1
|
Soliman MY, Idris OAF, Momtaz M, Kortam MA, ELNoury MA, Saleh HA, Abulnour A, Ali AA, Abbas M, Shaaban OM, Din ASSE, Gaafar H, Orief Y, Safwat M. Expert consensus on the role of supplementation in obstetrics and gynecology using modified delphi method. Arch Gynecol Obstet 2024; 309:639-650. [PMID: 38153519 PMCID: PMC10808492 DOI: 10.1007/s00404-023-07310-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/13/2023] [Indexed: 12/29/2023]
Abstract
PURPOSE To reach a consensus among obstetrics and gynecology experts on the effects of micronutrient supplementation on fertility and pregnancy to aid clinicians in decision-making and create a unified approach to managing micronutrient deficiencies in women, by performing a modified Delphi study. METHODS A three-round modified Delphi process was conducted among a Delphi panel of 38 Egyptian experts to define recommendations regarding the role of supplementation on fertility and pregnancy in women of reproductive age. A literature review was performed and supporting evidence was graded to help guide the recommendations based on available evidence. RESULTS A total of 62 statements were developed for discussion and voting. Out of the 62 statements, 60 statements reached expert consensus. Statements were divided into two domains. The first domain discussed the role of supplementation in fertility: optimizing natural fertility, polycystic ovary syndrome (PCOS), in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI), unexplained infertility, and endometriosis, whereas the second domain was concerned with the role of supplementation in pregnancy during the prenatal, antenatal, and postnatal periods. CONCLUSION In this work, a modified Delphi methodology was implemented to reach a consensus on the use of micronutrient supplementation in women of reproductive age. These recommendations can help clinicians in their practice, guide future research, and identify gaps in the market for the pharmaceutical industry. This clinical guidance can be extrapolated to similar communities.
Collapse
Affiliation(s)
| | | | - Mohamed Momtaz
- Obstetrics and Gynecology, Al Kasr Al Aini, Cairo University, Cairo, Egypt
| | | | | | - Hisham Ali Saleh
- Obstetrics and Gynecology, Alexandria University, Alexandria, Egypt
| | - Ayman Abulnour
- Obstetrics and Gynecology, Ain Shams University, Cairo, Egypt
| | - Ashraf Abo Ali
- Obstetrics and Gynecology, Alexandria University, Alexandria, Egypt
- El-Madina Fertility Centers, Alexandria, Egypt
| | - Mostafa Abbas
- Obstetrics and Gynecology, Zagazig University, Zagazig, Egypt
| | - Omar M Shaaban
- Obstetrics and Gynecology, Assiut University, Assiut, Egypt
| | | | - Hassan Gaafar
- Obstetrics and Gynecology, Al Kasr Al Aini, Cairo University, Cairo, Egypt
| | - Yasser Orief
- Obstetrics and Gynecology, Alexandria University, Alexandria, Egypt.
| | - Michael Safwat
- Medical Affairs Department, Eva Pharma for Pharmaceuticals and Medical Appliances, Cairo, Egypt
| |
Collapse
|
2
|
Nie X, Dong X, Hu Y, Xu F, Hu C, Shu C. Coenzyme Q10 Stimulate Reproductive Vatality. Drug Des Devel Ther 2023; 17:2623-2637. [PMID: 37667786 PMCID: PMC10475284 DOI: 10.2147/dddt.s386974] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/15/2023] [Indexed: 09/06/2023] Open
Abstract
Female infertility and pregnancy maintenance are associate with various factors, including quantity and quality of oocytes, genital inflammation, endometriosis, and other diseases. Women are even diagnosed as unexplained infertility or unexplained recurrent spontaneous abortion when failed to achieve pregnancy with current treatment, which are urgent clinical issues need to be addressed. Coenzyme Q10 (CoQ10) is a lipid-soluble electron carrier in the mitochondrial electron transport chain. It is not only essential for the mitochondria to produce energy, but also function as an antioxidant to maintain redox homeostasis in the body. Recently, the capacity of CoQ10 to reduce oxidative stress (OS), enhance mitochondrial activity, regulate gene expression and inhibit inflammatory responses, has been discovered as a novel adjuvant in male reproductive performance enhancing in both animal and human studies. Furthermore, CoQ10 is also proved to regulate immune balance, antioxidant, promote glucose and lipid metabolism. These properties will bring highlight for ovarian dysfunction reversing, ovulation ameliorating, oocyte maturation/fertilization promoting, and embryonic development optimizing. In this review, we systematically discuss the pleiotropic effects of CoQ10 in female reproductive disorders to investigate the mechanism and therapeutic potential to provide a reference in subsequent studies.
Collapse
Affiliation(s)
- Xinyu Nie
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xinru Dong
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yuge Hu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Fangjun Xu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Cong Hu
- Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Chang Shu
- Obstetrics and Gynecology Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
3
|
Jiang H, Chen L, Tian T, Shi H, Huang N, Chi H, Yang R, Long X, Qiao J. Inflammation mediates the effect of adiposity and lipid metabolism indicators on the embryogenesis of PCOS women undergoing in vitro fertilization/intracytoplasmic sperm injection. Front Endocrinol (Lausanne) 2023; 14:1198602. [PMID: 37560312 PMCID: PMC10408295 DOI: 10.3389/fendo.2023.1198602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a complex reproductive endocrine and metabolic disease affecting women of reproductive age. The low-grade chronic inflammation in PCOS is considered to be associated with obesity and dyslipidemia. We aim to investigate the potential mediating role of white blood cell (WBC) count, a representative inflammatory marker, in the effect of adiposity and lipid metabolism indicators on IVF/ICSI outcomes in PCOS women. Methods We conducted a retrospective cohort study of 1,534 PCOS women who underwent their first IVF/ICSI cycles with autologous oocytes at a reproductive center from January 2018 to December 2020. The associations between PCOS women's adiposity and lipid metabolism indicators and WBC count and IVF/ICSI outcomes were examined using multivariable generalized linear models. Mediation analyses were conducted to evaluate the possible mediating role of WBC count. Results We found significant dose-dependent correlations between adiposity and lipid metabolism indicators and IVF/ICSI outcomes (i.e., hormone levels on the ovulatory triggering day, oocyte development outcomes, fertilization, early embryo development outcomes, and pregnancy outcomes) (all p < 0.05), as well as between adiposity and lipid metabolism indicators and WBC count (all p < 0.001). Increasing WBC count was associated with adverse oocyte and embryonic development outcomes (all p < 0.05). Mediation analyses suggested that increasing serum TG and LDL-C levels and decreasing serum HDL-C level were significantly associated with reduced high-quality Day 3 embryo count in PCOS women, with 21.51%, 9.75%, and 14.10% mediated by WBC count, respectively (all p < 0.05). Conclusions We observed significant associations between lipid metabolism indicators and high-quality Day 3 embryo count in PCOS women, partially mediated by inflammation-related mechanisms, suggesting the potential intervention target for improving embryo quality in PCOS women.
Collapse
Affiliation(s)
- Huahua Jiang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Lixue Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Tian Tian
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Huifeng Shi
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Centre for Healthcare Quality Management in Obstetrics, Beijing, China
| | - Ning Huang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Hongbin Chi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rui Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Xiaoyu Long
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
4
|
Agrawal S, Sanap SN, Bisen AC, Biswas A, Choudhury AD, Verma SK, Jaiswal S, Narender T, Bhatta RS. Preclinical pharmacokinetics of 4-hydroxy isoleucine using LC-MS/MS: a potential polycystic ovary syndrome phytopharmaceutical therapeutics. Bioanalysis 2023; 15:711-725. [PMID: 37354099 DOI: 10.4155/bio-2023-0074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2023] Open
Abstract
Aim: To study the preclinical pharmacokinetics of 4-hydroxy isoleucine (4-HIL) targeted for polycystic ovary syndrome. Methodology: The quantitative bioanalysis of 4-HIL in different biological matrices in female Sprage-Dawley rats using LC-MS/MS. Results: At 50 mg/kg, 4-HIL had 56.8% absolute oral bioavailability. It was quickly absorbed and distributed in various tissues in order of small intestine > kidney > ovary > spleen > lung > liver > heart > brain after oral administration. Moreover, 11.07% of 4-HIL was recovered in urine and feces within 72 h. Conclusion: 4-HIL levels in vital organs were found safe, as per tissue distribution results. Hence, 4-HIL could be used as promising therapeutics for management of polycystic ovary syndrome.
Collapse
Affiliation(s)
- Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
| | - Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
| | - Abhijit Deb Choudhury
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
| | - Sarvesh Kumar Verma
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
| | - Shubham Jaiswal
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
- Division of Medicinal & Process Chemistry, CSIR - Central Drug Research Institute, Lucknow, 226031, India
| | - Tadigoppula Narender
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
- Division of Medicinal & Process Chemistry, CSIR - Central Drug Research Institute, Lucknow, 226031, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR - Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific & Innovative Research, Ghaziabad, 201002, India
| |
Collapse
|
5
|
Zhang T, He Q, Xiu H, Zhang Z, Liu Y, Chen Z, Hu H. Efficacy and Safety of Coenzyme Q10 Supplementation in the Treatment of Polycystic Ovary Syndrome: a Systematic Review and Meta-analysis. Reprod Sci 2023; 30:1033-1048. [PMID: 35941510 DOI: 10.1007/s43032-022-01038-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022]
Abstract
The aim of this study is to evaluate the efficacy and safety of coenzyme Q10 supplementation in the treatment of polycystic ovary syndrome (PCOS). We first searched PubMed, Wanfang Data, CNKI, Embase, ClinicalTrial.gov, and other databases. The retrieval time from the establishment of the database to January 2021. We collected relevant randomized controlled trials (RCTs) about coenzyme Q10 in the treatment of PCOS. Risk of bias assessment and meta-analysis of RCTs were performed using RevMan 5.0 software. This systematic review and meta-analysis include a total of 9 RCTs involving 1021 patients. The results show that the addition of coenzyme Q10 may improve insulin resistance (HOMA-IR (WMD - 0.67 [- 0.87, - 0.48], P < 0.00001); fasting insulin (WMD - 1.75 [- 2.65, - 0.84], P = 0.0002); fasting plasma glucose (WMD - 5.20 [- 8.86, - 1.54], P = 0.005)), improve sex hormone levels (FSH (SMD - 0.45 [0.11, 0.78], P = 0.009); testosterone (SMD - 0.28 [- 0.49, - 0.06], P = 0.01)), and improve blood lipids (triglycerides (SMD - 0.49 [- 0.89, - 0.09], P = 0.02); total cholesterol (SMD - 0.35 [- 0.56, - 0.14], P = 0.001); LDL-C (SMD - 0.22 [- 0.43, - 0.01], P = 0.04); HDL-C (SMD 0.22 [0.01, 0.43], P = 0.04)). Only one RCT reported adverse events, and they found that patients had no adverse effects or symptoms following supplementation. Based on the current evidence, it could be considered that the addition of CoQ10 is a safe therapy to improve PCOS by improving insulin resistance (reduce HOMA-IR, FINS, FPG), increasing sex hormone levels (increase FSH, reduce testosterone), and improving blood lipids (reduce TG, TC, LDL-C, and increased HDL-C).
Collapse
Affiliation(s)
- Tianqing Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Qi He
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Hao Xiu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - ZiZhu Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Yao Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Zhenrong Chen
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China
| | - Hengjing Hu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hunan Province, Hengyang, China.
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
6
|
Coenzyme Q10 and Endocrine Disorders: An Overview. Antioxidants (Basel) 2023; 12:antiox12020514. [PMID: 36830072 PMCID: PMC9952344 DOI: 10.3390/antiox12020514] [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: 01/03/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Mitochondrial dysfunction and oxidative stress have been implicated in the pathogenesis of a number of endocrine disorders; this, in turn, suggests a potential role for the vitamin-like substance coenzyme Q10 (CoQ10) in the pathogenesis and treatment of these disorders, on the basis of its key roles in mitochondrial function, and as an antioxidant. In this article we have therefore reviewed the role of CoQ10 deficiency and supplementation in disorders of the thyroid, pancreas, gonads, pituitary and adrenals, with a particular focus on hyperthyroidism, type II diabetes, male infertility and polycystic ovary syndrome.
Collapse
|
7
|
Qi S, Liang Q, Yang L, Zhou X, Chen K, Wen J. Effect of Coenzyme Q10 and transcutaneous electrical acupoint stimulation in assisted reproductive technology: a retrospective controlled study. Reprod Biol Endocrinol 2022; 20:167. [PMID: 36476305 PMCID: PMC9730642 DOI: 10.1186/s12958-022-01043-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To investigate the effects of coenzyme Q10 (CoQ10) and transcutaneous electrical acupoint stimulation (TEAS) pretreatment on pregnancy in patients with poor ovarian response (POR). METHODS A total of 330 POR patients who were pretreated with CoQ10 or CoQ10 combined with TEAS before their in vitro fertilization/intracytoplasmic sperm injection and embryo transfer (IVF/ICSI-ET) cycles and who were not pretreated were selected and divided into CoQ10 group (group A, n = 110), CoQ10 + TEAS group (group B, n = 110) and control group (group C, n = 110). For patients with 2 or more transfer cycles, only the information of the first cycle was included. Ovarian function, response to gonadotropin (Gn) stimulation, and pregnancy outcomes of the three groups were compared in the IVF/ICSI-ET cycles. RESULTS After pretreatment, basal FSH, total Gn dosage and duration were comparable among the three groups (all p-value > 0.05), basal E2 in group B decreased significantly compared with the control group (p = 0.022). Endometrial thickness on the human chorionic gonadotropin (hCG) day, antral follicle counts (AFC), the numbers of oocytes, metaphase II (MII) eggs and excellent embryos in the two pretreatment groups were significantly increased compared with group C (all p-value < 0.001), but the rates of MII oocytes, fertilization and excellent embryos had no apparent change. The endometrial thickness on the day of hCG, the numbers of MII eggs and excellent embryos in group B were higher than those in group A (p < 0.001; p = 0.020; p = 0.027; respectively). The embryo implantation rate (IR), clinical pregnancy rate (CPR) and live birth rate (LBR) in group B were significantly higher than those in group C (p = 0.022; p = 0.010; p = 0.019; respectively), but not significantly different from group A. CONCLUSION CoQ10 alone or in combination with TEAS are effective methods for IVF/ICSI-ET adjuvant therapy, which can significantly improve ovarian reactivity, increase the numbers of retrieved eggs and superior embryos, and improve endometrial receptivity. Adjuvant TEAS on the basis of CoQ10 can significantly enhance pregnancy rates, but CoQ10 alone failed to present such an obvious effect.
Collapse
Affiliation(s)
- Shanqin Qi
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, 250355, People's Republic of China
| | - Qi Liang
- Reproductive Medical Center, the Second Hospital affiliated to Shandong University of Traditional Chinese Medicine, Jingba Road, Jinan, 250001, People's Republic of China
| | - Lixia Yang
- Reproductive Medical Center, the Second Hospital affiliated to Shandong University of Traditional Chinese Medicine, Jingba Road, Jinan, 250001, People's Republic of China
| | - Xueyuan Zhou
- Reproductive Medical Center, the Second Hospital affiliated to Shandong University of Traditional Chinese Medicine, Jingba Road, Jinan, 250001, People's Republic of China
| | - Kun Chen
- Reproductive Medical Center, the Second Hospital affiliated to Shandong University of Traditional Chinese Medicine, Jingba Road, Jinan, 250001, People's Republic of China
| | - Ji Wen
- Reproductive Medical Center, the Second Hospital affiliated to Shandong University of Traditional Chinese Medicine, Jingba Road, Jinan, 250001, People's Republic of China.
| |
Collapse
|
8
|
Hornos Carneiro MF, Colaiácovo MP. Beneficial antioxidant effects of Coenzyme Q10 on reproduction. VITAMINS AND HORMONES 2022; 121:143-167. [PMID: 36707133 DOI: 10.1016/bs.vh.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This chapter focuses on preclinical and clinical studies conducted in recent years that contribute to increasing knowledge on the role of Coenzyme Q10 in female reproductive health. General aspects of CoQ10, such as its role as an antioxidant and in mitochondrial bioenergetics are considered. The age-dependent decline in human female reproductive potential is associated with cellular mitochondrial dysfunction and oxidative stress, and in some cases accompanied by a decrease in CoQ10 levels. Herein, we discuss experimental and clinical evidence on CoQ10 protective effects on reproductive health. We also address the potential of supplementation with this coenzyme to rescue reprotoxicity induced by exposure to environmental xenobiotics. This review not only contributes to our general understanding of the effects of aging on female reproduction but also provides new insights into strategies promoting reproductive health. The use of CoQ10 supplementation can improve reproductive performance through the scavenging of reactive oxygen species and free radicals. This strategy can constitute a low-risk and low-cost strategy to attenuate the impact on fertility related to aging and exposure to environmental chemicals.
Collapse
Affiliation(s)
| | - Monica P Colaiácovo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
9
|
Cheng X, He B. Clinical and Biochemical Potential of Antioxidants in Treating Polycystic Ovary Syndrome. Int J Womens Health 2022; 14:467-479. [PMID: 35392500 PMCID: PMC8982783 DOI: 10.2147/ijwh.s345853] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of infertility in reproductive-age women. Increased reactive oxygen species levels and decreased antioxidant capacity in PCOS patients can lead to metabolic disorders and damage the ovarian tissues, resulting in the occurrence of related symptoms. Antioxidants have been used in the treatment of PCOS and have yielded satisfactory outcomes due to their ability to counter oxidative stress. Many experiments on PCOS patients have proved that antioxidants can not only improve the ovarian environment, promote follicular maturation, and elevate oocyte quantities but can also regulate lipid and glucose metabolism as well as vascular endothelial cell function in PCOS patients, thereby attenuating adiposity and reducing the occurrence rate of chronic complications to ensure that patients can obtain long-term benefits. This review describes the use of antioxidants in PCOS, which have been used in the treatment.
Collapse
Affiliation(s)
- Xiangyi Cheng
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Bing He
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| |
Collapse
|
10
|
Karamali M, Gholizadeh M. The effects of coenzyme Q10 supplementation on metabolic profiles and parameters of mental health in women with polycystic ovary syndrome. Gynecol Endocrinol 2022; 38:45-49. [PMID: 34664527 DOI: 10.1080/09513590.2021.1991910] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Evaluating the impact of coenzyme Q10 (CoQ10) supplementation on hormonal indices, mental health, and biomarkers of inflammatory responses and oxidative stress among female patients suffering from polycystic ovary syndrome (PCOS). METHODS The present double-blinded, placebo-controlled randomized clinical trial consisted of 55 PCOS women (aged 18-40 years old), who were randomized into groups receiving 100 mg/day of CoQ10 (28 cases) or placebo (27 cases) for 12 weeks. RESULTS The supplementation of CoQ10 decreased significantly the scores of Beck Depression Inventory (BDI) (p = .03) and Beck Anxiety Inventory (BAI) (p = .01) and high-sensitivity C-reactive protein (hs-CRP) level (p = .005) when comparing with the placebo group. Moreover, CoQ10 group exhibited a significant drop in total testosterone (p = .004), dehydroepiandrosterone sulfate (DHEAS) (p < .001), hirsutism (p = .002) and malondialdehyde (MDA) (p = .001) levels in the serum, and a significant rise in sex hormone-binding globulin (SHBG) (p < .001) and total antioxidant capacity (TAC) (p < .001) levels in the serum than the placebo group. CONCLUSIONS 12-week supplementation of CoQ10 to PCOS women showed beneficial impact on BDI, BAI, hs-CRP, total testosterone, DHEAS, hirsutism, SHBG, TAC and MDA levels.
Collapse
Affiliation(s)
- Maryam Karamali
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Gholizadeh
- Department of Gynecology and Obstetrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Okamoto M, Nakamura A, Maeda A, Kameda Y, Sugawara K, Takekawa M, Ogino M, Makino E, Ishii N, Fujisawa A, Yamamoto Y, Kashiba M. Coenzyme Q10 levels increase with embryonic development in medaka. J Clin Biochem Nutr 2022; 70:231-239. [PMID: 35692673 PMCID: PMC9130062 DOI: 10.3164/jcbn.21-148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022] Open
Abstract
Coenzyme Q10 is an important molecule for mitochondrial respiration and as an antioxidant. Maintenance of the ovum in a good condition is considered to be important for successful fertilization and development, which has been reported to be promoted by coenzyme Q10. In this study, we investigated the level of coenzyme Q10 during ovum fertilization and maturation. We attempted to analyze coenzyme Q10 levels during ovum development in species that use coenzyme Q10 but not coenzyme Q9. It was shown that medaka produces coenzyme Q10. We then measured the amount of coenzyme Q10 after fertilization of medaka ovum and found that it increased. The amount of free cholesterol biosynthesized from acetyl CoA as well as coenzyme Q10 increased during development, but the increase in coenzyme Q10 was more pronounced. The mRNA expression level of coq9 also increased during embryonic development, but the mRNA expression levels of other coenzyme Q10 synthases did not. These results suggest that the coq9 gene is upregulated during the development of medaka ovum after fertilization, resulting in an increase in the amount of coenzyme Q10 in the ovum. Medaka, which like humans has coenzyme Q10, is expected to become a model animal for coenzyme Q10 research.
Collapse
Affiliation(s)
| | | | - Ayaka Maeda
- School of Bionics, Tokyo University of Technology
| | - Yuka Kameda
- School of Bionics, Tokyo University of Technology
| | | | | | - Momoko Ogino
- School of Bionics, Tokyo University of Technology
| | - Eriko Makino
- School of Bionics, Tokyo University of Technology
| | - Nagisa Ishii
- School of Bionics, Tokyo University of Technology
| | | | | | | |
Collapse
|
12
|
Gharaei R, Mahdavinezhad F, Samadian E, Asadi J, Ashrafnezhad Z, Kashani L, Amidi F. Antioxidant supplementations ameliorate PCOS complications: a review of RCTs and insights into the underlying mechanisms. J Assist Reprod Genet 2021; 38:2817-2831. [PMID: 34689247 PMCID: PMC8609065 DOI: 10.1007/s10815-021-02342-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most important gynecological disorders of women in the age of reproduction. Different hormonal and inflammatory cross-talks may play in the appearance of its eventual complications as a leading cause of infertility. Excessive production of reactive oxygen species over the power of the antioxidant system as oxidative stress is known to contribute to a variety of diseases like PCOS. Thus, the utilization of antioxidants can be efficient in preventing or assistant in treating these diseases. In this review, we describe the clinical trial studies that have examined the efficiency of antioxidant strategies against PCOS and the possible underlying mechanisms. The investigations presented here lead us to consider that targeting oxidative stress pathways is probably a powerful promising therapeutic approach towards PCOS. There is preparatory evidence of the effectiveness of antioxidant interventions in ameliorating some of the PCOS complications, including metabolic and hormonal disorders. Due to limited data and relatively few clinical trials, many of these interventions need further investigation before they can be considered effective agents for routine clinical use.
Collapse
Affiliation(s)
- Roghaye Gharaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Mahdavinezhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Samadian
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Jahanbakhsh Asadi
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zhaleh Ashrafnezhad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ladan Kashani
- Department of Obstetrics and Gynecology, School of Medicine, Arash Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Rodríguez-Varela C, Labarta E. Does Coenzyme Q10 Supplementation Improve Human Oocyte Quality? Int J Mol Sci 2021; 22:ijms22179541. [PMID: 34502447 PMCID: PMC8431086 DOI: 10.3390/ijms22179541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/19/2022] Open
Abstract
Acquiring oocyte competence requires optimal mitochondrial function and adequate ATP levels. In this context, CoQ10 supplementation may improve human oocyte quality and subsequent reproductive performance given its role in ATP synthesis and mitochondrial protection from ROS oxidative damage. In infertility treatments, CoQ10 therapy can be orally supplied to promote a more favorable environment for oocyte development in vivo or by its addition to culture media in an attempt to improve its quality in vitro. Human clinical studies evaluating the impact of CoQ10 on reproductive performance are summarized in this review, although the available data do not clearly prove its ability to improve human oocyte quality. The main objective is to provide readers with a complete overview of this topic's current status as well as the keys for potential future research lines that may help to take this therapy to clinical practice. Indeed, further clinical trials are needed to confirm these results along with molecular studies to evaluate the impact of CoQ10 supplementation on oxidative stress status and mitochondrial function in human gametes.
Collapse
Affiliation(s)
| | - Elena Labarta
- IVI Foundation—IIS La Fe, 46026 Valencia, Spain;
- IVIRMA Valencia, 46015 Valencia, Spain
| |
Collapse
|
14
|
Ammar IMM, Salem MAA. Amelioration of polycystic ovary syndrome-related disorders by supplementation of thymoquinone and metformin. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00076-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is one of the most common endocrine disorders of reproductive age women with a prevalence of 4-12%. The study aimed to investigate the potential benefit of adding thymoquinone to metformin in alleviating symptoms of polycystic ovarian syndrome. Two hundred seven overweight and obese PCOS patients were divided into two groups. Patients in group A received metformin 500 mg three times daily for 6 months. Patients in group B received a combination of metformin 500 mg and thymoquinone in the form of black cumin oil 500 mg capsules three times daily for 6 months. Follow-up was done after 3 and 6 months from the beginning of the study for evaluation of menstrual cycle pattern, body mass index, waist circumference, hip circumference, and waist:hip ratio, oral glucose tolerance test, glycosylated hemoglobin A1C, superoxide dismutase activity, and malondialdehyde concentration.
Results
Patients who received a combination of black cumin oil capsules and metformin, showed a significant decrease in number of patients suffering from amenorrhea or oligomenorrhea (P = 0.031), significant (P = 0.00) weight reduction (reduced BMI), and body fat redistribution (reduced W/H ratio), regaining oxidative balance with significant increase of SOD activity (P = 0.00) and decrease of MDA concentrations (P = 0.03). Regarding the number of prediabetic patients, there was no statistically significant difference (P = 0.89) when both groups were compared at the end of study period.
Conclusions
Black cumin oil supplementation constitutes a beneficial added value to metformin in ameliorating PCOS-related disorders with resumption of menstrual regularity, weight reduction, change of body fat distribution, and regaining oxidative balance.
Collapse
|
15
|
Ammar IMM, Abdou AM. Effect of Ubiquinol supplementation on ovulation induction in Clomiphene Citrate resistance. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2021. [DOI: 10.1186/s43043-021-00070-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Clomiphene Citrate is considered the gold-standard for induction of ovulation and has been used for several years to treat PCOS related infertility. Unfortunately, 15-40% of women with PCOS are resistant to Clomiphene Citrate. The study aimed to evaluate potential benefits of adding the active form of Coenzyme Q10 (Ubiquinol) to Clomiphene Citrate compared with Human Menopausal Gonadotropins (hMG) in Clomiphene Citrate resistant PCOS patients. 148 PCOS Patients with Clomiphene Citrate resistance were randomized into two groups (A and B). In group A, controlled ovarian stimulation was done by Clomiphene Citrate 150 mg daily (from 2nd till 6th day of cycle) together with Ubiquinol starting from 2nd day till day of hCG triggering in a dose of 100 mg orally once daily. In group B, hMG was given from 2nd day of the cycle in a dose ranging from 75 to 225 IU. Serial transvaginal ultrasonography was done starting on cycle day 8 and continued till size of leading follicle reaches 18 mm or more then ovulation triggering was done. Thereafter, patients were advised for a timed intercourse (TI) after 36 hours. A blood sample was withdrawn seven days after hCG triggering, for measurement of serum progesterone. If the Patient presented with a missed period for one week, a serum sample was sent for β-hCG.
Results
There were no statistically significant differences (P > 0.05) between studied groups regarding; number of cases reaching mature follicular size, number of stimulated cycles, endometrial thickness on the day of hCG triggering, mid-luteal serum progesterone, positive serum pregnancy test and clinical pregnancy rate.
Conclusions
Addition of Ubiquinol to Clomiphene Citrate improved ovarian responsiveness in Clomiphene Citrate resistant patients with results comparable to conventional hMG stimulation protocol.
Collapse
|
16
|
Chronopoulou E, Seifalian A, Stephenson J, Serhal P, Saab W, Seshadri S. Preconceptual care for couples seeking fertility treatment, an evidence-based approach. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.xfnr.2020.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Palomba S, Piltonen TT, Giudice LC. Endometrial function in women with polycystic ovary syndrome: a comprehensive review. Hum Reprod Update 2020; 27:584-618. [PMID: 33302299 DOI: 10.1093/humupd/dmaa051] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/29/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility. An endometrial component has been suggested to contribute to subfertility and poor reproductive outcomes in affected women. OBJECTIVE AND RATIONALE The aim of this review was to determine whether there is sufficient evidence to support that endometrial function is altered in women with PCOS, whether clinical features of PCOS affect the endometrium, and whether there are evidence-based interventions to improve endometrial dysfunction in PCOS women. SEARCH METHODS An extensive literature search was performed from 1970 up to July 2020 using PubMed and Web of Science without language restriction. The search included all titles and abstracts assessing a relationship between PCOS and endometrial function, the role played by clinical and biochemical/hormonal factors related to PCOS and endometrial function, and the potential interventions aimed to improve endometrial function in women with PCOS. All published papers were included if considered relevant. Studies having a specific topic/hypothesis regarding endometrial cancer/hyperplasia in women with PCOS were excluded from the analysis. OUTCOMES Experimental and clinical data suggest that the endometrium differs in women with PCOS when compared to healthy controls. Clinical characteristics related to the syndrome, alone and/or in combination, may contribute to dysregulation of endometrial expression of sex hormone receptors and co-receptors, increase endometrial insulin-resistance with impaired glucose transport and utilization, and result in chronic low-grade inflammation, immune dysfunction, altered uterine vascularity, abnormal endometrial gene expression and cellular abnormalities in women with PCOS. Among several interventions to improve endometrial function in women with PCOS, to date, only lifestyle modification, metformin and bariatric surgery have the highest scientific evidence for clinical benefit. WIDER IMPLICATIONS Endometrial dysfunction and abnormal trophoblast invasion and placentation in PCOS women can predispose to miscarriage and pregnancy complications. Thus, patients and their health care providers should advise about these risks. Although currently no intervention can be universally recommended to reverse endometrial dysfunction in PCOS women, lifestyle modifications and metformin may improve underlying endometrial dysfunction and pregnancy outcomes in obese and/or insulin resistant patients. Bariatric surgery has shown its efficacy in severely obese PCOS patients, but a careful evaluation of the benefit/risk ratio is warranted. Large scale randomized controlled clinical trials should address these possibilities.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Obstetrics and Gynecology, Grande Ospedale Metropolitano of Reggio Calabria, Reggio Calabria, Italy
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Linda C Giudice
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
18
|
Rodríguez-Varela C, Labarta E. Clinical Application of Antioxidants to Improve Human Oocyte Mitochondrial Function: A Review. Antioxidants (Basel) 2020; 9:antiox9121197. [PMID: 33260761 PMCID: PMC7761442 DOI: 10.3390/antiox9121197] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondria produce adenosine triphosphate (ATP) while also generating high amounts of reactive oxygen species (ROS) derived from oxygen metabolism. ROS are small but highly reactive molecules that can be detrimental if unregulated. While normally functioning mitochondria produce molecules that counteract ROS production, an imbalance between the amount of ROS produced in the mitochondria and the capacity of the cell to counteract them leads to oxidative stress and ultimately to mitochondrial dysfunction. This dysfunction impairs cellular functions through reduced ATP output and/or increased oxidative stress. Mitochondrial dysfunction may also lead to poor oocyte quality and embryo development, ultimately affecting pregnancy outcomes. Improving mitochondrial function through antioxidant supplementation may enhance reproductive performance. Recent studies suggest that antioxidants may treat infertility by restoring mitochondrial function and promoting mitochondrial biogenesis. However, further randomized, controlled trials are needed to determine their clinical efficacy. In this review, we discuss the use of resveratrol, coenzyme-Q10, melatonin, folic acid, and several vitamins as antioxidant treatments to improve human oocyte and embryo quality, focusing on the mitochondria as their main hypothetical target. However, this mechanism of action has not yet been demonstrated in the human oocyte, which highlights the need for further studies in this field.
Collapse
Affiliation(s)
- Cristina Rodríguez-Varela
- IVI Foundation—IIS La Fe, Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026 Valencia, Spain;
- Correspondence:
| | - Elena Labarta
- IVI Foundation—IIS La Fe, Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026 Valencia, Spain;
- IVIRMA Valencia, Plaza de la Policía Local 3, 46015 Valencia, Spain
| |
Collapse
|
19
|
Florou P, Anagnostis P, Theocharis P, Chourdakis M, Goulis DG. Does coenzyme Q 10 supplementation improve fertility outcomes in women undergoing assisted reproductive technology procedures? A systematic review and meta-analysis of randomized-controlled trials. J Assist Reprod Genet 2020; 37:2377-2387. [PMID: 32767206 PMCID: PMC7550497 DOI: 10.1007/s10815-020-01906-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/28/2020] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE Increased oxidative stress has been identified as a pathogenetic mechanism in female infertility. However, the effect of specific antioxidants, such as coenzyme Q10 (CoQ10), on the outcomes after assisted reproductive technologies (ART) has not been clarified. The aim of this study was to systematically review and meta-analyze the best available evidence regarding the effect of CoQ10 supplementation on clinical pregnancy (CPR), live birth (LBR), and miscarriage rates (MR) compared with placebo or no-treatment in women with infertility undergoing ART. METHODS A comprehensive literature search was conducted in PubMed (MEDLINE), Cochrane, and Scopus, from inception to March 2020. Data were expressed as odds ratio (OR) with 95% confidence intervals (CI). The I2 index was employed for heterogeneity. RESULTS Five randomized-controlled trials fulfilled eligibility criteria (449 infertile women; 215 in CoQ10 group and 234 in placebo/no treatment group). Oral supplementation of CoQ10 resulted in an increase of CPR when compared with placebo or no-treatment (28.8% vs. 14.1%, respectively; OR 2.44, 95% CI 1.30-4.59, p = 0.006; I2 32%). This effect remained significant when women with poor ovarian response and polycystic ovarian syndrome were analyzed separately. No difference between groups was observed regarding LBR (OR 1.67, 95% CI 0.66-4.25, p = 0.28; I2 34%) and MR (OR 0.61, 95% CI 0.13-2.81, p = 0.52; I2 0%). CONCLUSIONS Oral supplementation of CoQ10 may increase CPR when compared with placebo or no-treatment, in women with infertility undergoing ART procedures, without an effect on LBR or MR.
Collapse
Affiliation(s)
- Panagiota Florou
- Department of Endocrinology, Police Medical Center of Thessaloniki, 326 Monastiriou Street, 54121, Thessaloniki, Greece
| | - Panagiotis Anagnostis
- Department of Endocrinology, Police Medical Center of Thessaloniki, 326 Monastiriou Street, 54121, Thessaloniki, Greece
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Ring Road, Nea Efkarpia, 54601, Thessaloniki, Greece
| | - Patroklos Theocharis
- Department of Endocrinology, Police Medical Center of Thessaloniki, 326 Monastiriou Street, 54121, Thessaloniki, Greece
| | - Michail Chourdakis
- Laboratory of Hygiene, Social and Preventive Medicine and Medical Statistics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, "Papageorgiou" General Hospital, Aristotle University of Thessaloniki, Ring Road, Nea Efkarpia, 54601, Thessaloniki, Greece
| |
Collapse
|
20
|
Morsy AA, Sabri NA, Mourad AM, Mojahed EM, Shawki MA. Randomized controlled open-label study of the effect of vitamin E supplementation on fertility in clomiphene citrate-resistant polycystic ovary syndrome. J Obstet Gynaecol Res 2020; 46:2375-2382. [PMID: 32885585 DOI: 10.1111/jog.14467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/07/2020] [Accepted: 08/23/2020] [Indexed: 11/25/2022]
Abstract
AIM To evaluate the effect of vitamin E on ovulation and pregnancy in women with clomiphene citrate (CC)-resistant polycystic ovary syndrome (PCOS). METHODS A prospective, randomized, controlled, open label study was conducted on women with CC-resistant PCOS. Patients were randomized, to either control group (n = 30), who received metformin 500 mg thrice daily, in addition to 150 mg/day CC for 5 days starting from day 3 of menstruation for three menstruation cycles, or vitamin E group (n = 30) who received vitamin E 1500 IU/day for the whole study period in addition to metformin and CC with the same previous regimen. The primary outcome was cumulative ovulation rate, while secondary outcomes were pregnancy rate, serum midluteal progesterone, mean follicular diameter, number of dominant follicles and endometrial thickness. RESULTS Ovulation was reported in 57 (64.8%) of 88 cycles in the control group and 63 (73.3%) of 86 cycles in the vitamin E group (P = 0.227), while pregnancy was reported in 4 (4.5%) of 88 cycles in the control group and 6 (7%) of 86 cycles in the vitamin E group (P = 0.491).There were nonsignificant differences between groups regarding serum midluteal progesterone, number of dominant follicles and mean follicular diameter. Endometrial thickness was significantly higher in the vitamin E group compared to the control group. CONCLUSION The findings of this trial do not support the hypothesis that vitamin E may increase the ovulation and pregnancy rates in women with clomiphene citrate-resistant PCOS.
Collapse
Affiliation(s)
- Ahmed A Morsy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Misr University for Science and Technology, Giza, Egypt
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Abdelrehim M Mourad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Misr University for Science and Technology, Giza, Egypt
| | - Eman M Mojahed
- Department of Obstetrics and Gynecology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - May A Shawki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
21
|
Abstract
EDITORIAL NOTE M.G. Showell, R. Mackenzie‐Proctor, V. Jordan, and R.J. Hart, “Antioxidants for Female Subfertility,” Cochrane Database of Systematic Reviews, no. 8 (2020): CD007807, https://doi.org/10.1002/14651858.CD007807.pub4 This Editorial Note is for the above article, published online on August 27, 2020, in Cochrane Library (cochranelibrary.com), and has been issued by the Publisher, John Wiley & Sons Ltd, in agreement with Cochrane. The Editorial note has been agreed due to concerns discovered by the Cochrane managing editor regarding the retraction of six studies in the Review (Badawy et al. 2006, 10.1016/j.fertnstert.2006.02.097; El Refaeey et al. 2014, 10.1016/j.rbmo.2014.03.011; El Sharkwy & Abd El Aziz 2019a, https://doi.org/10.1002/ijgo.12902; Gerli et al. 2007, https://doi.org/10.26355/eurrev_202309_33752, full text: https://europepmc.org/article/MED/18074942; Ismail et al. 2014, http://dx.doi.org/10.1016/j.ejogrb.2014.06.008; Hashemi et al. 2017, https://doi.org/10.1080/14767058.2017.1372413). In addition, expressions of concern have been published for two studies (Jamilian et al. 2018, https://doi.org/10.1007/s12011-017-1236-3; Zadeh Modarres 2018, https://doi.org/10.1007/s12011-017-1148-2). The retracted studies will be moved to the Excluded Studies table, and their impact on the review findings will be investigated and acted on accordingly in a future update. Initial checks indicate that removal of the six retracted studies did not make an appreciable difference to the results. Likewise, the studies for which Expressions of Concern were issued will be moved to the Awaiting classification table; they did not report any review outcomes, so removal will have no impact on the review findings. BACKGROUND A couple may be considered to have fertility problems if they have been trying to conceive for over a year with no success. This may affect up to a quarter of all couples planning a child. It is estimated that for 40% to 50% of couples, subfertility may result from factors affecting women. Antioxidants are thought to reduce the oxidative stress brought on by these conditions. Currently, limited evidence suggests that antioxidants improve fertility, and trials have explored this area with varied results. This review assesses the evidence for the effectiveness of different antioxidants in female subfertility. OBJECTIVES To determine whether supplementary oral antioxidants compared with placebo, no treatment/standard treatment or another antioxidant improve fertility outcomes for subfertile women. SEARCH METHODS We searched the following databases (from their inception to September 2019), with no language or date restriction: Cochrane Gynaecology and Fertility Group (CGFG) specialised register, CENTRAL, MEDLINE, Embase, PsycINFO, CINAHL and AMED. We checked reference lists of relevant studies and searched the trial registers. SELECTION CRITERIA We included randomised controlled trials (RCTs) that compared any type, dose or combination of oral antioxidant supplement with placebo, no treatment or treatment with another antioxidant, among women attending a reproductive clinic. We excluded trials comparing antioxidants with fertility drugs alone and trials that only included fertile women attending a fertility clinic because of male partner infertility. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. The primary review outcome was live birth; secondary outcomes included clinical pregnancy rates and adverse events. MAIN RESULTS We included 63 trials involving 7760 women. Investigators compared oral antioxidants, including: combinations of antioxidants, N-acetylcysteine, melatonin, L-arginine, myo-inositol, carnitine, selenium, vitamin E, vitamin B complex, vitamin C, vitamin D+calcium, CoQ10, and omega-3-polyunsaturated fatty acids versus placebo, no treatment/standard treatment or another antioxidant. Only 27 of the 63 included trials reported funding sources. Due to the very low-quality of the evidence we are uncertain whether antioxidants improve live birth rate compared with placebo or no treatment/standard treatment (odds ratio (OR) 1.81, 95% confidence interval (CI) 1.36 to 2.43; P < 0.001, I2 = 29%; 13 RCTs, 1227 women). This suggests that among subfertile women with an expected live birth rate of 19%, the rate among women using antioxidants would be between 24% and 36%. Low-quality evidence suggests that antioxidants may improve clinical pregnancy rate compared with placebo or no treatment/standard treatment (OR 1.65, 95% CI 1.43 to 1.89; P < 0.001, I2 = 63%; 35 RCTs, 5165 women). This suggests that among subfertile women with an expected clinical pregnancy rate of 19%, the rate among women using antioxidants would be between 25% and 30%. Heterogeneity was moderately high. Overall 28 trials reported on various adverse events in the meta-analysis. The evidence suggests that the use of antioxidants makes no difference between the groups in rates of miscarriage (OR 1.13, 95% CI 0.82 to 1.55; P = 0.46, I2 = 0%; 24 RCTs, 3229 women; low-quality evidence). There was also no evidence of a difference between the groups in rates of multiple pregnancy (OR 1.00, 95% CI 0.63 to 1.56; P = 0.99, I2 = 0%; 9 RCTs, 1886 women; low-quality evidence). There was also no evidence of a difference between the groups in rates of gastrointestinal disturbances (OR 1.55, 95% CI 0.47 to 5.10; P = 0.47, I2 = 0%; 3 RCTs, 343 women; low-quality evidence). Low-quality evidence showed that there was also no difference between the groups in rates of ectopic pregnancy (OR 1.40, 95% CI 0.27 to 7.20; P = 0.69, I2 = 0%; 4 RCTs, 404 women). In the antioxidant versus antioxidant comparison, low-quality evidence shows no difference in a lower dose of melatonin being associated with an increased live-birth rate compared with higher-dose melatonin (OR 0.94, 95% CI 0.41 to 2.15; P = 0.89, I2 = 0%; 2 RCTs, 140 women). This suggests that among subfertile women with an expected live-birth rate of 24%, the rate among women using a lower dose of melatonin compared to a higher dose would be between 12% and 40%. Similarly with clinical pregnancy, there was no evidence of a difference between the groups in rates between a lower and a higher dose of melatonin (OR 0.94, 95% CI 0.41 to 2.15; P = 0.89, I2 = 0%; 2 RCTs, 140 women). Three trials reported on miscarriage in the antioxidant versus antioxidant comparison (two used doses of melatonin and one compared N-acetylcysteine versus L-carnitine). There were no miscarriages in either melatonin trial. Multiple pregnancy and gastrointestinal disturbances were not reported, and ectopic pregnancy was reported by only one trial, with no events. The study comparing N-acetylcysteine with L-carnitine did not report live birth rate. Very low-quality evidence shows no evidence of a difference in clinical pregnancy (OR 0.81, 95% CI 0.33 to 2.00; 1 RCT, 164 women; low-quality evidence). Low quality evidence shows no difference in miscarriage (OR 1.54, 95% CI 0.42 to 5.67; 1 RCT, 164 women; low-quality evidence). The study did not report multiple pregnancy, gastrointestinal disturbances or ectopic pregnancy. The overall quality of evidence was limited by serious risk of bias associated with poor reporting of methods, imprecision and inconsistency. AUTHORS' CONCLUSIONS In this review, there was low- to very low-quality evidence to show that taking an antioxidant may benefit subfertile women. Overall, there is no evidence of increased risk of miscarriage, multiple births, gastrointestinal effects or ectopic pregnancies, but evidence was of very low quality. At this time, there is limited evidence in support of supplemental oral antioxidants for subfertile women.
Collapse
Affiliation(s)
- Marian G Showell
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | | | - Vanessa Jordan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Roger J Hart
- School of Women's and Infants' Health, The University of Western Australia, King Edward Memorial Hospital and Fertility Specialists of Western Australia, Subiaco, Perth, Australia
| |
Collapse
|
22
|
Kyei G, Sobhani A, Nekonam S, Shabani M, Ebrahimi F, Qasemi M, Salahi E, Fardin A. Assessing the effect of MitoQ 10 and Vitamin D3 on ovarian oxidative stress, steroidogenesis and histomorphology in DHEA induced PCOS mouse model. Heliyon 2020; 6:e04279. [PMID: 32760818 PMCID: PMC7393412 DOI: 10.1016/j.heliyon.2020.e04279] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/13/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) continues to be one of the most complex reproductive and endocrine disorder among women of reproductive age. Recent reports have identified close interaction of Vitamin D deficiency and oxidative stress (OS) in exacerbating the pathophysiology of PCOS. This current study aims at assessing the combine effect of MitoQ10 and Vitamin D3 on dehydroepiandrosterone (DHEA) induced PCOS. Following successful induction of PCOS using DHEA, mice were organized into five groups (n = 8) namely: Negative Control (NC), Vitamin D3 Vehicle (VDV), Vitamin D3 (VD), MitoQ10 (MQ), Vitamin D3 plus MitoQ10 (V+M) and DHEA, ethanol and distilled water, Vitamin D3, MitoQ10 and Vitamin D3 plus MitoQ10 were respectively administered for 20 consecutive days. The study also included positive control (PC) group (n = 8) in which no treatment was applied. Treatment effects were assessed using hormonal assays, biochemical assays, Real-Time PCR, western blotting and histological analysis. Combination of Vitamin D3 and MitoQ10 significantly reduced levels of estradiol, progesterone, FSH, LH, LH/FSH, SOD and MDA. The expression rate of mRNAs of 3β-HSD, Cyp19a1, Cyp11a1, StAR, Keap1, HO-1 and Nrf2 were also significantly low in V+M group. Moreover, the histomorphological inspection of ovaries from this group revealed many healthy follicles at various stages of development including few atretic follicles, pre-antral and antral follicles and many corpora lutea. The characteristics observed in this group were in many ways similar to that of the PC group. The combination of MitoQ10 and Vitamin D3 may be potential candidate to ameliorate PCOS.
Collapse
Affiliation(s)
- Gordon Kyei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aligholi Sobhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Nekonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ebrahimi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Qasemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elnaz Salahi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amidi Fardin
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Ma L, Cai L, Hu M, Wang J, Xie J, Xing Y, Shen J, Cui Y, Liu XJ, Liu J. Coenzyme Q10 supplementation of human oocyte in vitro maturation reduces postmeiotic aneuploidies. Fertil Steril 2020; 114:331-337. [PMID: 32646587 DOI: 10.1016/j.fertnstert.2020.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To evaluate the effect of coenzyme Q10 (CoQ10) supplementation on oocyte maturation rates and postmeiotic aneuploidy rates during in vitro maturation (IVM) of human oocytes. DESIGN Clinical laboratory observation. SETTING Hospital and university laboratories. PATIENT(S) Forty-five patients aged ≥38 years and 18 patients aged ≤30 years undergoing in vitro fertilization. INTERVENTION(S) The germinal vesicle-stage oocytes and associated cumulus cells were cultured in IVM media for 24-48 hours with or without 50 μmol/L CoQ10. Oocyte maturation rates were determined based on the presence or absence of the first polar body. Postmeiotic aneuploidies were determined using next-generation sequencing analyses of biopsied polar bodies. MAIN OUTCOME MEASURE(S) Oocyte maturation rates, postmeiotic oocyte aneuploidy rates, and chromosome aneuploidy frequencies. RESULT(S) In women aged 38-46 years, 50 μmol/L CoQ10 significantly increased oocyte maturation rates (82.6% vs. 63.0%; P=.035), reduced oocyte aneuploidy rates (36.8% vs. 65.5%; P=.020), and reduced chromosome aneuploidy frequencies (4.1% vs. 7.0%; P=.012. In women aged ≤30 years, we failed to demonstrate an effect of CoQ10 on oocyte maturation rates or postmeiotic aneuploidies. CONCLUSION(S) CoQ10 supplementation during IVM increased oocyte maturation rates and reduced postmeiotic aneuploidies for older women.
Collapse
Affiliation(s)
- Long Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China; The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Lingbo Cai
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Mengting Hu
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jiazi Xie
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yan Xing
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jiandong Shen
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yugui Cui
- The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - X Johné Liu
- Ottawa Hospital Research Institute, The Ottawa Hospital - General Campus, Ottawa, Ontario, Canada; Department of Obstetrics and Gynaecology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jiayin Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, People's Republic of China; The State Key Laboratory of Reproductive Medicine, Clinical Center for Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
24
|
Bordewijk EM, Wang R, Askie LM, Gurrin LC, Thornton JG, van Wely M, Li W, Mol BW. Data integrity of 35 randomised controlled trials in women’ health. Eur J Obstet Gynecol Reprod Biol 2020; 249:72-83. [DOI: 10.1016/j.ejogrb.2020.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 11/28/2022]
|
25
|
Blumenfeld Z. What Is the Best Regimen for Ovarian Stimulation of Poor Responders in ART/IVF? Front Endocrinol (Lausanne) 2020; 11:192. [PMID: 32362870 PMCID: PMC7180183 DOI: 10.3389/fendo.2020.00192] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
The infertile patients with aging ovaries-also sometimes referred to as impending premature ovarian insufficiency (POI), impending premature ovarian failure (POF), or poor ovarian responders (POR), constitute a significant and increasing bulk of the patients appealing to IVF/ART. Different causes have been cited in the literature, among the identified etiologies, including chromosomal and genetic etiology, metabolic, enzymatic, iatrogenic, toxic, autoimmune, and infectious causes. Although the most successful and ultimate treatment of POI/POF/POR patients is egg donation (ED), many, if not most, of these infertile women are reluctant to consent to ED upon the initial diagnostic interview, requesting alternative solutions despite the low odds for success. Despite anecdotal case reports, no unequivocal treatment proved to be successful for these patients in prospective randomized controlled trials. Nevertheless, the addition of growth hormone (GH) to ovarian stimulation in POR with GH deficiency may improve the results of controlled ovarian hyperstimulation (COH) and the IVF success. In patients with autoimmune etiology for POR/POI, the combination of glucocorticosteroids, pituitary-ovarian suppression, and COH may be successful in achieving the desired conception.
Collapse
|
26
|
Izadi A, Shirazi S, Taghizadeh S, Gargari BP. Independent and Additive Effects of Coenzyme Q10 and Vitamin E on Cardiometabolic Outcomes and Visceral Adiposity in Women With Polycystic Ovary Syndrome. Arch Med Res 2019; 50:1-10. [DOI: 10.1016/j.arcmed.2019.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/29/2019] [Indexed: 02/05/2023]
|
27
|
Lans C, Taylor-Swanson L, Westfall R. Herbal fertility treatments used in North America from colonial times to 1900, and their potential for improving the success rate of assisted reproductive technology. REPRODUCTIVE BIOMEDICINE & SOCIETY ONLINE 2018; 5:60-81. [PMID: 30023440 PMCID: PMC6047296 DOI: 10.1016/j.rbms.2018.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 09/19/2016] [Accepted: 03/12/2018] [Indexed: 06/08/2023]
Abstract
This paper serves to fill a gap in the literature regarding evidence for the use of botanical remedies in the promotion of fertility. It examines the botanical remedies that were used in North America (1492-1900) for all stages of reproduction from preconception to birth, and discusses their potential for future use with present-day infertility treatments. Each medicinal plant discussed in this paper is assessed using an ethnomedicinal methodology that entails examining the published ethnobotanical, phytochemical and pharmacological data. A few clinical trials have shown that there is potential for medicinal plants to improve the success rate of assisted reproductive technology (ART) treatment if used in an integrated manner, similar to the integrated use of traditional Chinese medicine with ART treatment. For example, research has shown that older women who become pregnant have a high miscarriage rate, and this is one area that complementary and alternative medicines can address.
Collapse
Affiliation(s)
- Cheryl Lans
- Institute for Ethnobotany and Zoopharmacognosy, Beek, The Netherlands
| | - Lisa Taylor-Swanson
- College of Nursing University of Utah, Annette Cummings Building, 10 South 2000 East Salt Lake City, UT, USA
| | - Rachel Westfall
- Yukon Bureau of Statistics, Government of Yukon, Yukon, Canada
| |
Collapse
|
28
|
Xu Y, Nisenblat V, Lu C, Li R, Qiao J, Zhen X, Wang S. Pretreatment with coenzyme Q10 improves ovarian response and embryo quality in low-prognosis young women with decreased ovarian reserve: a randomized controlled trial. Reprod Biol Endocrinol 2018; 16:29. [PMID: 29587861 PMCID: PMC5870379 DOI: 10.1186/s12958-018-0343-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/06/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Management of women with reduced ovarian reserve or poor ovarian response (POR) to stimulation is one of the major challenges in reproductive medicine. The primary causes of POR remain elusive and oxidative stress was proposed as one of the important contributors. It has been suggested that focus on the specific subpopulations within heterogeneous group of poor responders could assist in evaluating optimal management strategies for these patients. This study investigated the effect of anti-oxidant treatment with coenzyme Q10 (CoQ10) on ovarian response and embryo quality in young low-prognosis patients with POR. METHODS This prospective, randomized controlled study included 186 consecutive patients with POR stratified according to the POSEIDON classification group 3 (age < 35, poor ovarian reserve parameters). The participants were randomized to the CoQ10 pre-treatment for 60 days preceding IVF-ICSI cycle or no pre-treatment. The number of high quality embryos was a primary outcome measure. RESULTS A total of 169 participants were evaluated (76 treated with CoQ10 and 93 controls); 17 women were excluded due to low compliance with CoQ10 administration. The baseline demographic and clinical characteristics were comparable between the groups. CoQ10 pretreatment resulted in significantly lower gonadotrophin requirements and higher peak E2 levels. Women in CoQ10 group had increased number of retrieved oocytes (4, IQR 2-5), higher fertilization rate (67.49%) and more high-quality embryos (1, IQR 0-2); p < 0.05. Significantly less women treated with CoQ10 had cancelled embryo transfer because of poor embryo development than controls (8.33% vs. 22.89%, p = 0.04) and more women from treatment group had available cryopreserved embryos (18.42% vs. 4.3%, p = 0.012). The clinical pregnancy and live birth rates per embryo transfer and per one complete stimulation cycle tended to be higher in CoQ10 group but did not achieve statistical significance. CONCLUSION Pretreatment with CoQ10 improves ovarian response to stimulation and embryological parameters in young women with poor ovarian reserve in IVF-ICSI cycles. Further work is required to determine whether there is an effect on clinical treatment endpoints.
Collapse
Affiliation(s)
- Yangying Xu
- Department of Reproduction, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Victoria Nisenblat
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Cuiling Lu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Rong Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Jie Qiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Xiumei Zhen
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100123, China.
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China.
| | - Shuyu Wang
- Department of Reproduction, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China.
| |
Collapse
|
29
|
Gadalla MA, Huang S, Wang R, Norman RJ, Abdullah SA, El Saman AM, Ismail AM, van Wely M, Mol BWJ. Effect of clomiphene citrate on endometrial thickness, ovulation, pregnancy and live birth in anovulatory women: systematic review and meta-analysis. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2018; 51:64-76. [PMID: 29055102 DOI: 10.1002/uog.18933] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/08/2017] [Accepted: 10/11/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES To compare the impact of clomiphene citrate (CC) vs other drug regimens on mid-cycle endometrial thickness (EMT), ovulation, pregnancy and live birth rates in women with World Health Organization (WHO) group II ovulatory disorders. METHODS We searched MEDLINE, EMBASE, Scopus, Web of Science, The Cochrane Central Register of Clinical Trials (CENTRAL) and the non-MEDLINE subset of PubMed from inception to December 2016 and cross-checked references of relevant articles. We included only randomized controlled trials (RCTs) comparing CC used alone vs other drug regimens for ovulation induction in women with WHO group II anovulation. Outcomes were mid-cycle EMT, ovulation, pregnancy and live birth rates. We pooled weighted mean differences (WMD) with 95% confidence intervals (CI) for continuous variables (EMT) and risk ratios (RR) with 95% CI for binary variables (ovulation, pregnancy and live birth rates). RESULTS We retrieved 1718 articles of which 33 RCTs (4349 women, 7210 ovulation induction cycles) were included. In 15 RCTs that compared CC with letrozole, EMT was lower in the CC group (1957 women, 3892 cycles; WMD, -1.39; 95% CI, -2.27 to -0.51; I2 = 100%), ovulation rates after CC and letrozole were comparable (1710 women, 3217 cycles; RR, 0.97; 95% CI, 0.90-1.04; I2 = 47%), while CC led to a lower pregnancy rate (1957 women, 3892 cycles; RR, 0.78; 95% CI, 0.63-0.95; I2 = 43%) and a lower live birth rate (RR, 0.70; 95% CI, 0.49-0.98; I2 = 35%). In two RCTs that compared CC with CC plus metformin, EMT, ovulation and pregnancy rates were comparable (101 women, 140 cycles; WMD, -0.23; 95% CI, -0.92 to 0.45; I2 = 78%; RR, 0.84; 95% CI, 0.67-1.06; I2 = 0%; and RR, 0.79; 95% CI, 0.33-1.87; I2 = 0%). In three studies that compared CC with CC plus N-acetyl cysteine (NAC), EMT was lower in the CC group (340 women, 300 cycles; WMD, -1.51; 95% CI, -1.98 to -1.04; I2 = 45%). In two studies that compared CC with CC + nitric oxide (NO) donor, EMT was lower in the CC group (120 women, 304 cycles; WMD, -1.75; 95% CI, -2.08 to -1.41; I2 = 0%). Compared with CC plus NO donor or NAC, CC showed statistically significant lower ovulation and pregnancy rates. Compared with tamoxifen in three studies, CC showed a tendency towards lower EMT (571 women, 844 cycles; WMD, -1.34; 95% CI, -2.70 to 0.01; I2 = 96%) with comparable ovulation and pregnancy rates. CONCLUSIONS In women with WHO group II ovulatory disorders, ovulation induction with CC might result in lower EMT than other ovulation induction regimens. Whether the lower EMT caused the lower pregnancy and live birth rates remains to be elucidated. Letrozole seems to be beneficial for these women. However, our findings should be interpreted with caution as the quality of evidence was very low. Copyright © 2017 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- M A Gadalla
- Women's Health Hospital, Department of Obstetrics and Gynecology, Assiut University, Assiut, Egypt
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - S Huang
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Reproductive Medicine Centre, Peking University Third Hospital, Beijing, China
| | - R Wang
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - R J Norman
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - S A Abdullah
- Women's Health Hospital, Department of Obstetrics and Gynecology, Assiut University, Assiut, Egypt
| | - A M El Saman
- Women's Health Hospital, Department of Obstetrics and Gynecology, Assiut University, Assiut, Egypt
| | - A M Ismail
- Women's Health Hospital, Department of Obstetrics and Gynecology, Assiut University, Assiut, Egypt
| | - M van Wely
- Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - B W J Mol
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
30
|
Abstract
BACKGROUND A couple may be considered to have fertility problems if they have been trying to conceive for over a year with no success. This may affect up to a quarter of all couples planning a child. It is estimated that for 40% to 50% of couples, subfertility may result from factors affecting women. Antioxidants are thought to reduce the oxidative stress brought on by these conditions. Currently, limited evidence suggests that antioxidants improve fertility, and trials have explored this area with varied results. This review assesses the evidence for the effectiveness of different antioxidants in female subfertility. OBJECTIVES To determine whether supplementary oral antioxidants compared with placebo, no treatment/standard treatment or another antioxidant improve fertility outcomes for subfertile women. SEARCH METHODS We searched the following databases (from their inception to September 2016) with no language or date restriction: Cochrane Gynaecology and Fertility Group (CGFG) specialised register, the Cochrane Central Register of Studies (CENTRAL CRSO), MEDLINE, Embase, PsycINFO, CINAHL and AMED. We checked reference lists of appropriate studies and searched for ongoing trials in the clinical trials registers. SELECTION CRITERIA We included randomised controlled trials (RCTs) that compared any type, dose or combination of oral antioxidant supplement with placebo, no treatment or treatment with another antioxidant, among women attending a reproductive clinic. We excluded trials comparing antioxidants with fertility drugs alone and trials that only included fertile women attending a fertility clinic because of male partner infertility. DATA COLLECTION AND ANALYSIS Two review authors independently selected eligible studies, extracted the data and assessed the risk of bias of the included studies. The primary review outcome was live birth; secondary outcomes included clinical pregnancy rates and adverse events. We pooled studies using a fixed-effect model, and calculated odds ratios (ORs) with 95% confidence intervals (CIs) for the dichotomous outcomes of live birth, clinical pregnancy and adverse events. We assessed the overall quality of the evidence by applying GRADE criteria. MAIN RESULTS We included 50 trials involving 6510 women. Investigators compared oral antioxidants, including combinations of antioxidants, N-acetyl-cysteine, melatonin, L-arginine, myo-inositol, D-chiro-inositol, carnitine, selenium, vitamin E, vitamin B complex, vitamin C, vitamin D+calcium, CoQ10, pentoxifylline and omega-3-polyunsaturated fatty acids versus placebo, no treatment/standard treatment or another antioxidant.Very low-quality evidence suggests that antioxidants may be associated with an increased live birth rate compared with placebo or no treatment/standard treatment (OR 2.13, 95% CI 1.45 to 3.12, P > 0.001, 8 RCTs, 651 women, I2 = 47%). This suggests that among subfertile women with an expected live birth rate of 20%, the rate among women using antioxidants would be between 26% and 43%.Very low-quality evidence suggests that antioxidants may be associated with an increased clinical pregnancy rate compared with placebo or no treatment/standard treatment (OR 1.52, 95% CI 1.31 to 1.76, P < 0.001, 26 RCTs, 4271 women, I2 = 66%). This suggests that among subfertile women with an expected clinical pregnancy rate of 22%, the rate among women using antioxidants would be between 27% and 33%. Heterogeneity was moderately high.There was insufficient evidence to determine whether there was a difference between the groups in rates of miscarriage (OR 0.79, 95% CI 0.58 to 1.08, P = 0.14, 18 RCTs, 2834 women, I2 = 23%, very low quality evidence). This suggests that, among subfertile women with an expected miscarriage rate of 7%, use of antioxidants would be expected to result in a miscarriage rate of between 4% and 7%. There was also insufficient evidence to determine whether there was a difference between the groups in rates of multiple pregnancy (OR 1.00, 95% CI 0.73 to 1.38, P = 0.98, 8 RCTs, 2163 women, I2 = 4%, very low quality evidence). This suggests that among subfertile women with an expected multiple pregnancy rate of 8%, use of antioxidants would be expected to result in a multiple pregnancy rate between 6% and 11%. Likewise, there was insufficient evidence to determine whether there was a difference between the groups in rates of gastrointestinal disturbances (OR 1.55, 95% CI 0.47 to 5.10, P = 0.47, 3 RCTs, 343 women, I2 = 0%, very low quality evidence). This suggests that among subfertile women with an expected gastrointestinal disturbance rate of 2%, use of antioxidants would be expected to result in a rate between 1% and 11%. Overall adverse events were reported by 35 trials in the meta-analysis, but there was insufficient evidence to draw any conclusions.Only one trial reported on live birth, clinical pregnancy or adverse effects in the antioxidant versus antioxidant comparison, and no conclusions could be drawn.Very low-quality evidence suggests that pentoxifylline may be associated with an increased clinical pregnancy rate compared with placebo or no treatment (OR 2.07, 95% CI 1.20 to 3.56, P = 0.009, 3 RCTs, 276 women, I2 = 0%). This suggests that among subfertile women with an expected clinical pregnancy rate of 25%, the rate among women using pentoxifylline would be between 28% and 53%.There was insufficient evidence to determine whether there was a difference between the groups in rates of miscarriage (OR 1.34, 95% CI 0.46 to 3.90, P = 0.58, 3 RCTs, 276 women, I2 = 0%) or multiple pregnancy (OR 0.78, 95% CI 0.20 to 3.09, one RCT, 112 women, very low quality evidence). This suggests that among subfertile women with an expected miscarriage rate of 4%, the rate among women using pentoxifylline would be between 2% and 15%. For multiple pregnancy, the data suggest that among subfertile women with an expected multiple pregnancy rate of 9%, the rate among women using pentoxifylline would be between 2% and 23%.The overall quality of evidence was limited by serious risk of bias associated with poor reporting of methods, imprecision and inconsistency. AUTHORS' CONCLUSIONS In this review, there was very low-quality evidence to show that taking an antioxidant may provide benefit for subfertile women, but insufficient evidence to draw any conclusions about adverse events. At this time, there is limited evidence in support of supplemental oral antioxidants for subfertile women.
Collapse
Affiliation(s)
- Marian G Showell
- University of AucklandDepartment of Obstetrics and GynaecologyPark Road GraftonAucklandNew Zealand1142
| | | | - Vanessa Jordan
- University of AucklandDepartment of Obstetrics and GynaecologyPark Road GraftonAucklandNew Zealand1142
| | - Roger J Hart
- The University of Western Australia, King Edward Memorial Hospital and Fertility Specialists of Western AustraliaSchool of Women's and Infants' Health374 Bagot RoadSubiaco, PerthAustralia6008
| |
Collapse
|
31
|
Obesity and female infertility: potential mediators of obesity's impact. Fertil Steril 2017; 107:840-847. [DOI: 10.1016/j.fertnstert.2017.01.017] [Citation(s) in RCA: 306] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 01/01/2023]
|
32
|
The association between coenzyme Q10 concentrations in follicular fluid with embryo morphokinetics and pregnancy rate in assisted reproductive techniques. J Assist Reprod Genet 2017; 34:599-605. [PMID: 28185121 DOI: 10.1007/s10815-017-0882-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 01/24/2017] [Indexed: 10/20/2022] Open
Abstract
PURPOSE This study seeks to evaluate the association between follicular fluid (FF) coenzyme Q10 (CoQ10) levels, embryo morphokinetics, and pregnancy rate. METHODS Sixty infertile patients who underwent intracytoplasmic sperm injection (ICSI) cycles were included in the study. For each patient, CoQ10 level of the follicular fluid was measured by high-performance liquid chromatography system. After the ICSI of each oocyte, the relationship between the level of CoQ10 content of each follicular fluid, the subsequent embryo quality, and embryo morphokinetics was investigated. The relationship between the level of CoQ10 content of each follicle and optimal time-lapse parameters for the embryos of these follicles including t5, s2, and cc2 was also analyzed. The embryos were further classified into four categories, namely, grades A, B, C, and D, according to morphokinetic parameters using t5-t2 and t5-t3 (cc3). Each follicular fluid analysis was performed for a single oocyte of a single embryo which was transferred to the patients. Additionally, follicular fluid CoQ10 levels and pregnancy rates were evaluated. RESULTS Follicular fluid CoQ10 levels were significantly higher in grades A and B than grades C and D embryos (p < 0.05). The concentration of CoQ10 levels was significantly higher in the pregnant group (p < 0.05). There was no significant correlation between optimal t5 and s2 morphokinetic parameters and CoQ10 levels. However, CoQ10 levels were significantly higher in follicular fluid of embryos which had optimal cc2 (p < 0.05). CONCLUSION High follicular fluid CoQ10 level is associated with optimal embryo morphokinetic parameters and higher pregnancy rates.
Collapse
|
33
|
Gat I, Blanco Mejia S, Balakier H, Librach CL, Claessens A, Ryan EAJ. The use of coenzyme Q10 and DHEA during IUI and IVF cycles in patients with decreased ovarian reserve. Gynecol Endocrinol 2016; 32:534-7. [PMID: 26829445 DOI: 10.3109/09513590.2015.1137095] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The objective of this study is to compare the combination of dehydroepiandrosterone (DHEA) and coenzyme Q10 (CoQ10) (D + C) with DHEA alone (D) in intrauterine insemination (IUI) and in vitro fertilization (IVF) cycles among patients with decreased ovarian reserve. METHODS We retrospectively extracted data from patients charts treated by DHEA with/without CoQ10 during IUI or IVF between February 2006 and June 2014. Prestimulation parameters included age, BMI, day 3 FSH and antral follicular count (AFC). Ovarian response parameters included total gonadotropins dosage, peak serum estradiol, number of follicles > 16 mm and fertilization rate. Clinical outcomes included clinical and ongoing pregnancy rates. RESULTS Three hundred and thirty IUI cycles involved D + C compared with 467 cycles of D; 78 IVF cycles involved D + C and 175 D. In both IUI and IVF, AFC was higher with D + C compared with D (7.4 ± 5.7 versus 5.9 ± 4.7, 8.2 ± 6.3 versus 5.2 ± 5, respectively, p < 0.05). D + C resulted in a more follicles > 16 mm during IUI cycles (3.3 ± 2.3 versus 2.9 ± 2.2, respectively, p = 0.01), while lower mean total gonadotropin dosage was administered after D + C supplementation compared with D (3414 ± 1141 IUs versus 3877 ± 1143 IUs respectively, p = 0.032) in IVF cycles. Pregnancy and delivery rates were similar for both IUI and IVF. CONCLUSION D + C significantly increases AFC and improves ovarian responsiveness during IUI and IVF without a difference in clinical outcome.
Collapse
Affiliation(s)
- Itai Gat
- a CReATe Fertility Centre , Toronto , Canada
- b Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center , Tel HaShomer , Ramat Gan , Israel
| | - Sonia Blanco Mejia
- c Toronto West Fertility Center , Etobicoke , Canada
- d Risk Factor Modification Center, St. Michael's Hospital , Toronto , Canada
| | | | - Clifford L Librach
- a CReATe Fertility Centre , Toronto , Canada
- e Department of Obstetrics & Gynecology , University of Toronto , Canada
- f Department of Physiology , University of Toronto , Canada , and
- g Department of Gynecology , Women's College Hospital , Toronto , Canada
| | - Anne Claessens
- c Toronto West Fertility Center , Etobicoke , Canada
- e Department of Obstetrics & Gynecology , University of Toronto , Canada
| | - Edward A J Ryan
- c Toronto West Fertility Center , Etobicoke , Canada
- e Department of Obstetrics & Gynecology , University of Toronto , Canada
| |
Collapse
|
34
|
Özcan P, Fıçıcıoğlu C, Kizilkale O, Yesiladali M, Tok OE, Ozkan F, Esrefoglu M. Can Coenzyme Q10 supplementation protect the ovarian reserve against oxidative damage? J Assist Reprod Genet 2016; 33:1223-30. [PMID: 27255570 DOI: 10.1007/s10815-016-0751-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/25/2016] [Indexed: 01/11/2023] Open
Abstract
PURPOSE We investigated antioxidant effects of CoQ10 supplementation on the prevention of OS-induced ovarian damage and to evaluate the protective effect of such supplementation against OS-related DNA damage. METHODS Twenty-four adult female Sprague-Dawley rats were randomly divided into three groups (8 rats per group): group 1 (control): saline, ip, and orally; group 2 (cisplatin group): cisplatin, 4.5 mg/kg ip, two times with an interval of 7 days; and group 3 (cisplatin + CoQ10 group): cisplatin, 4.5 mg/kg ip, two times with an interval of 7 days, and 24 h before cisplatin, 150 mg/kg/day orally in 1 mL of saline daily for 14 days. Serum concentrations of anti-Mullerian hormone (AMH), number of AMH-positive follicles, the assessment of the intensity of 8'OHdG immunoreactivity, the primordial, antral and atretic follicle counts in the ovary were assessed. RESULT(S) The mean serum AMH concentrations were 1.3 ± 0.19, 0.16 ± 0.03, and 0.27 ± 0.20 ng/mL in groups 1, 2, and 3, respectively (p < 0.01). Serum AMH levels were significantly higher in group 1 compared to groups 2 and 3 (p < 0.01 and p = 0.01, respectively). There was a statistically significant difference in AMH-positive follicle count between the groups (p < 0.01). Group 1 showed higher numbers of AMH-positive granulosa cells compared to group 2 (p = 0.01). A significant difference was found in the primordial, the atretic, and antral follicle counts between the three groups (p < 0.01, p < 0.01, and p < 0.01, respectively). The atretic follicle count was significantly lower in the cisplatin plus CoQ10 group compared to the cisplatin group (p < 0.01). The antral follicle counts were significantly higher in the cisplatin plus CoQ10 group compared with the cisplatin group (p < 0.01). There was a statistically significant difference in the intensity of staining of the follicles that were positive for anti-8'OHdG between the groups (p = 0.02). Group 1 showed a significant lower intensity of staining of the follicles positive for anti-8'OHdG compared with group 2 (p = 0.03). CONCLUSION(S) CoQ10 supplementation may protect ovarian reserve by counteracting both mitochondrial ovarian ageing and physiological programmed ovarian ageing although the certain effect of OS in female infertility is not clearly known.
Collapse
Affiliation(s)
- Pınar Özcan
- Department of Obstetrics and Gynecology, Bezmialem University Faculty of Medicine, İstanbul, Turkey. .,Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Cem Fıçıcıoğlu
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Ozge Kizilkale
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mert Yesiladali
- Department of Obstetrics and Gynecology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Olgu Enis Tok
- Department of Histology and Embryology, Bezmialem University Faculty of Medicine, İstanbul, Turkey
| | - Ferda Ozkan
- Department of Pathology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mukaddes Esrefoglu
- Department of Histology and Embryology, Bezmialem University Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
35
|
Mendez Lozano DH, Lenero MV, Gonzalez RL, Scheffer JB, Gonzalez MT, Barron Y, Frydman R. Tadalafil for Endometrial Growth in Clomiphene Citrate stimulated cycles in an IUI programma: A pilot study. Facts Views Vis Obgyn 2015; 7:231-237. [PMID: 27729968 PMCID: PMC5058412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
AIM OF THE STUDY The objective of this study was to assess the impact of tadalafil on endometrial growth, the uterine artery pulsatility index (PI) and the uterine artery resistance index (RI) in patients under clomiphene ovarian stimulation for intrauterine insemination (IUI). METHODS This randomized crossover study included 30 patients with a normal endometrium over 53 cycles, and 46 of those cycles in 23 patients were included in the analysis. In group A the patients were under 100 mg clomiphene daily for five days (2-6) and 5 mg tadalafil daily for 7 days (4-10). For Group B (control) the patients only received clomiphene. Measurements of the endometrium, PI, RI and estradiol determinations were taken on cycle days 4, 8 and 10. RESULTS We observed a better endometrial growth in Group A compared to Group B: 7.5 ± 2.1 mm vs 5.5 ± 1.2 mm, P < 0.0002 and 8.9 ± 1.8 mm vs 6.3 ± 1.8 mm, P < 0.0002 on days 8 and 10, respectively. Additionally, a progressive decrease in the RI was observed in Group A but not in Group B from day 8 (0.77 ± 0.15 vs 0.85 ± 0.18, P = 0.059) to day 10 (0.74 ± 0.20 vs 0.87 ± 0.14, P < 0.017). However, no differences were observed in PI or serum estradiol between Group A and Group B. CONCLUSION The use of tadalafil improved endometrial growth in patients under clomiphene ovarian stimulation with no significant effect on the uterine artery Pulsatility Index and serum estradiol.
Collapse
Affiliation(s)
- DH Mendez Lozano
- School of Medicine, Tecnológico de Monterrey, Ave. Morones Prieto No. 3000 Pte, Col. Los Doctores, 64710, Monterrey, Nuevo León, México.,Center for Reproductive Medicine CREASIS San Pedro, Monterrey, México
| | - MV Lenero
- School of Medicine, Tecnológico de Monterrey, Ave. Morones Prieto No. 3000 Pte, Col. Los Doctores, 64710, Monterrey, Nuevo León, México
| | - RL Gonzalez
- School of Medicine, Tecnológico de Monterrey, Ave. Morones Prieto No. 3000 Pte, Col. Los Doctores, 64710, Monterrey, Nuevo León, México.,Center for Reproductive Medicine CREASIS San Pedro, Monterrey, México
| | - JB Scheffer
- IBRRA – Instituto Brasileiro de Reprodução Assistida, Belo Horizonte, MG, Brazil
| | | | - Y Barron
- School of Medicine, Tecnológico de Monterrey, Ave. Morones Prieto No. 3000 Pte, Col. Los Doctores, 64710, Monterrey, Nuevo León, México
| | - R Frydman
- Departement d’Obstetrics et Gynecology, Hôpital Foch de Suresnes, 40 rue Worth, Suresnes, France
| |
Collapse
|
36
|
Rocca ML, Venturella R, Mocciaro R, Di Cello A, Sacchinelli A, Russo V, Trapasso S, Zullo F, Morelli M. Polycystic ovary syndrome: chemical pharmacotherapy. Expert Opin Pharmacother 2015; 16:1369-93. [PMID: 26001184 DOI: 10.1517/14656566.2015.1047344] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is the most common reproductive endocrine disease among women of childbearing age. The clinical features are heterogeneous and vary in intensity. Hirsutism, menstrual disorders and infertility are the most frequent conditions observed; however, long-term complications (dyslipidemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, endometrial cancer) are also often described. Each disorder may be managed by tailored strategies, employing sequential or combined pharmacological and/or non-pharmacological treatment. AREAS COVERED The authors review the drugs used for PCOS management and discuss new approaches. A systematic MEDLINE search regarding the randomized controlled trials, retrospective and observational studies about medical treatments of PCOS, the Cochrane library for reviews and also search for registered trials on ClinicalTrials.gov is performed. EXPERT OPINION A uniform treatment for PCOS patients does not exist. Clinicians should perform an accurate evaluation of patients' characteristics, identifying the phenotypic target and, subsequently, the best-tailored treatment to manage one or more clinical issues. Lifestyle intervention should always be the first recommended approach unless other issues indicate that drug or hormonal interventions are superior.
Collapse
Affiliation(s)
- Morena Luigia Rocca
- 'Magna Graecia' University, Cancer Center of Excellence "Tommaso Campanella" of Germaneto, Department of Experimental and Clinical Medicine, Unit of Obstetrics and Gynaecology, Oncology Unit , Viale Europa, loc. Germaneto, 88100, Catanzaro , Italy +39 328 5692428 ; +39 0961 883234 ;
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Talukdar A, Sharma KA, Rai R, Deka D, Rao DN. Effect of Coenzyme Q10 on Th1/Th2 Paradigm in Females with Idiopathic Recurrent Pregnancy Loss. Am J Reprod Immunol 2015; 74:169-80. [PMID: 25800618 DOI: 10.1111/aji.12376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/19/2015] [Indexed: 01/12/2023] Open
Abstract
PROBLEM Recurrent pregnancy loss is characterized by predominant Th1-type immunity and increased reactive oxygen species. Low levels of Coenzyme Q10 are found in the plasma of RPL as compared to healthy pregnant females. Our aim was to investigate whether in vitro supplementation of PBMCs from such females with CoQ10 could change the observed Th1 bias. METHOD OF STUDY PBMCs were isolated from 20 RPL pregnant and non-pregnant females and 16 healthy pregnant females and incubated with CoQ10 in in vitro conditions. Phenotyping of Th1, Th2, and Th17 cells was performed by flow cytometry. Cytokine levels were determined by ELISA. RESULTS PBMCs treated with CoQ10 showed significantly decreased percentage of Th1 cells (P < 0.005) in pregnant females with history of RPL than in the untreated ones. Also, levels of IFN-γ and TNF-α were significantly decreased in the culture supernatant of treated PBMCs from RPL. DCFDA staining showed significantly reduced production of ROS in the treated PBMCs in RPL females. CONCLUSION CoQ10 was effective in maintaining the immune homeostasis by reducing the proportion of IFN-γ-producing T cells and proinflammatory cytokine levels in the RPL pregnant females. This property could be attributed to the capability of CoQ10 in reducing oxidative stress by decreasing ROS production.
Collapse
Affiliation(s)
- Ayantika Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Kandala Aparna Sharma
- Department of Obstetrics & Gynecology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Reeta Rai
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Dipika Deka
- Department of Obstetrics & Gynecology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | |
Collapse
|