabolism, Nutrition, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Mark Sherman
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Nagarajan Kannan
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
- Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN 55902, USA
- Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal-576104, Karnataka, India
- Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
39
|
Zhang Y, Xu L. Comparative study of DHEA and letrozole induced polycystic ovary syndrome in post-pubertal rats. Gynecol Endocrinol 2022; 38:425-431. [PMID: 35311605 DOI: 10.1080/09513590.2022.2052843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AIM In order to select a suitable rat model for studying polycystic ovary syndrome (PCOS). METHODS Post-pubertal Sprague Dawley (SD) rats were allocated to four groups, abbreviated as the control 1 group, DHEA subcutaneous injection (DHEA) group, control 2 group, letrozole intragastric injection (letrozole) group. Body weight and the estrous cycle were monitored during the modeling. Serum stress hormones and reproductive hormones were evaluated by ELISA. The uterus and ovaries were weighed, ovarian follicle counts were calculated, and AR, LHR, FSHR, and AMH expression were assessed by immunohistochemistry. RESULTS In both model group subjected to the 30-day protocol of DHEA or letrozole, the mean body weights were significantly increased. Rats from both model group exhibited a significant increase of the mean length of the diestrous phase of the estrous cycle, the DHEA group had a longer diestrous phase. There was also a significant increase of serum T, E2, LH, insulin, and a significant decrease of AMH in both model group. There was a significant decrease of ovarian weight, P, and serum FSH, and a significant increase of uterus weight, serum LH/FSH in DHEA model group. There was no significant difference in serum P, FSH, and LH/FSH in letrozole model group. Both two model groups showed significantly higher AR, LHR, and AMH protein expression in rats' ovaries. FSHR protein expression in DHEA group was lower, and in letrozole model group was not different. CONCLUSION Compared with letrozole intragastric injection group, the pathological characteristics of DHEA subcutaneous injection group rats were closer to the characteristics of PCOS.
Collapse
Affiliation(s)
- Yuxuan Zhang
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingling Xu
- Department of Pharmacy, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
40
|
Wang X, Gu L, Zhang Y, Xiong C, Peng Y, Ding X. Effects of dehydroepiandrosterone alone or in combination with a high-fat diet and antibiotic cocktail on the heterogeneous phenotypes of PCOS mouse models by regulating gut microbiota. Front Endocrinol (Lausanne) 2022; 13:1030151. [PMID: 36619569 PMCID: PMC9813398 DOI: 10.3389/fendo.2022.1030151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine and metabolic disease. The gut microbiota is highly correlated with androgen secretion and insulin resistance (IR), which are two potential major pathogenic mechanisms of PCOS. Currently, an antibiotic cocktail (ABX) is often used to construct pseudo germ-free mouse models for studies on the gut microbiota and PCOS. Our work aimed to study the effects of dehydroepiandrosterone (DHEA), a high-fat diet (HFD) and ABX on the heterogeneous phenotypes of PCOS mouse models by regulating the gut microbiota. METHODS PCOS mouse models were established by subcutaneous injection of DHEA alone or in combination with a HFD in wild-type and pseudo germ-free mice. The changes in ovary morphology and sex hormonal and glycolipid metabolic parameters were evaluated. RESULTS Wild-type mice treated with DHEA or DHEA+HFD showed a PCOS-like phenotype of hyperandrogenism, anovulation and polycystic ovaries. The former was combined with hyperinsulinemia and IR, while the latter was combined with glucolipid metabolic disorders, extremely heterogeneous hyperinsulinemia and IR. The phenotype of PCOS mice, especially the metabolic parameters, was correlated with the gut microbiota. The pseudo germ-free mice treated with DHEA or DHEA+HFD also showed a PCOS-like phenotype. However, DHEA could not induce hyperinsulinemia or IR in pseudo germ-free mice. Pseudo germ-free mice treated with DHEA+HFD exhibited decreased serum AMH level, glucolipid metabolic disorders and IR. Compared with the wild-type mice, the pseudo germ-free mice treated with DHEA showed significantly higher testosterone and lipid levels and lower blood glucose levels, and they did not present with hyperinsulinemia or IR. CONCLUSION A better and stabilized mouse model simulating the pathophysiological defects of PCOS was induced by DHEA alone rather than by DHEA+HFD. The ABX intervention improved glucose metabolic disorders and hyperinsulinemia but aggravated the hyperandrogenism and lipid metabolic disorders of the PCOS mice. This study suggests that the gut microbiota plays an important role in the heterogeneous phenotypes of PCOS mouse models.
Collapse
Affiliation(s)
| | | | | | | | - Yongde Peng
- *Correspondence: Xiaoying Ding, ; Yongde Peng,
| | | |
Collapse
|