1
|
Tătaru I, Gardikiotis I, Dragostin OM, Confederat L, Gîrd C, Zamfir AS, Morariu ID, Chiţescu CL, Dinu (Iacob) A, Popescu LC, Zamfir CL. Multilevel Assessment of Glycemic, Hormonal, and Oxidative Parameters in an Experimental Diabetic Female Rat Model. Biomedicines 2025; 13:922. [PMID: 40299518 PMCID: PMC12024543 DOI: 10.3390/biomedicines13040922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Diabetes mellitus induces profound metabolic and endocrine alterations, impacting reproductive function through oxidative stress and hormonal imbalances. This study investigated the effects of alloxan-induced diabetes on hormonal status and oxidative stress in female Wistar rats. Methods: A synthetic sulfonamide derivative (compound S) was obtained via chemical synthesis and characterized by elemental and spectral analysis. Salvia officinalis extract was phytochemically profiled using UHPLC-HRMS and assessed for antioxidant potential using DPPH, ABTS, and FRAP assays. The synthetic compound and the plant extract, along with metformin were evaluated in vivo for their potential antihyperglycemic, hormone-regulating, and antioxidant properties., Serum levels of progesterone, estradiol, and follicle-stimulating hormone (FSH) were evaluated alongside oxidative stress biomarkers transforming growth factor-beta 1 (TGF-β1) and glutathione peroxidase 3 (GPX3). Results: Diabetic rats (untreated) exhibited a significant decrease in estradiol (22.00 ± 4.1 pg/mL vs. 54.74 ± 17.5 pg/mL in controls, p < 0.001) and an increase in progesterone levels (17.38 ± 9.6 ng/mL vs. 3.59 ± 0.90 ng/mL in controls, p < 0.05), suggestive for ovarian dysfunction. TGF-β1 levels were elevated in diabetic rats (27.73 ± 19.4 ng/mL vs. 21.55 ± 13.15 ng/mL in controls, p < 0.05), while increased serum GPX3 (61.50 ± 11.3 ng/mL vs. 38.20 ± 12.84 ng/mL in controls, p < 0.05) indicates enhanced oxidative stress. Statistical analysis revealed a correlation between serum GPX3 levels, FSH (p = -0.039), and estradiol (p = -0.025) in the diabetic group (L2). Conclusions: These findings contribute new evidence regarding the effects of diabetes on reproductive hormones and oxidative stress in female models.
Collapse
Affiliation(s)
- Iulian Tătaru
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.T.); (C.L.Z.)
| | - Ioannis Gardikiotis
- CEMEX—Advanced Center for Research and Development in Experimental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700454 Iasi, Romania;
| | - Oana-Maria Dragostin
- Research Centre in the Medical-Pharmaceutical Field, Department of Pharmaceutical Science, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800201 Galati, Romania; (C.L.C.); (A.D.)
| | - Luminita Confederat
- Department of Biomedical Sciences, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania;
| | - Cerasela Gîrd
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.G.); (L.C.P.)
| | - Alexandra-Simona Zamfir
- Department of Medical Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Ionela Daniela Morariu
- Department of Environmental and Food Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Carmen Lidia Chiţescu
- Research Centre in the Medical-Pharmaceutical Field, Department of Pharmaceutical Science, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800201 Galati, Romania; (C.L.C.); (A.D.)
| | - Ancuța Dinu (Iacob)
- Research Centre in the Medical-Pharmaceutical Field, Department of Pharmaceutical Science, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800201 Galati, Romania; (C.L.C.); (A.D.)
| | - Liliana Costea Popescu
- Department of Pharmacognosy, Phytochemistry and Phytotherapy, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.G.); (L.C.P.)
| | - Carmen Lăcrămioara Zamfir
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (I.T.); (C.L.Z.)
| |
Collapse
|
2
|
Smail NL, Adnane M, Wagener K, Drillich M, Chapwanya A. Roadmap to Dystocia Management-Guiding Obstetric Interventions in Cattle. Life (Basel) 2025; 15:457. [PMID: 40141801 PMCID: PMC11943763 DOI: 10.3390/life15030457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Dystocia, or difficult labor, is a common complication during parturition in cattle that poses substantial risks to both dam and fetus. When the incidence is high on a farm level, it is a significant economic burden for dairy and beef enterprises. This review paper presents a comprehensive roadmap strategy to enhance decision-making in the management of dystocia in cows. The strategy encompasses early recognition and assessment, utilization of advanced diagnostic tools, and a range of medical and surgical interventions tailored to specific maternal and fetal causes of dystocia. The roadmap also integrates preventive measures to reduce the incidence of dystocia through genetic selection and optimized nutrition. By addressing the key challenges in dystocia management, such as resource constraints, timely intervention, and the need for continuous education, this strategy aims to improve health outcomes for cows and calves and reduce economic losses. Implementing this structured approach can facilitate better preparedness, efficient resource utilization, and improved overall livestock management, thereby promoting the sustainability and productivity of the cattle industry and addressing animal welfare aspects.
Collapse
Affiliation(s)
- Nasreddine Larbi Smail
- Department of Biomedicine, Institute of Veterinary Sciences, University of Tiaret, Tiaret 14000, Algeria;
| | - Mounir Adnane
- Department of Biomedicine, Institute of Veterinary Sciences, University of Tiaret, Tiaret 14000, Algeria;
| | - Karen Wagener
- Clinical Unit for Herd Health Management, Clinical Centre for Ruminant and Camelid Medicine, Clinical Department for Farm Animals and Food System Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Marc Drillich
- Unit for Reproduction Medicine and Udder Health, Faculty of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Aspinas Chapwanya
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre 00265, Saint Kitts and Nevis;
| |
Collapse
|
3
|
Motafeghi F, Amiri M, Noroozzadeh M, Tehrani FR. The impact of GABA and GABAergic pathway in polycystic ovary syndrome: a systematic review. Obstet Gynecol Sci 2025; 68:93-108. [PMID: 39935052 PMCID: PMC11976924 DOI: 10.5468/ogs.24255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Emerging evidence indicates that dysfunction of the gamma-aminobutyric acid (GABA)ergic pathway may contribute to the pathophysiology of polycystic ovary syndrome (PCOS), and GABA demonstrates potential in the management of PCOS symptoms. This systematic review aimed to determine the role of the GABAergic pathway in PCOS and evaluate the impact of GABA on improving the condition. Web of Science, Embase, Scopus, Cochrane, and PubMed databases were systematically searched for experimental studies, clinical trials, animal studies, and cellular investigations. The search was conducted for relevant English-language manuscripts, published up to February 2024, using keywords, such as "polycystic ovary syndrome", PCOS, "gamma-aminobutyric acid" and GABA. Quality assessment of the included studies was performed using the Cochrane Collaboration's tool and the Newcastle-Ottawa scale. The results indicate that GABAergic dysfunction adversely affects gonadotrophin-releasing hormone neuronal activity, leading to hormonal imbalances and reproductive issues. Prenatal androgen exposure and kisspeptin signaling influence GABAergic transmission to GnRH neurons, thereby linking GABA to the pathogenesis of PCOS. Additionally, GABAergic signaling affects peripheral tissues relevant to PCOS, including the immune system, gut-brain axis, and ovaries. GABA supplementation has demonstrated potential benefits in enhancing metabolic and reproductive health, such as reducing insulin resistance and modulating sex hormone levels, as supported by animal models and clinical studies involving females with PCOS. The GABAergic signaling pathway may represent a promising therapeutic target for the management of PCOS. Nevertheless, further studies are required to validate these findings and deepen our understanding of the role of GABA in the pathogenesis and treatment of PCOS.
Collapse
Affiliation(s)
- Farzaneh Motafeghi
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
| | - Mina Amiri
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
- Foundation for Research & Education Excellence, Vestavia Hills, AL,
USA
| | - Mahsa Noroozzadeh
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Molecular Biology, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran,
Iran
- Foundation for Research & Education Excellence, Vestavia Hills, AL,
USA
| |
Collapse
|
4
|
Zhang F, Liu R, Xu Z, Chen W, Lu X, Wang Y. Chaihu Shugan San and Zuogui Yin synergistically improve premature ovarian failure in a rat model via the PI3K/Akt/mTOR pathway. Transl Oncol 2025; 53:102298. [PMID: 39933394 PMCID: PMC11869595 DOI: 10.1016/j.tranon.2025.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND premature ovarian failure (POF) is a significant condition characterized by the early loss of ovarian function, often leading to infertility and other health complications. Traditional Chinese Medicine (TCM) formulations, such as Chaihu Shugan San and Zuogui Yin, have been used to address various gynecological disorders. This study aims to evaluate the therapeutic effects of Chaihu Shugan San, Zuogui Yin, and their combination on POI in a rat model and to elucidate the underlying mechanisms involving the PI3K/Akt/mTOR signaling pathway. METHODS In this study, a premature ovarian failure (POF) rat model was established using chronic unpredictable mild stress (CUMS) combined with cyclophosphamide injections. Rats were treated with Chaihu Shugan San, Zuogui Yin, or their combination. Key outcomes, including ovarian granulosa cell proliferation, follicular integrity, and hormonal levels (FSH and E2), were assessed. Histopathological changes in ovarian tissue were evaluated using hematoxylin and eosin (H&E) staining, while apoptosis was detected through immunohistochemistry and Western blot analysis of proteins such as Bcl-2, Bax, Caspase3, and Caspase9. The activity of the PI3K/Akt/mTOR signaling pathway was quantified using Western blot, and statistical significance was determined (P < 0.05). RESULTS The results showed that all three treatments significantly improved ovarian function in the POF rat model. Compared to the model group, Chaihu Shugan San, Zuogui Yin, and their combination increased granulosa cell proliferation (P < 0.01), reduced follicular atresia (P < 0.05), and normalized hormone levels (FSH: reduced by 36.5 %; E2: increased by 42.8 %, P < 0.05). Histopathological analysis confirmed preserved follicular structure and reduced apoptosis in treated groups. Western blot results revealed enhanced PI3K/Akt/mTOR pathway activity, with upregulated Bcl-2 expression (P < 0.01), downregulated Bax expression (P < 0.01), and decreased Caspase3 and Caspase9 levels (P < 0.05). Additionally, CyclinD2 and P70S6 K expression were significantly increased in treated groups, suggesting enhanced cell cycle progression and protein synthesis. CONCLUSION Chaihu Shugan San, Zuogui Yin, and their combination exhibit potential efficacy as treatments for POF in rats by enhancing granulosa cell proliferation, preserving follicular integrity, and inhibiting apoptosis. These therapeutic effects are mediated through the activation of the PI3K/Akt/mTOR signaling pathway and regulation of apoptosis-related proteins. This study highlights the importance of further exploring apoptosis-related pathways to understand the therapeutic mechanisms of these TCM formulations, providing a foundation for new clinical strategies to manage POF and related reproductive health issues.
Collapse
Affiliation(s)
- Fei Zhang
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China
| | - RuiFang Liu
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China.
| | - ZhenLing Xu
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China.
| | - WenJie Chen
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China.
| | - XiYa Lu
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China.
| | - Yang Wang
- Fujian University of Traditional Chinese Medicine, Fujian 350122, China; Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fujian 350122, China.
| |
Collapse
|
5
|
Su P, Chen C, Sun Y. Physiopathology of polycystic ovary syndrome in endocrinology, metabolism and inflammation. J Ovarian Res 2025; 18:34. [PMID: 39980043 PMCID: PMC11841159 DOI: 10.1186/s13048-025-01621-6] [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: 08/26/2024] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder characterized by elevated androgen levels, ovarian cysts, and impaired ovulation in females. This condition is closely linked with various reproductive health issues and has significant impacts on endocrine and metabolic pathways. Patients with PCOS commonly exhibit hyperandrogenaemia and insulin resistance, leading to complications such as acne, hirsutism, weight fluctuations, and metabolic disturbances, as well as an increased risk for type 2 diabetes, cardiovascular disease, and endometrial cancer. Although extensive research has identified several mechanistic aspects of PCOS, a thorough understanding of its pathophysiology remains incomplete. This review aims to provide a detailed analysis of the physiological and pathological aspects of PCOS, covering endocrine, metabolic, and inflammatory dimensions, to better elucidate its etiological framework.
Collapse
Affiliation(s)
- Pingping Su
- Wenzhou Graduate Joint Training Base, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gynecology, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Chao Chen
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yun Sun
- Department of Gynecology, Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China.
| |
Collapse
|
6
|
Gao H, Cao X, Hua H, Zhu H. The Functions and Implications of MicroRNAs in Premature Ovarian Insufficiency. Mol Genet Genomic Med 2025; 13:e70074. [PMID: 39959946 PMCID: PMC11831191 DOI: 10.1002/mgg3.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/02/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a condition in women characterized by the premature decline of ovarian function before age 40, evident through menstrual irregularities such as amenorrhea or oligomenorrhea, elevated FSH levels exceeding 25 U/L, and a progressive decrease in estrogen levels. Despite considerable research, the exact pathogenic mechanisms underlying POI remain unclear. This study focuses on the role of microRNAs (miRNAs) in the development of POI. As critical regulators of gene expression, miRNAs may play significant roles in diagnosis and the development of innovative therapeutic approaches for POI. METHODS A comprehensive literature search was conducted on PubMed for this review. We included studies published in English up to September 2024. Our search utilized a combination of the following keywords: microRNA, premature ovarian insufficiency (POI), and premature ovarian failure (POF). Articles were filtered by title and subsequently through full-text review, selecting only those pertinent to our topics of interest and their related areas. RESULTS miRNAs have emerged as critical regulators in POI, mediating a range of biological processes that contribute to the disease's progression. Their involvement offers promising insights for early diagnosis, prognostic assessments, and therapeutic interventions, highlighting their potential as biomarkers and therapeutic targets. CONCLUSION Elucidating miRNAs' roles in POI opens new avenues for managing the disease. By understanding how miRNAs influence the molecular mechanisms of POI, innovative strategies can be developed for diagnosis and treatment, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Hui Gao
- Department of Reproductive Health, Nanjing Maternity and Child Health Care HospitalThe Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical UniversityNanjingChina
| | - Xi‐Xia Cao
- Department of Reproductive Health, Nanjing Maternity and Child Health Care HospitalThe Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical UniversityNanjingChina
| | - Hua Hua
- Department of Reproductive Health, Nanjing Maternity and Child Health Care HospitalThe Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical UniversityNanjingChina
| | - Hui Zhu
- Department of Reproductive Health, Nanjing Maternity and Child Health Care HospitalThe Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
7
|
Han Y, Dai Y, Wang K, Zhang X, Shao Z, Zhu X. Post-pandemic insights on COVID-19 and premature ovarian insufficiency. Open Life Sci 2025; 20:20221028. [PMID: 39886482 PMCID: PMC11780258 DOI: 10.1515/biol-2022-1028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 02/01/2025] Open
Abstract
The COVID-19 pandemic has raised concerns regarding its potential impact on premature ovarian insufficiency (POI). This overview examines the possible interactions between COVID-19 and POI, while also suggesting preventive measures. The viral infection's inflammatory response and immune dysregulation may adversely affect ovarian tissues, leading to inflammation and damage. Additionally, alterations in vascular function could impair ovarian blood flow and hormonal imbalances may disrupt normal ovarian function. Long-term health effects, such as "long COVID," may exacerbate these issues through chronic inflammation and immune dysfunction. Public health measures, such as vaccination and home isolation, may indirectly protect ovarian health by reducing systemic inflammation. Vaccines could mitigate the severity of COVID-19's impact on ovarian function, while isolation may reduce stress and inflammation. However, further research is needed to validate these mechanisms.
Collapse
Affiliation(s)
- Yaguang Han
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yang Dai
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Kexin Wang
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xin Zhang
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Zishen Shao
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xiaolin Zhu
- Department of Medicine, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 411 Guogeli Street, Nangang District, Harbin, Heilongjiang, 150006, P.R. China
| |
Collapse
|
8
|
Zhang J, Yu X, Li W, Jiang Y, Zhang L, Wang S. Bisphenol S impairs oocyte quality by inducing gut microbiota dysbiosis. mSystems 2025; 10:e0091224. [PMID: 39704538 PMCID: PMC11748550 DOI: 10.1128/msystems.00912-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
A good quality egg is essential for a successful pregnancy and early embryo development. Oocyte development is vulnerable to environmental exposures. Bisphenol S (BPS) is widely used as a replacement for its analog bisphenol A, but the reproductive toxicity of BPS has been of great concern. In this study, we showed that BPS exposure induces dysbiosis of the gut microbiota, which further leads to intestinal permeability and inflammation, and ultimately impairs oocyte quality. More importantly, we found that alginate oligosaccharide reshapes the gut microbiota to improve gut homeostasis, thereby preventing the deleterious effects of BPS on the gut and oocytes. Overall, this study not only demonstrates that BPS exposure impairs the intestine and oocytes by inducing dysbiosis of the gut microbiota but also develops a preventive strategy. IMPORTANCE Oocyte development is vulnerable to stimulation by intrinsic and extrinsic factors, particularly many environmental pollutants and chemicals in daily life. The reproductive toxicity of bisphenol S has been of great concern, although it is widely used as a safe substitute for its analog bisphenol A. However, it is not known how bisphenol S impairs oocyte quality. This work presents the exciting finding that bisphenol S induces gut microbiota dysbiosis, which further leads to increased intestinal permeability and inflammation and ultimately damages oocytes. More importantly, we show that alginate oligosaccharide improves gut homeostasis by reshaping the gut microbiota, therefore preventing the bisphenol S-induced gut microbiota dysbiosis and gut and oocyte damage. These findings present a major advance in the understanding of bisphenol S toxicity to oocytes and also provide a preventive strategy.
Collapse
Affiliation(s)
- Jiaming Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, Jinan, China
| | - Xiaoxia Yu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, Jinan, China
| | - Weidong Li
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Yunjing Jiang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Liangran Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Shunxin Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children, and Reproductive Health, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong, China
| |
Collapse
|
9
|
Zhu Q, Du J, Li Y, Qin X, He R, Ma H, Liang X. Downregulation of glucose-energy metabolism via AMPK signaling pathway in granulosa cells of diminished ovarian reserve patients. Gene 2025; 933:148979. [PMID: 39366473 DOI: 10.1016/j.gene.2024.148979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/15/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Glucose metabolism plays a crucial role in the function of granulosa cells (GCs) and the development of follicles. In cases of diminished ovarian reserve (DOR), alterations in these processes can impact female fertility. This study aims to investigate changes in glucose-energy metabolism in GCs of young DOR patients aged 20 to 35 years and their correlation with the onset and progression of DOR. 72 DOR cases and 75 women with normal ovarian reserve (NOR) as controls were included based on the POSEIDON and Bologna criteria. Samples of GCs and follicular fluid (FF) were collected for a comprehensive analysis involving transcriptomics, metabolomics, RT-qPCR, JC-1 staining, and flow cytometry. The study identified differentially expressed genes and metabolites in GCs of DOR and NOR groups, revealing 7 common pathways related to glucose-energy metabolism, along with 11 downregulated genes and 14 metabolites. Key substances in the glucose-energy metabolism pathway, such as succinate, lactate, NADP, ATP, and ADP, showed decreased levels, with the DOR group exhibiting a reduced ADP/ATP ratio. Downregulation of genes involved in glycolysis (HK, PGK, LDH1), the TCA cycle (CS), and gluconeogenesis (PCK) was observed, along with reduced glucose content and expression of glucose transporter genes (GLUT1 and GLUT3) in DOR GCs. Additionally, decreased AMPK pathway activity and impaired mitochondrial function in DOR suggest a connection between mitochondrial dysfunction and disrupted energy metabolism. Above all, the decline in glucose-energy metabolism in DOR is closely associated with its onset and progression. Reduced glucose uptake and impaired mitochondrial function in DOR GCs lead to internal energy imbalances, hindering the AMPK signaling pathway, limiting energy production and supply, and ultimately impacting follicle development and maturation.
Collapse
Affiliation(s)
- Qinying Zhu
- Department of Obstetrics and Gynecology, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Peking Union Medical College Hospital, Beijing, China; The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Hao Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, the First Hospital of Lanzhou University, Gansu Provincial Clinical Research Center for Gynecological Oncology, Lanzhou, China.
| |
Collapse
|
10
|
Zhang Y, Hu TT, Cheng YR, Zhang ZF, Su J. Global, regional, and national burden of anxiety disorders during the perimenopause (1990-2021) and projections to 2035. BMC Womens Health 2025; 25:11. [PMID: 39773442 PMCID: PMC11706191 DOI: 10.1186/s12905-025-03547-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
PURPOSE Perimenopause is associated with an increased risk of anxiety disorders, largely due to hormonal changes affecting the body's regulatory feedback mechanisms. This study aims to provide a comprehensive analysis of the global burden of anxiety disorders among perimenopausal women. METHODS Data from the 2021 Global Burden of Disease (GBD) database were utilized to assess disability-adjusted life years associated with anxiety disorders linked to perimenopause. We calculated trends using the estimated average percent change, and future projections were made using the Bayesian age-period-cohort model to estimate disability-adjusted life year trends for anxiety disorders from 2022 to 2035. RESULTS Between 1990 and 2021, the global age-standardized disability-adjusted life year rate for anxiety disorders among perimenopausal women increased from 625.51 (95% uncertainty interval: 429.1-891.09) to 677.15 (95% uncertainty interval: 469.45-952.72), indicating a rising trend with an estimated average percent change of 0.081 (95% confidence interval: 0.0043-0.143). Regional differences were noted, with anxiety disorder burdens varying across areas with different sociodemographic index levels. Projections suggest that by 2035, the global burden of anxiety disorders in perimenopausal women will rise to 1,180.43 per 100,000, a 40.67% increase compared with 2021 levels. CONCLUSION The burden of anxiety disorders during perimenopause is a growing global concern, with a significant increase anticipated in the coming years. Targeted prevention and intervention strategies are urgently needed to mitigate this rising burden and improve mental health outcomes during perimenopause.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Psychosomatic Diseases (II), Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Ting-Ting Hu
- Laboratory Department, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, 310008, China
| | - Yong-Ran Cheng
- School of Public Health, Hangzhou Medical College, Hangzhou, 311300, China
| | - Zhi-Fen Zhang
- Gynecological Endocrinology Department, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, 310008, China.
| | - Jun Su
- School of Public Health, Hangzhou Normal University, Hangzhou, 311121, China.
- Intensive Care Medicine Department, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, 310008, China.
| |
Collapse
|
11
|
Wilson MA, Lee KMN, Ehrlich DE, Rogers‐LaVanne MP, Jasienska G, Galbarczyk A, Clancy KBH. Cycle Effects Are Not Universal: A Case Study of Urinary C-Reactive Protein Concentrations in Rural Polish and Polish American Samples. Am J Hum Biol 2025; 37:e24207. [PMID: 39794910 PMCID: PMC11724170 DOI: 10.1002/ajhb.24207] [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: 06/17/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 01/13/2025] Open
Abstract
OBJECTIVES We need to better understand how the menstrual cycle interacts with other biological systems, such the inflammation and immune response. One way to study this interaction is through C-reactive protein (CRP). Studies of CRP concentrations across the menstrual cycle have been inconsistent. This study explores menstrual cycle CRP variation in two geographically different samples of Polish and Polish American individuals. METHODS Analyses were conducted on 76 Polish and 22 Polish American daily urine samples collected on the first day of menstruation until the start of their next period. CRP, estrone-3-glucuronide, and pregnanediol-3-glucuronide were assayed. Sample-specific linear mixed models were used to examine cycle effects and median CRP concentrations across cycle phases and between the start of menses and remainder of the cycle were compared using Kruskal-Wallace and Dunn tests. RESULTS Polish and Polish American samples had distinct menstrual cycle CRP phenotypes. The Polish sample did not show cycle effects. The Polish American LMM demonstrated that CRP decreases after the first 3 days of menses (estimate -0.17, t-value -5.2). The KW and Dunn tests supported this. CRP concentrations were higher during the early follicular (median 0.406, p < 0.05), specifically the first 3 days of menstruation (median 0.466, p < 0.01), and lower in the luteal (median 0.277, p < 0.05). CONCLUSIONS Results suggest that changes in CRP during menstrual cycle are not universal across populations. In the Polish American sample, CRP was highest during the early follicular, specifically the first 3 days of menstruation, suggesting a potential relationship between the menstrual cycle and inflammation.
Collapse
Affiliation(s)
- M. A. Wilson
- Department of AnthropologyUniversity of IllinoisUrbana‐ChampaignIllinoisUSA
| | - K. M. N. Lee
- Department of AnthropologyTulane UniversityNew OrleansLouisianaUSA
| | - D. E. Ehrlich
- Institute for Social Research and Data InnovationUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - M. P. Rogers‐LaVanne
- Carl R. Woese Institute for Genomic BiologyUniversity of IllinoisUrbana‐ChampaignIllinoisUSA
| | - G. Jasienska
- Department of Environmental Health, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
| | - A. Galbarczyk
- Department of Environmental Health, Faculty of Health SciencesJagiellonian University Medical CollegeKrakowPoland
- Department of Human Behavior, Ecology and CultureMax Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - K. B. H. Clancy
- Department of AnthropologyUniversity of IllinoisUrbana‐ChampaignIllinoisUSA
| |
Collapse
|
12
|
Pan B, Chai J, Fei K, Zheng T, Jiang Y. Dynamic changes in the transcriptome and metabolome of pig ovaries across developmental stages and gestation. BMC Genomics 2024; 25:1193. [PMID: 39695358 DOI: 10.1186/s12864-024-11122-3] [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: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The ovary is a central organ in the reproductive system that produces oocytes and synthesizes and secretes steroid hormones. Healthy development and regular cyclical change in the ovary is crucial for regulating reproductive processes. However, the key genes and metabolites that regulate ovarian development and pregnancy have not been fully elucidated. This study conducted high-throughput RNA sequencing and untargeted metabolite profiling of the ovarian tissues from Chenghua pigs at four stages, including postnatal day 3 (D3), puberty at the age of about 125 days (Pub), sexual maturity at the age of about 365 days (Y1), and 105 days after pregnancy at the age of about 360 days (Pre). RESULTS A total of 9,264 and 1,593 differentially expressed genes (DEGs) were identified during ovarian development and pregnancy. Several key genes involved in ovarian development, including SQLE, HMGCS1, MSMO1, SCARB1, CYP11A1, HSD3B1, HSD17B1, and SERPINE1 were identified. Similarly, LUM, FN1, PLAUR, SELP, SDC1, and VCAN were considered to be associated with pregnancy maintenance. Overexpression of HSD17B1 in granulosa cells significantly upregulated estrogen synthesis-related genes (HSD3B1, CYP11A1, and STAR); meanwhile, overexpression of PLAUR promotes granulosa cell proliferation. Furthermore, 66, 24, 77, and 7 differentially expressed miRNAs (DEMis) were found, leading to the selection of key miRNAs such as ssc-miR-206, ssc-miR-107, ssc-miR-429, ssc-miR-210, and ssc-miR-133a-3p by differential miRNA-targeted mRNA interaction network; meanwhile, ssc-miR-133a-3p was validated to have a targeting relationship with KCNA1 by dual-luciferase reporter systems assay. At the metabolic levels, androstenedione, 17a-hydroxyprogesterone, dehydroepiandrosterone, and progesterone were identified, with their synthesis regulated by these DEGs in the ovarian steroidogenesis pathway. Furthermore, treatment of cells with androstenedione upregulated the expression of HSD3B1, CYP11A1, and STAR. CONCLUSIONS This study revealed the dynamic changes in the transcriptome and metabolome of pig ovaries across developmental stages and gestation, indicating that it may provide new theoretical insights for improving sow fertility.
Collapse
Affiliation(s)
- Binyun Pan
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China
| | - Jin Chai
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China
| | - Kaixin Fei
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China
| | - Ting Zheng
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China
| | - Yanzhi Jiang
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, 625014, China.
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, , Chengdu, Sichuan, 611130, China.
| |
Collapse
|
13
|
Lakshmanan M, Saini M, Nune M. Exploring the innovative application of cerium oxide nanoparticles for addressing oxidative stress in ovarian tissue regeneration. J Ovarian Res 2024; 17:241. [PMID: 39633503 PMCID: PMC11619646 DOI: 10.1186/s13048-024-01566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024] Open
Abstract
The female reproductive system dysfunction considerably affects the overall health of women and children on a global scale. Over the decade, the incidence of reproductive disorders has become a significant source of suffering for women. Infertility in women may be caused by a range of acquired and congenital abnormalities. Ovaries play a central role in the female reproductive function. Any defect in the normal functioning of these endocrine organs causes health issues and reproductive challenges extending beyond infertility, as the hormones interact with other tissues and biological processes in the body. The complex pathophysiology of ovarian disorders makes it a multifactorial disease. The key etiological factors associated with the diseases include genetic factors, hormonal imbalance, environmental and lifestyle factors, inflammatory conditions, oxidative stress, autoimmune diseases, metabolic factors, and age. Oxidative stress is a major contributor to disease development and progression affecting the oocyte quality, fertilization, embryo development, and implantation. The choice of treatment for ovarian disorders varies among individuals and has associated complications. Reproductive tissue engineering holds great promise for overcoming the challenges associated with the current therapeutic approach to tissue regeneration. Furthermore, incorporating nanotechnology into tissue engineering could offer an efficient treatment strategy. This review provides an overview of incorporating antioxidant nanomaterials for engineering ovarian tissue to address the disease recurrence and associated pathophysiology. Cerium oxide nanoparticles (CeO2 NPs) are prioritized for evaluation primarily due to their antioxidant properties. In conclusion, the review explores the potential applications of CeO2 NPs for effective and clinically significant ovarian tissue regeneration.
Collapse
Affiliation(s)
- Maya Lakshmanan
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Monika Saini
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029, India
| | - Manasa Nune
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
14
|
SantaBarbara K, Helms E, Harris N. The associations between calorie tracking, body image dissatisfaction, eating disorders, and menstrual cycle characteristics in resistance-trained athletes. J Int Soc Sports Nutr 2024; 21:2433743. [PMID: 39579199 PMCID: PMC11587730 DOI: 10.1080/15502783.2024.2433743] [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: 10/27/2023] [Accepted: 11/11/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND While body image dissatisfaction (BID) and eating disorders (EDs) are relatively common in athletes (ranging from 11% to 67% of athletes, depending on the sport) [1], they are also prevalent in weight-class restricted sports (a common format in strength sports), and among physique athletes [2]. These athletes manipulate their nutrition to reach aesthetic or body weight standards and, in that process, may undergo prolonged periods of low energy availability. Low energy availability, defined as consuming insufficient energy for one's lean mass and exercise activity, can lead to Relative Energy Deficiency in sport (REDs), a syndrome that can impact menstrual cycle (MC) symptoms (and many other aspects of physiology and psychology) [3]. There has not been an investigation into the relationships between these resistance-trained (RT) athletes' nutritional habits, MC-related symptoms, BIDs, and EDs. METHODS A survey was implemented to explore the dieting habits, MC characteristics, BID, and EDs in RT females. RESULTS 64.6% (n = 469) of participants reported tracking calories, with a slightly higher percentage of competitive athletes tracking calories 71.8% (n = 181) than recreational-level athletes. Competitive athletes were significantly more likely to track calories than recreational-level athletes (p = 0.003). When asked what the primary purpose of calorie restriction was, most participants selected weight loss for aesthetic purposes 58.8% (n = 356). Competitive athletes were less likely to select weight loss for aesthetic purposes 35.7% (n = 77), but weight loss for the purpose of a weight class-based sport was higher at 43.5% (n = 94). There were no significant associations between BID and MC characteristics or most MC symptoms and limited associations between EDs and MC characteristics and symptoms. CONCLUSION RT athletes exhibited a higher prevalence of calorie tracking than the general population. Competitive RT athletes were less likely to calorie restrict for aesthetic purposes than non-athletes, but more likely to calorie restrict for the purpose of weight-class-based sports. There were limited significant associations between BID and MC characteristics or MC symptoms, as well as between EDs and MC characteristics. However, there was a significant association between amenorrhea and EDs, which aligns with previous research in this area. Both BID and EDs were significantly associated with MC-based mental health effects; this is likely due to the interconnected nature of mental health concerns, such as EDs with depression and anxiety.
Collapse
Affiliation(s)
- Kimberly SantaBarbara
- Auckland University of Technology, Sport Performance Research Institute New Zealand (SPRINZ), Auckland, New Zealand
- California State Polytechnic University, Department of Kinesiology and Health Promotion, Pomona, CA, USA
| | - Eric Helms
- Auckland University of Technology, Sport Performance Research Institute New Zealand (SPRINZ), Auckland, New Zealand
- Florida Atlantic University, Department of Exercise Science and Health Promotion, Muscle Physiology Laboratory, Boca Raton, FL, USA
| | - Nigel Harris
- Auckland University of Technology, Human Potential Centre, Auckland, New Zealand
| |
Collapse
|
15
|
Liu Q, Lin Y, Zhang W. Psychological stress dysfunction in women with premenstrual syndrome. Heliyon 2024; 10:e40233. [PMID: 39748962 PMCID: PMC11693916 DOI: 10.1016/j.heliyon.2024.e40233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 01/04/2025] Open
Abstract
Premenstrual syndrome (PMS) encompasses a range of emotional, physiological, and behavioral symptoms that occur during the luteal phase of the menstrual cycle (MC) and resolve with the onset of menstruation. These symptoms, which can include fatigue, physical pain, anxiety, irritability, and depression, significantly affect women's daily lives and overall well-being. In severe cases, PMS can progress to premenstrual dysphoric disorder (PMDD), profoundly impairing quality of life. Despite its prevalence, the neural mechanisms underlying PMS-particularly those related to stress-are not fully understood.This review aims to explore the complex interactions between PMS and stress, with a focus on the hormonal pathways involved. We propose that abnormal stress coping styles and stress reactivity patterns, collectively referred to as stress dysfunction, are crucial factors influencing women's vulnerability to PMS. We examine the relationship between PMS and stress from four perspectives: (1) PMS shares neuroendocrine metabolic circuits based on hormonal fluctuations with stress reactivity systems; (2) there is comorbidity between PMS and stress-related disorders; (3) PMS itself may act as a stressor, potentially creating a negative feedback loop that exacerbates symptoms; and (4) biofeedback training used for stress disorders may be effective in treating PMS. By providing a detailed analysis of stress-related hormonal changes and their effects on PMS, this review offers new insights into the physiological processes underlying PMS. Understanding these interactions may inform the development of targeted interventions and improve the quality of life for women affected by PMS.
Collapse
Affiliation(s)
- Qing Liu
- College of Education, Zhejiang University of Technology, Hangzhou, China
| | - Yuhang Lin
- College of Education, Zhejiang University of Technology, Hangzhou, China
| | - Wenjuan Zhang
- Mental Health Education Center, Xidian University, Xi'an, China
| |
Collapse
|
16
|
Mashhadi Meighani E, Yahyaei A, Ghaffari F. Fertility Preservation in Female Patients with Cancer. Part I: Challenges and Future Prospects in Developing Countries; A Narrative Review Study. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2024; 18:293-304. [PMID: 39564819 PMCID: PMC11589969 DOI: 10.22074/ijfs.2024.2015626.1569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/18/2024] [Accepted: 05/20/2024] [Indexed: 11/21/2024]
Abstract
While international guidelines on oncofertility practice have been published in developed countries, there is limited information available on oncofertility practice in developing countries, which often face limitations in their cancer health support networks. As survival rates improve in the field of cancer and other diseases, there is a growing need for stronger oncofertility services in developing countries. Given that 50% of cancer patients are under the age of 65, many of these patients are of reproductive age and preserving their fertility health prior to starting treatment is valuable. However, due to resource limitations and the heavy burden of treatment costs imposed on these patients and their families, fertility preservation (FP) may not be a top priority for them. As a result, many healthcare providers and their patients focus on eliminating cancer and its related treatments while overlooking the importance of fertility as a factor that can significantly impact their future quality of life. In this review, we have tried to increase the knowledge of healthcare providers involved in the oncofertility network by reviewing current cancer treatments, FP options in female cancer patients, and their challenges in developing countries.
Collapse
Affiliation(s)
- Esmat Mashhadi Meighani
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Azar Yahyaei
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Firouzeh Ghaffari
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Emails: ,
| |
Collapse
|
17
|
Naveed M, Hill JW. The Underlying Effect of Urate Levels on Female Infertility. Metabolites 2024; 14:564. [PMID: 39452945 PMCID: PMC11509475 DOI: 10.3390/metabo14100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Female infertility is a complex and multifaceted condition that affects millions of women globally [...].
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA;
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, School of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA;
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
18
|
Leng D, Zeng B, Wang T, Chen BL, Li DY, Li ZJ. Single nucleus/cell RNA-seq of the chicken hypothalamic-pituitary-ovarian axis offers new insights into the molecular regulatory mechanisms of ovarian development. Zool Res 2024; 45:1088-1107. [PMID: 39245652 PMCID: PMC11491784 DOI: 10.24272/j.issn.2095-8137.2024.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-ovarian (HPO) axis represents a central neuroendocrine network essential for reproductive function. Despite its critical role, the intrinsic heterogeneity within the HPO axis across vertebrates and the complex intercellular interactions remain poorly defined. This study provides the first comprehensive, unbiased, cell type-specific molecular profiling of all three components of the HPO axis in adult Lohmann layers and Liangshan Yanying chickens. Within the hypothalamus, pituitary, and ovary, seven, 12, and 13 distinct cell types were identified, respectively. Results indicated that the pituitary adenylate cyclase activating polypeptide (PACAP), follicle-stimulating hormone (FSH), and prolactin (PRL) signaling pathways may modulate the synthesis and secretion of gonadotropin-releasing hormone (GnRH), FSH, and luteinizing hormone (LH) within the hypothalamus and pituitary. In the ovary, interactions between granulosa cells and oocytes involved the KIT, CD99, LIFR, FN1, and ANGPTL signaling pathways, which collectively regulate follicular maturation. The SEMA4 signaling pathway emerged as a critical mediator across all three tissues of the HPO axis. Additionally, gene expression analysis revealed that relaxin 3 (RLN3), gastrin-releasing peptide (GRP), and cocaine- and amphetamine regulated transcripts (CART, also known as CARTPT) may function as novel endocrine hormones, influencing the HPO axis through autocrine, paracrine, and endocrine pathways. Comparative analyses between Lohmann layers and Liangshan Yanying chickens demonstrated higher expression levels of GRP, RLN3, CARTPT, LHCGR, FSHR, and GRPR in the ovaries of Lohmann layers, potentially contributing to their superior reproductive performance. In conclusion, this study provides a detailed molecular characterization of the HPO axis, offering novel insights into the regulatory mechanisms underlying reproductive biology.
Collapse
Affiliation(s)
- Dong Leng
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Bo Zeng
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China
| | - Bin-Long Chen
- College of Animal Science, Xichang University, Xichang, Sichuan 615000, China. E-mail:
| | - Di-Yan Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan 610106, China. E-mail:
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, Henan 450046, China. E-mail:
| |
Collapse
|
19
|
Caserta S, Cancemi G, Murdaca G, Stagno F, Di Gioacchino M, Gangemi S, Allegra A. The Effects of Cancer Immunotherapy on Fertility: Focus on Hematological Malignancies. Biomedicines 2024; 12:2106. [PMID: 39335619 PMCID: PMC11428457 DOI: 10.3390/biomedicines12092106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/07/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
In recent years, cancer management has benefitted from new effective treatments, including immunotherapy. While these therapies improve cancer survival rates, they can alter immune responses and cause long-term side effects, of which gonadotoxic effects and the potential impact on male and female fertility are growing concerns. Immunotherapies, such as immune checkpoint inhibitors, immunomodulators, monoclonal antibodies, and CAR-T, can lead to elevated levels of proinflammatory cytokines and immune-related adverse events that may exacerbate fertility problems. Immunotherapy-related inflammation, characterized by cytokine imbalances and the activation of pathways such as AMPK/mTOR, has been implicated in the mechanisms of fertility impairment. In men, hypospermatogenesis and aspermatogenesis have been observed after treatment with immune checkpoint inhibitors, by direct effects on the gonads, particularly through the inhibition of cytotoxic T lymphocyte antigen-4. In women, both damage to ovarian reserves, recurrent pregnancy loss, and implantation failure have been documented, secondary to a complex interplay between immune cells, such as T cells and uterine NK cells. In this review, the impact of immunotherapy on fertility in patients with hematological cancers was analyzed. While this area is still underexplored, fertility preservation methods remain crucial. Future studies should investigate immunotherapy's effects on fertility and establish standardized preservation protocols.
Collapse
Affiliation(s)
- Santino Caserta
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.C.); (F.S.); (A.A.)
| | - Gabriella Cancemi
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.C.); (F.S.); (A.A.)
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genova, 16126 Genova, Italy
- Allergology and Clinical Immunology, San Bartolomeo Hospital, 19038 Sarzana, Italy
| | - Fabio Stagno
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.C.); (F.S.); (A.A.)
| | - Mario Di Gioacchino
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy;
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, via Consolare Valeria, 98125 Messina, Italy; (S.C.); (G.C.); (F.S.); (A.A.)
| |
Collapse
|
20
|
Li X, Lin S, Yang X, Chen C, Cao S, Zhang Q, Ma J, Zhu G, Zhang Q, Fang Q, Zheng C, Liang W, Wu X. When IGF-1 Meets Metabolic Inflammation and Polycystic Ovary Syndrome. Int Immunopharmacol 2024; 138:112529. [PMID: 38941670 DOI: 10.1016/j.intimp.2024.112529] [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: 04/12/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder associated with insulin resistance (IR) and hyperandrogenaemia (HA). Metabolic inflammation (MI), characterized by a chronic low-grade inflammatory state, is intimately linked with chronic metabolic diseases such as IR and diabetes and is also considered an essential factor in the development of PCOS. Insulin-like growth factor 1 (IGF-1) plays an essential role in PCOS pathogenesis through its multiple functions in regulating cell proliferation metabolic processes and reducing inflammatory responses. This review summarizes the molecular mechanisms by which IGF-1, via MI, participates in the onset and progression of PCOS, aiming to provide insights for studies and clinical treatment of PCOS.
Collapse
Affiliation(s)
- Xiushen Li
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China; Department of Traditional Chinese Medicine, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Sailing Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Xiaolu Yang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Can Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Shu Cao
- Xin'an Academy, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Jingxin Ma
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Guli Zhu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qi Zhang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Qiongfang Fang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China.
| | - Xueqing Wu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China; Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
21
|
Ru M, Liang H, Ruan J, Haji RA, Cui Y, Yin C, Wei Q, Huang J. Chicken ovarian follicular atresia: interaction network at organic, cellular, and molecular levels. Poult Sci 2024; 103:103893. [PMID: 38870615 PMCID: PMC11225904 DOI: 10.1016/j.psj.2024.103893] [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: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Most of follicles undergo a degenerative process called follicular atresia. This process directly affects the egg production of laying hens and is regulated by external and internal factors. External factors primarily include nutrition and environmental factors. In follicular atresia, internal factors are predominantly regulated at 3 levels; organic, cellular and molecular levels. At the organic level, the hypothalamic-pituitary-ovary (HPO) axis plays an essential role in controlling follicular development. At the cellular level, gonadotropins and cytokines, as well as estrogens, bind to their receptors and activate different signaling pathways, thereby suppressing follicular atresia. By contrast, oxidative stress induces follicular atresia by increasing ROS levels. At the molecular level, granulosa cell (GC) apoptosis is not the only factor triggering follicular atresia. Autophagy is also known to give rise to atresia. Epigenetics also plays a pivotal role in regulating gene expression in processes that seem to be related to follicular atresia, such as apoptosis, autophagy, proliferation, and steroidogenesis. Among these processes, the miRNA regulation mechanism is well-studied. The current review focuses on factors that regulate follicular atresia at organic, cellular and molecular levels and evaluates the interaction network among these levels. Additionally, this review summarizes atretic follicle characteristics, in vitro modeling methods, and factors preventing follicular atresia in laying hens.
Collapse
Affiliation(s)
- Meng Ru
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Haiping Liang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jiming Ruan
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Ramlat Ali Haji
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Yong Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Chao Yin
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Qing Wei
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China
| | - Jianzhen Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Economic and Technological Development District, Nanchang 330045, China.
| |
Collapse
|
22
|
Deng ZM, Dai FF, Wang RQ, Deng HB, Yin TL, Cheng YX, Chen GT. Organ-on-a-chip: future of female reproductive pathophysiological models. J Nanobiotechnology 2024; 22:455. [PMID: 39085921 PMCID: PMC11290169 DOI: 10.1186/s12951-024-02651-w] [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: 08/02/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
The female reproductive system comprises the internal and external genitalia, which communicate through intricate endocrine pathways. Besides secreting hormones that maintain the female secondary sexual characteristics, it also produces follicles and offspring. However, the in vitro systems have been very limited in recapitulating the specific anatomy and pathophysiology of women. Organ-on-a-chip technology, based on microfluidics, can better simulate the cellular microenvironment in vivo, opening a new field for the basic and clinical research of female reproductive system diseases. This technology can not only reconstruct the organ structure but also emulate the organ function as much as possible. The precisely controlled fluidic microenvironment provided by microfluidics vividly mimics the complex endocrine hormone crosstalk among various organs of the female reproductive system, making it a powerful preclinical tool and the future of pathophysiological models of the female reproductive system. Here, we review the research on the application of organ-on-a-chip platforms in the female reproductive systems, focusing on the latest progress in developing models that reproduce the physiological functions or disease features of female reproductive organs and tissues, and highlighting the challenges and future directions in this field.
Collapse
Affiliation(s)
- Zhi-Min Deng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Fang-Fang Dai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui-Qi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hong-Bing Deng
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan, Hubei, 430060, China
| | - Tai-Lang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
23
|
Zheng B, Hu X, Hu Y, Dong S, Xiao X, Qi H, Wang Y, Wang W, Wang Z. Type III adenylyl cyclase is essential for follicular development in female mice and their reproductive lifespan. iScience 2024; 27:110293. [PMID: 39050703 PMCID: PMC11267094 DOI: 10.1016/j.isci.2024.110293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/15/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
Premature ovarian failure (POF) is a complex and heterogeneous disease that causes infertility and subfertility. However, the molecular mechanism of POF has not been fully elucidated. Here, we show that the loss of adenylyl cyclase III (Adcy3) in female mice leads to POF and a shortened reproductive lifespan. We found that Adcy3 is abundantly expressed in mouse oocytes. Adcy3 knockout mice exhibited the excessive activation of primordial follicles, progressive follicle loss, follicular atresia, and ultimately POF. Mechanistically, we found that mitochondrial oxidative stress in oocytes significantly increased with age in Adcy3-deficient mice and was accompanied by oocyte apoptosis and defective folliculogenesis. In contrast, compared with wild-type female mice, humanized ADCY3 knock-in female mice exhibited improved fertility with age. Collectively, these results reveal that the previously unrecognized Adcy3 signaling pathway is tightly linked to female ovarian aging, providing potential pharmaceutical targets for preventing and treating POF.
Collapse
Affiliation(s)
- Baofang Zheng
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Xiaoyu Hu
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
- Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding 071002, China
| | - Yuanhui Hu
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Sheng Dong
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Xin Xiao
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Haoming Qi
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Yongdi Wang
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Weina Wang
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
| | - Zhenshan Wang
- School of Life Sciences, Institute of Life Science, Hebei University, Baoding 071002, China
- Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding 071002, China
| |
Collapse
|
24
|
Jiao J, Hao J, Hou L, Luo Z, Shan S, Ding Y, Ma L, Huang Y, Ying Q, Wang F, Zhou J, Ning Y, Song P, Xu L. Age at natural menopause and associated factors with early and late menopause among Chinese women in Zhejiang province: A cross-sectional study. PLoS One 2024; 19:e0307402. [PMID: 39012896 PMCID: PMC11251616 DOI: 10.1371/journal.pone.0307402] [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: 02/26/2024] [Accepted: 06/29/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVES Menopause is a significant life transition for women, impacting their physical and psychological health. The age at natural menopause (ANM) and its associated factors have differed by race and region. This study aimed to investigate ANM and associated factors of early and late menopause among Chinese women in Zhejiang province. METHODS A cross-sectional study was conducted using a multi-stage stratified cluster sampling method to recruit 8,006 women aged 40-69 years who had resided in Zhejiang province for over 6 months between July 2019 and December 2021. Self-reported ANM and sociodemographics, lifestyle behaviors, reproductive history, and health-related factors were collected using questionnaires in face-to-face surveys. ANM were categorized into three groups: early menopause (<45 years), normal menopause (45-54 years), and late menopause (≥55 years). Kaplan-Meier survival analysis was utilized to calculate the median ANM. Multivariable multinomial logistic regression was employed to explore the associated factors of early menopause and late menopause. RESULTS A total of 6,047 women aged 40-69 years were included for survival analysis, with 3,176 of them for the regression analysis. The overall median ANM was 51 years (Inter-quartile range [IQR]: 51-52). Women who were smokers (odds ratio [OR]:4.54, 95% confidence interval [CI]:1.6-12.84), had irregular menstrual cycles (OR:1.78, 95% CI:1.12-2.83) and hypertension (OR:1.55, 95% CI:1.09-2.21) had a higher odds ratio of early menopause, while central obesity (OR:1.33, 95% CI:1.03-1.73) and hyperlipidemia (OR:1.51, 95% CI:1.04-2.18) were factors associated with late menopause. CONCLUSIONS This study revealed the associations between ANM and various factors among Chinese women. These factors included socio-demographic factors such as age; life behavior factors like current or prior smoking status; reproductive history factors such as irregular menstrual cycles, miscarriages, and breastfeeding; and health-related factors like central adiposity, hypertension, and hyperlipidemia. These findings provided a basis for understanding factors associated with ANM.
Collapse
Affiliation(s)
- Jie Jiao
- Zhejiang Maternal, Child and Reproductive Health Center, Hangzhou, Zhejiang, China
| | - Jiajun Hao
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Leying Hou
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zeyu Luo
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shiyi Shan
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuehong Ding
- Zhejiang Maternal, Child and Reproductive Health Center, Hangzhou, Zhejiang, China
| | - Linjuan Ma
- Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yizhou Huang
- Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian Ying
- Institute of Basic Medicine and Cancer (IBMC), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Feixue Wang
- Zhejiang Maternal, Child and Reproductive Health Center, Hangzhou, Zhejiang, China
| | - Jianhong Zhou
- Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yumei Ning
- Zhejiang Maternal, Child and Reproductive Health Center, Hangzhou, Zhejiang, China
| | - Peige Song
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ling Xu
- Zhejiang Maternal, Child and Reproductive Health Center, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Ruiz-González D, Cavero-Redondo I, Hernández-Martínez A, Baena-Raya A, Martínez-Forte S, Altmäe S, Fernández-Alonso AM, Soriano-Maldonado A. Comparative efficacy of exercise, diet and/or pharmacological interventions on BMI, ovulation, and hormonal profile in reproductive-aged women with overweight or obesity: a systematic review and network meta-analysis. Hum Reprod Update 2024; 30:472-487. [PMID: 38627233 PMCID: PMC11215161 DOI: 10.1093/humupd/dmae008] [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: 05/17/2023] [Revised: 03/01/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND The increasing prevalence of obesity worldwide poses a significant threat to reproductive function owing, in part, to hormonal disturbances caused by negative feedback between excess adiposity and the hypothalamic-pituitary-ovarian axis. Consequently, finding the most appropriate strategies to lose weight and improve ovulation in women with overweight or obesity is a clinically relevant matter that needs to be investigated. A comprehensive comparison of the independent and combined efficacy of lifestyle and/or pharmacological interventions on BMI, ovulation, and hormonal profile in women with overweight or obesity at risk of anovulatory infertility would facilitate improving fertility strategies in this population. OBJECTIVE AND RATIONALE This study aimed to evaluate the comparative efficacy of exercise, diet, and pharmacological interventions on BMI, ovulation, and hormonal profile in reproductive-aged women with overweight or obesity. SEARCH METHODS A systematic review was performed by searching PubMed, Scopus, Web of Science, PsycINFO, and Cochrane Library up to 14 December 2023, for randomized controlled trials assessing the effects of exercise, diet and/or pharmacological interventions (i.e. weight-lowering drugs or ovulation inducers) on BMI, ovulation, and/or hormonal profile in reproductive-aged women with overweight or obesity. We performed frequentist random-effect network meta-analyses and rated the certainty of the evidence. The primary outcomes were BMI and ovulation rate, and the secondary outcomes were serum reproductive hormone levels (gonadotrophins, androgens, or oestrogens). We performed sensitivity analyses, including the studies that only involved women with PCOS. OUTCOMES Among 1190 records screened, 148 full texts were assessed for eligibility resulting in 95 trials (9910 women), of which 53% presented a high or unclear risk of bias. The network meta-analyses revealed that, compared to control: diet combined with weight-lowering drugs (mean difference (MD) -2.61 kg/m2; 95% CI -3.04 to -2.19; τ2 = 0.22) and adding exercise (MD -2.35 kg/m2; 95% CI -2.81 to -1.89; τ2 = 0.22) led to the greatest decrease in BMI; exercise combined with diet and ovulation inducers (risk ratio (RR) 7.15; 95% CI 1.94-26.40; τ2 = 0.07) and exercise combined with diet and weight-lowering drugs (RR 4.80; 95% CI 1.67-13.84; τ2 = 0.07) produced the highest increase in ovulation rate; and exercise combined with diet and weight-lowering drugs was the most effective strategy in reducing testosterone levels (standardized mean difference (SMD) -2.91; 95% CI -4.07 to -1.74; τ2 = 2.25), the third most effective strategy in increasing sex hormone-binding globulin levels (SMD 2.37; 95% CI 0.99-3.76; τ2 = 2.48), and it was coupled with being ranked first in terms of free androgen index reduction (SMD -1.59; 95% CI -3.18 to 0.01; τ2 = 1.91). The surface under the cumulative ranking curve scores suggested that: diet combined with weight-lowering drugs is the strategy most likely (94%) to produce the highest BMI reduction; and exercise combined with diet and ovulation inducers is the strategy most likely (89%) to produce the highest ovulation rate improvement. The sensitivity analyses, which exclusively included studies involving women diagnosed with PCOS, were consistent with the results presented above. WIDER IMPLICATIONS Overall, the findings of this network meta-analysis indicate that the combination of exercise, diet, and pharmacological interventions is effective for weight loss, improving ovulation, and normalizing the androgen levels of women with overweight or obesity. Although higher quality studies are needed, these results support that the optimal treatment strategy for women with overweight or obesity wishing to conceive must consider exercise, diet, and pharmacological interventions during the shared decision-making process.
Collapse
Affiliation(s)
- David Ruiz-González
- Department of Education, Faculty of Education Sciences, and SPORT Research Group (CTS-1024), CIBIS (Centro de Investigación para el Bienestar y la Inclusión Social) Research Center, University of Almería, Almería, Spain
| | - Iván Cavero-Redondo
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Alba Hernández-Martínez
- Department of Education, Faculty of Education Sciences, and SPORT Research Group (CTS-1024), CIBIS (Centro de Investigación para el Bienestar y la Inclusión Social) Research Center, University of Almería, Almería, Spain
| | - Andrés Baena-Raya
- Department of Education, Faculty of Education Sciences, and SPORT Research Group (CTS-1024), CIBIS (Centro de Investigación para el Bienestar y la Inclusión Social) Research Center, University of Almería, Almería, Spain
| | - Sonia Martínez-Forte
- Obstetrics and Gynaecology Unit, Torrecárdenas University Hospital, Almería, Spain
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | | | - Alberto Soriano-Maldonado
- Department of Education, Faculty of Education Sciences, and SPORT Research Group (CTS-1024), CIBIS (Centro de Investigación para el Bienestar y la Inclusión Social) Research Center, University of Almería, Almería, Spain
| |
Collapse
|
26
|
Jiang L, Liu X, Deng F, Wang Y, Fan Q. Edible bird's nest improves the premature ovarian failure induced by tripterygium glycosides. Food Sci Nutr 2024; 12:4713-4722. [PMID: 39055185 PMCID: PMC11266920 DOI: 10.1002/fsn3.4119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/03/2024] [Accepted: 03/09/2024] [Indexed: 07/27/2024] Open
Abstract
Premature ovarian failure (POF) is a common disease in the field of gynecological endocrinology that seriously affects the physical and mental health of patients. Previous studies found that edible bird's nest (EBN) could improve uterine function. These suggested that EBN might also have an ameliorating effect on POF. Therefore, in this study, tripterygium glycosides (TGs) were used to induce POF in rats, and the effect of EBN on the improvement of POF was investigated. After the administration of EBN for 14 days, ovarian index and uterine index, serum hormone levels, apoptosis rate of ovarian granulosa cells, follicle-stimulating hormone receptor (FSHR) protein expression level, and the histopathological examination of the ovaries were determined. It was found that administration of medium and high EBN dose groups increased the ovarian index and granular layer thickness of rats with POF. Particularly, higher follicle-stimulating hormone levels and lower corpus luteum content were observed in the high EBN dose group. In addition, there were lower luteinizing hormone levels and fewer atretic follicles but higher progesterone levels in the medium EBN dose group. These results indicated that EBN had preventive and curative effects on POF induced by TGs. Its mechanism of action might be related to the reduction of ovarian granulosa cell apoptosis, regulation of hormones and receptors, and inhibition of follicle closure.
Collapse
Affiliation(s)
- Lin Jiang
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhouChina
| | - Xuncai Liu
- Bird's Nest Research Institute, Xiamen Yan Palace Seelong Biotechnology Co., Ltd.XiamenChina
| | - Fenghong Deng
- Bird's Nest Research Institute, Xiamen Yan Palace Seelong Biotechnology Co., Ltd.XiamenChina
| | - Yaxin Wang
- Bird's Nest Research Institute, Xiamen Yan Palace Seelong Biotechnology Co., Ltd.XiamenChina
| | - Qunyan Fan
- Bird's Nest Research Institute, Xiamen Yan Palace Seelong Biotechnology Co., Ltd.XiamenChina
| |
Collapse
|
27
|
Savastano MC, Fossataro C, Carlà MM, Cestrone V, Biagini I, Sammarco L, Giannuzzi F, Fasciani R, Apa R, Lanzone A, Diterlizzi A, Policriti M, Di Stasio E, Killian R, Rizzo C, Rizzo S. Chorioretinal biomarkers in hypothalamic amenorrhea. Graefes Arch Clin Exp Ophthalmol 2024; 262:2057-2065. [PMID: 38407592 PMCID: PMC11222264 DOI: 10.1007/s00417-023-06346-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/14/2023] [Accepted: 12/12/2023] [Indexed: 02/27/2024] Open
Abstract
PURPOSE The aim of our study was to evaluate changes in the retinal and choriocapillaris circulations in patients with hypothalamic amenorrhea. METHODS Prospective, cross-sectional observational study on 25 patients (50 eyes) diagnosed with hypothalamic amenorrhea and 25 age-matched healthy women. Optical coherence tomography angiography (OCTA) was used to evaluate the vessel density (VD) of superficial capillary plexus (SCP), deep capillary plexus (DCP), and choriocapillaris VD layers in whole 6.4 × 6.4-mm image and in fovea grid-based image. In patients' group, systemic parameters were collected: body mass index (BMI), endometrial rhyme thickness, follicle stimulating hormone (FSH), luteinizing hormone (LH), prolactin, insulin, and cortisol. RESULTS SCP and DCP did not show any statistical difference when comparing patients and controls (all p > 0.05). Differently, choriocapillaris VD in the whole region showed a non-significant tendency toward higher values in the patients group in both eyes (p = 0.038 for right eye [RE], p = 0.044 for left eye [LE]). Foveal choriocapillaris VD was higher in hypothalamic amenorrhea women vs. healthy controls (66.0 ± 2.4 vs. 63.7 ± 6.6%, p = 0.136 for RE; 65.0 ± 2.4 vs. 61.6 ± 7.0%, p = 0.005 for LE). Focusing on correlation with systemic parameters, SCP and DCP foveal density had a medium/high effect size with endometrial rhyme, along with DCP in the fovea area vs. cortisol and SCP in the whole area vs. FSH. CONCLUSION When comparing hypothalamic amenorrhea patients to healthy subjects, OCTA detected changes in the choriocapillaris layer, showing increased VD in the early stage of the systemic pathology, suggesting that microvascular "compaction" could be a first phase of hypoestrogenism adaptation.
Collapse
Affiliation(s)
- Maria Cristina Savastano
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Claudia Fossataro
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Matteo Mario Carlà
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy.
- Catholic University "Sacro Cuore,", Rome, Italy.
| | - Valentina Cestrone
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Ilaria Biagini
- Department NEUROFARBA, University of Florence, Florence, Italy
| | - Leonardo Sammarco
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Federico Giannuzzi
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Romina Fasciani
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
| | - Rosanna Apa
- Catholic University "Sacro Cuore,", Rome, Italy
- Obstetric Pathology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", 00168, Rome, Italy
| | - Antonio Lanzone
- Catholic University "Sacro Cuore,", Rome, Italy
- Obstetric Pathology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", 00168, Rome, Italy
| | - Alice Diterlizzi
- Catholic University "Sacro Cuore,", Rome, Italy
- Obstetric Pathology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", 00168, Rome, Italy
| | - Martina Policriti
- Catholic University "Sacro Cuore,", Rome, Italy
- Obstetric Pathology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", 00168, Rome, Italy
| | - Enrico Di Stasio
- Department of Basic Biotechnological Sciences, Intensive Care and Perioperative Clinics Research, Catholic University of the Sacred Heart, Milan, Italy
- "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", 00168, Rome, Italy
| | - Raphael Killian
- Ophthalmic Unit, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Clara Rizzo
- Ophthalmology Unit, Department of Surgery, University Hospital, 56100, Pisa, Italy
| | - Stanislao Rizzo
- Ophthalmology Unit, "Fondazione Policlinico Universitario A. Gemelli, IRCCS,", Largo A. Gemelli, 8, 00168, Rome, Italy
- Catholic University "Sacro Cuore,", Rome, Italy
- Neuroscience Institute, Italian National Research Council, CNR, Pisa, Italy
| |
Collapse
|
28
|
Mussa I, Jibro U, Sertu A, Deressa A, Mohammed F, Regassa LD, Cheru A, Mohammed E, Abdurahman D, Balis B. Burden of irregular menstrual cycle and its predictors among reproductive-age women in Ethiopia: Systematic review and meta-analysis. SAGE Open Med 2024; 12:20503121241259623. [PMID: 38895544 PMCID: PMC11185000 DOI: 10.1177/20503121241259623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction Irregular menstrual cycle has negative health and psychosocial repercussions for women of reproductive age worldwide. However, there is no national data for policymakers and health planners in Ethiopia. Therefore, this review aimed to determine the overall burden of irregular menstrual cycle and predictors among reproductive-age women in Ethiopia. Methods International databases (SCOPUS, CINAHL, CAB Abstract, EMBASE, PubMed, Web of Science, Google, and Google Scholar) and lists of references were employed to search literature in Ethiopia. The random-effects model was used to calculate the odds ratios of the outcome variable using STATA version 18. The heterogeneity of the studies was measured by computing I 2 and p-values. In addition, sensitivity analysis and funnel plots were performed to test the stability of pooled data in the presence of outliers and publication bias. Results The review includes 21 studies and 9109 populations. The overall burden of irregular menstrual cycles among reproductive-age women was 35% (95% CI: 30-41) with I 2 = 96.96%. Sleeping for <5 h a day (AOR: 2.49; 95% CI: 1.49-3.49) and a stressful life (AOR: 3.15; 95% CI: 1.44-4.85) were predictors of irregular menstrual cycles. Conclusion More than one in every three reproductive-age women in Ethiopia experience irregular menstrual cycles. Sleeping for <5 h a day and stress increase the likelihood of an irregular menstrual cycle, which can be modified by improving sleeping hours and decreasing stress stimulators through psychotherapy.
Collapse
Affiliation(s)
- Ibsa Mussa
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Usmael Jibro
- School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Addisu Sertu
- School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Alemayehu Deressa
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Fethia Mohammed
- Department of Psychiatry, School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Lemma Demissie Regassa
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Abera Cheru
- School of Environmental Health Science, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Eptisam Mohammed
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Dureti Abdurahman
- School of Public Health, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Bikila Balis
- School of Nursing and Midwifery, College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| |
Collapse
|
29
|
Bakhsh H. Retrospective Analysis of Effective Management Strategies for Primary Amenorrhea of Reproductive Age in Saudi Arabia. Life (Basel) 2024; 14:772. [PMID: 38929754 PMCID: PMC11204569 DOI: 10.3390/life14060772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Primary amenorrhea, the absence of menstruation by age 15, can have significant implications for reproductive health and overall well-being. This retrospective study aimed to evaluate the effectiveness of various management strategies for primary amenorrhea among women of reproductive age in Saudi Arabia. Medical records of 63 eligible patients from 2018 to 2023 were analyzed, assessing diagnostic methods, treatment modalities, and associated outcomes. The findings revealed that hormonal therapy was the most commonly employed management strategy (50.0%) and demonstrated the highest rate of achieving menstrual regularity (62.5%). Surgical interventions were utilized in 28.1% of cases, with a 50.0% rate of symptom resolution. Lifestyle modifications were less frequent (21.9%) but showed a moderate rate of symptom resolution (35.7%). Logistic regression analysis identified age, underlying etiology, and management strategy as significant predictors of treatment success. Subgroup analyses highlighted the efficacy of hormonal therapy and lifestyle modifications for genetic etiologies, while surgical interventions were more effective for anatomical causes. The study underscores the importance of a comprehensive diagnostic approach and personalized treatment plans tailored to individual patient characteristics. Despite limitations, the findings contribute to the understanding of optimal management strategies for primary amenorrhea and emphasize the need for multidisciplinary collaboration in addressing this complex condition.
Collapse
Affiliation(s)
- Hanadi Bakhsh
- Obstetrics and Gynecology Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh 11564, Saudi Arabia;
- Department of Obstetrics and Gynecology, King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| |
Collapse
|
30
|
Cuffaro F, Russo E, Amedei A. Endometriosis, Pain, and Related Psychological Disorders: Unveiling the Interplay among the Microbiome, Inflammation, and Oxidative Stress as a Common Thread. Int J Mol Sci 2024; 25:6473. [PMID: 38928175 PMCID: PMC11203696 DOI: 10.3390/ijms25126473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Endometriosis (EM), a chronic condition in endometrial tissue outside the uterus, affects around 10% of reproductive-age women, significantly affecting fertility. Its prevalence remains elusive due to the surgical confirmation needed for diagnosis. Manifesting with a range of symptoms, including dysmenorrhea, dyschezia, dysuria, dyspareunia, fatigue, and gastrointestinal discomfort, EM significantly impairs quality of life due to severe chronic pelvic pain (CPP). Psychological manifestations, notably depression and anxiety, frequently accompany the physical symptoms, with CPP serving as a key mediator. Pain stems from endometrial lesions, involving oxidative stress, neuroinflammation, angiogenesis, and sensitization processes. Microbial dysbiosis appears to be crucial in the inflammatory mechanisms underlying EM and associated CPP, as well as psychological symptoms. In this scenario, dietary interventions and nutritional supplements could help manage EM symptoms by targeting inflammation, oxidative stress, and the microbiome. Our manuscript starts by delving into the complex relationship between EM pain and psychological comorbidities. It subsequently addresses the emerging roles of the microbiome, inflammation, and oxidative stress as common links among these abovementioned conditions. Furthermore, the review explores how dietary and nutritional interventions may influence the composition and function of the microbiome, reduce inflammation and oxidative stress, alleviate pain, and potentially affect EM-associated psychological disorders.
Collapse
Affiliation(s)
- Francesca Cuffaro
- Division of Interdisciplinary Internal Medicine, Careggi University Hospital of Florence, 50134 Florence, Italy;
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 50139 Florence, Italy
| |
Collapse
|
31
|
Zhao M, Xie Y, Xu X, Zhang Z, Shen C, Chen X, Zhu B, Yang L, Zhou B. Reproductive and transgenerational toxicity of bisphenol S exposure in pregnant rats: Insights into hormonal imbalance and steroid biosynthesis pathway disruption. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172379. [PMID: 38614345 DOI: 10.1016/j.scitotenv.2024.172379] [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: 01/23/2024] [Revised: 03/23/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
Bisphenol S (BPS) is an alternative chemical to bisphenol A commonly used in food packaging materials. It raises concerns due to potential adverse effects on human health. However, limited evidence exists regarding reproductive toxicity from BPS exposure, and the mechanism of associated transgenerational toxicity remains unclear. In this study, pregnant SD rats were exposed to two different doses of BPS (0.05 or 20 mg/kg) from GD6 to PND21. The objective was to investigate reproductive and transmissible toxicity induced by BPS, explore endocrine effects, and uncover potential underlying mechanisms in rats. Perinatal exposure to BPS in the F0 generation significantly decreased the rate of body weight, ovarian organ coefficient, and growth and development of the F1 generation. Notably, these changes included abnormal increases in body weight and length, estrous cycle disruption, and embryonic dysplasia in F1. 4D-DIA proteomic and PRM analyses revealed that exposure to 20 mg/kg group significantly altered the expression of proteins, such as Lhcgr and Akr1c3, within the steroid biosynthetic pathway. This led to elevated levels of FSH and LH in the blood. The hypothalamic-pituitary-ovarian (HPO) axis, responsible for promoting fertility through the cyclic secretion of gonadotropins and steroid hormones, was affected. RT-qPCR and Western blot results demonstrated that the expression of GnRH in the hypothalamus was decreased, the GnRHR in the pituitary gland was decreased, and the expression of FSHβ and LHβ in the pituitary gland was increased. Overall, BPS exposure disrupts the HPO axis, hormone levels, and steroid biosynthesis in the ovaries, affecting offspring development and fertility. This study provides new insights into the potential effects of BPS exposure on the reproductive function of the body and its relevant mechanisms of action.
Collapse
Affiliation(s)
- Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Ying Xie
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Xiuqin Xu
- Wuhan Biobank Co., Ltd., Wuhan 430070, China
| | - Zequan Zhang
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Can Shen
- School of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xianglin Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Biran Zhu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
32
|
Tang W, Yan H, Chen X, Pu Y, Qi X, Dong L, Su C. hUCMSC-derived extracellular vesicles relieve cisplatin-induced granulosa cell apoptosis in mice by transferring anti-apoptotic miRNAs. J Biomed Res 2024; 39:36-49. [PMID: 38808551 PMCID: PMC11873591 DOI: 10.7555/jbr.37.20230310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/16/2024] [Accepted: 04/22/2024] [Indexed: 05/30/2024] Open
Abstract
Premature ovarian insufficiency (POI) caused by chemotherapy is a common complication in female cancer survivors of childbearing age. Traditional methods, including mesenchymal stem cell (MSC) transplant and hormone replacement therapy, have limited clinical application because of their drawbacks, and more methods need to be developed. In the current study, the potential effects and underlying mechanisms of human umbilical cord MSC-derived extracellular vesicles (hUCMSC-EVs) were investigated in a cisplatin (CDDP)-induced POI mouse model and a human granulosa cell (GC) line. The results showed that hUCMSC-EVs significantly attenuated body weight loss, ovarian weight loss, ovary atrophy, and follicle loss in moderate-dose (1.5 mg/kg) CDDP-induced POI mice, similar to the effects observed with hUCMSCs. We further found that the hUCMSC-EVs inhibited CDDP-induced ovarian GC apoptosis by upregulating anti-apoptotic miRNA levels in GCs, thereby downregulating the mRNA levels of multiple pro-apoptotic genes. In general, our findings indicate that the moderate-dose chemotherapy may be a better choice for clinical oncotherapy, considering effective rescue of the oncotherapy-induced ovarian damage with hUCMSC-EVs. Additionally, multiple miRNAs in hUCMSC-EVs may potentially be used to inhibit the chemotherapy-induced ovarian GC apoptosis, thereby restoring ovarian function and improving the life quality of female cancer patients.
Collapse
Affiliation(s)
- Wenjing Tang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Haiyan Yan
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaojun Chen
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yanan Pu
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xin Qi
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Liyang Dong
- Department of Nuclear Medicine, the Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | - Chuan Su
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Pathogen Biology & Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
33
|
Ding M, Lu Y, Wen Q, Xing C, Huang X, Zhang Y, Wang W, Zhang C, Zhang M, Meng F, Liu K, Liu G, Song L. Ovarian PERK/NRF2/CX43/StAR/progesterone pathway activation mediates female reproductive dysfunction induced by cold exposure. Sci Rep 2024; 14:10248. [PMID: 38702372 PMCID: PMC11068861 DOI: 10.1038/s41598-024-60907-9] [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: 09/20/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
Ambient air temperature is a key factor affecting human health. Female reproductive disorders are representative health risk events under low temperature. However, the mechanism involving in cold-induced female reproductive disorders remains largely unknown. Female mice were intermittently exposed to cold conditions (4 °C) to address the health risk of low temperature on female reproductive system. Primary granulosa cells (GCs) were prepared and cultured under low temperature (35 °C) or exposed to β3-adrenoreceptor agonist, isoproterenol, to mimic the condition of cold exposure. Western-blot, RT-PCR, co-IP, ELISA, pharmacological inhibition or siRNA-mediated knockdown of target gene were performed to investigate the possible role of hormones, gap conjunction proteins, and ER stress sensor protein in regulating female reproductive disorders under cold exposure. Cold exposure induced estrous cycle disorder and follicular dysplasia in female mice, accompanying with abnormal upregulation of progesterone and its synthetic rate-limiting enzyme, StAR, in the ovarian granulosa cells. Under the same conditions, an increase in connexin 43 (CX43) expressions in the GCs was also observed, which contributed to elevated progesterone levels in the ovary. Moreover, ER stress sensor protein, PERK, was activated in the ovarian GCs after cold exposure, leading to the upregulation of downstream NRF2-dependent CX43 transcription and aberrant increase in progesterone synthesis. Most importantly, blocking PERK expression in vivo significantly inhibited NRF2/CX43/StAR/progesterone pathway activation in the ovary and efficiently rescued the prolongation of estrous cycle and the increase in follicular atresia of the female mice induced by cold stress. We have elucidated the mechanism of ovarian PERK/NRF2/CX43/StAR/progesterone pathway activation in mediating female reproductive disorder under cold exposure. Targeting PERK might be helpful for maintaining female reproductive health under cold conditions.
Collapse
Affiliation(s)
- Mengnan Ding
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yarong Lu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Henan University Joint National Laboratory for Antibody Drug Engineering, Henan, 465004, China
| | - Qing Wen
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Chen Xing
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yifan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Wei Wang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- School of Pharmacy, Jiamusi University, Jiamusi, 154007, China
| | - Chongchong Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Henan University Joint National Laboratory for Antibody Drug Engineering, Henan, 465004, China
| | - Min Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Fanfei Meng
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Kun Liu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Guangchao Liu
- Henan University Joint National Laboratory for Antibody Drug Engineering, Henan, 465004, China
| | - Lun Song
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
- College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, 473007, China.
- School of Pharmacy, Jiamusi University, Jiamusi, 154007, China.
- Anhui Medical University, 81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
34
|
Santacruz-Márquez R, Neff AM, Mourikes VE, Fletcher EJ, Flaws JA. The effects of inhaled pollutants on reproduction in marginalized communities: a contemporary review. Inhal Toxicol 2024; 36:286-303. [PMID: 37075037 PMCID: PMC10584991 DOI: 10.1080/08958378.2023.2197941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/25/2023] [Indexed: 04/20/2023]
Abstract
Important differences in health that are closely linked with social disadvantage exist within and between countries. According to the World Health Organization, life expectancy and good health continue to increase in many parts of the world, but fail to improve in other parts of the world, indicating that differences in life expectancy and health arise due to the circumstances in which people grow, live, work, and age, and the systems put in place to deal with illness. Marginalized communities experience higher rates of certain diseases and more deaths compared to the general population, indicating a profound disparity in health status. Although several factors place marginalized communities at high risk for poor health outcomes, one important factor is exposure to air pollutants. Marginalized communities and minorities are exposed to higher levels of air pollutants than the majority population. Interestingly, a link exists between air pollutant exposure and adverse reproductive outcomes, suggesting that marginalized communities may have increased reproductive disorders due to increased exposure to air pollutants compared to the general population. This review summarizes different studies showing that marginalized communities have higher exposure to air pollutants, the types of air pollutants present in our environment, and the associations between air pollution and adverse reproductive outcomes, focusing on marginalized communities.
Collapse
Affiliation(s)
| | - Alison M. Neff
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| | | | - Endia J. Fletcher
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| | - Jodi A. Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| |
Collapse
|
35
|
Jin Y, Di-si D, Ke-ming W. XinJiaCongRongTuSiZiWan protects triptolide-induced rats from oxidative stress injury via mitophagy mediated PINK1/Parkin signaling pathway. Acta Cir Bras 2024; 39:e391424. [PMID: 38511762 PMCID: PMC10953615 DOI: 10.1590/acb391424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/15/2023] [Indexed: 03/22/2024] Open
Abstract
PURPOSE XinJiaCongRongTuSiZiWan (XJCRTSZW) is a traditional Chinese medicine compound for invigorating the kidney, nourishing blood, and promoting blood circulation. This study aimed to explore the effect of XJCRTSZW on triptolide (TP)-induced oxidative stress injury. METHODS Adult female Sprague-Dawley rats and human ovarian granulosa cell lines were treated with TP and XJCRTSZW. Hematoxylin and eosin staining, enzyme-linked immunosorbent assay, flow cytometry, CCK-8, JC-1 staining, transmission electron microscopy, reverse transcription-quantitative polymerase chain reaction, and Western blotting were performed in this study. RESULTS XJCRTSZW treatment observably ameliorated the TP-induced pathological symptoms. Furthermore, XJCRTSZW treatment observably enhanced the TP-induced reduction of estradiol, anti-Mullerian hormone, progesterone, superoxide dismutase, ATP content, mitochondrial membrane potential, p62, and Hsp60 mRNA, and protein levels in vivo and in vitro (p < 0.05). However, TP-induced elevation of follicle stimulating hormone and luteinizing hormone concentrations, malondialdehyde levels, reactive oxygen species levels, apoptosis rate, mitophagy, and the mRNA and protein expressions of LC3-II/LC3-I, PTEN-induced kinase 1 (PINK1), and Parkin were decreased (p < 0.05). In addition, XJCRTSZW treatment markedly increased cell viability in vitro (p < 0.05). CONCLUSIONS XJCRTSZW protects TP-induced rats from oxidative stress injury via the mitophagy-mediated PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Yan Jin
- Shaanxi University of Chinese Medicine – Xianyang, China
- Shaanxi Key Laboratory of Chinese Medicine – Research on Physical Constitution and Diseases – Xianyang, China
| | - Deng Di-si
- Hospital of Chengdu University of Traditional Chinese Medicine – Department of Gynecology – Chengdu, China
| | - Wu Ke-ming
- Hospital of Chengdu University of Traditional Chinese Medicine – Department of Gynecology – Chengdu, China
| |
Collapse
|
36
|
Lonardo MS, Cacciapuoti N, Guida B, Di Lorenzo M, Chiurazzi M, Damiano S, Menale C. Hypothalamic-Ovarian axis and Adiposity Relationship in Polycystic Ovary Syndrome: Physiopathology and Therapeutic Options for the Management of Metabolic and Inflammatory Aspects. Curr Obes Rep 2024; 13:51-70. [PMID: 38172476 PMCID: PMC10933167 DOI: 10.1007/s13679-023-00531-2] [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] [Accepted: 09/27/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE OF REVIEW The goal of the present review is to address the main adiposity-related alterations in Polycystic Ovary Syndrome (PCOS) focusing on hypothalamic-pituitary-ovarian (H-P-O) axis and to provide an overview of nutraceutical and pharmacological therapeutic strategies. RECENT FINDINGS Female reproduction is a complex and delicate interplay between neuroendocrine signals involving the H-P-O axis. Elements that disrupt the balance of these interactions can lead to metabolic and reproductive disorders, such as PCOS. This disorder includes menstrual, metabolic, and biochemical abnormalities as well as hyperandrogenism, oligo-anovulatory menstrual cycles, insulin resistance, and hyperleptinemia which share an inflammatory state with other chronic diseases. Moreover, as in a self-feeding cycle, high androgen levels in PCOS lead to visceral fat deposition, resulting in insulin resistance and hyperinsulinemia, further stimulating ovarian and adrenal androgen production. In fact, regardless of age and BMI, women with PCOS have more adipose tissue and less lean mass than healthy women. Excessive adiposity, especially visceral adiposity, is capable of affecting female reproduction through direct mechanisms compromising the luteal phase, and indirect mechanisms as metabolic alterations able to affect the function of the H-P-O axis. The intricate crosstalk between adiposity, inflammatory status and H-P-O axis function contributes to the main adiposity-related alterations in PCOS, and alongside currently available hormonal treatments, nutraceutical and pharmacological therapeutic strategies can be exploited to treat these alterations, in order to enable a more comprehensive synergistic and tailored treatment.
Collapse
Affiliation(s)
- Maria Serena Lonardo
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy.
| | - Nunzia Cacciapuoti
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Bruna Guida
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Mariana Di Lorenzo
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Martina Chiurazzi
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Simona Damiano
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, Physiology Nutrition Unit, Federico II University of Naples, Via Sergio Pansini 5, 80131, Napoli, Italy
| |
Collapse
|
37
|
Dang C, Chen Z, Chai Y, Liu P, Yu X, Liu Y, Liu J. Assessing the relationship between gut microbiota and endometriosis: a bidirectional two-sample mendelian randomization analysis. BMC Womens Health 2024; 24:123. [PMID: 38365715 PMCID: PMC10873948 DOI: 10.1186/s12905-024-02945-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND An increasing body of observational studies have indicated an association between gut microbiota and endometriosis. However, the causal relationship between them is not yet clear. In this study, we employed Mendelian randomization method to investigate the causal relationship between 211 gut microbiota taxa and endometriosis. METHODS Independent genetic loci significantly associated with the relative abundance of 211 gut microbiota taxa, based on predefined thresholds, were extracted as instrumental variables. The primary analytical approach employed was the IVW method. Effect estimates were assessed primarily using the odds ratio and 95% confidence intervals. Supplementary analyses were conducted using MR-Egger regression, the weighted median method, the simple mode and the weighted mode method to complement the IVW results. In addition, we conducted tests for heterogeneity, horizontal pleiotropy, sensitivity analysis, and MR Steiger to assess the robustness of the results and the strength of the causal relationships. RESULTS Based on the IVW method, we found that the family Prevotellaceae, genus Anaerotruncus, genus Olsenella, genus Oscillospira, and order Bacillales were identified as risk factors for endometriosis, while class Melainabacteria and genus Eubacterium ruminantium group were protective factors. Additionally, no causal relationship was observed between endometriosis and gut microbiota. Heterogeneity tests, pleiotropy tests, and leave-one-out sensitivity analyses did not detect any significant heterogeneity or pleiotropic effects. CONCLUSIONS Our MR study has provided evidence supporting a potential causal relationship between gut microbiota and endometriosis, and it suggests the absence of bidirectional causal effects. These findings could potentially offer new insights for the development of novel strategies for the prevention and treatment of endometriosis.
Collapse
Affiliation(s)
- Chunxiao Dang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Zhenting Chen
- Department of eugenic genetics, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, 257091, Shandong, China
| | - Yuyan Chai
- Department of obstetrics, The People's Hospital of Dongying Distric, Dongying, 257091, Shandong, China
| | - Pengfei Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Xiao Yu
- Department of gynaecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Yan Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250000, Shandong, China.
| | - Jinxing Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China.
| |
Collapse
|
38
|
Szukiewicz D. Current Insights in Prolactin Signaling and Ovulatory Function. Int J Mol Sci 2024; 25:1976. [PMID: 38396659 PMCID: PMC10889014 DOI: 10.3390/ijms25041976] [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: 01/10/2024] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Prolactin (PRL) is a pleiotropic hormone released from lactotrophic cells of the anterior pituitary gland that also originates from extrapituitary sources and plays an important role in regulating lactation in mammals, as well as other actions. Acting in an endocrine and paracrine/autocrine manner, PRL regulates the hypothalamic-pituitary-ovarian axis, thus influencing the maturation of ovarian follicles and ovulation. This review provides a detailed discussion of the current knowledge on the role of PRL in the context of ovulation and ovulatory disorders, particularly with regard to hyperprolactinemia, which is one of the most common causes of infertility in women. Much attention has been given to the PRL structure and the PRL receptor (PRLR), as well as the diverse functions of PRLR signaling under normal and pathological conditions. The hormonal regulation of the menstrual cycle in connection with folliculogenesis and ovulation, as well as the current classifications of ovulation disorders, are also described. Finally, the state of knowledge regarding the importance of TIDA (tuberoinfundibular dopamine), KNDγ (kisspeptin/neurokinin B/dynorphin), and GnRH (gonadotropin-releasing hormone) neurons in PRL- and kisspeptin (KP)-dependent regulation of the hypothalamic-pituitary-gonadal (HPG) axis in women is reviewed. Based on this review, a rationale for influencing PRL signaling pathways in therapeutic activities accompanying ovulation disorders is presented.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
39
|
Cui J, Wang Y. Premature ovarian insufficiency: a review on the role of tobacco smoke, its clinical harm, and treatment. J Ovarian Res 2024; 17:8. [PMID: 38191456 PMCID: PMC10775475 DOI: 10.1186/s13048-023-01330-y] [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: 09/18/2023] [Accepted: 12/17/2023] [Indexed: 01/10/2024] Open
Abstract
Premature ovarian insufficiency (POI) is a condition in which the quantity of follicles and the quality of oocytes gradually decrease. This results in an estrogen secretion disorder and abnormal follicle development, which can lead to related diseases, early onset of menopause, sexual dysfunction, and an increased risk of cardiovascular issues, osteoporosis, and depression, among others. This disease significantly impacts the physical and mental health and overall quality of life of affected women. Factors such as genetic abnormalities, oophorectomy, radiotherapy for malignancy, idiopathic conditions, and an unhealthy lifestyle, including smoking, can accelerate the depletion of the follicular pool and the onset of menopause. Extensive research has been conducted on the detrimental effects of tobacco smoke on the ovaries. This article aims to review the advancements in understanding the impact of tobacco smoke on POI, both in vivo and in vitro. Furthermore, we explore the potential adverse effects of common toxicants found in tobacco smoke, such as polycyclic aromatic hydrocarbons (PAHs), heavy metals like cadmium, alkaloids like nicotine and its major metabolite cotinine, benzo[a]pyrene, and aromatic amines. In addition to discussing the toxicants, this article also reviews the complications associated with POI and the current state of research and application of treatment methods. These findings will contribute to the development of more precise treatments for POI, offering theoretical support for enhancing the long-term quality of life for women affected by this condition.
Collapse
Affiliation(s)
- Jinghan Cui
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|
40
|
Shi K, Liu X, Duan Y, Ding J, Jia Y, Jiang Z, Feng C. Multi-omics analysis reveals associations between host gene expression, gut microbiota, and metabolites in chickens. J Anim Sci 2024; 102:skae263. [PMID: 39243135 PMCID: PMC11457126 DOI: 10.1093/jas/skae263] [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: 05/16/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024] Open
Abstract
Egg-laying is an important trait in chickens, and it is affected by many factors, such as hormones regulated by the hypothalamic-pituitary axis and precursors synthesized by the liver. Recent studies showed that gut microbiota was associated with egg-laying, however, its underlying mechanism remains unclear. We comprehensively analyzed the host transcriptome, gut microbiota, and metabolome in broiler breeder hens during the pre-laying, peak-laying, and late-laying periods. The transcriptome analysis of the tissues related to the hypothalamic-pituitary-liver (HPL) axis revealed dynamic gene expression during egg-laying periods. Differentially expressed genes (DEGs) (i.e., PENK, NPY, AVP, PRL, RLN3, and FST) from the hypothalamus and pituitary gland were involved in female gonadal development, hormone secretion, response to endogenous stimulus, liver development, and amide metabolism. In liver, DEGs (i.e., FABP3, VTG1, LPL, APOA5, APOV1, and RBP5) were enriched in efferocytosis, sphingolipid metabolism, amide, and peptide biosynthesis. Alpha and beta diversity changed significantly in cecum microbiota during different laying periods. The abundance of Firmicutes was decreased and the abundance of Bacteroidota was increased during the peak-laying period. Functional analysis showed that the biosynthesis of secondary metabolites, amino acids, purine, and steroid hormones was altered during laying. The metabolome analysis from cecal contents showed that amino acid metabolism and steroid hormone biosynthesis changed during laying. Integrated analysis of the cecal microbiota and metabolites showed the genus Megasphaera was involved in amino acid metabolism, which included 3-phenyllatic acid, quinic acid, caffeic acid, and folic acid, and the genus Hungatella participated in steroid hormone biosynthesis through its strong correlation with estradiol. These results explored the dynamic changes in tissues related to the HPL axis and cecal microbiota and provided new insights into the interaction between the host and microbiota during egg-laying in chickens.
Collapse
Affiliation(s)
- Kai Shi
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiangping Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ying Duan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jiqiang Ding
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yimin Jia
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ziqin Jiang
- Department of Breeding Research, Guangdong Wens South Poultry Breeding Co. Ltd, Yunfu, China
| | - Chungang Feng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
41
|
Yousif A, Ebeid A, Kacsoh B, Bazzaro M, Chefetz I. The Ovary-Brain Connection. Cells 2024; 13:94. [PMID: 38201298 PMCID: PMC10778337 DOI: 10.3390/cells13010094] [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: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The brain and the ovaries are in a state of continuous communication [...].
Collapse
Affiliation(s)
- Abdelrahman Yousif
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA
| | - Ahmed Ebeid
- Department of Obstetrics and Gynecology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Balint Kacsoh
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| | - Martina Bazzaro
- Masonic Cancer Center and Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Biomedical and Clinical Science, Linköping University, SE-581 85 Linköping, Sweden
| | - Ilana Chefetz
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31207, USA
| |
Collapse
|
42
|
Su BQ, Yang GY, Wang J, Ming SL, Chu BB. Pseudorabies virus inhibits progesterone-induced inactivation of TRPML1 to facilitate viral entry. PLoS Pathog 2024; 20:e1011956. [PMID: 38295116 PMCID: PMC10829982 DOI: 10.1371/journal.ppat.1011956] [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: 09/19/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
Viral infection is a significant risk factor for fertility issues. Here, we demonstrated that infection by neurotropic alphaherpesviruses, such as pseudorabies virus (PRV), could impair female fertility by disrupting the hypothalamus-pituitary-ovary axis (HPOA), reducing progesterone (P4) levels, and consequently lowering pregnancy rates. Our study revealed that PRV exploited the transient receptor potential mucolipin 1 (TRPML1) and its lipid activator, phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2), to facilitate viral entry through lysosomal cholesterol and Ca2+. P4 antagonized this process by inducing lysosomal storage disorders and promoting the proteasomal degradation of TRPML1 via murine double minute 2 (MDM2)-mediated polyubiquitination. Overall, the study identifies a novel mechanism by which PRV hijacks the lysosomal pathway to evade P4-mediated antiviral defense and impair female fertility. This mechanism may be common among alphaherpesviruses and could contribute significantly to their impact on female reproductive health, providing new insights for the development of antiviral therapies.
Collapse
Affiliation(s)
- Bing-Qian Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou, Henan Province, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou, Henan Province, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan Province, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou, Henan Province, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan Province, China
| | - Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou, Henan Province, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Growth and Development of Henan Province, Henan Agricultural University, Zhengzhou, Henan Province, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan Province, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan Province, China
- Longhu Advanced Immunization Laboratory, Zhengzhou, Henan Province, China
| |
Collapse
|
43
|
Boychuk AV, Miklashevska OA, Khlibovska OI, Yakymchuk YB, Nikitina IM, Herevych NV. Comorbid pathology of the mammary glands and endometriosis: risk factors and prognosis. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:247-253. [PMID: 38592985 DOI: 10.36740/wlek202402109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
OBJECTIVE Aim: based on a retrospective analysis, the relationship between external genital endometriosis and comorbid breast pathology was established and risk factors were identified, their comparison and the formation of a prognostic risk criterion were determined. PATIENTS AND METHODS Materials and Methods: to address the objectives of the study, a retrospective analysis of 470 cases of patients treated for external genital endometriosis after surgical treatment and comorbid breast pathology was conducted. The control group included 30 healthy non-pregnant women. Statistical processing was performed on a personal computer using the statistical software package Statistica 10. RESULTS Results: As a result of the analysis, the age of the patients ranged from 23 to 40 years. The average age of patients in the study group was (32.2}1.18) years, and in the control group (31.1}1.35) (p>0.05). The groups were homogeneous in terms of age (p>0.05), marital status (p>0.05) and level of education (p>0.05). Close relatives in 208 (44.25}2.18) % (OR=8.86; 95 % CI: (0.68-10.53); p<0.002) cases suffered from benign (hormone-dependent) tumours and tumour-like diseases of the uterus and appendages in isolation or in various combinations (fibroids, adenomyosis, endometrial hyperplasia). It was also found that 102 (21.70}1.67) % of patients had endometriosis, which may indicate a genetic predisposition to this disease. In the closest relatives of EM patients: in 118 (25.10}2.01) % of the examined parents, breast problems were noted, in 66 (14.04}1.12) % - diabetes mellitus, and in 98 (20.85}1.22) % thyroid diseases were detected, which in total amounted to (60.00}2.23) % (OR=9.12; 95 % CI: (0.58-11.54); p<0.002). Early menarche almost tripled the risk of EM (OR=2.72; 95% CI: (1.02-5.11); p<0.002), and menstrual irregularities doubled it (OR=2.04; 95% CI: (1.09-3.14); p<0.05), higher education, urban residents - 2.2 times higher (OR= 2.27; 95 % CI: (1.11-3.63); p<0.05), diseases of the gastrointestinal tract and hepatobiliary complex - 5.2 times higher (OR=5.27; 95 % CI: (1.89-12.03); p<0.05), frequently recurrent inflammatory diseases of the appendages - 3 times higher (OR=3.14; 95 % CI: (0.91-5.14); p<0.05), dysmetabolic manifestations (thyroid dysfunction) - 5 times higher (OR=5.11; 95 % CI: (1.61-9.503); p<0.002). CONCLUSION Conclusions: Thus, in endometriosis and dyshormonal diseases of the mammary glands, menstrual and generative function disorders, along with clinical symptoms of pelvic pain, dysmenorrhoea, autonomic nervous system disorders and sexual dysfunction, are significant components of this problem, initiating comorbidity processes in target organs in the setting of hormonal maladaptation. Therefore, these comorbidities become a trigger for the activation of systemic hormonal imbalance and become an urgent interdisciplinary problem that requires further study.
Collapse
Affiliation(s)
- Alla V Boychuk
- I. HORBACHEVSKY TERNOPIL NATIONAL MEDICAL UNIVERSITY, TERNOPIL, UKRAINE
| | | | | | | | | | | |
Collapse
|
44
|
van Lohuizen R, Paungarttner J, Lampl C, MaassenVanDenBrink A, Al-Hassany L. Considerations for hormonal therapy in migraine patients: a critical review of current practice. Expert Rev Neurother 2023; 24:1-21. [PMID: 38112066 PMCID: PMC10791067 DOI: 10.1080/14737175.2023.2296610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
INTRODUCTION Migraine, a neurovascular headache disorder, is a leading cause of disability worldwide. Within the multifaceted pathophysiology of migraine, hormonal fluctuations play an evident triggering and exacerbating role, pointing toward the need for identification and proper usage of both existing and new hormonal targets in migraine treatment. AREAS COVERED With a threefold higher incidence of migraine in women than in men, the authors delve into sex hormone-related events in migraine patients. A comprehensive overview is given of existing hormonal therapies, including oral contraceptives, intrauterine devices, transdermal and subcutaneous estradiol patches, gnRH-agonists, oral testosterone, and 5α reductase inhibitors. The authors discuss their effectiveness and risks, noting their suitability for different patient profiles. Next, novel evolving hormonal treatments, such as oxytocin and prolactin, are explored. Lastly, the authors cover hormonal conditions associated with migraine, such as polycystic ovary syndrome, endometriosis, and transgender persons receiving gender affirming hormone therapy, aiming to provide more personalized and effective solutions for migraine management. EXPERT OPINION Rigorous research into both existing and new hormonal targets, as well as the underlying pathophysiology, is needed to support a tailored approach in migraine treatment, in an ongoing effort to alleviate the impact of migraine on individuals and society.
Collapse
Affiliation(s)
- Romy van Lohuizen
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Christian Lampl
- Headache Medical Center Linz, Linz, Austria
- Department of Neurology and Stroke Unit, Koventhospital Barmherzige Brüder Linz, Linz, Austria
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Linda Al-Hassany
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
45
|
Panda SP, Kesharwani A, Singh GD, Prasanth D, Vatchavai BR, Kumari PVK, Panda SK, Mallick SP. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer's disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023; 92:102086. [PMID: 37821047 DOI: 10.1016/j.arr.2023.102086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The Kisspeptin1 (KISS1)/neurokinin B (NKB)/Dynorphin (Dyn) [KNDy] neurons in the hypothalamus regulate the reproduction stage in human beings and rodents. KNDy neurons co-expressed all KISS1, NKB, and Dyn peptides, and hence commonly regarded as KISS1 neurons. KNDy neurons contribute to the "GnRH pulse generator" and are implicated in the regulation of pulsatile GnRH release. The estradiol (E2)-estrogen receptor (ER) interactions over GnRH neurons in the hypothalamus cause nitric oxide (NO) discharge, in addition to presynaptic GABA and glutamate discharge from respective neurons. The released GABA and glutamate facilitate the activity of GnRH neurons via GABAA-R and AMPA/kainate-R. The KISS1 stimulates MAPK/ERK1/2 signaling and cause the release of Ca2+ from intracellular store, which contribute to neuroendocrine function, increase apoptosis and decrease cell proliferation and metastasis. The ageing in women deteriorates KISS1/KISS1R interaction in the hypothalamus which causes lower levels of GnRH. Because examining the human brain is so challenging, decades of clinical research have failed to find the causes of KNDy/GnRH dysfunction. The KISS1/KISS1R interactions in the brain have a neuroprotective effect against Alzheimer's disease (AD). These findings modulate the pathophysiological role of the KNDy/GnRH neural network in polycystic ovarian syndrome (PCOS) associated with ageing and, its protective role in cancer and AD. This review concludes with protecting effect of the steroid-derived acute regulatory enzyme (StAR) against neurotoxicity in the hippocampus, and hypothalamus, and these measures are fundamental for delaying ageing with PCOS. StAR could serve as novel diagnostic marker and therapeutic target for the most prevalent hormone-sensitive breast cancers (BCs).
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | | | - Dsnbk Prasanth
- KVSR Siddhartha College of Pharmaceutical Sciences, Vijayawada, Andhrapradesh, India
| | - Bhaskara Raju Vatchavai
- Sri Vasavi Institute of Pharmaceutical Sciences, Pedatadepalli, Tadepalligudem, Andhrapradesh, India
| | - P V Kamala Kumari
- Vignan Institute of Pharmaceutical Technology, Duvvada, Visakhapatnam, Andhrapradesh, India
| | | | | |
Collapse
|
46
|
Thaker N, Dhande R, Parihar P. Role of Transvaginal Sonography in the Diagnosis of Female Infertility: A Comprehensive Review. Cureus 2023; 15:e50048. [PMID: 38186406 PMCID: PMC10767472 DOI: 10.7759/cureus.50048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Female infertility, a complex and emotionally challenging condition, impacts millions of women worldwide. Timely and accurate diagnosis is crucial for tailoring effective solutions to overcome fertility challenges. Transvaginal sonography, a real-time and non-invasive imaging modality, is pivotal in this diagnostic process. This review focuses on the structural abnormalities of the female reproductive system related to female infertility, particularly highlighting the capabilities of transvaginal sonography in assessing ovulatory disorders, structural anomalies, endometrial conditions, ovarian reserve, and other contributing factors. It is important to note that while transvaginal sonography excels in detecting structural abnormalities, it may not effectively identify lifestyle and hormonal changes. This limitation underscores the necessity for a comprehensive diagnostic approach that includes additional modalities to address the multifaceted nature of female infertility. Despite acknowledging the inherent limitations and operator dependence of transvaginal sonography, we emphasize its significance in guiding clinicians toward well-informed decisions and personalized treatment plans. Looking forward, we anticipate the continual evolution of sonographic technology, offering enhanced diagnostic capabilities. The commitment to improving fertility outcomes for individuals and couples navigating the intricate path toward parenthood remains paramount. In conclusion, a holistic diagnostic approach incorporating various modalities is essential for a thorough understanding and effective management of female infertility.
Collapse
Affiliation(s)
- Nirja Thaker
- Radiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Rajasbala Dhande
- Radiodiagnosis, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Pratapsingh Parihar
- Radiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
47
|
Pant P, Sircar R, Prasad R, Prasad HO, Chitme HR. Protein Expression and Bioinformatics Study of Granulosa Cells of Polycystic Ovary Syndrome Expressed Under the Influence of DHEA. Clin Med Insights Endocrinol Diabetes 2023; 16:11795514231206732. [PMID: 38023736 PMCID: PMC10644732 DOI: 10.1177/11795514231206732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Background The reproductive system is heavily dependent on ovarian follicles, which are made up of germ cells (oocytes) and granulosa cells (GCs), including cumulus granulosa cells (CGCs) and mural granulosa cells (MGCs). Understanding their normal and steroid-induced functions is the key to understanding the pathophysiology of endocrinal diseases in women. Objective This study investigated the differentially expressed proteins by CGCs and MGCs of patients with polycystic ovarian syndrome (PCOS) and without subsequent exposure to dehydroepiandrosterone sulfate (DHEAS) and functional differentiation. Design The present study was observational and experimental study carried out in hospital involving 80 female patients undergoing IVF for infertility. Methods In this study, we isolated CGCs and MGCs from the follicular fluid of both PCOS and non-PCOS patients undergoing in vitro fertilization (IVF). The cells were cultured and treated with DHEAS for 48 hours, and these cells were extracted, digested, and analyzed by tandem mass spectrometry followed by processing of the results using open-source bioinformatics tools. Results The present investigation discovered 276 and 341 proteins in CGCs and MGCs, respectively. DHEAS reduced the number of proteins expressed by CGCs and MGCs to 34 and 57 from 91 and 94, respectively. Venn results of CGCs revealed 49, 53, 36, and 21 proteins in normal CGCs, PCOS-CGCs, post-DHEAS, and PCOS-CGCs, respectively. Venn analysis of MGCs showed 51 proteins specific to PCOS and 29 shared by normal and PCOS samples after DHEAS therapy. MGCs express the most binding and catalytic proteins, whereas CGCs express transporter-related proteins. A protein pathway study demonstrated considerable differences between normal and PCOS samples, while DHEAS-treated samples of both cell lines showed distinct pathways. String findings identified important network route components such as albumin, actin, apolipoprotein, complement component C3, and heat shock protein. Conclusion This is the first study to show how DHEAS-induced stress affects the expression of proteins by MGCs and CGCs isolated from normal and PCOS patients. Further studies are recommended to identify PCOS biomarkers from CGCs and MGCs expressed under the influence of DHEAS.
Collapse
Affiliation(s)
- Pankaj Pant
- Faculty of Pharmacy, DIT University, Dehradun, Uttarakhand, India
| | - Reema Sircar
- Indira IVF Hospital, Dehradun, Uttarakhand, India
| | - Ritu Prasad
- Morpheus Prasad International Hospital, Dehradun, Uttarakhand, India
| | - Hari Om Prasad
- Morpheus Prasad International Hospital, Dehradun, Uttarakhand, India
| | | |
Collapse
|
48
|
Bakhsh H, Alqntash N, Almajed E. The Successful Management of Primary Amenorrhea in Woodhouse-Sakati Syndrome: A Case Report and a Literature Review. Life (Basel) 2023; 13:2022. [PMID: 37895404 PMCID: PMC10608343 DOI: 10.3390/life13102022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Woodhouse-Sakati syndrome (WSS) is a rare multisystemic disease resulting from an autosomal recessive gene mutation characterized by distinctive facial appearance, alopecia, impaired HbA1c, and hypogonadism. PURPOSE To present the successful management of primary amenorrhea in a WSS patient. CASE PRESENTATION We report a 19-year-old Saudi female referred to the gynecology clinic at the age of 16 as a case of primary amenorrhea. The patient underwent a genetic analysis, which revealed mutations in the DCAF17 gene, confirming the diagnosis of WSS. Treatment includes hormonal replacement therapy for the induction of puberty. CONCLUSIONS Careful and detailed medical and physical examination led to appropriate testing confirming the WSS diagnosis. Genetic tests for family members and the offspring of the patient are strongly recommended. Treatment timing and dosage are determined by the patient's individual needs, which take into consideration the patient's potential for growth, the family's readiness, and any comorbidities.
Collapse
Affiliation(s)
- Hanadi Bakhsh
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia (E.A.)
- Department of Obstetrics and Gynecology, King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
| | - Norah Alqntash
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia (E.A.)
| | - Ebtesam Almajed
- Clinical Sciences Department, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia (E.A.)
| |
Collapse
|
49
|
Mosavi S, Rostami A, Pooladi M, ShojaeiBaghini M, Poudineh S, Poudineh M, Behzadi E. Icariin, A Novel Promising Complementary Therapeutic Strategy in the Management of Female Infertility: A Literature Review:. Galen Med J 2023; 12:e2528. [PMID: 37706169 PMCID: PMC10497255 DOI: 10.31661/gmj.v12i.2528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 09/15/2023] Open
Abstract
The prevalence of pre-menopausal female infertility is increasing considerably due to various causes such as environmental pollutants, increased administration of chemotherapeutics and radiation exposure, microbial infections, and genetic/epigenetic alterations. However, the current therapeutical strategies remain unfavorite as the disadvantages are strongly challenging. Icariin (ICI) is a phytoestrogen that exerts some promising properties in order to alleviate female infertility. Therefore, the current literature review aimed to evaluate the conducted studies regarding the beneficial impacts of ICI on the female reproductive system and female fecundity. The findings of the present study revealed that ICI is able to modulate the levels of reproductive hormones as it causes a significant decrement in the levels of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) while increasing the levels of estrogen and progesterone. Furthermore, the administration of ICI results in a dramatic alteration in the expression of sex steroids’ receptors, particularly in female reproductive tissues. In addition, preserving ovarian follicular reserve, improving the ovarian and uterine histoarchitecture, elongating the estrous cycle duration, and eventually advancing the female fecundity are other major effects of ICI on the female reproductive system. Despite these desired beneficial properties, the current knowledge appears to be insufficient, hence further investigations, particularly on humans, are encouraged. To the best of our knowledge, this review provides a comprehensive information regarding the beneficial effects of Icariin on female infertility for the firs time.
Collapse
Affiliation(s)
- Sima Mosavi
- Department of Obstetrics and Gynecology, School of medicine, Urmia university of medical sciences, Urmia, Iran
| | - Amirabbas Rostami
- Department of Internal Medicine, Faculty of General Medicine, Yerevan State Medical University aer Mkhitar Heratsi, Yerevan, Armenia
| | - Marzieh Pooladi
- Department of Anatomical Sciences, School Of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdie ShojaeiBaghini
- Medical Informatics Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Sahar Poudineh
- School of Medicine, Mashhad Azad University, Mashhad, Iran
| | | | - Esmaeil Behzadi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
50
|
Femi-Olabisi JF, Ishola AA, Olujimi FO. Effect of Parquetina nigrescens (Afzel.) Leaves on Letrozole-Induced PCOS in Rats: a Molecular Insight into Its Phytoconstituents. Appl Biochem Biotechnol 2023; 195:4744-4774. [PMID: 37171758 DOI: 10.1007/s12010-023-04537-3] [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] [Accepted: 04/11/2023] [Indexed: 05/13/2023]
Abstract
Polycystic ovarian syndrome (PCOS) is one of the common causes of female infertility in women of reproductive age. P. nigrescens is a plant used in the treatment of various diseases including menstrual disorders. This study investigated the effect of ethanolic extracts of P. nigrescens leaves on the estrous cycle, fasting blood glucose, and hormonal and lipid profile in letrozole-induced PCOS rats and also evaluated the molecular mechanism of the active constituents using computational methods. After the induction of PCOS with letrozole, rats were treated orally for 14 days with distilled water (1 mg/kg/day), clomiphene citrate (2 mg/kg/day), metformin (7.14 mg/kg/day), and ethanolic extract of P. nigrescens (50 and 100 mg). Thereafter, selected biochemical parameters were assayed to determine the extract's effect on the estrous cycle. Molecular docking and molecular dynamics simulation (MDS) were carried out to determine the binding affinity and relative stability of the ligand-receptor complexes. Letrozole-induced PCOS rats showed irregular estrous cyclicity, elevated (p > 0.05) triglycerides, low-density lipoprotein cholesterol (LDL), total cholesterol, insulin, testosterone, and luteinizing hormone (LH) concentration, low (p > 0.05) progesterone, low follicle-stimulating hormone (FSH), high-density lipoprotein cholesterol (HDL), and high fasting blood glucose concentration compared to that of the control group. The reproductive, biochemical, and structural alterations were reversed by the administration of ethanolic extract of P. nigrescens leaves (50 mg/kg) which restored the estrous cycle after 14 days of treatment. However, the ethanolic extracts of P. nigrescens (100 mg/kg) significantly increased (p > 0.05) FSH, HDL, and progesterone concentrations but decreased the LH, progesterone, and total cholesterol. Of all 44 compounds identified in GCMS analysis of an ethanolic extract of P. nigrescens leaves, only 2-ethylbutyl heptyl ester (CID 91705405) had a higher binding affinity for hormonal receptors and enzymes responsible for hepatic gluconeogenesis compared to standard drugs used in the study. CID 91705405 was also relatively stable over 100 ns of MDS. This compound is therefore revealed to have the potential to modulate both endocrine and metabolic pathways involved in PCOS. The ethanolic extract of P. nigrescens leaves can therefore be considered in the management/treatment of the reproductive and metabolic disorders related to PCOS subject to further experimental validation.
Collapse
Affiliation(s)
- Joy Fehintoluwa Femi-Olabisi
- Department of Biochemistry, College of Basic & Applied Sciences, Mountain Top University, Makogi Oba, Ogun State, Nigeria.
| | - Ahmed Adebayo Ishola
- Central Research Laboratory, 132B, University Road, Tanke, Ilorin, Kwara State, Nigeria
| | - Folakemi Omolara Olujimi
- Department of Biochemistry, College of Basic & Applied Sciences, Mountain Top University, Makogi Oba, Ogun State, Nigeria
| |
Collapse
|