1
|
Jia M, Wang J, Lin C, Zhang Q, Xue Y, Huang X, Ren Y, Chen C, Liu Y, Xu Y. Hydrogel Strategies for Female Reproduction Dysfunction. ACS NANO 2024. [PMID: 39437800 DOI: 10.1021/acsnano.4c05634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Infertility is an important issue for human reproductive health, with over half of all cases of infertility associated with female factors. Dysfunction of the complex female reproductive system may cause infertility. In clinical practice, female infertility is often treated with oral medications and/or surgical procedures, and ultimately with assisted reproductive technologies. Owing to their excellent biocompatibility, low immunogenicity, and adjustable mechanical properties, hydrogels are emerging as valuable tools in the reconstruction of organ function, supplemented by tissue engineering techniques to increase their structure and functionality. Hydrogel-based female reproductive reconstruction strategies targeting the pathological mechanisms of female infertility may provide alternatives for the treatment of ovarian, endometrium/uterine, and fallopian tube dysfunction. In this review, we provide a general introduction to the basic physiology and pathology of the female reproductive system, the limitations of current infertility treatments, and the lack of translation from animal models to human reproductive physiology. We further provide an overview of the current and future potential applications of hydrogels in the treatment of female reproductive system dysfunction, highlighting the great prospects of hydrogel-based strategies in the field of translational medicine, along with the significant challenges to be overcome.
Collapse
Affiliation(s)
- Minxuan Jia
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou, Guangdong 510535, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jiamin Wang
- Reproductive Medical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- The Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong 510080, China
| | - Chubing Lin
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou, Guangdong 510535, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Qingyan Zhang
- Reproductive Medical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- The Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong 510080, China
| | - Yueguang Xue
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou, Guangdong 510535, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Xin Huang
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
- GBA National Institute for Nanotechnology Innovation, Guangzhou, Guangdong 510535, China
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Yan Ren
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Chunying Chen
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Ying Liu
- New Cornerstone Science Laboratory, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Yanwen Xu
- Reproductive Medical Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- The Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, Guangdong 510080, China
| |
Collapse
|
2
|
Feng S, Luo Y, Chen Y, Zhu H, Zhao T, Ma F, Lin Y, Ning Y, Wu J. Individualized responses to acupuncture in premature ovarian insufficiency: A study protocol for a nested case-control trial with transcriptome analysis. Heliyon 2024; 10:e37859. [PMID: 39328559 PMCID: PMC11425121 DOI: 10.1016/j.heliyon.2024.e37859] [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: 01/11/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Background Premature ovarian insufficiency (POI), a modifiable cause of infertility with substantial implications for women's well-being, prompts the exploration of efficacious adjunctive treatments. Acupuncture emerges as a promising therapeutic avenue; however, the nuanced effects of acupuncture in POI warrant more comprehensive investigation. The intricate mechanisms dictating individualized responses remain elusive. This trial seeks to assess the effectiveness of acupuncture as an adjunctive treatment for POI, concurrently delving into the impact of transcriptome analysis on peripheral blood to unravel the underpinnings of these individual variations. The overarching objective is to enrich our comprehension of acupuncture's therapeutic potential in the context of POI, with a view to advancing holistic patient care. Methods/design Constituting an open-label, nested case-control study, this research endeavors to enroll 108 women diagnosed with POI. Participants will be randomly assigned in a 1:1 ratio to either the study group or the control group, each comprising 54 subjects. Ten patients from each group meeting specific criteria will partake in transcriptome analysis. An additional 10 subjects meeting the study criteria will form a healthy control group. The study group will exclusively undergo acupuncture treatment, while the control group will solely receive Fenmutong. Acupuncture sessions, administered thrice weekly across three menstrual cycles from the fifth day of menstruation, constitute the intervention. Primary outcome measurement rests on Follicle-Stimulating Hormone (FSH) levels, supplemented by secondary assessments encompassing biometric features, hormone biomarkers, anxiety and depression scores, and transcriptome analysis. Baseline measurements precede intervention, with post-intervention evaluations following. The study endeavors to discern specific genes linked to individualized responses to acupuncture. Data analysis, employing SPSS 25.0 software, incorporates a meticulous examination of peripheral blood samples for transcriptome analysis. The investigation aspires to shed light on genetic determinants influencing the effects of acupuncture on women with POI, thereby fostering elevated standards in patient care and management. Discussion This study pivots on the principal objective of scrutinizing the efficacy of acupuncture as an adjunctive treatment for POI. Beyond effectiveness, it undertakes an exploration of the intricate mechanisms underlying the diverse responses exhibited by individuals in the context of acupuncture, augmenting the depth of understanding in this therapeutic domain.
Collapse
Affiliation(s)
- Shiyu Feng
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu Luo
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yan Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Haimin Zhu
- Southern Medical University, Guangzhou 510515, China
| | - Tianqi Zhao
- Southern Medical University, Guangzhou 510515, China
| | - Fei Ma
- Department of Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Southern Medical University, Guangzhou 510515, China
| | - Yanting Lin
- Department of Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
| | - Yan Ning
- Department of Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Southern Medical University, Guangzhou 510515, China
| | - Jiaman Wu
- Department of Chinese Medicine, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen 518028, China
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
- Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
3
|
Wang YL, Yin LX, Li M. Assessment of left ventricular myocardial systolic dysfunction in premature ovarian insufficiency patients using echocardiographic layer-specific myocardial strain imaging. Echo Res Pract 2024; 11:20. [PMID: 39218983 PMCID: PMC11367989 DOI: 10.1186/s44156-024-00056-x] [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: 02/15/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Due to the lack of oestrogen, premature ovarian insufficiency (POI) is an independent risk factor for ischaemic heart disease and overall cardiovascular disease. This study aimed to apply layer-specific myocardial strain for early quantitative evaluation of subclinical left ventricular myocardial systolic function changes in patients with POI. METHODS Forty-eight newly diagnosed, untreated patients with POI (POI group) and fifty healthy female subjects matched for age, height and weight (control group) were enrolled. Standard transthoracic echocardiography was used to measure conventional parameters and layer-specific strain parameters.The layer-specific strain parameters included subendomyocardial global longitudinal strain (GLSendo), mid-layer myocardial global longitudinal strain (GLSmid), subepimyocardial global longitudinal strain (GLSepi), subendomyocardial global circumferential strain (GCSendo), mid-layer myocardial global circumferential strain (GCSmid), and subepimyocardial global circumferential strain (GCSepi). RESULTS There were no significant differences in age, body mass index (BMI), blood pressure, or left ventricular ejection fraction (LVEF) between the two groups. The end-diastolic interventricular septal thickness (IVST) was greater in the POI group (8.29 ± 1.32 vs. 7.66 ± 0.82, P = 0.008), and the POI group had lower E, E/A, and lateral e' (all P < 0.05). As for systolic functions,the POI group had lower GLSendo, GLSmid, GLSepi, GCSendo, GCSmid, and GCSepi (all P < 0.05).The intraobserver and interobserver coefficients of GLSendo, GLSmid, GLSepi, GCSendo, GCSmid, and GCSepi were greater than 0.900. CONCLUSIONS POI patients with normal LVEF may suffer from subclinical left ventricular myocardial systolic dysfunction. Echocardiography of layer-specific myocardial strain could more sensitively detect subclinical impairment of left ventricular systolic function in POI patients.
Collapse
Affiliation(s)
- Yu-Lin Wang
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Li-Xue Yin
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Mei Li
- Department of Gynaecology and Obstetrics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Rosa E Silva PH, Ferreira SR, Pravatta Rezende G, Angerame Yela D, Benetti-Pinto CL. Premature ovarian insufficiency: knowledge, attitudes, and quality of care offered by gynecologists. Menopause 2024; 31:796-800. [PMID: 38954487 DOI: 10.1097/gme.0000000000002389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
OBJECTIVE Premature ovarian insufficiency (POI) affects up to 3% of the global female population, influencing metabolic, cardiovascular, and reproductive health. Medical expertise in diagnosis, effects, and treatment strategies, particularly for gynecologists, is crucial for ensuring improved healthcare for women. The objective of this study is to assess the state of medical knowledge regarding the diagnosis, treatment, and follow-up of POI among Brazilian gynecologists. METHODS A cross-sectional study was conducted using online questionnaires administered to 16,000 members of the Brazilian Federation of Gynecology and Obstetrics. RESULTS In total, 460 questionnaires were received from gynecologists who had an average age of 44.49 ± 12.57 years and 19.37 ± 12.95 years of professional experience. Fifty-three percent of gynecologists diagnosed POI correctly, and 49% requested karyotype analysis, while fewer than 10% identified all POI etiologies. Over 90% of gynecologists understood the long-term consequences of POI for bone and cardiovascular health. Despite being a consequence of hypoestrogenism, hormone therapy was recommended only by 20% of doctors, with no more than 50% of them prescribing appropriate doses for young women. Regarding self-perception, 60% of gynecologists declared deficient knowledge regarding how to offer care and guidance to women, with hormone therapy being reported as the most important reason (47%). CONCLUSIONS Current concepts and guidelines for POI are not adequately understood or applied in Brazilian clinical practice, leading to suboptimal care.
Collapse
Affiliation(s)
- Pedro Henrique Rosa E Silva
- From the Department of Obstetrics and Gynecology, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | | | | | | | | |
Collapse
|
5
|
Kunicki M, Rzewuska N, Gross-Kępińska K. Immunophenotypic profiles and inflammatory markers in Premature Ovarian Insufficiency. J Reprod Immunol 2024; 164:104253. [PMID: 38776714 DOI: 10.1016/j.jri.2024.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/01/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Premature Ovarian Insufficiency (POI), also known as Premature Ovarian Failure (POF), is a heterogeneous disorder characterized by the cessation of ovarian function before age 40. Clinical symptoms include menstrual disorders: amenorrhea/oligomenorrhea or symptoms of estrogen deficiency. This review aims to provide the most important summary of the immunophenotypic profile of premature ovarian failure syndrome, along with a review of the latest reports on the usefulness of inflammatory markers. The inflammatory microenvironment in POI applies to many levels. Concomitants of autoimmune ovarian inflammation and impaired cellular immune response may be a picture of impaired regulation in autoimmune ovarian disease. The serum concentration of pro-inflammatory cytokines, like IL-6, IL-8, IL-17, tumor necrosis factor α (TNF-α), and interferon-gamma (IFN-γ), tend to increase, whereas levels of the anti-inflammatory cytokine, IL-10, tend to decrease. In our review, we focus on whether the measured immunological parameters could help in the diagnosis and prognosis of the syndrome. Among the inflammatory markers, neutrophil-to-lymphocyte ratio (NLR) is noteworthy, as it is decreased in patients with POI. It is important to stress that besides case series, we need properly powered studies with randomization to answer which treatment is effective, and how to deal with concurrent autoimmunity. In this review, we emphasize the importance of the premature ovarian failure syndrome immunoprofile for a proper understanding of the complexity of this syndrome, potential diagnostic points, and therapeutic targets.
Collapse
Affiliation(s)
- Michał Kunicki
- Department of Gynecological Endocrinology, Medical University of Warsaw, Warsaw 00-315, Poland; INVICTA Fertility and Reproductive Center, Warsaw 00-019, Poland
| | - Natalia Rzewuska
- Department of Gynecological Endocrinology, Medical University of Warsaw, Warsaw 00-315, Poland.
| | | |
Collapse
|
6
|
Yi Y, Feng Y, Shi Y, Xiao J, Liu M, Wang K. Per- and Polyfluoroalkyl Substances (PFASs) and Their Potential Effects on Female Reproductive Diseases. TOXICS 2024; 12:539. [PMID: 39195641 PMCID: PMC11358978 DOI: 10.3390/toxics12080539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Per- and polyfluoroalkyl substances (PFASs) are a class of anthropogenic organic compounds widely present in the natural and human living environments. These emerging persistent pollutants can enter the human body through multiple channels, posing risks to human health. In particular, exposure to PFASs in women may cause a series of reproductive health hazards and infertility. Based on a review of the existing literature, this study preliminarily summarizes the effects of PFAS exposure on the occurrence and development of female reproductive endocrine diseases, such as polycystic ovary syndrome (PCOS), endometriosis, primary ovarian insufficiency (POI), and diminished ovarian reserve (DOR). Furthermore, we outline the relevant mechanisms through which PFASs interfere with the physiological function of the female ovary and finally highlight the role played by nutrients in reducing the reproductive health hazards caused by PFASs. It is worth noting that the physiological mechanisms of PFASs in the above diseases are still unclear. Therefore, it is necessary to further study the molecular mechanisms of PFASs in female reproductive diseases and the role of nutrients in this process.
Collapse
Affiliation(s)
- Yuqing Yi
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yang Feng
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Yuechen Shi
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Jiaming Xiao
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ming Liu
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, Dalian 116044, China
| | - Ke Wang
- Department of Clinical Nutrition, Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China; (Y.Y.); (Y.F.); (Y.S.); (J.X.); (M.L.)
| |
Collapse
|
7
|
Franza L, Caldarelli M, Villani ER, Cianci R. Sex Differences in Cardiovascular Diseases: Exploring the Role of Microbiota and Immunity. Biomedicines 2024; 12:1645. [PMID: 39200110 PMCID: PMC11352091 DOI: 10.3390/biomedicines12081645] [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: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of mortality and morbidity in Western countries, thus representing a global health concern. CVDs show different patterns in terms of the prevalence and presentation in men and women. The role of sex hormones has been extensively implicated in these sex-specific differences, due to the presence of the menstrual cycle and menopause in women. Moreover, the gut microbiota (GM) has been implicated in cardiovascular health, considering the growing evidence that it is involved in determining the development of specific diseases. In particular, gut-derived metabolites have been linked to CVDs and kidney disorders, which can in turn promote the progression of CVDs. Considering the differences in the composition of GM between men and women, it is possible that gut microbiota act as a mediator in regard to the sex disparities in CVDs. This narrative review aims to comprehensively review the interplay between sex, GM, and CVDs, discussing potential mechanisms and therapeutic options.
Collapse
Affiliation(s)
- Laura Franza
- Emergency, Anesthesiological and Reanimation Sciences Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS of Rome, 00168 Rome, Italy;
- Emergency Department, Azienda Ospedaliero-Universitaria di Modena, Largo del Pozzo, 71, 41125 Modena, Italy
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Emanuele Rocco Villani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- UOC Geriatra-Disturbi Cognitivi e Demenze, Dipartimento di Cure Primarie, AUSL Modena, 41012 Modena, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
8
|
Yang K, Wu L, Zeng L, Xiang W, Chen J, Yan Y, Hao M, Song T, Zhai E, Zhang G, Liu H. Exploring the effect of Gouqi Nuzhen Liuhe decoction on the PI3K/mTOR signaling pathway for premature ovarian insufficiency based on system pharmacology. Heliyon 2024; 10:e33105. [PMID: 38994091 PMCID: PMC11238132 DOI: 10.1016/j.heliyon.2024.e33105] [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: 02/21/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Objective To explore the effect of Gouqi Nuzhen Liuhe Decoction (GNLHD) on the PI3K/mTOR Signaling Pathway for Premature Ovarian Insufficiency (POI) based on system pharmacology. Methods First, the system pharmacology approach was used to predict the mechanism of GNLHD. Then, mice were randomly divided into model group, positive group, GNLHD high-dose group, GNLHD medium-dose group, and GNLHD low-dose group. Hematoxylin-eosin (HE) staining was used to observe the pathological changes of ovarian tissue under light microscope. The expression levels of estradiol (E2), follicle-stimulating hormone (FSH) and luteinizing hormone (LH) were detected by enzyme-linked immunosorbent assay. The expressions of PI3K, AKT1 and mTOR proteins in ovarian tissue were detected by immunohistochemistry. Results The results of system pharmacology showed that GNLHD may regulate biological processes and signaling pathways such as: reproductive structure development, reproductive system development, Oocyte meiosis and so on. Compared with the model group, the levels of E2 in the GNLHD group were increased, and the levels of FSH and LH were decreased (P < 0.05). Compared with the model group, the number of mature follicles in the GNLHD group was significantly increased, the number of atretic follicles was relatively decreased, and the expressions of PI3K, AKT1, and MTOR proteins in the GNLHD group were significantly increased (P < 0.05). Conclusion GNLHD may improve the ovarian function of POI mice by affecting the expression of PI3K, AKT1 and mTOR proteins, promote the growth and development of follicles, increase the E2 level, reduce FSH and LH level, and maintain the stability of the ovarian internal environment.
Collapse
Affiliation(s)
- Kailin Yang
- Hunan Provincial Key Laboratory of Translational Medicine for Traditional Chinese Medicine Prescription Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, China
| | - Lingyu Wu
- Hunan Provincial Key Laboratory of Translational Medicine for Traditional Chinese Medicine Prescription Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Liuting Zeng
- Hunan Provincial Key Laboratory of Translational Medicine for Traditional Chinese Medicine Prescription Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde City, China
| | - Junpeng Chen
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, China
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, China
| | - Tian Song
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Enjian Zhai
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao, China
| | - Guomin Zhang
- Hunan Provincial Key Laboratory of Translational Medicine for Traditional Chinese Medicine Prescription Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Huiping Liu
- Hunan Provincial Key Laboratory of Translational Medicine for Traditional Chinese Medicine Prescription Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
9
|
Wang R, Lv Y, Dou T, Yang Q, Yu C, Guan Q. Autoimmune thyroid disease and ovarian hypofunction: a review of literature. J Ovarian Res 2024; 17:125. [PMID: 38877588 PMCID: PMC11177435 DOI: 10.1186/s13048-024-01451-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: 09/23/2023] [Accepted: 06/10/2024] [Indexed: 06/16/2024] Open
Abstract
Thyroid hormones(THs) are essential for the proper functioning of the ovaries, and multiple studies have shown that thyroid abnormalities, especially during adolescence and reproductive age, can lead to lifelong ovarian dysfunction. Autoimmune thyroid disease (AITD), one of the most common organ specific autoimmune diseases, is mainly mediated by cellular autoimmune reactions, and has strong inflammatory infiltration and immune active cells, including chemokines and cytokines, which are important components of ovarian aging. This suggests that autoimmune and inflammatory molecular processes may play a role in the emergence of ovarian dysfunction. The purpose of this review is to summarize recent in vivo and in vitro evidence of a complex relationship between AITD and ovarian dysfunction. AITD is closely related to the decline of ovarian function from the perspective of antibody, cytokine, oxidative stress, and genetic factors. Finally, some of the currently known treatments for AITD and hypo ovarian disease are summarized.
Collapse
Affiliation(s)
- Ru Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging,Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital of Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Youyuan Lv
- Internal Medicine Department of the Second Affiliated Hospital of Shandong University, Jinan, 250021, Shandong, China
| | - Tao Dou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging,Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital of Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Qian Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging,Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital of Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Chunxiao Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging,Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital of Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
| | - Qingbo Guan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging,Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital of Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, 250021, Shandong, China.
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
| |
Collapse
|
10
|
Blümel JE, Chedraui P, Vallejo MS, Dextre M, Elizalde A, Escalante C, Monterrosa-Castro A, Ñañez M, Ojeda E, Rey C, Rodríguez D, Rodrigues MA, Salinas C, Tserotas K. Genitourinary symptoms and sexual function in women with primary ovarian insufficiency. Climacteric 2024; 27:269-274. [PMID: 38308574 DOI: 10.1080/13697137.2024.2306278] [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: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 02/05/2024]
Abstract
OBJECTIVE There are limited studies on urogenital symptoms in women who experience menopause before the age of 40 years due to primary ovarian insufficiency (POI) or bilateral oophorectomy (surgical POI). This study aimed to compare the urogenital symptoms, including sexuality, of women with POI to those without the condition. METHODS This cross-sectional study conducted was in seven Latin American countries, in which postmenopausal women (with POI and non-POI) were surveyed with a general questionnaire, the Menopause Rating Scale (MRS) and the six-item Female Sexual Function Index (FSFI-6). The association of premature menopause with more urogenital symptoms and lower sexual function was evaluated with logistic regression analysis. RESULTS Women with POI experience more urogenital symptoms (MRS urogenital score: 3.54 ± 3.16 vs. 3.15 ± 2.89, p < 0.05) and have lower sexual function (total FSFI-6 score: 13.71 ± 7.55 vs. 14.77 ± 7.57 p < 0.05) than women who experience menopause at a normal age range. There were no significant differences in symptoms when comparing women based on the type of POI (idiopathic or surgical). After adjusting for covariates, our logistic regression model determined that POI is associated with more urogenital symptoms (odds ratio [OR]: 1.38, 95% confidence interval [CI] 1.06-1.80) and lower sexual function (OR: 1.67, 95% CI 1.25-2.25). CONCLUSION POI, whether idiopathic or secondary to bilateral oophorectomy, is associated with symptoms that affect vaginal and sexual health.
Collapse
Affiliation(s)
- J E Blümel
- Departamento de Medicina Interna Sur, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - P Chedraui
- Escuela de Posgrado en Salud, Universidad Espíritu Santo, Samborondón, Ecuador
- Facultad de Ciencias de la Salud, Universidad Católica 'Nuestra Señora de la Asunción', Asunción, Paraguay
| | - M S Vallejo
- Servicio de Obstetricia y Ginecología, Hospital Clínico de la Universidad de Chile, Santiago de Chile, Chile
| | - M Dextre
- Ginecología Obstetricia, Clínica Internacional, Lima, Perú
| | - A Elizalde
- Departamento de la Mujer, Niñez y Adolescencia, Facultad de Medicina de la Universidad Nacional del Nordeste, Corrientes, Argentina
| | - C Escalante
- Departamento de Ginecología, Facultad de Medicina, Universidad de Costa Rica, Costa Rica
| | - A Monterrosa-Castro
- Grupo de Investigación Salud de la Mujer, Universidad de Cartagena, Cartagena, Colombia
| | - M Ñañez
- II Cátedra de Ginecología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - E Ojeda
- Departamento Académico de Medicina Humana, Universidad Andina del Cusco, Cusco, Perú
| | - C Rey
- Asociación Argentina para el Estudio del Climaterio, Buenos Aires, Argentina
| | - D Rodríguez
- Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M A Rodrigues
- Gynecology and Obstetrics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - C Salinas
- Obstetricia y Ginecología, Hospital Ángeles, Puebla, México
| | - K Tserotas
- Clínica Tserotas, Ciudad de Panamá, Panamá
| |
Collapse
|
11
|
Greene DN, Goldstein Z, Krasowski MD. Uncertainty in Hormone Monitoring of Transgender and Non-Binary People. J Appl Lab Med 2024; 9:426-429. [PMID: 38452237 DOI: 10.1093/jalm/jfae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 03/09/2024]
Affiliation(s)
- Dina N Greene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Zil Goldstein
- Callen-Lorde Community Health Center, New York, NY, United States
- City University of New York, Graduate School of Public Health & Health Policy, New York, NY, United States
| | - Matthew D Krasowski
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
12
|
Disha B, Mathew RP, Dalal AB, Mahato AK, Satyamoorthy K, Singh KK, Thangaraj K, Govindaraj P. Mitochondria in biology and medicine - 2023. Mitochondrion 2024; 76:101853. [PMID: 38423268 DOI: 10.1016/j.mito.2024.101853] [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: 11/27/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Mitochondria are an indispensable part of the cell that plays a crucial role in regulating various signaling pathways, energy metabolism, cell differentiation, proliferation, and cell death. Since mitochondria have their own genetic material, they differ from their nuclear counterparts, and dysregulation is responsible for a broad spectrum of diseases. Mitochondrial dysfunction is associated with several disorders, including neuro-muscular disorders, cancer, and premature aging, among others. The intricacy of the field is due to the cross-talk between nuclear and mitochondrial genes, which has also improved our knowledge of mitochondrial functions and their pathogenesis. Therefore, interdisciplinary research and communication are crucial for mitochondrial biology and medicine due to the challenges they pose for diagnosis and treatment. The ninth annual conference of the Society for Mitochondria Research and Medicine (SMRM)- India, titled "Mitochondria in Biology and Medicine" was organized at the Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India, on June 21-23, 2023. The latest advancements in the field of mitochondrial biology and medicine were discussed at the conference. In this article, we summarize the entire event for the benefit of researchers working in the field of mitochondrial biology and medicine.
Collapse
Affiliation(s)
- B Disha
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India; Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Rohan Peter Mathew
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India; Manipal Academy of Higher Education, Manipal 576104, India
| | - Ashwin B Dalal
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India
| | - Ajay K Mahato
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India
| | - Kapaettu Satyamoorthy
- Shri Dharmasthala Manjunatheshwara (SDM) University, SDM College of Medical Sciences and Hospital, Manjushree Nagar, Sattur, Dharwad 580009, India
| | - Keshav K Singh
- Department of Genetics, School of Medicine, The University of Alabama at Birmingham, Kaul Genetics Building, Rm. 620, 720 20th St. South, Birmingham, AL, 35294, USA
| | - Kumarasamy Thangaraj
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| | - Periyasamy Govindaraj
- Centre for DNA Fingerprinting and Diagnostics, Uppal, Hyderabad 500039, India; Department of Neuropathology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru 560029, India.
| |
Collapse
|
13
|
Xing Z, Kirby RS. Age at natural or surgical menopause, all-cause mortality, and lifespan among postmenopausal women in the United States. Menopause 2024; 31:176-185. [PMID: 38385730 DOI: 10.1097/gme.0000000000002314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
OBJECTIVE This study investigated the association of age at natural menopause with or without undergoing hysterectomy and/or bilateral oophorectomy after menopause and age at surgical menopause with all-cause mortality and lifespan in postmenopausal women. METHODS The data stemmed from the National Health and Nutrition Examination Survey (NHANES) (1999-2018) and NHANES III (1988-1994), including 14,161 postmenopausal women over 40. Cox proportional hazard models were used to estimate unadjusted and adjusted hazard ratios (HRs) (95% confidence intervals [CIs]). We also used Cox proportional hazard models with penalized splines to depict the association between continuous age at menopause and all-cause mortality and nonparametric regression with smoothing splines to illustrate the association between age at menopause and lifespan in deceased participants. RESULTS The adjusted HRs (95% CIs) for age at natural menopause of <40, 40 to 44, and 55+ years in women without undergoing hysterectomy or bilateral oophorectomy after menopause were 1.48 (1.15-1.91), 1.16 (1.00-1.35), and 0.91 (0.77-1.07) compared with age at natural menopause of 45 to 54, respectively. The respective HRs (95% CIs) for age at surgical menopause were 1.39 (1.11-1.75), 1.09 (0.86-1.38), and 0.83 (0.53-1.32). However, no significant association was found between age at natural menopause and all-cause mortality among women undergoing hysterectomy and/or bilateral oophorectomy after menopause. When treated as continuous variables, age at natural menopause without undergoing hysterectomy or bilateral oophorectomy after menopause presented inverse and nonlinear associations with all-cause mortality, whereas age at surgical menopause was linearly inversely associated with all-cause mortality. The association between age at menopause and lifespan was linearly positive regardless of menopausal type. CONCLUSION Young age at menopause was associated with increased risks of all-cause mortality. The later menopause age was related to a longer lifespan.
Collapse
Affiliation(s)
- Zailing Xing
- From the Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida
| | - Russell S Kirby
- Chiles Center, College of Public Health, University of South Florida, Tampa, FL
| |
Collapse
|
14
|
Kim HK, Kim TJ. Current Status and Future Prospects of Stem Cell Therapy for Infertile Patients with Premature Ovarian Insufficiency. Biomolecules 2024; 14:242. [PMID: 38397479 PMCID: PMC10887045 DOI: 10.3390/biom14020242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Premature ovarian insufficiency (POI), also known as premature menopause or premature ovarian failure, signifies the partial or complete loss of ovarian endocrine function and fertility before 40 years of age. This condition affects approximately 1% of women of childbearing age. Although 5-10% of patients may conceive naturally, conventional infertility treatments, including assisted reproductive technology, often prove ineffective for the majority. For infertile patients with POI, oocyte donation or adoption exist, although a prevalent desire persists among them to have biological children. Stem cells, which are characterized by their undifferentiated nature, self-renewal capability, and potential to differentiate into various cell types, have emerged as promising avenues for treating POI. Stem cell therapy can potentially reverse the diminished ovarian endocrine function and restore fertility. Beyond direct POI therapy, stem cells show promise in supplementary applications such as ovarian tissue cryopreservation and tissue engineering. However, technological and ethical challenges hinder the widespread clinical application of stem cells. This review examines the current landscape of stem cell therapy for POI, underscoring the importance of comprehensive assessments that acknowledge the diversity of cell types and functions. Additionally, this review scrutinizes the limitations and prospects associated with the clinical implementation of stem cell treatments for POI.
Collapse
Affiliation(s)
- Hye Kyeong Kim
- Department of Obstetrics & Gynecology, Infertility Center, CHA University Ilsan Medical Center, Goyang 10414, Republic of Korea;
| | - Tae Jin Kim
- Department of Urology, CHA University Ilsan Medical Center, CHA University School of Medicine, Goyang 10414, Republic of Korea
| |
Collapse
|
15
|
Zhang S, Yahaya BH, Pan Y, Liu Y, Lin J. Menstrual blood-derived endometrial stem cell, a unique and promising alternative in the stem cell-based therapy for chemotherapy-induced premature ovarian insufficiency. Stem Cell Res Ther 2023; 14:327. [PMID: 37957675 PMCID: PMC10644549 DOI: 10.1186/s13287-023-03551-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: 12/16/2022] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Chemotherapy can cause ovarian dysfunction and infertility since the ovary is extremely sensitive to chemotherapeutic drugs. Apart from the indispensable role of the ovary in the overall hormonal milieu, ovarian dysfunction also affects many other organ systems and functions including sexuality, bones, the cardiovascular system, and neurocognitive function. Although conventional hormone replacement therapy can partly relieve the adverse symptoms of premature ovarian insufficiency (POI), the treatment cannot fundamentally prevent deterioration of POI. Therefore, effective treatments to improve chemotherapy-induced POI are urgently needed, especially for patients desiring fertility preservation. Recently, mesenchymal stem cell (MSC)-based therapies have resulted in promising improvements in chemotherapy-induced ovary dysfunction by enhancing the anti-apoptotic capacity of ovarian cells, preventing ovarian follicular atresia, promoting angiogenesis and improving injured ovarian structure and the pregnancy rate. These improvements are mainly attributed to MSC-derived biological factors, functional RNAs, and even mitochondria, which are directly secreted or indirectly translocated with extracellular vesicles (microvesicles and exosomes) to repair ovarian dysfunction. Additionally, as a novel source of MSCs, menstrual blood-derived endometrial stem cells (MenSCs) have exhibited promising therapeutic effects in various diseases due to their comprehensive advantages, such as periodic and non-invasive sample collection, abundant sources, regular donation and autologous transplantation. Therefore, this review summarizes the efficacy of MSCs transplantation in improving chemotherapy-induced POI and analyzes the underlying mechanism, and further discusses the benefit and existing challenges in promoting the clinical application of MenSCs in chemotherapy-induced POI.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, , China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| |
Collapse
|
16
|
Li HF, Zhang JX, Chen WJ. Dissecting the efficacy of the use of acupuncture and Chinese herbal medicine for the treatment of premature ovarian insufficiency (POI): A systematic review and metaanalysis. Heliyon 2023; 9:e20498. [PMID: 37818004 PMCID: PMC10560776 DOI: 10.1016/j.heliyon.2023.e20498] [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: 12/15/2022] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/12/2023] Open
Abstract
Premature ovarian insufficiency is a multi-factor gynecological disease that has become a major global health problem. In recent years, several trials have explored the treatment of premature ovarian insufficiency using Chinese herbal medicine and acupuncture, but the efficacy and safety of this combination remains controversial. This systematic review and meta-analysis aimed to comprehensively evaluate the efficacy and safety of combining Chinese herbal medicine with acupuncture to treat premature ovarian insufficiency. From eight different databases, we retrieved randomized controlled trials wherein Chinese herbal medicine and acupuncture had been compared with western medicine in the treatment of premature ovarian insufficiency. The bias risk assessment stipulated by the Cochrane Collaboration's tool was utilized to evaluate the quality of the chosen randomized controlled trials. This meta-analysis was executed with the help of Review Manager 5.3 and Stata 10.0. The quality of evidence was assessed using the Grading of Recommendations, Assessment, Development, and Evaluation framework. A total of 10 randomized controlled trials involving 594 premature ovarian insufficiency patients were included in the analysis. Compared with western medicine, co-treatment with acupuncture and Chinese herbal medicine exhibited a significantly higher total effective rate (relative risk: 1.21; 95% confidence interval: 1.12-1.31; P < 0.01, I2 = 0%), but lower levels of luteinizing hormone (standardized mean difference: -0.57; 95% confidence interval: -1.06, -0.08; P < 0.05, I2 = 80%), follicle-stimulating hormone, and Kupperman index score. Moreover, the combined intervention increased estradiol level in the serum. Overall, the data demonstrate that acupuncture plus Chinese herbal medicine is an efficacious and safe treatment option for POI patients. These findings must be verified by conducting large-scale, multicenter, high-quality, and long-term randomized controlled trials.
Collapse
Affiliation(s)
- Hui-fang Li
- Department of Gynecology, Tongxiang Maternal and Child Health-Care Center, Tongxiang, Zhejiang, 314500, PR China
| | - Jin-xia Zhang
- Department of Gynecology, Tongxiang Maternal and Child Health-Care Center, Tongxiang, Zhejiang, 314500, PR China
| | - Wen-jun Chen
- Hangzhou Medical College, Hangzhou, Zhejiang, 311399, China
- Department of Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
17
|
Shen C, Jiang Y, Lin J, He Y, Liu Y, Fang D. SIRT6 reduces the symptoms of premature ovarian failure and alleviates oxidative stress and apoptosis in granulosa cells by degrading p66SHC via H3K9AC. Gynecol Endocrinol 2023; 39:2250003. [PMID: 37634527 DOI: 10.1080/09513590.2023.2250003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
CONTEXT Substantial evidence suggests that ovarian oxidative stress can result in severe ovarian dysfunction. OBJECTIVE The purpose of this article is to investigate the potential of SIRT6 in alleviating premature ovarian failure (POF) by inhibiting oxidative stress. METHODS To mimic POF, mice were administered daily subcutaneous injections of d-galactose. The levels of E2, FSH, LH, AMH, and progesterone in serum were measured, along with changes in follicles and SIRT6 levels. Mice were treated with the SIRT6 agonist MDL-800, SIRT6 levels, follicles, and aforementioned hormones were reassessed. The effects of MDL-800 on oxidative stress and apoptosis were subsequently identified. Primary granulosa cells were isolated from mice, and the effects of H2O2 and MDL-800 on cell viability, oxidative stress, SIRT6 level, and apoptosis were evaluated. In addition, the regulation of SIRT6 on H3K9AC/p66SHC was verified by examining changes in protein levels, promoter activity, and the reversal effects of p66SHC overexpression. RESULTS MDL-800 mitigated hormone fluctuations, reduced follicle depletion in ovarian tissue, and attenuated oxidative stress and apoptosis in mice. In vitro experiments demonstrated that MDL-800 enhanced the resilience of primary granulosa cells against H2O2, as evidenced by increased cell viability and reduced oxidative stress and apoptosis. Furthermore, SIRT6 was found to decrease H3K9AC and p66SHC levels, as well as attenuate p66SHC promoter activity. The protective effects of MDL-800 on cells were reversed upon p66SHC overexpression. CONCLUSION In summary, this study highlights that activation of SIRT6 can alleviate POF and reduce oxidative stress by degrading H3K9AC and suppressing p66Shc levels in granulosa cells.
Collapse
Affiliation(s)
- Chuan Shen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| | - Jia Lin
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yibei He
- Department of Laboratory Medicine, Chengdu Chenghua District Maternal and Child Health Hospital, Chengdu, Sichuan, P.R. China
| | - Yue Liu
- Department of Laboratory Medicine, Chengdu Chenghua District Maternal and Child Health Hospital, Chengdu, Sichuan, P.R. China
| | - Dingzhi Fang
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
18
|
Fisher SA, Grijalva M, Guo R, Johnston SA, Laurent LC, Nguyen H, Renz J, Rosario JG, Rudich S, Gregory BD, Kim J, O’Neill K. Systematic Sampling of the Female Reproductive System for Molecular Characterization. Curr Protoc 2023; 3:e848. [PMID: 37584588 PMCID: PMC10575691 DOI: 10.1002/cpz1.848] [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] [Indexed: 08/17/2023]
Abstract
As part of the National Institutes of Health Human BioMolecular Atlas Program to develop a global platform to map the 37 trillion cells in the adult human body, we are generating a comprehensive molecular characterization of the female reproductive system. Data gathered from multiple single-cell/single-nucleus and spatial molecular assays will be used to build a 3D molecular atlas. Herein, we describe our multistep protocol, beginning with an optimized organ procurement workflow that maintains functional characteristics of the uterus, ovaries, and fallopian tubes by perfusing these organs with preservation solution. We have also developed a structured tissue sampling procedure that retains information on individual-level anatomic, physiologic, and individual diversity of the female reproductive system, toward full exploration of the function and structure of female reproductive cells. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Preparation and preservation of the female reproductive system (ovaries, fallopian tubes, and uterus) prior to procurement Basic Protocol 2: Removal of the female reproductive system en bloc Basic Protocol 3: Postsurgical dissection of ovaries Basic Protocol 4: Postsurgical dissection of fallopian tubes Basic Protocol 5: Postsurgical dissection of cervix Basic Protocol 6: Postsurgical dissection of uterine body Support Protocol 1: OCT-embedded tissue protocol Support Protocol 2: Tissue fixation protocol Support Protocol 3: Snap-frozen tissue protocol Basic Protocol 7: Tissue slice preparation for Visium analysis Support Protocol 4: Hematoxylin and eosin staining for 10X Visium imaging Basic Protocol 8: Manual tissue dissociation for Multiome analysis Basic Protocol 9: Tissue dissociation for Multiome analysis using S2 Singulator.
Collapse
Affiliation(s)
- Stephen A Fisher
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Marielena Grijalva
- 421 Curie Blvd., BRB II/III rm 1342, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Rong Guo
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sarah A Johnston
- 421 Curie Blvd., BRB II/III rm 1342, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - Hieu Nguyen
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - John Renz
- Gift of Life, 5901 Broken Sound Pkwy NW, Suite 600, Boca Raton, FL 33487 USA
| | - Jean G Rosario
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Steven Rudich
- Gift of Life, 5901 Broken Sound Pkwy NW, Suite 600, Boca Raton, FL 33487 USA
| | - Brian D Gregory
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Junhyong Kim
- 433 S. University Ave., Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Kathleen O’Neill
- 421 Curie Blvd., BRB II/III rm 1342, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
19
|
Lu Y, Wei Y, Shen X, Tong Y, Lu J, Zhang Y, Ma Y, Zhang R. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles improve ovarian function in rats with primary ovarian insufficiency by carrying miR-145-5p. J Reprod Immunol 2023; 158:103971. [PMID: 37329866 DOI: 10.1016/j.jri.2023.103971] [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: 02/27/2023] [Revised: 05/17/2023] [Accepted: 05/28/2023] [Indexed: 06/19/2023]
Abstract
OBJECTIVE Stem cell/exosome therapy is a novel strategy for primary ovarian insufficiency (POI). This paper is to examine the role of human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hUCMSC-EVs) in POI. METHODS hUCMSC-EVs were extracted and identified. POI rats were induced by cyclophosphamide for 15 days and treated with EV or GW4869 every 5 days and euthanized 28 days later. Vaginal smears were observed for 21 days. Serum hormone levels (FSH/E2/AMH) were measured by ELISA. Ovarian morphology, follicle numbers, and granulosa cell (GC) apoptosis were observed by HE and TUNEL staining. GCs extracted from Swiss albino rats were cyclophosphamide-induced to establish the POI cell model, followed by oxidative injury and apoptosis evaluation with the help of DCF-DA fluorescence, ELISA, and flow cytometry. The relation between miR-145-5p and XBP1 was predicted on StarBase and validated by dual-luciferase assay. miR-145-5p and XBP1 levels were measured by RT-qPCR and Western blot. RESULTS EV treatment reduced irregular estrus cycle incidence since day 7, increased E2 and AMH levels and all-stage follicle numbers, reduced FSH level, GC apoptosis, and atretic follicle numbers in POI rats. EV treatment diminished GC oxidative injury and apoptosis in vitro. miR-145-5p knockdown in hUCMSC-EVs partly abolished hUCMSC-EV-mediated effects on GCs and ovarian function in vivo and on GC oxidative injury and apoptosis in vitro. Silencing XBP1 partially negated miR-145-5p knockdown-exerted effects on GCs in vitro. CONCLUSION miR-145-5p carried by hUCMSC-EVs attenuates GC oxidative injury and apoptosis and thus extenuates ovarian injury and improves ovarian function in POI rats.
Collapse
Affiliation(s)
- Yanyang Lu
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Ying Wei
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Xiaoqin Shen
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Yixi Tong
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Jin Lu
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Yahui Zhang
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Yun Ma
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China
| | - Rong Zhang
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, N0.1055, Sanxiang Road, Suzhou 215000, China.
| |
Collapse
|
20
|
Pu X, Zhang L, Zhang P, Xu Y, Wang J, Zhao X, Dai Z, Zhou H, Zhao S, Fan A. Human UC-MSC-derived exosomes facilitate ovarian renovation in rats with chemotherapy-induced premature ovarian insufficiency. Front Endocrinol (Lausanne) 2023; 14:1205901. [PMID: 37564988 PMCID: PMC10411896 DOI: 10.3389/fendo.2023.1205901] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
Premature ovarian insufficiency (POI) induced by chemotherapy is an intractable disorder with a considerable incidence that commonly results in insufficient fertility and concomitant complications in female patients. Due to limitations in the current progress in POI diagnosis and treatment, there is an urgent need to develop novel remedies to improve ovarian function and protect fertility. The ameliorative effect of human umbilical cord mesenchymal stem cells (hUCMSCs) and exosomes derived from them in POI treatment could be a new hope for patients. Herein, we identified exosomes from hUCMSCs (hUCMSC-Exos). Then, systematic infusion of hUCMSC-Exos was accomplished via tail intravenous injection to investigate the feasibility of the treatment of rats with chemotherapy-induced POI by intraperitoneal injection of cyclophosphamide (CTX) and busulfan (BUS). Ovarian functions in the indicated group were evaluated, including oestrous cycle, serum sex hormone levels, follicle counts, ovarian pathological changes, proliferation and apoptosis of granulosa cells (GCs), and reproductive ability testing. Furthermore, the potential influence of hUCMSC-Exos on ovarian tissues was illuminated by conducting RNA-seq and multifaceted bioinformatics analyses. POI rats with hUCMSC-Exos transplantation exhibited a decrease in follicle-stimulating hormone (FSH) and apoptosis of GCs but an increase in oestradiol (E2), anti-Müllerian hormone (AMH), and the number of ovarian follicles and foetuses in the uterus. And the immunomodulation- and cellular vitality-associated gene sets in rats had also undergone moderate changes. Our data indicated the feasibility of hUCMSC-Exos in improving ovarian function and protecting fertility in chemotherapy-induced POI rats. HUCMSC-Exos can improve the local microenvironment of ovarian tissue in POI rats by participating in immune regulation, cellular viability, inflammation regulation, fibrosis and metabolism, and other related signal pathways.
Collapse
Affiliation(s)
- Xiaodi Pu
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Leisheng Zhang
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Peiyu Zhang
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Yaqiong Xu
- Department of Obstetrics and Gynecology, Guizhou Medical University, Guiyang, China
| | - Jun Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaomei Zhao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhihua Dai
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd., Guiyang, China
| | - Hua Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shuyun Zhao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology of the Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Anran Fan
- Key Laboratory of Reproductive Medicine, Stem Cell and Tissue Engineering Research Center in Guizhou Province, Guizhou Medical University, Guiyang, China
| |
Collapse
|
21
|
Meng X, Zhang S, Zhao L, Wang Y. Hydrogen-rich water treatment targets RT1-Db1 and RT1-Bb to alleviate premature ovarian failure in rats. PeerJ 2023; 11:e15564. [PMID: 37397014 PMCID: PMC10314742 DOI: 10.7717/peerj.15564] [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: 02/14/2023] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
Background Premature ovarian failure (POF) is defined as the cessation of ovarian function before the age of 40 years, imposing a significant health burden on patients. However, effective etiological therapy for POF is scarce. Thus, we aimed to explore the protective role and targets of hydrogen-rich water (HRW) in POF. Methods Based on cyclophosphamide (CTX)-induced POF rat models, the protective role of HRW treatment was mainly determined through serum 17-β-estradiol (E2), follicle-stimulating hormone (FSH), anti-mullerian hormone (AMH) levels, ovarian histomorphological analysis, and TUNEL assay. Tandem mass tag (TMT)-based quantitative proteomic analysis was then conducted on ovarian tissues, and the targets of HRW in POF were identified integrating differential expression analysis, functional enrichment analysis, and interaction analysis. Results In HRW treatment of POF rats, the serum AMH and E2 levels significantly increased, and FSH level significantly reduced, indicating the protective role of HRW. After TMT quantitative proteomic analysis, a total of 16 candidate differentially expressed proteins (DEPs) were identified after the cross analysis of DEPs from POF vs. control and POF+HRW vs. POF groups, which were found to be significantly enriched in 296 GO terms and 36 KEGG pathways. The crucial targets, RT1-Db1 and RT1-Bb, were finally identified based on both protein-protein interaction network and GeneMANIA network. Conclusions The HRW treatment could significantly alleviate the ovarian injury of POF rats; RT1-Db1 and RT1-Bb are identified as two crucial targets of HRW treatment in POF rats.
Collapse
Affiliation(s)
- Xiaoyin Meng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
- Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - Shuai Zhang
- Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - Lu Zhao
- Tianjin Central Hospital of Gynecology and Obstetrics, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin, China
| |
Collapse
|
22
|
Zhou Q, Jin X, Wang J, Li H, Yang L, Wu W, Chen W. 4-vinylcyclohexene diepoxide induces premature ovarian insufficiency in rats by triggering the autophagy of granule cells through regulating miR-144. J Reprod Immunol 2023; 157:103928. [PMID: 36889083 DOI: 10.1016/j.jri.2023.103928] [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: 01/04/2023] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
This research explored the pathological and molecular mechanisms of 4-vinylcyclohexene diepoxide (VCD)-induced POI model. QRT-PCR was exploited to detect miR-144 expression in the peripheral blood of POI patients. Rat and KGN cells were treated with VCD to construct POI rat or cell model, respectively. After miR-144 agomir or MK-2206 treatment, miR-144 level, follicle damage, autophagy level and expressions of key pathway-related proteins in rats were detected, and cell viability and autophagy in KGN cells were detected. MiR-144 was apparently down-regulated in the peripheral blood of POI patients. Decreased miR-144 was viewed in both the serum and ovary of rats, yet this trend was apparently reversed by miR-144 agomir. The increased concentration of Follicle-stimulating hormone (FSH) and Luteinizing hormone (LH), along with decreased concentration of E2 and AMH, was observed in the serum of model rats, which was conspicuously negated by control agomir or miR-144 agomir. Increased number of autophagosomes, up-regulated PTEN, and inactivated AKT/m-TOR pathway induced by VCD in ovary tissues were strikingly offset by miR-144 agomir. Results of cytotoxicity assay revealed that 2 mM VCD prominently repressed KGN cell viability. In vitro experiments confirmed that miR-144 interfered with the effect of VCD on autophagy in KGN cells through the AKT/mTOR pathway. Taken together, VCD triggers autophagy to induce POI after targeting the AKT pathway by inhibiting miR-144, it suggest that up-regulation the expression of miR-144 may have the potential to treat POI.
Collapse
Affiliation(s)
- Qun Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, China
| | - Xin Jin
- Department of Massage, The First Affiliated Hospital of Zhejiang Chinese Medical University, China
| | - Jiaxi Wang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, China
| | - Huifang Li
- Department of TCM Gynecology, Tongxiang Maternal and Child Health-Care Center, China
| | - Lijuan Yang
- Department of Gynecology, First School of Clinical Medicine,Yunnan University of Chinese Medicine, China
| | - Weibo Wu
- First School of Clinical Medicine, Zhejiang Chinese Medical University, China
| | - Wenjun Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, China.
| |
Collapse
|
23
|
Wang X, Li T, Bai X, Zhu Y, Zhang M, Wang L. Therapeutic prospect on umbilical cord mesenchymal stem cells in animal model with primary ovarian insufficiency: a meta-analysis. Front Med (Lausanne) 2023; 10:1211070. [PMID: 37324123 PMCID: PMC10264577 DOI: 10.3389/fmed.2023.1211070] [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: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Background Primary ovarian insufficiency (POI) leads to not only infertile but several adverse health events to women. Traditional treatment methods have their own set of limitations and drawbacks that vary in degree. Application of human umbilical cord mesenchymal stem cell (hUCMSC) is a promising strategy for POI. However, there is a lack of literatures on application of hUCMSC in human. Animal experimental model, however, can reflect the potential effectiveness of this employment. This study aimed to evaluate the curative effect of hUCMSC on animals with POI on a larger scale. Methods To gather data, Pubmed, Embase, and Cochrane Library were searched for studies published up to April 2022. Various indices, including the animals' estrous cycle, serum sex hormone levels, and follicle number in the ovary, were compared between the experimental group and those with Premature Ovarian Insufficiency (POI). Results The administration of human umbilical cord-derived mesenchymal stem cells (hUCMSC) has been shown to significantly improve the estrous cycle (RR: 3.32, 95% CI: [1.80, 6.12], I2 = 0%, P = 0.0001), but robustly decrease its length (SMD: -1.97, 95% CI: [-2.58, -1.36], I2 = 0%, P < 0.00001). It can also strikingly increase levels of serum estradiol (SMD: 5.34, 95% CI: [3.11, 7.57], I2 = 93%, P < 0.00001) and anti-müllerian hormone (SMD: 1.92, 95% CI: [0.60, 3.25], I2 = 68%, P = 0.004). Besides, it lowers levels of serum follicle-stimulating hormone (SMD: -3.02, 95% CI: [-4.88, -1.16], I2 = 93%, P = 0.001) and luteinising hormone (SMD: -2.22, 95% CI: [-3.67, -0.76], I2 = 78%, P = 0.003), and thus collectively promotes folliculogenesis (SMD: 4.90, 95% CI: [3.92, 5.88], I2 = 0%, P < 0.00001). Conclusions Based on the presented findings, it is concluded that the administration of hUCMSC in animal models with POI can result in significant improvements in several key indicators, including estrous cycle recovery, hormone level modulation, and promotion of folliculogenesis. These positive outcomes suggest that hUCMSC may have potential as a treatment for POI in humans. However, further research is needed to establish the safety and efficacy of hUCMSC in humans before their clinical application. Systematic review registration https://inplasy.com/inplasy-2023-5-0075/, identifier: INPLASY202350075.
Collapse
Affiliation(s)
- Xinrun Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuechai Bai
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Yun Zhu
- Center for Clinical Big Data and Analytics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Meiliang Zhang
- Department of Obstetrics and Gynecology, Yiwu Maternity and Children Hosptial, Yiwu Branch of Children's Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Liang Wang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Kakinuma K, Kakinuma T. Analysis of oxidative stress and antioxidative potential in premature ovarian insufficiency. World J Clin Cases 2023; 11:2684-2693. [PMID: 37214574 PMCID: PMC10198121 DOI: 10.12998/wjcc.v11.i12.2684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/20/2023] [Accepted: 03/23/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is characterized by an early decline in ovarian function, inducing secondary amenorrhea. While the cause of POI has not yet been identified, the function of mitochondria in the ovaries and the cytotoxicity associated with reactive oxygen species (ROS) have been implicated in follicle pool depletion and a decline in follicle quality. Recently developed tests have enabled easy measurement of diacron-reactive oxygen metabolites (d-ROMs) and biological antioxidant potential (BAP). The combination of these two tests is used to comprehensively assess oxidative stress in the blood.
AIM To comprehensively assess the oxidative stress of d-ROMs and BAP in POI.
METHODS Participants were classified into two groups: A POI group of 11 women aged < 40 years examined between January 2021 and June 2022 with a history of secondary amenorrhea for at least 4 mo in our hospital and an FSH value of ≥ 40 mIU/mL; and a control group of healthy women of the same age with normal ovarian function in our hospital. Plasma d-ROMs and BAP were measured in both these groups underwent. Differences between groups were assessed using the t-test.
RESULTS The mean age and mean body mass index (BMI) were 35.8 ± 3.0 years and 20.1 ± 1.9 kg/m2 in the control group and 35.8 ± 2.7 years and 19.4 ± 2.5 kg/m2 in the POI group, respectively. The mean gravidity and parity in control and POI groups were 0.6 ± 0.7 and 0.4 ± 0.5 and 0.6 ± 0.9 and 0.3 ± 0.5, respectively. The two groups did not differ significantly in terms of mean age, BMI, gravidity, or parity. The d-ROMs level was significantly higher in the POI group than in the control group (478.2 ± 58.7 vs 341.1 ± 35.1 U.CARR; P < 0.001); however, the BAP level did not significantly differ between the two groups (2078.5 ± 157.4 vs 2029.0 ± 186.4 μmol/L). The oxidase stress index (d-ROMs/BAP × 100) was significantly higher in the POI group than in the control group (23.7 ± 3.3 vs 16.5 ± 2.1; P < 0.001).
CONCLUSION Oxidative stress was significantly greater in the POI group than in the control group, suggesting oxidative stress as a factor that can serve as a POI biomarker.
Collapse
Affiliation(s)
- Kaoru Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| | - Toshiyuki Kakinuma
- Department of Obstetrics and Gynecology, International Health and Welfare Hospital, Nasushiobara 327-2763, Japan
| |
Collapse
|
25
|
Li M, Zhu Y, Wei J, Chen L, Chen S, Lai D. The global prevalence of premature ovarian insufficiency: a systematic review and meta-analysis. Climacteric 2023; 26:95-102. [PMID: 36519275 DOI: 10.1080/13697137.2022.2153033] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE The objective of this review was to answer the global prevalence of premature ovarian insufficiency (POI), and explore the associated factors including etiopathology and regions with POI. METHODS The search was conducted on reports from a total of eight databases that comprised Chinese National Knowledge Infrastructure (CNKI), Wanfang, China BioMedical Literature Database (CBM), PubMed, the Cochrane Library, Embase, Web of Science and Ovid MEDLINE® between 1946 and 2021. To analyze the source of heterogeneity, we performed subgroup analysis based on different etiologies and regions. Meta-analysis was carried out by Stata14.0 software. RESULTS The results showed that the global overall prevalence of POI among women was 3.5%. By subgroup analysis, the prevalence of POI among women with iatrogenic etiology was 11.2%, followed by autoimmunity (10.5%); the prevalence of POI by region was 11.3% at the highest in North America followed by South America (5.4%); and the prevalence of POI was 5.3% in a developing country, higher than 3.1% in a developed country. The trend of prevalence of POI over the past 20 years was on the rise (although p > 0.05). CONCLUSION We recommend that health and medical institutions strengthen public health awareness, achieve health-education goals related to POI and increase women's awareness of and attention to POI.
Collapse
Affiliation(s)
- M Li
- Department of Nosocomial Infection, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Y Zhu
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, P.R. China
- Department of Scientific Research, Children's Hospital of Fudan University, Shanghai, P.R. China
| | - J Wei
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai, P.R. China
| | - L Chen
- Department of Nosocomial Infection, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - S Chen
- Department of Nosocomial Infection, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - D Lai
- Department of Nosocomial Infection, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
26
|
Almeida GHDR, Iglesia RP, Rinaldi JDC, Murai MK, Calomeno CVAQ, da Silva Junior LN, Horvath-Pereira BDO, Pinho LBM, Miglino MA, Carreira ACO. Current Trends on Bioengineering Approaches for Ovarian Microenvironment Reconstruction. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36355603 DOI: 10.1089/ten.teb.2022.0171] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian tissue has a unique microarchitecture and a complex cellular and molecular dynamics that are essential for follicular survival and development. Due to this great complexity, several factors may lead to ovarian insufficiency, and therefore to systemic metabolic disorders and female infertility. Techniques currently used in the reproductive clinic such as oocyte cryopreservation or even ovarian tissue transplant, although effective, have several limitations, which impair their wide application. In this scenario, mimetic ovarian tissue reconstruction comes as an innovative alternative to develop new methodologies for germ cells preservation and ovarian functions restoration. The ovarian extracellular matrix (ECM) is crucial for oocyte viability maintenance, once it acts actively in folliculogenesis. One of the key components of ovarian bioengineering is biomaterials application that mimics ECM and provides conditions for cell anchorage, proliferation, and differentiation. Therefore, this review aims at describing ovarian tissue engineering approaches and listing the main limitations of current methods for preservation and reestablishment of ovarian fertility. In addition, we describe the main elements that structure this study field, highlighting the main advances and the challenges to overcome to develop innovative methodologies to be applied in reproductive medicine. Impact Statement This review presents the main advances in the application of tissue bioengineering in the ovarian tissue reconstruction to develop innovative solutions for ovarian fertility reestablishment.
Collapse
Affiliation(s)
| | - Rebeca Piatniczka Iglesia
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mikaelly Kiemy Murai
- Department of Morphological Sciences, State University of Maringa, Maringá, Brazil
| | | | | | | | - Letícia Beatriz Mazo Pinho
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Center of Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
| |
Collapse
|
27
|
Guo X, Zhu Y, Guo L, Qi Y, Liu X, Wang J, Zhang J, Cui L, Shi Y, Wang Q, Liu C, Lu G, Liu Y, Li T, Hong S, Qin Y, Xiong X, Wu H, Huang L, Huang H, Gu C, Li B, Li J. BCAA insufficiency leads to premature ovarian insufficiency via ceramide-induced elevation of ROS. EMBO Mol Med 2023; 15:e17450. [PMID: 36847712 PMCID: PMC10086587 DOI: 10.15252/emmm.202317450] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 03/01/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a disease featured by early menopause before 40 years of age, accompanied by an elevation of follicle-stimulating hormone. Though POI affects many aspects of women's health, its major causes remain unknown. Many clinical studies have shown that POI patients are generally underweight, indicating a potential correlation between POI and metabolic disorders. To understand the pathogenesis of POI, we performed metabolomics analysis on serum and identified branch-chain amino acid (BCAA) insufficiency-related metabolic disorders in two independent cohorts from two clinics. A low BCAA diet phenotypically reproduced the metabolic, endocrine, ovarian, and reproductive changes of POI in young C57BL/6J mice. A mechanism study revealed that the BCAA insufficiency-induced POI is associated with abnormal activation of the ceramide-reactive oxygen species (ROS) axis and consequent impairment of ovarian granulosa cell function. Significantly, the dietary supplement of BCAA prevented the development of ROS-induced POI in female mice. The results of this pathogenic study will lead to the development of specific therapies for POI.
Collapse
Affiliation(s)
- Xiao Guo
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yuemeng Zhu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Lu Guo
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yiwen Qi
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Xiaocheng Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jinhui Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jiangtao Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Linlin Cui
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
| | - Yueyang Shi
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qichu Wang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Cenxi Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Guangxing Lu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yilian Liu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tao Li
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shangyu Hong
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yingying Qin
- Shanghai First Maternity and Infant Hospital, Shanghai, China.,Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuelian Xiong
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hao Wu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Lin Huang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - He Huang
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Chao Gu
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Bin Li
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jin Li
- Obstetrics and Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital and Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Mirzaei R, Bidgoli SA, Khosrokhavar R, Shoeibi S, Ashtiani HA. Increased risk of primary ovarian insufficiency by high-fructose diet consumption: a 90-day study in female rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:7415-7426. [PMID: 36040692 DOI: 10.1007/s11356-022-22258-8] [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: 12/22/2021] [Accepted: 07/22/2022] [Indexed: 06/15/2023]
Abstract
There is ambiguous evidence that high-fructose diet can induce toxicity in different organ systems but its endocrine disrupting effects by abnormal changes in female reproductive organs is poorly evidenced. This study aimed to address the reproductive safety of high fructose diet through clinical, biochemical, hormonal, histopathological, and immunohistochemical analysis. For this purpose, 5-6 weeks mature female Wistar rats were divided in three groups and each five animals/group exposed to standard chow + water + HFCS-55, standard chow + water + sucrose 75%w/v and standard chow + water for 90 days. Remarkable increase in most lipid profile factors and total body weights of HFCS-55 fed rats and sucrose fed rats were detected in similar pattern compared to control. At the same time, a battery of differential signs and symptoms in HFCS-fed groups including squamous metaplasia in the uterine tissue and ovarian congestion, significant increase in FSH and LH levels, meaningful decreased serum testosterone and 17β-estradiol levels, and strong androgen receptor expression in ovaries and uterine of HFCS group of animals were recorded compared to other two study groups. These thought-provoking signs and signals of fructose induced reproductive toxicity in this model emphasis the contribution of HFCS-55 to deteriorated ovarian and endometrial health and increased risk primary ovarian insufficiency (POI) in women.
Collapse
Affiliation(s)
- Roya Mirzaei
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Arbabi Bidgoli
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Roya Khosrokhavar
- Food and Drug Laboratory Research Center, Food and Drug Administration, MOH&ME, Tehran, Iran
| | - Shahram Shoeibi
- Food and Drug Laboratory Research Center, Food and Drug Administration, MOH&ME, Tehran, Iran
| | - Hamidreza Ahmadi Ashtiani
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
29
|
Kapoor E, Faubion SS, Gazzuola Rocca L, Mielke MM, Smith CY, Rocca WA. Trajectories of metabolic parameters after bilateral oophorectomy in premenopausal women. Maturitas 2022; 165:38-46. [PMID: 35905571 PMCID: PMC9529838 DOI: 10.1016/j.maturitas.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 06/27/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study the trajectories of metabolic parameters after bilateral oophorectomy. STUDY DESIGN This population-based cohort study included a random sample of all premenopausal women who underwent bilateral oophorectomy at or before age 45 years from 1988 to 2007 in Olmsted County, Minnesota, and their age-matched (±1 year) referent women who did not undergo bilateral oophorectomy. MAIN OUTCOME MEASURES The medical records of all women were reviewed to collect the metabolic parameters over a 10-year period. We compared three groups of women: 1) referent women (n = 270), 2) women who underwent bilateral oophorectomy and received estrogen therapy (n = 163), and 3) women who underwent bilateral oophorectomy and did not receive estrogen therapy (n = 107). RESULTS Over 10 years of follow-up, the three groups had significantly different mean values of diastolic blood pressure, weight, body mass index (BMI), total cholesterol, triglycerides, and high-density lipoprotein cholesterol (HDL-C). However, women with and without bilateral oophorectomy were already different at baseline for hyperlipidemia, systolic blood pressure, weight, and BMI. Nevertheless, the trajectories of change over 10 years were significant for weight (group by time interaction p = 0.03), BMI (p = 0.03), and HDL-C (p = 0.004). The changes occurred primarily in the initial 4-5 years. Women who received estrogen therapy after bilateral oophorectomy were comparable to the referent women with respect to the weight and BMI trends, and they experienced an increase in HDL-C over time. CONCLUSION Women who underwent bilateral oophorectomy before menopause experienced unfavorable changes in some metabolic parameters possibly increasing their cardiovascular risk.
Collapse
Affiliation(s)
- Ekta Kapoor
- Menopause and Women's Sexual Health Clinic, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States; Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, United States; Center for Women's Health, Mayo Clinic, Rochester, MN, United States; Mayo Clinic Specialized Center of Research Excellence (SCORE) on Sex Differences, Mayo Clinic, Rochester, MN, United States.
| | - Stephanie S Faubion
- Center for Women's Health, Mayo Clinic, Rochester, MN, United States; Division of General Internal Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Liliana Gazzuola Rocca
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Michelle M Mielke
- Mayo Clinic Specialized Center of Research Excellence (SCORE) on Sex Differences, Mayo Clinic, Rochester, MN, United States; Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States; Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Carin Y Smith
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Walter A Rocca
- Mayo Clinic Specialized Center of Research Excellence (SCORE) on Sex Differences, Mayo Clinic, Rochester, MN, United States; Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States; Department of Neurology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
30
|
Loose JA, Amrit FRG, Patil T, Yanowitz JL, Ghazi A. Meiotic dysfunction accelerates somatic aging in Caenorhabditis elegans. Aging Cell 2022; 21:e13716. [PMID: 36176234 PMCID: PMC9649607 DOI: 10.1111/acel.13716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 08/07/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023] Open
Abstract
An expanding body of evidence, from studies in model organisms to human clinical data, reveals that reproductive health influences organismal aging. However, the impact of germline integrity on somatic aging is poorly understood. Moreover, assessing the causal relationship of such an impact is challenging to address in human and vertebrate models. Here, we demonstrate that disruption of meiosis, a germline restricted process, shortened lifespan, impaired individual aspects of healthspan, and accelerated somatic aging in Caenorhabditis elegans. Young meiotic mutants exhibited transcriptional profiles that showed remarkable overlap with the transcriptomes of old worms and shared similarities with transcriptomes of aging human tissues as well. We found that meiosis dysfunction caused increased expression of functionally relevant longevity determinants whose inactivation enhanced the lifespan of normal animals. Further, meiotic mutants manifested destabilized protein homeostasis and enhanced proteasomal activity partially rescued the associated lifespan defects. Our study demonstrates a role for meiotic integrity in controlling somatic aging and reveals proteostasis control as a potential mechanism through which germline status impacts overall organismal health.
Collapse
Affiliation(s)
- Julia A. Loose
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Francis R. G. Amrit
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Thayjas Patil
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Judith L. Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee‐Womens Research InstituteUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Arjumand Ghazi
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Developmental Biology, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Cell Biology & PhysiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
31
|
Cai Y, Zhang Q, Yu K, Wang Q. Study of Serum Anti-Mullerian Hormone in the Diagnosis of Ovarian Reserve Function in Patients with Premature Ovarian Insufficiency. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3878359. [PMID: 36277874 PMCID: PMC9584692 DOI: 10.1155/2022/3878359] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022]
Abstract
Objective To explore the diagnostic value of serum anti-Mullerian hormone (AMH) for patients with premature ovarian insufficiency (POI) and premature ovarian failure (POF). Methods Totally, 125 women with menstrual disorder treated in the Obstetrics and Gynecology Department of Ningbo Women & Children Hospital between January 2020 and December 2021 were enrolled. Among them, based on the follicle-stimulating hormone (FSH) level, 54 patients (25 IU/L < FSH ≤ 40 IU/L) were assigned to the POI group, and 71 patients (FSH > 40 IU/L) were assigned to the POF group. In addition, 72 individuals who went physical examination in the hospital and showed normal menstrual cycle were selected as the control (CON) group. Serum AMH in each group was quantified via enzyme-linked immunosorbent assay (ELISA) and Beckman Coulter Access active immunoassay analyzer, and the levels of serum neutral hormones [luteinizing hormone (LH), FSH, as well as estradiol (E2)] in each group were detected through the electrochemiluminescence method. The difference between AMH level acquired by the latest automatic method and that acquired by the traditional manual ELISA was compared, and the correlation of serum AMH with sex hormones was analyzed. In addition, receiver-operating characteristic (ROC) curves were drawn for determining the diagnostic value of serum AHM for POI ad POF. Results Beckman Coulter Access quantified AMH more accurately and fastly (Beckman Coulter Access: 35 minutes; manual quantification: 3-4 hours) and was more sensitive than ELISA, with a requirement to less serum. The levels of serum AMH and E2 in the POF group were 0.04 ± 0.10 ng/mL and 35.16 ± 53.06 ng/mL, respectively, which were notably lower than those in the POI group ((0.69 ± 1.46) ng/mL and (3.96 ± 2.82) ng/mL) and CON group ((76.31 ± 97.84) ng/mL and (113.19 ± 114.84) ng/mL). The LH and FSH levels in the POF group were 37.86 ± 19.44 IU/L and 75.05 ± 35.31 IU/L, which were higher than those in POI group ((22.66 ± 26.15) IU/L and (11.30 ± 17.05) IU/L) and the CON group ((29.81 ± 4.45) IU/L and (6.78 ± 3.45) IU/L) (P < 0.05). The POI group showed a notably lower serum AMH level and notably higher LH and FSH levels than the CON group (P < 0.05), and the POI group was similar to the CON group in the E2 level (P > 0.05). Serum AMH showed a positive correlation with E2 (r = 0.291, P < 0.05) and a negative association with both FSH (r = -0.476, P < 0.05) and LH (r = -0.143, P < 0.1). The optimal cut-off value of serum AMH in predicting POI was 0.83 ng/mL, and the corresponding sensitivity and specificity were 95.8% and 85.2%. The optimal cut-off of serum AMH in predicting POF was 0.075 ng/mL, and the corresponding sensitivity and specificity were 81.7% and 94.4%. The area under ROC curve (AUC) of serum AMH + FSH in the diagnosis of POF was close to 1. Conclusion Beckman Coulter Access AMH test is the latest automatic electrochemiluminescence sandwich immunoassay, with higher sensitivity and reproducibility than traditional manual ELISA and with ability to produce repeatable results. With the decline of ovarian function, the serum AMH of POI patients decreases gradually, and the serum AMH of POF patients decreases obviously, so serum AMH level has great value in predicting POI and POF and can be used as a sensitive index for early diagnosis of the two. Serum AMH combined with FSH can lift the diagnostic efficiency of POF.
Collapse
Affiliation(s)
- Yan Cai
- Department of Gynecology, Ningbo Women and Children's Hospital in Zhejiang Province, Ningbo, 315012 Zhejiang Province, China
| | - Qiong Zhang
- Clinical Laboratory, Ningbo Women and Children's Hospital in Zhejiang Province, Ningbo, 315012 Zhejiang Province, China
| | - Kewen Yu
- Department of Gynecology, Ningbo Women and Children's Hospital in Zhejiang Province, Ningbo, 315012 Zhejiang Province, China
| | - Qiming Wang
- Department of Gynecology, Ningbo Women and Children's Hospital in Zhejiang Province, Ningbo, 315012 Zhejiang Province, China
| |
Collapse
|
32
|
Dou X, Jin X, Chen X, Zhou Q, Chen H, Wen M, Chen W. Bu-Shen-Ning-Xin decoction alleviates premature ovarian insufficiency (POI) by regulating autophagy of granule cells through activating PI3K/AKT/mTOR pathway. Gynecol Endocrinol 2022; 38:754-764. [PMID: 35989579 DOI: 10.1080/09513590.2022.2112941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022] Open
Abstract
PURPOSE To explore the therapeutic effects of Bu-Shen-Ning-Xin decoction (BSNXD) on POI and the underlying mechanism. METHODS VCD was used to induce the in vivo and in vitro POI model. HE staining was used to evaluate the pathological state of ovarian tissues. ELISA was used to detect the production of hormones in the serum and granule cells (GCs). An immunohistochemical assay was used to determine the expression of ATG7 and p-AKT in the ovarian tissues. The number of oocytes in POI rats was counted. The mitochondrial membrane potential (MMP) in oocytes and GCs was detected by flow cytometry. A Western blot assay was used to measure the expression of AKT, p-AKT, p-mTOR, mTOR, S6K, p-S6K, ULK1, p-ULK1, Beclin-1, Bcl-2, LC3-II, LC3-I, ATG7, and cleaved Caspase3. The numbers of autophagosomes were detected by transmission electron microscope and autophagic flux assay. The CCK-8 assay was used to detect the cell viability. RESULTS Decreased primary follicles in the ovarian tissues, elevated concentration of FSH, and LH, suppressed concentration of E2 and AMH in the serum, reduced number of oocytes, and mitochondrial dysfunction in oocytes induced by VCD were significantly reversed by BSNXD. Activated autophagy state and inhibited PI3K/AKT/mTOR pathway stimulated by VCD in both ovarian tissues and GCs were dramatically reversed by BSNXD. The protective effect of BSNXD on VCD-treated GCs was abolished by LY294002, an inhibitor of the PI3K/AKT/mTOR pathway. CONCLUSION Our data revealed that BSNXD alleviated POI by regulating autophagy of granule cells through activating PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Xiaoqing Dou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Xin Jin
- Department of Massage, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Xingbei Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Qun Zhou
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Hanyu Chen
- School of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing City, Jiangsu Province,China
| | - Mingxiao Wen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| | - Wenjun Chen
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
33
|
Gottfried S. Women: Diet, Cardiometabolic Health, and Functional Medicine. Phys Med Rehabil Clin N Am 2022; 33:621-645. [DOI: 10.1016/j.pmr.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Chen Y, Fan X, Ma K, Wang K, Tian C, Li M, Gong L. Bushen Culuan Decoction Ameliorates Premature Ovarian Insufficiency by Acting on the Nrf2/ARE Signaling Pathway to Alleviate Oxidative Stress. Front Pharmacol 2022; 13:857932. [PMID: 35462905 PMCID: PMC9019758 DOI: 10.3389/fphar.2022.857932] [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: 01/19/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Premature ovarian insufficiency (POI) can result in lower fertility and shorten the female reproductive span. Bushen-Culuan Decoction (BCD) is a traditional Chinese medication utilized for treating POI for many years. We previously observed that BCD protects against further deterioration of the ovarian reserve of POI patients, however, the underlying mechanism has not been well studied. Our investigation seeks to evaluate the effect of BCD on POI induced by Tripterygium wilfordii polyglycosidium (TWP) and the likely mechanistic pathways, which we hypothesize may involve the Nrf2/ARE pathway. The body weights, estrous cycle, serum hormone levels, histological follicular analysis and quantification, levels of oxidative stress biomarkers in the ovarian tissue of POI mice models were evaluated. Western blotting and RT-PCR enabled quantification of the components of the Nrf2/ARE pathway. Our results showed that BCD restored hormonal profiles and estrous cycles of POI mice similar to those observed in healthy controls. BCD reduced the numbers of atretic follicles while increasing the number of primordial follicles. BCD facilitated lower 8-OHdG and MDA levels while increasing levels of key antioxidant enzymes including GSH-Px, CAT, and SOD. Furthermore, TWP increased Bach 1, Nrf2, and Keap 1 expressions at the translational level, while decreased that of HO-1. BCD treatment also promoted nuclear translocation rates of Bach 1 and Nrf2, suppressed Keap 1 protein expression, as well as raised HO-1 protein expression. Taken together, BCD likely augments ovarian reserve by activating the Nrf2/ARE signaling pathway, which stimulated higher levels of antioxidants and suppressed oxidative stress. BCD may be an important therapeutic compound in POI.
Collapse
Affiliation(s)
- Yanxia Chen
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Xiaodi Fan
- Institute of Basic Medical Science of Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Kun Ma
- China Academy of Chinese Medical Science, Beijing, China
| | - Kaili Wang
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Caidie Tian
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China
| | - Min Li
- China Academy of Chinese Medical Science, Beijing, China
| | - Linjuan Gong
- Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China
| |
Collapse
|
35
|
Does the Value of FSH Predict Severity of Metabolic Complications in Females with POI? J Clin Med 2022; 11:jcm11072024. [PMID: 35407635 PMCID: PMC8999648 DOI: 10.3390/jcm11072024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 01/22/2023] Open
Abstract
Premature ovarian insufficiency (POI) is defined as a cessation of ovarian function before the age of 40. Such early deprivation of estrogens in women may be associated with several adverse cardiovascular and metabolic consequences. The aim of this retrospective study was to investigate whether women with POI and a serum follicle-stimulating hormone (FSH) level of 25−40 I/U (Group A) have the same metabolic profile as women with POI and a serum FSH level of >40 I/U (Group B). One hundred twenty-three women were included in the study group (Group A; n = 41; Group B; n = 82). The control group comprised 77 healthy women with regular menstruation. In the age- and BMI-adjusted model, no differences were found between the groups with respect to total cholesterol, high-density lipoproteins, triglycerides, HOMA-IR, glucose, and insulin. The only significant difference was found in terms of low-density lipoprotein cholesterol (LDL-C). The highest serum concentration was found in Group B, the second highest was found in Group A, and the lowest was in the controls. In conclusion, changing the threshold of FSH required to establish a POI diagnosis may have an impact on the level of serum LDL-C.
Collapse
|
36
|
Ruan X, Mueck AO. Optimizing menopausal hormone therapy: for treatment and prevention, menstrual regulation, and reduction of possible risks. GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
37
|
Secomandi L, Borghesan M, Velarde M, Demaria M. The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update 2022; 28:172-189. [PMID: 34918084 PMCID: PMC8888999 DOI: 10.1093/humupd/dmab038] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/28/2021] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Advanced maternal age is associated with decreased oocyte quantity and quality as well as uterine and placental dysfunctions. These changes lead to infertility, pregnancy complications and birth defects in the offspring. As the mean age of giving birth is increasing worldwide, prevention of age-associated infertility and pregnancy complications, along with the more frequent use of ART, become extremely important. Currently, significant research is being conducted to unravel the mechanisms underlying female reproductive aging. Among the potential mechanisms involved, recent evidence has suggested a contributing role for cellular senescence, a cellular state of irreversible growth arrest characterized by a hypersecretory and pro-inflammatory phenotype. Elucidating the role of senescence in female reproductive aging holds the potential for developing novel and less invasive therapeutic measures to prevent or even reverse female reproductive aging and increase offspring wellbeing. OBJECTIVE AND RATIONALE The review will summarize the positive and negative implications of cellular senescence in the pathophysiology of the female reproductive organs during aging and critically explore the use of novel senotherapeutics aiming to reverse and/or eliminate their detrimental effects. The focus will be on major senescence mechanisms of the ovaries, the uterus, and the placenta, as well as the potential and risks of using senotherapies that have been discovered in recent years. SEARCH METHODS Data for this review were identified by searches of MEDLINE, PubMed and Google Scholar. References from relevant articles using the search terms 'Cellular Senescence', 'Aging', 'Gestational age', 'Maternal Age', 'Anti-aging', 'Uterus', 'Pregnancy', 'Fertility', 'Infertility', 'Reproduction', 'Implant', 'Senolytic', 'Senostatic', 'Senotherapy' and 'Senotherapeutic' where selected. A total of 182 articles published in English between 2005 and 2020 were included, 27 of which focus on potential senotherapies for reproductive aging. Exclusion criteria were inclusion of the terms 'male' and 'plants'. OUTCOMES Aging is a major determinant of reproductive wellbeing. Cellular senescence is a basic aging mechanism, which can be exploited for therapeutic interventions. Within the last decade, several new strategies for the development and repurposing of drugs targeting senescent cells have emerged, such as modulators of the anti-inflammatory response, oxidative stress, DNA damage, and mitochondria and protein dysfunctions. Several studies of female reproductive aging and senotherapies have been discussed that show promising results for future interventions. WIDER IMPLICATIONS In most countries of the Organization for Economic Co-operation and Development, the average age at which women give birth is above 30 years. Currently, in countries such as the Netherlands, Australia, Spain, Finland, Germany and the UK, birth rates among 30- to 34-year-olds are now higher than in any other age groups. This review will provide new knowledge and scientific advancement on the senescence mechanisms during female reproductive aging, and benefit fundamental and clinical scientists and professionals in the areas of reproduction, cancer, immunobiology and fibrosis.
Collapse
Affiliation(s)
- Laura Secomandi
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michela Borghesan
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| | - Michael Velarde
- Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, PH 1101, Philippines
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), 9713AV Groningen, The Netherlands
| |
Collapse
|
38
|
Systematic Pharmacology-Based Strategy to Explore the Molecular Network Mechanism of Modified Taohong Siwu Decoction in the Treatment of Premature Ovarian Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3044463. [PMID: 35096106 PMCID: PMC8799328 DOI: 10.1155/2022/3044463] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To explore the molecular network mechanism of modified Taohong Siwu Decoction (MTHSWD) to interfere with premature ovarian failure based on systematic pharmacological strategy. METHODS The network pharmacology strategy was used to explore the potential mechanism of MTHSWD intervention in POF, and then it was verified through animal experiments. Mouse zona pellucida 3 was used as an antigen to subcutaneously immunize BALB/c female mice to establish an immune POF model. Mice were divided into MTHSWD low-, medium-, and high-dose groups, positive control group, model group, and normal group. After 30 days of drug intervention, ovarian tissue was taken for pathological hematoxylin-eosin (HE) staining, and immunohistochemical methods were used to detect the expression of TGF-β1 and TGF-βRII and Smad2/3 protein expression in follicular wall granular cells and ovarian tissue, respectively. RESULTS Network pharmacology studies have shown that MTHSWD may interfere with the TGF-β signaling pathway. Animal experimental research shows that, compared with the model group, the number of ovarian mature follicles in the MTHSWD groups and the positive group was significantly increased, and the number of atresia follicles decreased. Immunohistochemistry showed that, compared with the control group, the expression of TGF-β1, TGF-βRII, and Smad2/3 in the follicular wall granulosa cells and ovarian tissues of MTHSWD groups was significantly higher than that of the model group (P < 0.05). CONCLUSION MTHSWD may improve the ovarian function of POF mice by upregulating the protein expression of granulosa cells TGF-β1, TGF-βRII, and Smad2/3.
Collapse
|
39
|
Liu Y, Zhu X, Wu C, Lang Y, Zhao W, Li Y. Melatonin protects against ovarian damage by inhibiting autophagy in granulosa cells in rats. Clinics (Sao Paulo) 2022; 77:100119. [PMID: 36194922 PMCID: PMC9531038 DOI: 10.1016/j.clinsp.2022.100119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVES This study sought to further verify the protective mechanism of Melatonin (MT) against ovarian damage through animal model experiments and to lay a theoretical and experimental foundation for exploring new approaches for ovarian damage treatment. METHOD The wet weight and ovarian index of rat ovaries were weighted, and the morphology of ovarian tissues and the number of follicles in the pathological sections of collected ovarian tissues were recorded. And the serum sex hormone levels, the key proteins of the autophagy pathway (PI3K, AKT, mTOR, LC3II, LC3I, and Agt5) in rat ovarian tissues, as well as the viability and mortality of ovarian granulosa cells in each group were measured by ELISA, western blotting, CCK8 kit and LDH kit, respectively. RESULTS The results showed that MT increased ovarian weight and improved the ovarian index in ovarian damage rats. Also, MT could improve autophagy-induced ovarian tissue injury, increase the number of primordial follicles, primary follicles, and sinus follicles, and decrease the number of atretic follicles. Furthermore, MT upregulated serum AMH, INH-B, and E2 levels downregulated serum FSH and LH levels in ovarian damage rats and activated the PI3K/AKT/mTOR signaling pathway. Besides, MT inhibited autophagic apoptosis of ovarian granulosa cells and repressed the expression of key proteins in the autophagic pathway and reduced the expression levels of Agt5 and LC3II/I. CONCLUSIONS MT inhibits granulosa cell autophagy by activating the PI3K/Akt/mTOR signaling pathway, thereby exerting a protective effect against ovarian damage.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gynecology, Weifang People's Hospital, Weifang, Shandong, China
| | - Xiaohe Zhu
- Department of Obstetrics, Weifang People's Hospital, Weifang, Shandong, China
| | - Chunli Wu
- Department of Obstetrics, Weifang People's Hospital, Weifang, Shandong, China
| | - Yan Lang
- Department of Obstetrics, Weifang People's Hospital, Weifang, Shandong, China
| | - Wenjie Zhao
- Department of Reproductive Medicine, Weifang People's Hospital, Weifang, Shandong, China
| | - Yanmin Li
- Department of Reproductive Medicine, Weifang People's Hospital, Weifang, Shandong, China.
| |
Collapse
|
40
|
Pellikaan K, Ben Brahim Y, Rosenberg AGW, Davidse K, Poitou C, Coupaye M, Goldstone AP, Høybye C, Markovic TP, Grugni G, Crinò A, Caixàs A, Eldar-Geva T, Hirsch HJ, Gross-Tsur V, Butler MG, Miller JL, van der Kuy PHM, van den Berg SAA, Visser JA, van der Lely AJ, de Graaff LCG. Hypogonadism in Women with Prader-Willi Syndrome-Clinical Recommendations Based on a Dutch Cohort Study, Review of the Literature and an International Expert Panel Discussion. J Clin Med 2021; 10:jcm10245781. [PMID: 34945077 PMCID: PMC8707541 DOI: 10.3390/jcm10245781] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/27/2022] Open
Abstract
Prader-Willi syndrome (PWS) is a rare neuroendocrine genetic syndrome. Characteristics of PWS include hyperphagia, hypotonia, and intellectual disability. Pituitary hormone deficiencies, caused by hypothalamic dysfunction, are common and hypogonadism is the most prevalent. Untreated hypogonadism can cause osteoporosis, which is already an important issue in PWS. Therefore, timely detection and treatment of hypogonadism is crucial. To increase understanding and prevent undertreatment, we (1) performed a cohort study in the Dutch PWS population, (2) thoroughly reviewed the literature on female hypogonadism in PWS and (3) provide clinical recommendations on behalf of an international expert panel. For the cohort study, we retrospectively collected results of a systematic health screening in 64 female adults with PWS, which included a medical questionnaire, medical file search, medical interview, physical examination and biochemical measurements. Our data show that hypogonadism is frequent in females with PWS (94%), but is often undiagnosed and untreated. This could be related to unfamiliarity with the syndrome, fear of behavioral changes, hygienic concerns, or drug interactions. To prevent underdiagnosis and undertreatment, we provide practical recommendations for the screening and treatment of hypogonadism in females with PWS.
Collapse
Affiliation(s)
- Karlijn Pellikaan
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Dutch Center of Reference for Prader-Willi Syndrome, 3015 GD Rotterdam, The Netherlands
- Academic Centre for Growth Disorders, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Yassine Ben Brahim
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Dutch Center of Reference for Prader-Willi Syndrome, 3015 GD Rotterdam, The Netherlands
- Academic Centre for Growth Disorders, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Anna G. W. Rosenberg
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Dutch Center of Reference for Prader-Willi Syndrome, 3015 GD Rotterdam, The Netherlands
- Academic Centre for Growth Disorders, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Kirsten Davidse
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Dutch Center of Reference for Prader-Willi Syndrome, 3015 GD Rotterdam, The Netherlands
- Academic Centre for Growth Disorders, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Christine Poitou
- Rare Diseases Center of Reference ‘Prader-Willi Syndrome and Obesity with Eating Disorders’ (PRADORT), Nutrition Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France; (C.P.); (M.C.)
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- ENDO-ERN (European Reference Network)
| | - Muriel Coupaye
- Rare Diseases Center of Reference ‘Prader-Willi Syndrome and Obesity with Eating Disorders’ (PRADORT), Nutrition Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France; (C.P.); (M.C.)
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- ENDO-ERN (European Reference Network)
| | - Anthony P. Goldstone
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- PsychoNeuroEndocrinology Research Group, Centre for Neuropsychopharmacology, Division of Psychiatry, and Computational, Cognitive and Clinical Neuroimaging Laboratory, Department of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London W12 0HS, UK
| | - Charlotte Høybye
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- ENDO-ERN (European Reference Network)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17176 Stockholm, Sweden
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Department of Endocrinology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Tania P. Markovic
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- Metabolism & Obesity Services, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Graziano Grugni
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- ENDO-ERN (European Reference Network)
- Divison of Auxology, Istituto Auxologico Italiano, IRCCS, 28824 Piancavallo, Italy
| | - Antonino Crinò
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- Reference Center for Prader-Willi Syndrome, Bambino Gesù Hospital, Research Institute, 00050 Palidoro, Italy
| | - Assumpta Caixàs
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- Endocrinology and Nutrition Department, Institut d’Investigació I Innovació Parc Taulí I3PT, Parc Taulí Hospital Universitari, Department of Medicine, Universitat Autònoma de Barcelona, 08208 Sabadell, Spain
- Correspondence: (A.C.); (L.C.G.d.G.)
| | - Talia Eldar-Geva
- The Israel Multidisciplinary Prader-Willi Syndrome Clinic, Jerusalem 9103102, Israel; (T.E.-G.); (H.J.H.); (V.G.-T.)
- Reproductive Endocrinology and Genetics Unit, Department of Obstetrics and Gynecology, Shaare-Zedek Medical Center, Jerusalem 9103102, Israel
- Hebrew University School of Medicine, Jerusalem 9112102, Israel
| | - Harry J. Hirsch
- The Israel Multidisciplinary Prader-Willi Syndrome Clinic, Jerusalem 9103102, Israel; (T.E.-G.); (H.J.H.); (V.G.-T.)
- Department of Pediatrics, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Varda Gross-Tsur
- The Israel Multidisciplinary Prader-Willi Syndrome Clinic, Jerusalem 9103102, Israel; (T.E.-G.); (H.J.H.); (V.G.-T.)
- Hebrew University School of Medicine, Jerusalem 9112102, Israel
- Neuropediatrics Unit, Department of Pediatrics, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Merlin G. Butler
- Departments of Psychiatry, Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Jennifer L. Miller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Paul-Hugo M. van der Kuy
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - Sjoerd A. A. van den Berg
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Department of Clinical Chemistry, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Jenny A. Visser
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
| | - Aart J. van der Lely
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
| | - Laura C. G. de Graaff
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (K.P.); (Y.B.B.); (A.G.W.R.); (K.D.); (S.A.A.v.d.B.); (J.A.V.); (A.J.v.d.L.)
- Center for Adults with Rare Genetic Syndromes, Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Dutch Center of Reference for Prader-Willi Syndrome, 3015 GD Rotterdam, The Netherlands
- Academic Centre for Growth Disorders, Erasmus University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- International Network for Research, Management & Education on adults with PWS (INfoRMEd-PWS); (A.P.G.); (C.H.); (T.P.M.); (G.G.); (A.C.)
- ENDO-ERN (European Reference Network)
- Correspondence: (A.C.); (L.C.G.d.G.)
| |
Collapse
|
41
|
Hyperin Alleviates Triptolide-Induced Ovarian Granulosa Cell Injury by Regulating AKT/TSC1/mTORC1 Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9399261. [PMID: 34707679 PMCID: PMC8545507 DOI: 10.1155/2021/9399261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/31/2021] [Accepted: 09/18/2021] [Indexed: 11/17/2022]
Abstract
Premature ovarian insufficiency (POI) is characterized by the loss of ovarian function before 40 years of age and affects approximately 1% of women worldwide. Caragana sinica is a traditional Miao (a Chinese ethnic minority) medicine that improves ovarian function and follicular development. In the present study, we aimed to investigate the effect of active ingredients of C. sinica on POI and determine underlying mechanisms. Herein, the chemical composition of the C. sinica compound was analyzed using ultra-high-performance liquid chromatography, which identified hyperin (HR) as one of the main ingredients in C. sinica. Then, interaction targets of HR and POI were predicted and analyzed using network pharmacology and bioinformatics. The effect of HR on triptolide (TP)-induced granulosa cell injury was evaluated, and the underlying mechanism was explored based on bioinformatic results. A total of 100 interaction targets for POI and HR were obtained. The protein-protein interaction network of identified interaction targets emphasized the topological importance of AKT1. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that HR might regulate POI by modulating the mechanistic target of rapamycin (mTOR) signaling pathway. In addition, the KEGG graph of the mTOR signaling pathway revealed that AKT phosphorylation inhibits the TSC1/2, while TSC1/2 activation inhibits the expression of mTORC1. The fundamental experiment revealed that HR increased proliferation, progesterone receptor levels, and estradiol levels decreased by TP in KGN cells. Additionally, HR alleviated TP-induced apoptosis and G1/G1 phase arrest in KGN cells. Western blotting demonstrated that HR increased the phosphorylation of AKT and mTORC1 and decreased TSC1 expression in TP-induced KGN cells. Collectively, our findings revealed that HR alleviates TP-induced granulosa cell injury by regulating AKT/TSC1/mTORC1 signaling, providing insight into the treatment of POI.
Collapse
|
42
|
Xie QE, Wang MY, Cao ZP, Du X, Ji DM, Liang D, Cao YX, Liu YJ. Melatonin protects against excessive autophagy-induced mitochondrial and ovarian reserve function deficiency though ERK signaling pathway in Chinese hamster ovary (CHO) cells. Mitochondrion 2021; 61:44-53. [PMID: 34571250 DOI: 10.1016/j.mito.2021.09.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023]
Abstract
Excessive autophagy-induced follicular atresia of ovarian granulosa cells might be one of the pathogenesis of Premature Ovarian Insufficiency (POI), and melatonin (MT) exerted many beneficial effects on mitochondria. However, there was little report regarding the beneficial effects of MT on excessive autophagy-induced mitochondrial and ovarian reserve function deficiency, and the mechanisms have not been clearly identified. Autophagy played a protective role in cells survival, however, high level of autophagy could lead to cell death. In this report, firstly, Chinese hamster ovary cell damage model stably expressing EGFP-LC3 was established. Next, we systematically investigated the protective effects of MT on mitochondrial and ovarian reserve function and molecular mechanisms using this cell damage model. Our results revealed that 10-9 M MT not only protected against the decline of anti-mullerian hormone (AMH) expression induced by excessive autophagy, but also rescued excessive autophagy-induced impairment of mitochondrial expression and mitochondrial membrane potential. Furthermore, MT protected against excessive autophagy-induced decrease of nucleus-encoded proteins including SDHA and mitofilin, and mitochondrial dynamic-related proteins including OPA1, MFN2, and DRP1. MT also decreased mitochondrial oxidative stress, increased antioxidant enzyme superoxide dismutase 2 (SOD2) expression and ameliorated the G2/M cell cycle arrest induced by excessive autophagy. Finally, MT inhibited excessive autophagy-induced activation of extracellular signal regulated kinase (ERK) signaling pathway. In conclusion, our study showed that MT rescued impairment of mitochondrial and ovarian reserve function, and production of mitochondrial ROS and cell cycle arrest induced by excessive autophagy through down-regulated ERK pathway, implying the potential therapeutic drug target for POI.
Collapse
Affiliation(s)
- Q E Xie
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Anhui Medical University, Hefei 230022, PR China; Department of Histology and Embryology, Anhui Medical University, Hefei 230032, Anhui, PR China
| | - M Y Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Anhui Medical University, Hefei 230022, PR China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, PR China; Anhui Provincial Institute of Translational Medicine, No 81 Meishan Road, Hefei 230032, Anhui, PR China
| | - Z P Cao
- The Third People's Hospital of Hefei, The Third Clinical Teaching Hospital of Anhui Medical University, Hefei 230022, Anhui, PR China
| | - X Du
- 901th hospital of PLA Joint Logistic Support Force, No 424 West Changjiang Road, Heifei 230031, Anhui, PR China
| | - D M Ji
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, PR China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, PR China; Anhui Provincial Institute of Translational Medicine, No 81 Meishan Road, Hefei 230032, Anhui, PR China
| | - D Liang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Anhui Medical University, Hefei 230022, PR China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, PR China.
| | - Y X Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Anhui Medical University, Hefei 230022, PR China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, PR China.
| | - Y J Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; NHC Key Laboratory of study on abnormal gametes and reproductive tract, Anhui Medical University, Hefei 230022, PR China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, PR China.
| |
Collapse
|
43
|
Yi A, Qin X, Du Z, Wang T, Liu F. Clinical Observation on the Improvement of Serum Sex Hormone and Ovarian Function in Premature Ovarian Failure Patients with Deficiency-Cold Syndrome by Combining Wenjing Decoction with Tiaobu Chongren Acupuncture and Moxibustion. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:3926822. [PMID: 34545290 PMCID: PMC8449719 DOI: 10.1155/2021/3926822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To explore the effect of Wenjing Decoction and Tiaobu Chongren acupuncture and moxibustion therapy on serum sex hormones and ovarian function recovery in premature ovarian failure (POF) patients with deficiency-cold syndrome. METHODS From February 2018 to October 2019, 119 POI patients with deficiency-cold syndrome who met the selection criteria were randomly divided into the control group (n = 59) and the observation group (n = 60). The control group was treated with hormone replacement therapy, and the observation group was treated with Wenjing Decoction and Tiaobu Chongren acupuncture and moxibustion therapy on the basis of the control group. Both groups were treated continuously for 3 months. After treatment, the clinical efficacy of the two groups was compared, the Chinese medicine syndrome score, serum estradiol (E 2), follicle-stimulating hormone (FSH), luteinizing hormone (LH), anti-mullerian hormone (AMH) and bilateral antral follicle count (AFC), mean ovarian volume, and endometrial thickness of the two groups before and after treatment were compared. RESULT The effective rate of the observation group (93.10%) was higher than that of the control group (77.59%) (P < 0.05). After treatment, the index scores of traditional Chinese medicine syndromes in two groups were lower than before (P < 0.05), and the observation group was lower than the control group (P < 0.05). The serum E 2 and AMH levels in the two groups increased, while FSH and LH levels decreased compared with before treatment, and the observation group improved significantly compared with the control group (P < 0.05). The bilateral AFC, mean ovarian volume, and endometrial thickness of the two groups increased compared with before treatment, and the observation group was higher than the control group (P < 0.05). CONCLUSION On the basis of hormone replacement therapy, the combination of Wenjing Decoction and Tiaobu Chongren acupuncture and moxibustion therapy is effective in treating POF patients with deficiency-cold syndrome, which can effectively regulate their serum sex hormone levels and promote the recovery of ovarian function.
Collapse
Affiliation(s)
- Anlun Yi
- Laboratory of Chongqing Hechuan Maternal and Child Health Hospital, Hechuan District, Chongqing 401520, China
| | - Xianbing Qin
- Chongqing Yongchuan District Hospital of Traditional Chinese Medicine Laboratory, No. 2, Yingbin Avenue, Yongchuan District, Chongqing 402160, China
| | - Zhiyin Du
- School of Information Management, Chongqing Medical University, Chongqing 400016, China
| | - Tingling Wang
- Laboratory of Chongqing Hechuan Maternal and Child Health Hospital, Hechuan District, Chongqing 401520, China
| | - Fang Liu
- Chongqing Red Cross Hospital (Jiangbei District People's Hospital), No. 1, Jialing Village 1, Huaxin Street, Jiangbei District, Chongqing 400020, China
| |
Collapse
|
44
|
Cloke B, Rymer J. Premature ovarian insufficiency - the need for a genomic map. Climacteric 2021; 24:444-452. [PMID: 34308731 DOI: 10.1080/13697137.2021.1945025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Premature ovarian insufficiency (POI) is a life-long disorder of heterogeneous etiology, presenting as adolescent primary amenorrhea in its most severe form, with an overall incidence of 1%. Idiopathic POI accounts for up to 70% of women with POI; and genomic, genetic, epidemiological, familial and cohort studies demonstrate a genetic component to this condition. Currently, the only genetic tests routinely performed in non-syndromic POI are FMR1 premutation and cytogenetics, the latter specifically for X-chromosome abnormalities. However, a myriad of genetic aberrations has been identified and implicated, some of which act in a monogenic Mendelian fashion. The presence of multiple genetic aberrations and the complexity of POI genomics are hardly surprising since the embryological formation of the primordial oocyte pool, postnatal oogenesis and folliculogenesis are all highly complex pathways. With this review, the aim is to discuss the current genetic etiologies in the emerging field of POI genomics. Promising candidate genes include STAG3, SYCE1, FIGLA, NOBOX, FSHR, BMP15 and INHA. This area has the potential to progress rapidly in light of advances in genomic technologies. The development of a POI genomic map not only will assist in understanding the underlying molecular mechanisms affecting ovarian function but will also be essential in designing predictive and diagnostic gene panels as well as future novel therapeutic strategies.
Collapse
Affiliation(s)
- B Cloke
- Menopause Research Unit, McNair Gynaecology Centre, Guy's Hospital, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| | - J Rymer
- Menopause Research Unit, McNair Gynaecology Centre, Guy's Hospital, Guy's and St Thomas' Hospitals NHS Trust, London, UK.,School of Medical Education, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
45
|
Stevenson JC, Collins P, Hamoda H, Lambrinoudaki I, Maas AHEM, Maclaran K, Panay N. Cardiometabolic health in premature ovarian insufficiency. Climacteric 2021; 24:474-480. [PMID: 34169795 DOI: 10.1080/13697137.2021.1910232] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Premature ovarian insufficiency (POI) is an increasing public health problem with a prevalence now approaching 4%. POI results in adverse effects on the skeleton and central nervous system as well as disturbances of metabolic and cardiological factors that predispose to a major increased risk of cardiovascular disease (CVD). This article reviews the effects of the premature loss of ovarian function on lipids and lipoproteins, glucose and insulin metabolism, body composition, hemostasis and blood pressure, together with effects on the development of metabolic syndrome and diabetes mellitus. The article examines the effects of POI on vascular endothelial function and inflammation that result in arterial disease, and reviews the effects of hormone replacement therapy (HRT) on these various metabolic processes and on cardiovascular outcomes. It is essential that women with POI receive hormonal treatment to help prevent the development of CVD, and that this treatment is continued at least until the normal age of menopause. It appears that HRT has a more favorable effect than the combined oral contraceptive, but larger clinical trials are needed to establish the optimal treatment. Other therapeutic measures may need to be added to correct existing metabolic abnormalities and, in particular, attention to lifestyle factors such as diet and exercise must be encouraged.
Collapse
Affiliation(s)
- J C Stevenson
- National Heart & Lung Institute, Imperial College London, Royal Brompton Hospital, London, UK
| | - P Collins
- National Heart & Lung Institute, Imperial College London, Royal Brompton Hospital, London, UK
| | - H Hamoda
- Department of Gynaecology, King's College Hospital, London, UK
| | - I Lambrinoudaki
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - A H E M Maas
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K Maclaran
- Department of Gynaecology, Chelsea & Westminster Hospital, London, UK
| | - N Panay
- Department of Gynaecology, Queen Charlotte's and Chelsea & Westminster Hospitals, Imperial College London, London, UK
| |
Collapse
|
46
|
Prevention and treatment of iatrogenic premature ovarian insufficiency: interpretation of the first Chinese guideline on ovarian tissue cryopreservation and transplantation. GLOBAL HEALTH JOURNAL 2021. [DOI: 10.1016/j.glohj.2021.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
47
|
Cheng J, Ruan X, Zhou Q, Li Y, Du J, Jin F, Wang H, Gu M, Mueck AO. How much total ovarian tissue can be removed without compromising ovarian function? An animal study. Gynecol Endocrinol 2021; 37:240-245. [PMID: 32367735 DOI: 10.1080/09513590.2020.1760242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Removal of ovarian tissue is a new option for fertility preservation for certain cancer patients in China. The aim was to investigate the impact of stepwise removed tissue on hormone levels and follicles in rats. METHODS Six to ten-week old rats were divided into six groups (% total ovarian tissue): 1 = control (100%), 2 (75%), 3 (50%), 4 (25%), 5 (12.5%), 6 (0%, bilateral ovariectomy). Blood test was carried out fortnightly to assess estradiol (E2), progesterone (P), follicle-stimulating hormone (FSH), anti-Müllerian hormone (AMH), and inhibin B (INHB). Ovaries are obtained from surgical resections and from rats sacrificed after 12 weeks. RESULTS During 12 weeks, groups 4, 5, and 6 had higher FSH and lower AMH and INHB values compared to control (p< .05), but in group 4 E2 and P was not significantly different from control (p> .05). All ovarian function parameters stopped in groups 5 and 6. Follicle morphology was not significantly different between baseline and 12 weeks after surgery in groups 1-5. CONCLUSIONS For the first time, we demonstrated that even up to 75% of total ovarian tissue can be removed without impact on E2 and P production in rats, which, if confirmed in women, would mean that hazardous (or possibly contraindicated in cancer patients) hormone therapy is not required to avoid the negative consequences of hormone depletion.
Collapse
Affiliation(s)
- Jiaojiao Cheng
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
- Research Centre for Women's Health and University Women's Hospital of Tuebingen, University of Tuebingen, Tuebingen, Germany
| | - Qi Zhou
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yanglu Li
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Juan Du
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Fengyu Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Husheng Wang
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Muqing Gu
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Alfred Otto Mueck
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
- Research Centre for Women's Health and University Women's Hospital of Tuebingen, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
48
|
Jeon MJ, Choi YS, Kim ID, Criswell T, Atala A, Yoo JJ, Jackson JD. Engineering Functional Rat Ovarian Spheroids Using Granulosa and Theca Cells. Reprod Sci 2021; 28:1697-1708. [PMID: 33511540 DOI: 10.1007/s43032-020-00445-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/20/2020] [Indexed: 11/25/2022]
Abstract
Although menopausal hormone therapy (MHT) is the most effective approach to managing the loss of ovarian activity, serious side effects have been reported. Cell-based therapy is a promising alternative for MHT. This study constructed engineered ovarian cell spheroids and investigated their endocrine function. Theca and granulosa cells were isolated from ovaries of 10-week-old rats. Two types of engineered ovarian cell spheroids were fabricated through forced aggregation in microwells, multilayered spheroids with centralized granulosa aggregates surrounded by an outer layer of theca cells and mixed ovarian spheroids lacking spatial rearrangement. The ovarian cell spheroids were encapsulated into a collagen gel. Non-aggregated ovarian cells served as controls. The endocrine function of the engineered ovarian spheroids was assessed over 30 days. The structure of the spheroids was well maintained during culture. The secretion of 17β-estradiol from both types of engineered ovarian cell spheroids was higher than in the control group and increased continuously in a time-dependent manner. Secretion of 17β-estradiol in the multi-layered ovarian cell spheroids was higher than in the non-layered constructs. Increased secretion of progesterone was detected in the multi-layered ovarian cell spheroids at day 5 of culture and was sustained during the culture period. The initial secretion level of progesterone in the non-layered ovarian cell spheroids was similar to those from the controls and increased significantly from days 21 to 30. An in vitro rat model of engineered ovarian cell spheroids was developed that was capable of secreting sex steroid hormones, indicating that the hormone secreting function of ovaries can be recapitulated ex vivo and potentially adapted for MHT.
Collapse
Affiliation(s)
- Myung Jae Jeon
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101, Daehak-ro, Jong-gu, Seoul, 110-799, Republic of Korea
| | - Young Sik Choi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, 120-752, Republic of Korea
| | - Il Dong Kim
- Department of Obstetrics and Gynecology, Bundang Jeaseng General Hospital, 20, Seohyeon-ro 180beon-gil, Bundang-gu, Seognam-si, Gyeonggi-do, 13590, Republic of Korea
| | - Tracy Criswell
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
49
|
Yang L, Chen Y, Liu Y, Xing Y, Miao C, Zhao Y, Chang X, Zhang Q. The Role of Oxidative Stress and Natural Antioxidants in Ovarian Aging. Front Pharmacol 2021; 11:617843. [PMID: 33569007 PMCID: PMC7869110 DOI: 10.3389/fphar.2020.617843] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
The ovarian system comprises vital organs in females and is of great significance for the maintenance of reproductive potential and endocrine stability. Although complex pathogenesis undoubtedly contributes to ovarian aging, increasing attention is being paid to the extensive influence of oxidative stress. However, the role of oxidative stress in ovarian aging is yet to be fully elucidated. Exploring oxidative stress-related processes might be a promising strategy against ovarian aging. In this review, compelling evidence is shown that oxidative stress plays a role in the etiology of ovarian aging and promotes the development of other ovarian aging-related etiologies, including telomere shortening, mitochondrial dysfunction, apoptosis, and inflammation. In addition, some natural antioxidants such as quercetin, resveratrol, and curcumin have a protective role in the ovaries through multiple mechanisms. These findings raise the prospect of oxidative stress modulator-natural antioxidants as therapeutic interventions for delaying ovarian aging.
Collapse
Affiliation(s)
- Liuqing Yang
- Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yun Chen
- Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Liu
- The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Xing
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chenyun Miao
- Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Zhao
- Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangwei Chang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qin Zhang
- Guangxing Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
50
|
Rossetti R, Moleri S, Guizzardi F, Gentilini D, Libera L, Marozzi A, Moretti C, Brancati F, Bonomi M, Persani L. Targeted Next-Generation Sequencing Indicates a Frequent Oligogenic Involvement in Primary Ovarian Insufficiency Onset. Front Endocrinol (Lausanne) 2021; 12:664645. [PMID: 34803902 PMCID: PMC8600266 DOI: 10.3389/fendo.2021.664645] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/22/2021] [Indexed: 01/12/2023] Open
Abstract
Primary ovarian insufficiency (POI) is one of the major causes of female infertility associated with the premature loss of ovarian function in about 3.7% of women before the age of 40. This disorder is highly heterogeneous and can manifest with a wide range of clinical phenotypes, ranging from ovarian dysgenesis and primary amenorrhea to post-pubertal secondary amenorrhea, with elevated serum gonadotropins and hypoestrogenism. The ovarian defect still remains idiopathic in some cases; however, a strong genetic component has been demonstrated by the next-generation sequencing (NGS) approach of familiar and sporadic POI cases. As recent evidence suggested an oligogenic architecture for POI, we developed a target NGS panel with 295 genes including known candidates and novel genetic determinants potentially involved in POI pathogenesis. Sixty-four patients with early onset POI (range: 10-25 years) of our cohort have been screened with 90% of target coverage at 50×. Here, we report 48 analyzed patients with at least one genetic variant (75%) in the selected candidate genes. In particular, we found the following: 11/64 patients (17%) with two variants, 9/64 (14%) with three variants, 9/64 (14%) with four variants, 3/64 (5%) with five variants, and 2/64 (3%) with six variants. The most severe phenotypes were associated with either the major number of variations or a worse prediction in pathogenicity of variants. Bioinformatic gene ontology analysis identified the following major pathways likely affected by gene variants: 1) cell cycle, meiosis, and DNA repair; 2) extracellular matrix remodeling; 3) reproduction; 4) cell metabolism; 5) cell proliferation; 6) calcium homeostasis; 7) NOTCH signaling; 8) signal transduction; 9) WNT signaling; 10) cell death; and 11) ubiquitin modifications. Consistently, the identified pathways have been described in other studies dissecting the mechanisms of folliculogenesis in animal models of altered fertility. In conclusion, our results contribute to define POI as an oligogenic disease and suggest novel candidates to be investigated in patients with POI.
Collapse
Affiliation(s)
- Raffaella Rossetti
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- *Correspondence: Raffaella Rossetti, ; Luca Persani,
| | - Silvia Moleri
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Fabiana Guizzardi
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- Molecular Biology Laboratory, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Davide Gentilini
- Bioinformatics and Statistical Genomics Unit, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Laura Libera
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
| | - Anna Marozzi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Costanzo Moretti
- Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Francesco Brancati
- Medical Genetics, Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Human Functional Genomics, IRCCS San Raffaele Pisana, Rome, Italy
| | - Marco Bonomi
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Luca Persani
- Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Research, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- *Correspondence: Raffaella Rossetti, ; Luca Persani,
| |
Collapse
|