1
|
Katyal G, Kaur G, Ashraf H, Bodapati A, Hanif A, Okafor DK, Khan S. Systematic Review of the roles of Inositol and Vitamin D in improving fertility among patients with Polycystic Ovary Syndrome. Clin Exp Reprod Med 2024; 51:181-191. [PMID: 38599886 PMCID: PMC11372307 DOI: 10.5653/cerm.2023.06485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/20/2023] [Indexed: 04/12/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder among reproductive-age women. As a leading cause of anovulatory infertility, it complicates fertility treatments, including in vitro fertilization. The widely accepted 2003 Rotterdam diagnostic criteria for PCOS include sub-phenotypes based on variations in androgen excess, ovulatory dysfunction, and polycystic ovarian morphology. In this systematic review, we examined the impacts of inositol and vitamin D on fertility in PCOS. Adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 guidelines, we used relevant keywords to comprehensively search databases including PubMed, Google Scholar, and MDPI. From an initial pool of 345 articles, 10 met the inclusion criteria. The articles suggest that vitamin D and inositol, particularly myo-inositol and D-chiro-inositol, may represent therapeutic options for PCOS. Vitamin D influences ovarian follicular development, glucose regulation, and insulin sensitivity. When combined with metformin therapy, it is associated with improved menstrual regularity and ovulation. Inositol is crucial for cellular signaling, energy metabolism, glucose regulation, and fertility. This systematic review underscores the importance of investigating inositol and vitamin D within a PCOS management strategy, given the disorder's prevalence and impacts on fertility and metabolic health. Although these agents show promise, additional research could clarify their mechanisms of action and therapeutic benefits. This review emphasizes the need for exploration of effective treatments to improve the quality of life among individuals with PCOS. Inositol and vitamin D represent potential options, but more studies are required to elucidate their roles in the management of this condition.
Collapse
Affiliation(s)
- Gitika Katyal
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Gursharan Kaur
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Hafsa Ashraf
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Adiprasad Bodapati
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Ayesha Hanif
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Donatus Kaine Okafor
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| | - Safeera Khan
- California Institute of Behavioral Neurosciences and Psychology, Fairfield, CA, USA
| |
Collapse
|
2
|
Fitz V, Graca S, Mahalingaiah S, Liu J, Lai L, Butt A, Armour M, Rao V, Naidoo D, Maunder A, Yang G, Vaddiparthi V, Witchel SF, Pena A, Spritzer PM, Li R, Tay C, Mousa A, Teede H, Ee C. Inositol for Polycystic Ovary Syndrome: A Systematic Review and Meta-analysis to Inform the 2023 Update of the International Evidence-based PCOS Guidelines. J Clin Endocrinol Metab 2024; 109:1630-1655. [PMID: 38163998 PMCID: PMC11099481 DOI: 10.1210/clinem/dgad762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
CONTEXT Insulin resistance is common in women with polycystic ovary syndrome (PCOS). Inositol may have insulin sensitizing effects; however, its efficacy in the management of PCOS remains indeterminate. OBJECTIVE To inform the 2023 international evidence-based guidelines in PCOS, this systematic review and meta-analysis evaluated the efficacy of inositol, alone or in combination with other therapies, in the management of PCOS. DATA SOURCES Medline, PsycInfo, EMBASE, All EBM, and CINAHL from inception until August 2022. STUDY SELECTION Thirty trials (n = 2230; 1093 intervention, 1137 control), with 19 pooled in meta-analyses were included. DATA EXTRACTION Data were extracted for hormonal, metabolic, lipids, psychological, anthropometric, reproductive outcomes, and adverse effects by 1 reviewer, independently verified by a second. DATA SYNTHESIS Thirteen comparisons were assessed, with 3 in meta-analyses. Evidence suggests benefits for myo-inositol or D-chiro-inositol (DCI) for some metabolic measures and potential benefits from DCI for ovulation, but inositol may have no effect on other outcomes. Metformin may improve waist-hip ratio and hirsutism compared to inositol, but there is likely no difference for reproductive outcomes, and the evidence is very uncertain for body mass indexI. Myo-inositol likely causes fewer gastrointestinal adverse events compared with metformin; however, these are typically mild and self-limited. CONCLUSION The evidence supporting the use of inositol in the management of PCOS is limited and inconclusive. Clinicians and their patients should consider the uncertainty of the evidence together with individual values and preferences when engaging in shared decision-making regarding the use of inositol for PCOS.
Collapse
Affiliation(s)
- Victoria Fitz
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sandro Graca
- School of Health and Society, Faculty of Education, Health and Wellbeing, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Shruthi Mahalingaiah
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jing Liu
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Lily Lai
- Primary Care Research Centre, University of Southampton, Southampton SO17 1BJ, UK
| | - Ali Butt
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Mike Armour
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Vibhuti Rao
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Dhevaksha Naidoo
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Alison Maunder
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | - Guoyan Yang
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
| | | | - Selma F Witchel
- UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Alexia Pena
- Discipline of Paediatrics, The University of Adelaide and Robinson Research Institute, Adelaide 5005, Australia
| | - Poli Mara Spritzer
- Gynecological Endocrinology Unit, Division of Endocrinology, Hospital de Clinicas de Porto Alegre; Department of Physiology, Universidade Federal do Rio Grande do Sul, Rio Grande do Sul 91509-900, Brazil
| | - Rong Li
- Department of OB & GYN, Reproductive Medical Center, Peking University Third Hospital, Beijing 100191, China
| | - Chau Tay
- Monash Centre for Health Research and Implementation, Monash University, Clayton 3800, Australia
| | - Aya Mousa
- Monash Centre for Health Research and Implementation, Monash University, Clayton 3800, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation, Monash University, Clayton 3800, Australia
| | - Carolyn Ee
- NICM Health Research Institute, Western Sydney University, Penrith 2751, Australia
- Caring Futures Institute, Flinders University, Bedford Park 5042, Australia
| |
Collapse
|
3
|
Etrusco A, Laganà AS, Chiantera V, Buzzaccarini G, Unfer V. Myo-inositol in assisted reproductive technology from bench to bedside. Trends Endocrinol Metab 2024; 35:74-83. [PMID: 37798243 DOI: 10.1016/j.tem.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
Inositols are insulin-sensitizing compounds of promising efficacy in the management of polycystic ovary syndrome (PCOS). On the one hand, myo-inositol (myo-ins) plays a regulatory role in male and female reproductive function, influencing the development of oocytes, spermatozoa, and embryos. On the other hand, high concentrations of D-chiro-inositol (D-chiro-ins) in the ovary may adversely affect oocyte quality. This review analyses the available literature, which encourages the clinical use of myo-ins in assisted reproductive technologies (ARTs) due to its beneficial effects on female and male reproduction.
Collapse
Affiliation(s)
- Andrea Etrusco
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Unit of Obstetrics and Gynecology, 'Paolo Giaccone' Hospital, Palermo, Italy
| | - Antonio Simone Laganà
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Unit of Obstetrics and Gynecology, 'Paolo Giaccone' Hospital, Palermo, Italy; The Experts Group on Inositol in Basic and Clinical Research (EGOI)
| | - Vito Chiantera
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Unit of Gynecologic Oncology, National Cancer Institute - IRCCS - Fondazione 'G. Pascale', Naples, Italy
| | - Giovanni Buzzaccarini
- Obstetrics and Gynaecology Unit, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI); UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy.
| |
Collapse
|
4
|
Bashiri Z, Sheibak N, Amjadi F, Zandieh Z. The role of myo-inositol supplement in assisted reproductive techniques. HUM FERTIL 2023; 26:1044-1060. [PMID: 35730666 DOI: 10.1080/14647273.2022.2073273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/20/2021] [Indexed: 11/04/2022]
Abstract
Assisted reproductive techniques can help many infertile couples conceive. Therefore, there is a need for an effective method to overcome the widespread problems of infertile men and women. Oocyte and sperm quality can increase the chances of successful in vitro fertilisation. The maturation environment in which gametes are present can affect their competency for fertilisation. It is well established that myo-inositol (MI) plays a pivotal role in reproductive physiology. It participates in cell membrane formation, lipid synthesis, cell proliferation, cardiac regulation, metabolic alterations, and fertility. This molecule also acts as a direct messenger of insulin and improves glucose uptake in various reproductive tissues. Evidence suggests that MI regulates events such as gamete maturation, fertilisation, and embryo growth through intracellular Ca2 + release and various signalling pathways. In addition to the in-vivo production of MI from glucose in the reproductive organs, its synthesis by in vitro-cultured sperm and follicles has also been reported. Therefore, MI is suggested as a therapeutic approach to maintain sperm and oocyte health in men and women with reproductive disorders and individuals of reproductive age.
Collapse
Affiliation(s)
- Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility and Infertility Clinic, Hamedan, Iran
| | - Nadia Sheibak
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amjadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Effects of myo-inositol plus folic acid on ovarian morphology and oocyte quality in PCOS mouse model. ZYGOTE 2023; 31:111-122. [PMID: 36617989 DOI: 10.1017/s0967199422000557] [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: 01/10/2023]
Abstract
Although the role of myo-inositol (MYO) in promoting the oocyte quality of PCOS patients has been documented in human studies; the cellular effects of this supplement on oocytes have not been directly examined due to ethical limitations. In the first phase of this study, MYO dosimetry was carried out simultaneously with the PCOS model development. An effective dose was obtained following the assessment of fasting insulin and testosterone levels using ELISA and ovarian morphology appraisal by histopathology. In the second phase, following the continuous administration of the effective dose of MYO and dehydroepiandrosterone (DHEA), cellular evaluation was performed. The quality of oocytes from superovulation was analyzed by examining maturity and normal morphology percentage using a stereomicroscope, intracellular reactive oxygen species (ROS) and glutathione (GSH) levels using fluorometry, and ATP count evaluation using ELISA. The results revealed that, among the four different MYO concentrations, the 0.36 mg/g dose compared with the DHEA group reduced testosterone levels and large atretic antral follicles (LAtAnF) diameter. This dose also increased the corpus luteum count and the granulosa:theca (G/T)layer thickness ratio in antral follicles. Furthermore, this dose increased mature oocytes and normal morphology percentage, ATP count, and GSH levels; however, it decreased ROS levels in mature oocytes. Our findings provide the grounds for further cellular and molecular studies on the PCOS mouse model, suggesting that the improvement in mitochondrial function and its antioxidant properties is probably one of the mechanisms by which MYO increases oocyte quality.
Collapse
|
6
|
Medenica S, Spoltore ME, Ormazabal P, Marina LV, Sojat AS, Faggiano A, Gnessi L, Mazzilli R, Watanabe M. Female infertility in the era of obesity: The clash of two pandemics or inevitable consequence? Clin Endocrinol (Oxf) 2023; 98:141-152. [PMID: 35644933 PMCID: PMC10084349 DOI: 10.1111/cen.14785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/07/2022] [Accepted: 05/25/2022] [Indexed: 01/04/2023]
Abstract
Obesity is an epidemic that has led to a rise in the incidence of many comorbidities: among others, reduced fertility is often under-evaluated in clinical practice. The mechanisms underlying the link between reduced fertility and obesity are numerous, with insulin resistance, hyperglycaemia and the frequent coexistence of polycystic ovary syndrome being the most acknowledged. However, several other factors concur, such as gut microbiome alterations, low-grade chronic inflammation and oxidative stress. Not only do women with obesity take longer to conceive, but in vitro fertilization (IVF) is also less likely to succeed. We herein provide an updated state-of-the-art regarding the molecular bases of what we could define as dysmetabolic infertility, focusing on the clinical aspects, as well as possible treatment.
Collapse
Affiliation(s)
- Sanja Medenica
- Department of Internal Medicine, Endocrinology Section, Clinical Center of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Maria Elena Spoltore
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Paulina Ormazabal
- Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
- Laboratory of Obesity and Metabolism in Geriatrics and Adults (OMEGA), Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Macul, Santiago, Chile
| | - Ljiljana V Marina
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Antoan Stefan Sojat
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, Rome, Italy
| | - Mikiko Watanabe
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
7
|
GPX3 Overexpression in Cumulus Cells Entails a Poor Prognosis for Uterine Implantation of Morphotype A Embryos. BIOLOGY 2022; 11:biology11091361. [PMID: 36138840 PMCID: PMC9495337 DOI: 10.3390/biology11091361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022]
Abstract
Morphological embryo quality is an accurate prognostic tool for the success of assisted reproduction implantation, although complete certainty cannot be guaranteed. The transcriptome of the cumulus cells could be monitored as a faithful reflex of the physiological state of the oocytes, given the molecular crosstalk between both types of cells. Here, we compare the expression of specific genes related to oocyte competence, such as hyaluronic acid synthase 2 (HAS2), cell division control protein 42 (CDC42), connexin 43 (CX43), and glutathione peroxidase 3 (GPX3), in cumulus cells from implanted versus non-implanted embryos in 25 women, using RT-qPCR. After embryo transfer, two cohorts were differentiated: the pregnant group (women with the implantation of 100% of embryos transferred) versus the non-pregnant group (with an absence of embryo implantation), aiming to compare the possible differential expression of the selected genes in the cumulus cells of embryos from each group. HAS2, CDC42 and CX43 did not reveal differential expression between the two cohorts. However, GPX3 showed significantly reduced expression in the cumulus belonging to the pregnant group. Interestingly, even cumulus cells belonging only to morphotype A embryos showed a significantly lower expression of GPX3 in the pregnancy group. GPX3 overexpression in cumulus cells could be a poor prognostic indicator of implantation, discriminating beyond the capacity of the morphokinetic score. Unveiling the cumulus transcriptome could improve successful implantation in assisted reproduction treatments.
Collapse
|
8
|
Tanhaye Kalate Sabz F, Amjadi FS, Zandieh Z, Hosseini E, Aflatoonian R, Tabatabaei M, Mohammadian M, Ashrafi M. GM-CSF (granulocyte-macrophage colony-stimulating factor) treatment improves sperm parameters in men with oligoasthenoteratospermia via PI3K/AKT pathway. Andrologia 2022; 54:1618-1630. [PMID: 35545511 DOI: 10.1111/and.14427] [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: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/20/2022] [Indexed: 11/28/2022] Open
Abstract
Poor sperm quality in oligoasthenoteratospermia patients negatively affects assisted reproductive technology outcomes. Therefore, the development of sperm media is necessary to improve sperm parameters. This study investigated the effect of GM-CSF via PI3K/AKT pathway on sperm quality in OAT patients. Semen samples were collected from 20 OAT patients, and each sample was divided into two groups: Experiment and Control. In the experimental group, the samples were incubated with medium containing GM-CSF, and control samples were incubated without GM-CSF. Sperm parameters, mitochondrial membrane potential, acrosome reaction and DFI were studied; in addition, gene expression of PI3KR1, PI3KCA, GLUT1, GLUT3 and AKT1 was analysed, evaluation of PAKT/TAKT, and expression of GLUT 1, 3 was examined; subsequent fertilization rate and embryo quality were assessed. Our data showed that GM-CSF supplementation could significantly increase motility, mitochondrial activity, gene expression of PI3KCA, AKT1, the protein level of PAKT/TAKT and expression of GLUT 1, 3 while it decreases DNA fragmentation. The fertilization rate and embryo quality significantly improved in the treatment group. LY294002 had adverse effects on sperm motility and the PAKT/TAKT ratio. GM-CSF can improve in vitro sperm quality and could be a suitable supplement to sperm media for OAT patients.
Collapse
Affiliation(s)
| | - Fatemeh Sadat Amjadi
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Akbar-Abadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomical Science, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Obstetrics and Gynecology, School of Medicine, Shahid Akbar-Abadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Hosseini
- Department of Obstetrics and Gynecology, Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Tabatabaei
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Akbar-Abadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Mohammadian
- Department of Obstetrics and Gynecology, Mousavi Hospital, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Microbiology, Biology Research Center, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Mahnaz Ashrafi
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Akbar-Abadi Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Knockdown of bone morphogenetic protein 4 gene induces apoptosis and inhibits proliferation of bovine cumulus cells. Theriogenology 2022; 188:28-36. [DOI: 10.1016/j.theriogenology.2022.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 12/11/2022]
|
10
|
The effect of Myo-Inositol supplement on molecular regulation of folliculogenesis, steroidogenesis, and assisted reproductive technique outcomes in patients with polycystic ovarian syndrome. Mol Biol Rep 2022; 49:875-884. [PMID: 35040006 DOI: 10.1007/s11033-021-06833-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/12/2021] [Indexed: 10/19/2022]
Abstract
RESEARCH QUESTION The mechanism of Myo-Inositol, as an adjuvant, on key signaling pathways related to oocyte maturation, fertilization rate, and embryo quality as well as ovarian steroidogenesis in cumulus cells of PCOS patients, is still unclear. DESIGN Infertile patients who were candidates for ART cycles were divided into three groups (n = 30 in each group), including group 1: PCOS patients only receiving folic acid, group 2: PCOS patients receiving daily Myo-Inositol combined with folic acid, and a control group (group 3): normal ovulatory women without PCOS receiving only folic acid from 1 month prior to IVF cycle until the day of ovum pick up. During the ART procedure, oocytes maturation, fertilization rate, and embryo quality were assessed. The gene expressions of FSHR, LHR, CYP11A1, CYP19A1, 3β-HSD2, and StAR were also analyzed using qRT-PCR. Western blot analysis was performed for the evaluation of AKT, ERK, CREB, and AMPK phosphorylation. RESULT Despite equal number of retrieved oocytes, the percentages of MII oocytes, fertilization rate, and embryo quality were found to be significantly higher in group 2 due to the administration of inofolic. The expressions of all the studied genes were significantly higher in the cumulus cells of group 1 compared to the group 2. Higher phosphorylation of ERK1/2 was found in the groups 2 and 3 compared to the group 1. On the other hand, p-Akt has significantly decreased in the group 2 compared to the group 1. CONCLUSION Our study provides new insight into the molecular mechanism underlying the positive effect of Myo-Inositol on intrinsic ovarian defects in PCOS, steroidogenesis, oocyte maturation, fertilization rate, and embryo quality.
Collapse
|
11
|
Mei Q, Li H, Liu Y, Wang X, Xiang W. Advances in the study of CDC42 in the female reproductive system. J Cell Mol Med 2021; 26:16-24. [PMID: 34859585 PMCID: PMC8742232 DOI: 10.1111/jcmm.17088] [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: 07/14/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
CDC42 is a member of the Rho‐GTPase family and is involved in a variety of cellular functions including regulation of cell cycle progression, constitution of the actin backbone and membrane transport. In particular, CDC42 plays a key role in the establishment of polarity in female vertebrate oocytes, and essential to this major regulatory role is its local occupation of specific regions of the cell to ensure that the contractile ring is assembled at the right time and place to ensure proper gametogenesis. The multifactor controlled ‘inactivation‐activation’ process of CDC42 also allows it to play an important role in the multilevel signalling network, and the synergistic regulation of multiple genes ensures maximum precision during gametogenesis. The purpose of this paper is to review the role of CDC42 in the control of gametogenesis and to explore its related mechanisms, with the aim of further understanding the great research potential of CDC42 in female vertebrate germ cells and its future clinical translation.
Collapse
Affiliation(s)
- Qiaojuan Mei
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiying Li
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Liu
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Wang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpei Xiang
- Institute of Reproductive Health and Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Melatonin and Myo-Inositol: Supporting Reproduction from the Oocyte to Birth. Int J Mol Sci 2021; 22:ijms22168433. [PMID: 34445135 PMCID: PMC8395120 DOI: 10.3390/ijms22168433] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
Human pregnancy is a sequence of events finely tuned by several molecular interactions that come with a new birth. The precise interlocking of these events affecting the reproductive system guarantees safe embryo formation and fetal development. In this scenario, melatonin and myo-inositol seem to be pivotal not only in the physiology of the reproduction process, but also in the promotion of positive gestational outcomes. Evidence demonstrates that melatonin, beyond the role of circadian rhythm management, is a key controller of human reproductive functions. Similarly, as the most representative member of the inositol’s family, myo-inositol is essential in ensuring correct advancing of reproductive cellular events. The molecular crosstalk mediated by these two species is directly regulated by their availability in the human body. To date, biological implications of unbalanced amounts of melatonin and myo-inositol in each pregnancy step are growing the idea that these molecules actively contribute to reduce negative outcomes and improve the fertilization rate. Clinical data suggest that melatonin and myo-inositol may constitute an optimal dietary supplementation to sustain safe human gestation and a new potential way to prevent pregnancy-associated pathologies.
Collapse
|
13
|
Abdel-Maboud M, Menshawy A, Hasabo EA, Abdelraoof MI, Alshandidy M, Eid M, Menshawy E, Outani O, Menshawy A. The comparative effectiveness of 55 interventions in obese patients with polycystic ovary syndrome: A network meta-analysis of 101 randomized trials. PLoS One 2021; 16:e0254412. [PMID: 34280195 PMCID: PMC8289030 DOI: 10.1371/journal.pone.0254412] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) affects up to 18% of reproductive-age females. The prevalence of obesity in PCOS patients reaches up to 80%, which is 2-fold higher than the general population. OBJECTIVE The present study aimed to compare the effectiveness of 55 pharmacological interventions across 17 different outcomes in overweight/obese PCOS patients with hyperandrogenism manifestations for both short- and long-term follow-ups. A comprehensive literature search was performed on PubMed, Scopus, Embase, Science Direct, Web of Science, and Cochrane CENTRAL for randomized controlled trials comparing any conventional pharmacological intervention as a monotherapy or a combination in overweight/obese patients with polycystic ovary syndrome and hyperandrogenism manifestations. Extracted data included three main parameters; I. Anthropometric parameters (BMI, Waist and Hip circumferences, and Waist/HIP ratio), II. Hormonal parameters (FSH, LH, FSG, SHBG, Estradiol, Total Testosterone, Free testosterone, DHEAS, Androstenedione), and III. Metabolic parameters (Total Cholesterol, LDL-C, HDL-C, Triglycerides, Fasting glucose, Fasting glucose, HOMA-IR). Critical appraisal and risk of bias assessments were performed using the modified Jadad scale, and the overall quality of this network meta-analysis was evaluated according to the CINeMA framework. We performed both a pairwise meta-analysis and a network meta-analysis to evaluate the effect sizes with 95% CI, and we calculated the surface under the cumulative ranking curve (SUCRA) for each intervention. RESULTS Our final search on May 15th 2021 retrieved 23,305 unique citations from searching six electronic databases. Eventually, 101 RCTs of 108 reports with a total of 8,765 patients were included in our systematic review and multi-treatments meta-analysis. 55 different interventions were included: 22 monotherapies, and 33 combinations. The two-dimensional cluster ranking of the average SUCRA values for metabolic and hormonal parameters with significant estimates revealed flutamide (77.5%, 70%; respectively) as the highest and rosiglitazone (38.2%, 26.3%; respectively) as the lowest, in terms of the overall efficacy in reducing weight and hyperandrogenism. However, cyproterone-acetate+ethinylestradiol exhibited a higher ranking in improving hormonal parameters (71.1%), but even a lower-ranking regarding metabolic parameters (34.5%). CONCLUSIONS AND RELEVANCE Current evidence demonstrated the superiority of flutamide in improving both metabolic and hormonal parameters, and the higher efficacy of cyproterone-acetate+ethinylestradiol only in improving hormonal parameters. Nearly all interventions were comparable in female hormones, FGS, HDL, glucose, and insulin levels improvements.
Collapse
Affiliation(s)
| | - Amr Menshawy
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | | | | | - Muhammad Eid
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | - Oumaima Outani
- Faculty of Medicine and Pharmacy of Rabat, Mohammed 5 University, Rabat, Morocco
| | - Ahmed Menshawy
- Department of Obstetrics and Gynecology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
14
|
Notch signaling pathway in cumulus cells reflecting zygote and embryo quality in polycystic ovary syndrome. Arch Gynecol Obstet 2021; 304:1097-1105. [PMID: 33772329 DOI: 10.1007/s00404-021-06039-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE The present study aimed to explore the associations between the expression pattern of molecules in the Notch pathway in the cumulus cells of polycystic ovary syndrome (PCOS) patients and the quality of zygotes and embryos. METHODS A total of 200 cumulus complexes surrounding mature oocytes were obtained from 40 patients with and without PCOS undergoing intracytoplasmic sperm injection (ICSI). The expressions of Notch-1, Notch-2, and Notch-3 genes were examined by Reverse Transcription Q-PCR assay. Moreover, immunocytochemistry was performed for the expressions of Jagged-1 and Jagged-2 proteins. The correlations between the Notch receptors and their ligand expressions and the qualities of the zygote and embryo were investigated. RESULTS The expression levels of Notch-2, Notch-3, Jagged-1, and Jagged-2 were significantly lower in patients with PCOS than in normal women (p < 0.05), while Notch-1 showed no meaningful difference between the groups. A positive correlation was found between Notch-1 and embryo quality. Furthermore, only Notch-2 and Jagged-2 marginally correlated with zygote quality. CONCLUSION The data of the present study indicated that evaluating the molecules in the Notch pathway in PCOS patients' cumulus cells provides a novel approach to predict the zygote and embryo quality. However, further studies on a larger population are needed to validate this finding.
Collapse
|
15
|
Merviel P, James P, Bouée S, Le Guillou M, Rince C, Nachtergaele C, Kerlan V. Impact of myo-inositol treatment in women with polycystic ovary syndrome in assisted reproductive technologies. Reprod Health 2021; 18:13. [PMID: 33468143 PMCID: PMC7816413 DOI: 10.1186/s12978-021-01073-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/05/2021] [Indexed: 02/02/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is marked in 30 to 40% by insulin resistance and hyperandrogenism. Myo-inositol (MI) increases insulin sensitivity, decreases hyperandrogenism and improves the menstrual cycle. Its effect during assisted reproductive technologies (ART) has been studied by many authors. We conducted a review of the literature on the impact of MI administration in PCOS women in assisted reproductive technologies. Myo-inositol is effective in normalizing ovarian function, improving oocyte and embryo quality in PCOS, however further evaluations by large multicentre randomized controlled trials are needed to assess the clinical pregnancy and live birth rates in ART.
Collapse
Affiliation(s)
- Philippe Merviel
- Fertility and ART Department, Brest University Hospital, 2 Avenue Foch, 29200, Brest, France.
| | - Pandora James
- Fertility and ART Department, Brest University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Sarah Bouée
- Fertility and ART Department, Brest University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Mathilde Le Guillou
- Fertility and ART Department, Brest University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Camille Rince
- Fertility and ART Department, Brest University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Charlotte Nachtergaele
- Endocrinology and Metabolism Department, Brest University Hospital, Boulevard Tanguy Prigent, 29200, Brest, France
| | - Véronique Kerlan
- Endocrinology and Metabolism Department, Brest University Hospital, Boulevard Tanguy Prigent, 29200, Brest, France
| |
Collapse
|
16
|
Novielli C, Anelli GM, Lisso F, Marzorati A, Parrilla B, Oneta M, Savasi VM, Cetin I, Mandò C. Effects of α-lipoic acid and myo-inositol supplementation on the oocyte environment of infertile obese women: A preliminary study. Reprod Biol 2020; 20:541-546. [PMID: 33371944 DOI: 10.1016/j.repbio.2020.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 11/19/2022]
Abstract
Obesity is becoming pandemic and is associated with impaired reproductive potential. Oxidative stress, low-grade chronic inflammation and mitochondrial dysfunctions, which characterize obesity, strongly affect oocyte environment and function. Supplementation with antioxidant and anti-inflammatory compounds has been suggested to improve fertility. Here we evaluated the effect of α-lipoic acid and myo-inositol supplementation on the oocyte environment of infertile obese women. Nineteen normal-weight and twenty-three obese women, infertile for non-ovarian reasons, were recruited. For two months before ovarian stimulation, all women received 400 μg/die folic acid, whereas 15 obese were additionally supplemented with 800 mg α-lipoic acid, 2 g myo-inositol/die. Antioxidant capacity was measured in follicular fluid by enzymatic assay; mitochondrial DNA (mtDNA) content and mRNA levels of two respiratory chain subunits were analyzed in granulosa cells by Real-time PCR. Pregnancy rate was similar between normal-weight and treated obese, and lower in untreated obese patients. Supplemented women showed significantly higher antioxidant levels in follicular fluid compared to the two groups taking only folic acid. Conversely, granulosa cells mtDNA content was decreased in treated and higher in untreated obese patients compared to normal-weight women, suggesting mtDNA increases to compensate for oxidative-stress damages. Reduced expression of respiratory subunits in untreated obese may confirm mitochondria impairment. Interestingly, mtDNA levels inversely correlated to both total and metaphase II oocyte number. In this preliminary study, combined supplementation of α-lipoic acid and myo-inositol in infertile obese women was associated with amelioration in the oxidative status of the oocyte environment, possibly contributing to a higher pregnancy rate.
Collapse
Affiliation(s)
- Chiara Novielli
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy
| | - Gaia M Anelli
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy
| | - Fabrizia Lisso
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy
| | - Anna Marzorati
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy
| | - Bina Parrilla
- ASST Fatebenefratelli Sacco, Unit of Obstetrics and Gynecology, Milano, Italy
| | - Monica Oneta
- ASST Fatebenefratelli Sacco, Unit of Obstetrics and Gynecology, Milano, Italy
| | - Valeria M Savasi
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy; ASST Fatebenefratelli Sacco, Unit of Obstetrics and Gynecology, Milano, Italy
| | - Irene Cetin
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy; ASST Fatebenefratelli Sacco, Unit of Obstetrics and Gynecology, Milano, Italy
| | - Chiara Mandò
- Università degli Studi di Milano, "Luigi Sacco" Department of Biomedical and Clinical Sciences, Milano, Italy.
| |
Collapse
|
17
|
Mikwar M, MacFarlane AJ, Marchetti F. Mechanisms of oocyte aneuploidy associated with advanced maternal age. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2020; 785:108320. [PMID: 32800274 DOI: 10.1016/j.mrrev.2020.108320] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/30/2022]
Abstract
It is well established that maternal age is associated with a rapid decline in the production of healthy and high-quality oocytes resulting in reduced fertility in women older than 35 years of age. In particular, chromosome segregation errors during meiotic divisions are increasingly common and lead to the production of oocytes with an incorrect number of chromosomes, a condition known as aneuploidy. When an aneuploid oocyte is fertilized by a sperm it gives rise to an aneuploid embryo that, except in rare situations, will result in a spontaneous abortion. As females advance in age, they are at higher risk of infertility, miscarriage, or having a pregnancy affected by congenital birth defects such as Down syndrome (trisomy 21), Edwards syndrome (trisomy 18), and Turner syndrome (monosomy X). Here, we review the potential molecular mechanisms associated with increased chromosome segregation errors during meiosis as a function of maternal age. Our review shows that multiple exogenous and endogenous factors contribute to the age-related increase in oocyte aneuploidy. Specifically, the weight of evidence indicates that recombination failure, cohesin deterioration, spindle assembly checkpoint (SAC) disregulation, abnormalities in post-translational modification of histones and tubulin, and mitochondrial dysfunction are the leading causes of oocyte aneuploidy associated with maternal aging. There is also growing evidence that dietary and other bioactive interventions may mitigate the effect of maternal aging on oocyte quality and oocyte aneuploidy, thereby improving fertility outcomes. Maternal age is a major concern for aneuploidy and genetic disorders in the offspring in the context of an increasing proportion of mothers having children at increasingly older ages. A better understanding of the mechanisms associated with maternal aging leading to aneuploidy and of intervention strategies that may mitigate these detrimental effects and reduce its occurrence are essential for preventing abnormal reproductive outcomes in the human population.
Collapse
Affiliation(s)
- Myy Mikwar
- Department of Biology, Carleton University, Ottawa, Ontario, Canada; Nutrition Research Division, Health Canada, Ottawa, Ontario, Canada
| | - Amanda J MacFarlane
- Department of Biology, Carleton University, Ottawa, Ontario, Canada; Nutrition Research Division, Health Canada, Ottawa, Ontario, Canada
| | - Francesco Marchetti
- Department of Biology, Carleton University, Ottawa, Ontario, Canada; Mechanistic Studies Division, Health Canada, Ottawa, Ontario, Canada.
| |
Collapse
|