1
|
Palomba S, Costanzi F, Caserta D, Vitagliano A. Pharmacological and non-pharmacological interventions for improving endometrial receptivity in infertile patients with polycystic ovary syndrome: a comprehensive review of the available evidence. Reprod Biomed Online 2024; 49:104381. [PMID: 39454320 DOI: 10.1016/j.rbmo.2024.104381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 10/28/2024]
Abstract
Direct and indirect evidence suggests that endometrial receptivity may play a crucial role in the reduced fertility rate of women with polycystic ovary syndrome (PCOS). Various pharmacological and non-pharmacological strategies with potential effects on endometrial receptivity in patients with PCOS have been proposed. The aim of this study was to summarize the rationale and the clinical and experimental evidence of interventions tested for improving endometrial receptivity in infertile patients with PCOS. A systematic review was conducted by consulting electronic databases. All interventions with a potential influence on endometrial receptivity in infertile patients with PCOS were evaluated, and their main biological mechanisms were analysed. In total, 24 interventions related to endometrial receptivity were identified. Notwithstanding a strong biological rationale, no intervention aimed at improving endometrial receptivity in women with PCOS is supported by an adequate body of evidence, limiting their use in clinical practice. Further high-quality research is needed in this field to limit potentially ineffective and unsafe add-on treatments in infertile patients with PCOS.
Collapse
Affiliation(s)
- Stefano Palomba
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Flavia Costanzi
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy; University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Donatella Caserta
- Unit of Gynaecology, Department of Medical-Surgical Sciences and Translational Medicine, University 'Sapienza' of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Amerigo Vitagliano
- Unit of Obstetrics and Gynaecology, Department of Interdisciplinary Medicine, University of Bari, Bari, Italy
| |
Collapse
|
2
|
Li L, Zhou X, Liu W, Chen Z, Xiao X, Deng G. Supplementation with NAD+ and its precursors: A rescue of female reproductive diseases. Biochem Biophys Rep 2024; 38:101715. [PMID: 38698835 PMCID: PMC11063225 DOI: 10.1016/j.bbrep.2024.101715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential coenzyme involved in many pathophysiological processes. Supplementation with NAD+ and its precursors has been demonstrated as an emerging therapeutic strategy for the diseases. NAD+ also plays an important role in the reproductive system. Here, we summarize the function of NAD+ in various reproductive diseases and review the application of NAD+ and its precursors in the preservation of reproductive capacity and the prevention of embryonic malformations. It is shown that NAD+ shows good promise as a therapeutic approach for saving reproductive capacity.
Collapse
Affiliation(s)
- Lan Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xin Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Wene Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Zhen Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Xiaoqin Xiao
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guiming Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| |
Collapse
|
3
|
Siemers KM, Klein AK, Baack ML. Mitochondrial Dysfunction in PCOS: Insights into Reproductive Organ Pathophysiology. Int J Mol Sci 2023; 24:13123. [PMID: 37685928 PMCID: PMC10488260 DOI: 10.3390/ijms241713123] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/15/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex, but relatively common endocrine disorder associated with chronic anovulation, hyperandrogenism, and micro-polycystic ovaries. In addition to reduced fertility, people with PCOS have a higher risk of obesity, insulin resistance, and metabolic disease, all comorbidities that are associated with mitochondrial dysfunction. This review summarizes human and animal data that report mitochondrial dysfunction and metabolic dysregulation in PCOS to better understand how mitochondria impact reproductive organ pathophysiology. This in-depth review considers all the elements regulating mitochondrial quantity and quality, from mitochondrial biogenesis under the transcriptional regulation of both the nuclear and mitochondrial genome to the ultrastructural and functional complexes that regulate cellular metabolism and reactive oxygen species production, as well as the dynamics that regulate subcellular interactions that are key to mitochondrial quality control. When any of these mitochondrial functions are disrupted, the energetic equilibrium within the cell changes, cell processes can fail, and cell death can occur. If this process is ongoing, it affects tissue and organ function, causing disease. The objective of this review is to consolidate and classify a broad number of PCOS studies to understand how various mitochondrial processes impact reproductive organs, including the ovary (oocytes and granulosa cells), uterus, placenta, and circulation, causing reproductive pathophysiology. A secondary objective is to uncover the potential role of mitochondria in the transgenerational transmission of PCOS and metabolic disorders.
Collapse
Affiliation(s)
- Kyle M. Siemers
- Physician Scientist (MD/Ph.D.) Program, Sanford School of Medicine, University of South Dakota, 414 E. Clark Street, Vermillion, SD 57069, USA;
| | - Abigail K. Klein
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Lee Medical Building, 414 E. Clark St., Sioux Falls, SD 57069, USA;
| | - Michelle L. Baack
- Department of Pediatrics, Division of Neonatology, Sanford School of Medicine, University of South Dakota, 1400 W. 22nd St., Sioux Falls, SD 57105, USA
- Environmental Influences on Health and Disease Group, Sanford Research, 2301 E. 60th St., Sioux Falls, SD 57104, USA
| |
Collapse
|
4
|
Huo P, Li M, Le J, Zhu C, Yao J, Zhang S. Resveratrol improves follicular development of PCOS rats via regulating glycolysis pathway and targeting SIRT1. Syst Biol Reprod Med 2022; 69:153-165. [PMID: 36268996 DOI: 10.1080/19396368.2022.2125855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a disease characterized by metabolic disorders. This study aimed to examine the effects of resveratrol treatment on ovulation in the PCOS rat model. Quantitative real-time PCR and immunohistochemistry were used to determine the mRNA and protein expression levels. TNUEL assay was used to evaluate cell apoptosis in ovary. The metabolites were evaluated by liquid chromatography with tandem mass spectrometry. Resveratrol alleviated disrupted estrous cycle and improved granular cell layers, and reversed the decreased proliferation and increased cell apoptosis of granulosa cells in the ovarian tissues of PCOS rats. Resveratrol restored the changes in the mRNA expression levels in the rate-limiting genes of glycolysis in the PCOS ovary. The expression of lactate dehydrogenase A (LDH-A), pyruvate kinase isozyme M2 (PKM2), and sirtuin 1 (SIRT1) was significantly downregulated in ovarian tissues of the PCOS rats; while the resveratrol treatment significantly increased the expression of LDH-A, PKM2, and SIRT1 in the ovarian tissues of PCOS rats. Collectively, the protective effects of resveratrol in the PCOS rats may be associated with the regulation of glycolysis-related mediators including PKM2, LDH-A, and SIRT1. Resveratrol may represent a good candidate in alleviating the development of PCOS.
Collapse
Affiliation(s)
- Peng Huo
- Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin, China
| | - Man Li
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jianghua Le
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chunjiang Zhu
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jun Yao
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
5
|
Enhanced glycolysis in granulosa cells promotes the activation of primordial follicles through mTOR signaling. Cell Death Dis 2022; 13:87. [PMID: 35087042 PMCID: PMC8795455 DOI: 10.1038/s41419-022-04541-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/09/2021] [Accepted: 01/11/2022] [Indexed: 11/20/2022]
Abstract
In mammals, nonrenewable primordial follicles are activated in an orderly manner to maintain the longevity of reproductive life. Mammalian target of rapamycin (mTOR)-KIT ligand (KITL) signaling in pre-granulosa cells and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-forkhead Box O3a (FOXO3a) signaling in oocytes are important for primordial follicle activation. The activation process is accompanied by the enhancement of energy metabolism, but the causal relationship is unclear. In the present study, the levels of glycolysis-related proteins GLUT4, HK1, PFKL, and PKM2 were significantly increased in granulosa cells but were decreased in oocytes during the mouse primordial-to-primary follicle transition. Both short-term pyruvate deprivation in vitro and acute fasting in vivo increased the glycolysis-related gene and protein levels, decreased AMPK activity, and increased mTOR activity in mouse ovaries. The downstream pathways Akt and FOXO3a were phosphorylated, resulting in mouse primordial follicle activation. The blockade of glycolysis by 2-deoxyglucose (2-DG), but not the blockade of the communication network between pre-granulosa cells and oocyte by KIT inhibitor ISCK03, decreased short-term pyruvate deprivation-promoted mTOR activity. Glycolysis was also increased in human granulosa cells during the primordial-to-primary follicle transition, and short-term pyruvate deprivation promoted the activation of human primordial follicles by increasing the glycolysis-related protein levels and mTOR activity in ovarian tissues. Taken together, the enhanced glycolysis in granulosa cells promotes the activation of primordial follicles through mTOR signaling. These findings provide new insight into the relationship between glycolytic disorders and POI/PCOS.
Collapse
|
6
|
Zhu Q, Yao Y, Xu L, Wu H, Wang W, He Y, Wang Y, Lu Y, Qi J, Ding Y, Li X, Huang J, Zhao H, Du Y, Sun K, Sun Y. Elevated SAA1 promotes the development of insulin resistance in ovarian granulosa cells in polycystic ovary syndrome. Reprod Biol Endocrinol 2022; 20:4. [PMID: 34980155 PMCID: PMC8721971 DOI: 10.1186/s12958-021-00873-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/06/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Insulin resistance (IR) contributes to ovarian dysfunctions in polycystic ovarian syndrome (PCOS) patients. Serum amyloid A1 (SAA1) is an acute phase protein produced primarily by the liver in response to inflammation. In addition to its role in inflammation, SAA1 may participate in IR development in peripheral tissues. Yet, expressional regulation of SAA1 in the ovary and its role in the pathogenesis of ovarian IR in PCOS remain elusive. METHODS Follicular fluid, granulosa cells and peripheral venous blood were collected from PCOS and non-PCOS patients with and without IR to measure SAA1 abundance for analysis of its correlation with IR status. The effects of SAA1 on its own expression and insulin signaling pathway were investigated in cultured primary granulosa cells. RESULTS Ovarian granulosa cells were capable of producing SAA1, which could be induced by SAA1 per se. Moreover, the abundance of SAA1 significantly increased in granulosa cells and follicular fluid in PCOS patients with IR. SAA1 treatment significantly attenuated insulin-stimulated membrane translocation of glucose transporter 4 and glucose uptake in granulosa cells through induction of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression with subsequent inhibition of Akt phosphorylation. These effects of SAA1 could be blocked by inhibitors for toll-like receptors 2/4 (TLR 2/4) and nuclear factor kappa light chain enhancer of activated B (NF-κB). CONCLUSIONS Human granulosa cells are capable of feedforward production of SAA1, which significantly increased in PCOS patients with IR. Excessive SAA1 reduces insulin sensitivity in granulosa cells via induction of PTEN and subsequent inhibition of Akt phosphorylation upon activation of TLR2/4 and NF-κB pathway. These findings highlight that elevation of SAA1 in the ovary promotes the development of IR in granulosa cells of PCOS patients.
Collapse
Affiliation(s)
- Qinling Zhu
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Yue Yao
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Lizhen Xu
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Hasiximuke Wu
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Yaqiong He
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Yuan Wang
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Yao Lu
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Jia Qi
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Ying Ding
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Xinyu Li
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Jiaan Huang
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Hanting Zhao
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Yanzhi Du
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China
| | - Kang Sun
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China.
| | - Yun Sun
- Center for Reproductive Medicine, Ren ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, People's Republic of China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, People's Republic of China.
| |
Collapse
|
7
|
Liang A, Huang L, Liu H, He W, Lei X, Li M, Li S, Liang H, Chen G, Tang J, Chen F, Cao X, Wang Y, Shen X, Chen X. Resveratrol Improves Follicular Development of PCOS Rats by Regulating the Glycolytic Pathway. Mol Nutr Food Res 2021; 65:e2100457. [PMID: 34664388 DOI: 10.1002/mnfr.202100457] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/05/2021] [Indexed: 01/23/2023]
Abstract
SCOPE Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder that can cause infertility; however, the underlying mechanisms remain ill-defined, and there are no available drugs or strategies for the treatment of PCOS. This study examined the therapeutic effect of resveratrol in a rat model of PCOS. METHODS AND RESULTS PCOS is induced in rats by administration of letrozole and a high fat diet to determine whether resveratrol has a protective effect. Oral administration of resveratrol significantly decreased body weight, as well as the serum levels of testosterone and follicle stimulating hormone. Resveratrol improved the estrous cycle by restoring the thickness and number of granular cells. Resveratrol increased the levels of lactate and ATP, decreased pyruvate levels, and restored the glycolytic process, upregulating LDHA, HK2, and PKM2. Resveratrol also upregulated SIRT2, thereby modulating the expression of rate-limiting enzymes of glycolysis. CONCLUSION Resveratrol suppressed damage to the ovaries in PCOS rats by restoring glycolytic activity, providing potential targets for the treatment of PCOS.
Collapse
Affiliation(s)
- Aihong Liang
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Lan'e Huang
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Huiqing Liu
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Weiguo He
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaocan Lei
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Meixiang Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Suyun Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hongxing Liang
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China
| | - Gang Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jinru Tang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Fengyu Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiting Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yiyao Wang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaobu Shen
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China
| | - Xi Chen
- Hunan Province Innovative Training Base for Medical Postgraduates, Hengyang Medical School, University of South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan, 416000, China.,Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
8
|
Palomba S. Is fertility reduced in ovulatory women with polycystic ovary syndrome? An opinion paper. Hum Reprod 2021; 36:2421-2428. [PMID: 34333641 DOI: 10.1093/humrep/deab181] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 06/22/2021] [Indexed: 01/13/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility; however, whether women with PCOS and spontaneous or drug-induced ovulatory cycles have the same reproductive potential as non-PCOS controls is a matter of debate. In the present opinion paper, the author takes the opportunity to summarize the collective evidence supporting the hypothesis of reduced fertility potential in women with PCOS, regardless of ovulatory status, and speculate that reduced reproductive potential may be caused by altered oocytes, embryo and endometrial competence, and infertility-related co-morbidities as well as an increased risk of pregnancy complications.
Collapse
Affiliation(s)
- Stefano Palomba
- Obstetrics and Gynecology, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
9
|
Masciangelo R, Chiti MC, Camboni A, Amorim CA, Donnez J, Dolmans MM. Mitochondrial content, activity, and morphology in prepubertal and adult human ovaries. J Assist Reprod Genet 2021; 38:2581-2590. [PMID: 34331619 DOI: 10.1007/s10815-021-02282-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/15/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To investigate whether mitochondrial content, activity, and morphology differ in prepubertal versus adult ovarian follicles. METHODS Ovarian tissue was collected from 7 prepubertal girls (age 1-10 years) and 6 adult women (age 20-35 years). Primordial and primary follicles were isolated from frozen-thawed prepubertal and adult ovarian tissue and their viability was assessed. Mitochondrial content was investigated by TOMM20 immunostaining of prepubertal and adult ovarian tissue, while mitochondrial activity in isolated follicles was analyzed by MitoTracker CM-H2XRos and JC-1. Frozen-thawed ovarian tissue from the same patients was also evaluated by transmission electron microscopy to examine mitochondrial morphology. RESULTS Higher TOMM20 staining was detected in prepubertal follicles compared to their adult counterparts, indicating the presence of more mitochondria in prepubertal follicles. Analysis of mitochondrial activity by MitoTracker showed higher fluorescence intensity in prepubertal follicles, suggesting that follicles in this group are more active than adult follicles. JC-1 analysis did not reveal any statistically significant difference in the inactive/active ratio between the two groups. Moreover, ultrastructural analysis by TEM detected morphological differences in the shape and cristae of prepubertal mitochondria, probably suggesting a mechanism of response to autophagy. CONCLUSION Differences in the number, activity, and morphology of mitochondria were reported, suggesting that consequential modifications might occur during puberty, which could be the window of opportunity required by mitochondria to undergo changes needed to reach maturity, and hence the capacity for ovulation and fertilization.
Collapse
Affiliation(s)
- Rossella Masciangelo
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200, Brussels, Belgium
| | - Maria Costanza Chiti
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200, Brussels, Belgium
| | - Alessandra Camboni
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200, Brussels, Belgium
| | - Christiani Andrade Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200, Brussels, Belgium
| | - Jacques Donnez
- Société de Recherche Pour L'Infertilité, Avenue Grandchamp 143, 1150, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte. B1.52.02, 1200, Brussels, Belgium.
- Département de Gynécologie, Cliniques Universitaires St. Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| |
Collapse
|
10
|
Effects of low-dose X-ray medical diagnostics on female gonads: Insights from large animal oocytes and human ovaries as complementary models. PLoS One 2021; 16:e0253536. [PMID: 34166427 PMCID: PMC8224917 DOI: 10.1371/journal.pone.0253536] [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: 01/05/2021] [Accepted: 06/08/2021] [Indexed: 11/19/2022] Open
Abstract
Diagnostic imaging has significantly grown over the last thirty years as indispensable support for diagnostic, prognostic, therapeutic and monitoring procedures of human diseases. This study explored the effects of low-dose X-ray medical diagnostics exposure on female fertility. To aim this, cumulus-oocyte complexes (COCs) recovered from the ovaries of juvenile sheep and human ovaries were used as complementary models for in vitro studies. In the sheep model, the effects of low-dose X-rays on oocyte viability and developmental competence were evaluated. In human ovaries originated from two age group (21–25 and 33–36 years old) subjects with gender dysphoria, X-rays effects on tissue morphology, follicular density and expression of apoptosis-related (NOXA, PUMA, Bcl2, Bak, γH2AX) and cell cycle-related genes (p21 and ki67) were investigated. It was noted that in sheep, the minimum dose of 10 mGy did not influence most of examined parameters at oocyte and embryo levels, whereas 50 and 100 mGy X-ray exposure reduced oocyte bioenergetic/oxidative activity but without any visible effects on oocyte and embryo development. In addition, blastocyst bioenergetic/oxidative status was reduced with all used doses. Overall data on human ovaries showed that low-dose X-rays, similarly as in sheep, did not alter any of examined parameters. However, in women belonging to the 33–36 year group, significantly reduced follicular density was observed after exposure to 50 and 100 mGy, and increased NOXA and Bax expression after exposure at 50 mGy. In conclusion, used low-doses of X-ray exposure, which resemble doses used in medical diagnostics, produce weak damaging effects on female fertility with increased susceptibility in advanced age.
Collapse
|
11
|
Wang F, Xie N, Wu Y, Zhang Q, Zhu Y, Dai M, Zhou J, Pan J, Tang M, Cheng Q, Shi B, Guo Q, Li X, Xie L, Wang B, Yang D, Weng Q, Guo L, Ye J, Pan M, Zhang S, Zhou H, Zhen C, Liu P, Ning K, Brackenridge L, Hardiman PJ, Qu F. Association between circadian rhythm disruption and polycystic ovary syndrome. Fertil Steril 2020; 115:771-781. [PMID: 33358334 DOI: 10.1016/j.fertnstert.2020.08.1425] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To explore the association of circadian rhythm disruption with polycystic ovary syndrome (PCOS) and the potential underlying mechanism in ovarian granulosa cells (GCs). DESIGN Multicenter questionnaire-based survey, in vivo and ex vivo studies. SETTING Twelve hospitals in China, animal research center, and research laboratory of a women's hospital. PATIENTS/ANIMALS A total of 436 PCOS case subjects and 715 control subjects were recruited for the survey. In vivo and ex vivo studies were conducted in PCOS-model rats and on ovarian GCs collected from women with PCOS and control subjects. INTERVENTION(S) The PCOS rat model was established with the use of testosterone propionate. MAIN OUTCOME MEASURE(S) Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), RNA sequencing, rhythmicity analysis, functional enrichment analysis. RESULT(S) There was a significant correlation between night shift work and PCOS. PCOS-model rats presented distinct differences in the circadian variation of corticotropin-releasing hormone, adrenocorticotropic hormone, prolactin, and a 4-h phase delay in thyrotropic hormone levels. The motif enrichment analysis of ATAC-seq revealed the absence of clock-related transcription factors in specific peaks of PCOS group, and RNA sequencing ex vivo at various time points over 24 hours demonstrated the differential rhythmic expression patterns of women with PCOS. Kyoto Encyclopedia of Genes and Genomes analysis further highlighted metabolic dysfunction, including both carbohydrate and amino acid metabolism and the tricarboxylic acid cycle. CONCLUSION(S) There is a significant association of night shift work with PCOS, and genome-wide chronodisruption exists in ovarian GCs.
Collapse
Affiliation(s)
- Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom
| | - Ningning Xie
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuhang Zhu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Minchen Dai
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jue Zhou
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China; Institute for Women's Health, University College London, London, United Kingdom
| | - Jiexue Pan
- First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Mengling Tang
- School of Public Health, Zhejiang University, Hangzhou, China
| | - Qi Cheng
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Biwei Shi
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qinyuan Guo
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Xinling Li
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Lifeng Xie
- Maternal and Child Healthcare Hospital of Liuzhou, Liuzhou, China
| | - Bing Wang
- Second Hospital of Jiaxing, Jiaxing, China
| | - Dongxia Yang
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qing Weng
- First People's Hospital of Yuhang District of Hangzhou, Hangzhou, China
| | - Lanzhong Guo
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Jisheng Ye
- Dongyang Women's and Children's Hospital, Dongyang, China
| | - Mingwo Pan
- Guangdong Women and Children Hospital, Guangzhou, China
| | - Shuyi Zhang
- Baiyin City Maternity and Childcare Hospital, Baiyin, China
| | - Hua Zhou
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cailan Zhen
- People's Hospital of Lucheng, Lucheng, China
| | - Ping Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of the Ministry of Education, West China Second Hospital, Chengdu, People's Republic of China
| | - Ke Ning
- Department of Social Science, Institute of Education, University College London, London, United Kingdom
| | - Lisa Brackenridge
- Institute for Women's Health, University College London, London, United Kingdom
| | - Paul J Hardiman
- Institute for Women's Health, University College London, London, United Kingdom
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Institute for Women's Health, University College London, London, United Kingdom.
| |
Collapse
|
12
|
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: 155] [Impact Index Per Article: 38.8] [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
|
13
|
Zhang S, Tu H, Yao J, Le J, Jiang Z, Tang Q, Zhang R, Huo P, Lei X. Combined use of Diane-35 and metformin improves the ovulation in the PCOS rat model possibly via regulating glycolysis pathway. Reprod Biol Endocrinol 2020; 18:58. [PMID: 32493421 PMCID: PMC7268382 DOI: 10.1186/s12958-020-00613-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/17/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex endocrine and metabolic disease with unknown pathogenesis. However, the treatment of Diane-35 combined with metformin can improve the endocrine and ovulation of PCOS. In this study, we investigated the effects of Diane-35 combined with metformin (DM) treatment on ovulation and glucose metabolism in a PCOS rat model. METHODS Sprague Dawley rats were divided into 3 groups, control group, model group (PCOS group) and Diane-35 combined with metformin (PCOS + DM group). The mRNA expression levels were determined by qRT-PCR. The hormone levels were determined by enzyme-linked immunosorbent assay. Immunostaining detected the protein levels of lactate dehydrogenase A (LDH-A), pyruvate kinase isozyme M2 (PKM2) and sirtuin 1 (SIRT1) in the ovarian tissues. TNUEL assay was performed to determine cell apoptosis in the PCOS rats. The metabolites in the ovarian tissues were analyzed by liquid chromatography with tandem mass spectrometry. RESULTS PCOS rats showed an increased in body weight, levels of luteinizing hormone and testosterone and insulin resistance, which was significantly attenuated by the DM treatment. The DM treatment improved disrupted estrous cycle and increased the granulosa cells of the ovary in the PCOS rats. The decreased proliferation and increased cell apoptosis of granulosa cells in the ovarian tissues of PCOS rats were significantly reversed by the DM treatment. The analysis of metabolics revealed that ATP and lactate levels were significantly decreased in PCOS rats, which was recovered by the DM treatment. Furthermore, the expression of LDH-A, PKM2 and SIRT1 was significantly down-regulated in ovarian tissues of the PCOS rats; while the DM treatment significantly increased the expression of LDH-A, PKM2 and SIRT1 in the ovarian tissues of the PCOS rats. CONCLUSION In conclusion, our study demonstrated that Diane-35 plus metformin treatment improved the pathological changes in the PCOS rats. Further studies suggest that Diane-35 plus metformin can improve the energy metabolism of the ovary via regulating the glycolysis pathway. The mechanistic studies indicated that the therapeutic effects of Diane-35 plus metformin treatment in the PCOS rats may be associated with the regulation of glycolysis-related mediators including PKM2, LDH-A and SIRT1.
Collapse
Affiliation(s)
- Shun Zhang
- grid.452806.dDepartment of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001 China
| | - Haoyan Tu
- grid.452806.dDepartment of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001 China
| | - Jun Yao
- grid.452806.dDepartment of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001 China
| | - Jianghua Le
- grid.452806.dDepartment of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, 541001 China
| | - Zhengxu Jiang
- grid.417409.f0000 0001 0240 6969School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000 China
| | - Qianqian Tang
- grid.417409.f0000 0001 0240 6969School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000 China
| | - Rongrong Zhang
- grid.417409.f0000 0001 0240 6969School of Basic Medical Sciences, Zunyi Medical University, Zunyi, 563000 China
| | - Peng Huo
- grid.443385.d0000 0004 1798 9548School of Public Health, Guilin Medical University, Guilin, 541004 China
| | - Xiaocan Lei
- grid.412017.10000 0001 0266 8918Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, University of South China, Hengyang, 421001 China
| |
Collapse
|
14
|
Delaroche L, Dupont C, Oger P, Aubriot FX, Lamazou F, Yazbeck C. [Polycystic ovary syndrome does not affect blastulation nor cumulative live birth rates]. ACTA ACUST UNITED AC 2019; 47:655-661. [PMID: 31336185 DOI: 10.1016/j.gofs.2019.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Polycystic ovarian syndrome (PCOS) brings complications in the management of the assisted reproductive technology (ART) because of an oocyte quality probably impaired due to modifications of intra- and extra-ovarian factors. Our study aimed to investigate the extended culture in PCOS patients and its influence on the cumulative live birth rates. METHODS Fifty-nine PCOS patients (as defined by the Rotterdam criteria) and 114 normo-ovulatory patients (i.e. with tubal, male or idiopathic infertility, regular cycles and AMH>2ng/mL) aged<37years old who underwent a 1st or 2nd ART attempt with extended culture to day 6 were included from October 2015 to December 2017. The blastulation and cumulative live birth rates were compared between the two groups. RESULTS The PCOS and control patients were 32.22 and 32.91years old respectively (P=0.05). The median number of oocytes retrieved was significantly higher in the PCOS group and the median oocyte maturity rate significantly lower compared with controls. The blastulation rates were similar between the PCOS and the control groups, respectively 57.8% vs. 58.6%, P=0.88. Because of the risks of hyperstimulation syndrome, a freeze all strategy was achieved for 38.9% of PCOS patients vs. 14.0% of the control patients (P<0.01). The cumulative live birth rates were not statistically different: 31.7% in the PCOS group vs. 37.2% in the control group, P=0.50. CONCLUSIONS PCOS was not observed to affect the extended culture nor the cumulative live birth rates in comparison to normo-ovulatory patients, supporting the blastocyst transfer strategy as a suitable option to PCOS patients.
Collapse
Affiliation(s)
- L Delaroche
- Laboratoire Eylau-Unilabs, 55-57, rue Saint-Didier, 75116 Paris, France.
| | - C Dupont
- Inserm équipe lipodystrophies génétiques et acquises, service de biologie de la reproduction-CECOS, Saint-Antoine Research center, hôpital Tenon, Sorbonne université, AP-HP, 75020 Paris, France
| | - P Oger
- Centre d'AMP de la clinique Pierre-Cherest, 5, rue Pierre-Cherest, 92200 Neuilly-sur-Seine, France
| | - F-X Aubriot
- Centre d'AMP de la clinique Pierre-Cherest, 5, rue Pierre-Cherest, 92200 Neuilly-sur-Seine, France
| | - F Lamazou
- Centre d'AMP de la clinique Pierre-Cherest, 5, rue Pierre-Cherest, 92200 Neuilly-sur-Seine, France
| | - C Yazbeck
- Centre d'AMP de la clinique Pierre-Cherest, 5, rue Pierre-Cherest, 92200 Neuilly-sur-Seine, France
| |
Collapse
|
15
|
Li Y, Wang L, Xu J, Niu W, Shi H, Hu L, Zhang Y, Zhang M, Bao X, Zhang N, Sun Y. Higher chromosomal aberration rate in miscarried conceptus from polycystic ovary syndrome women undergoing assisted reproductive treatment. Fertil Steril 2019; 111:936-943.e2. [PMID: 30922640 DOI: 10.1016/j.fertnstert.2019.01.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/31/2018] [Accepted: 01/16/2019] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To assess the potential association between polycystic ovary syndrome (PCOS) and chromosomally aberrant miscarriage during treatment with assisted reproductive technology (ART). DESIGN A retrospective, single-center study. SETTING University-affiliated reproductive center. PATIENT(S) A total of 328 patients sent their first trimester miscarried chorionic villus for genetic examination after ART in our center from January 2013 to September 2016, of which 119 cases were women with PCOS and 209 were non-PCOS controls. No known definite miscarriage-related concomitants existed in any study subject. INTERVENTION(S) Single nucleotide polymorphism array analysis was performed on all collected samples. MAIN OUTCOME MEASURE(S) Frequency of aberrant karyotype of miscarried conceptus and the correlation between PCOS and chromosomally aberrant miscarriage. RESULT(S) A total of 173 (52.7% of 328) conceptuses were identified as chromosomally aberrant by single nucleotide polymorphism array. Chromosomal aberrations were more frequent in conceptuses from PCOS patients compared with controls (61.3% vs. 47.8%). Furthermore, both univariate and multivariable analysis identified PCOS as a risk factor for an embryo/fetus to be chromosomally abnormal, with odds ratios of 1.957 (95% confidence interval, 1.067-3.590) and 2.008 (95% confidence interval, 1.038-3.883), respectively. CONCLUSION(S) Women with PCOS were at an increased risk of miscarrying a chromosomally aberrant embryo/fetus compared with non-PCOS controls during ART. Mechanisms require further investigation. Preimplantation genetic screening might be an effective approach to decrease the risk of spontaneous miscarriage for women with PCOS.
Collapse
Affiliation(s)
- Ying Li
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Linlin Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Jiawei Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Wenbin Niu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Hao Shi
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Linli Hu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Yile Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Meixiang Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Xiao Bao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Nan Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, and Henan Province Key Laboratory of Reproduction and Genetics, Henan, People's Republic of China.
| |
Collapse
|
16
|
Tsai TS, Tyagi S, St John JC. The molecular characterisation of mitochondrial DNA deficient oocytes using a pig model. Hum Reprod 2019; 33:942-953. [PMID: 29546367 DOI: 10.1093/humrep/dey052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/19/2018] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What are the molecular differences between mitochondrial DNA (mtDNA)-deficient and mtDNA-normal oocytes and how does mitochondrial supplementation alter these? SUMMARY ANSWER Changes to DNA methylation in a 5' cytosine-phosphate-guanine 3' (CpG) island in the mtDNA-specific replication factor (DNA polymerase gamma (POLG)) of mtDNA-deficient oocytes mediates an increase in mtDNA copy number by the 2-cell stage that positively modulates the expression of nuclear genes, which affect cellular and metabolic processes, following autologous mitochondrial supplementation. WHAT IS KNOWN ALREADY Too few copies of mtDNA in mature oocytes can lead to fertilisation failure or preimplantation embryo arrest. mtDNA-deficient oocytes that progress to blastocyst express genes associated with poor cellular and metabolic processes, transcriptional activation and mitochondrial biogenesis. STUDY DESIGN, SIZE, DURATION Using a pig oocyte model, we assessed mtDNA-deficient and mtDNA-normal oocytes during in vitro maturation for mtDNA variants and levels of DNA methylation in POLG. We supplemented mtDNA-deficient oocytes with autologous populations of mitochondria to determine if there were changes to DNA methylation in POLG that coincided with increases in mtDNA copy number. We assessed metaphase II mtDNA-deficient and mtDNA-normal oocytes by RNA sequencing to identify differentially expressed genes and compared their profiles to blastocysts derived from mtDNA-normal, mtDNA-deficient and supplemented mtDNA-deficient oocytes. PARTICIPANTS/MATERIALS, SETTING, METHODS mtDNA variant analysis (n = 24), mtDNA copy number (n = 60), POLG gene expression (n = 24), and RNA sequencing (n = 32 single; and 12 pooled cohorts of n = 5) were performed on oocytes and embryos. DNA methylation of a CpG island in POLG was determined quantitatively by pyrosequencing on oocytes to 2-cell embryos (n = 408). Bioinformatics tools were used to assess differences between mtDNA-normal and mtDNA-deficient oocytes and between mtDNA-normal and mtDNA-deficient oocytes and supplemented oocytes and their blastocyst stage equivalents. MAIN RESULTS AND THE ROLE OF CHANCE Whilst mtDNA-deficient oocytes regulated variants less stringently during maturation (P < 0.05), there were no differences in the ratio of variants in mature-stage oocytes. However, mtDNA-normal mature oocytes had significantly more molecules affected due to their higher copy number (P < 0.0001). Normal mature oocytes differently DNA methylated a CpG island in POLG compared with mtDNA-deficient oocytes (P < 0.01). Supplementation of mtDNA-deficient oocytes modulated DNA methylation at this CpG island leading to a mtDNA replication event prior to embryonic genome activation inducing significant increases in mtDNA copy number. RNA-Seq identified 57 differentially expressed genes (false discovery rate (FDR) < 0.05) between the two cohorts of oocytes with blastocyst stage gene expression altered by supplementation of mtDNA-deficient oocytes (P < 0.05) including genes associated with metabolic disorders. One key factor was branched chain amino acid transaminase 2 (BCAT2), a regulator of amino acid metabolism and associated with diabetes. LARGE SCALE DATA Sequence data are available on the NCBI Sequence Read Archive under the project number PRJNA422295. RNA sequencing data were deposited into NCBI Gene Expression Omnibus, under the accession number GSE108900. LIMITATIONS, REASONS FOR CAUTION Whilst this work was conducted in a species that is highly relevant to human reproduction, the outcomes need to be tested in human oocytes and blastocysts prior to clinical application. WIDER IMPLICATIONS OF THE FINDINGS The outcomes demonstrate a mechanism of action following mtDNA supplementation of mtDNA-deficient oocytes that results in improved gene expression at the blastocyst stage of development. STUDY FUNDING/COMPETING INTERESTS This work was funded by OvaScience Inc. OvaScience did not influence the study design, analysis of results or interpretation of the data.
Collapse
Affiliation(s)
- Te-Sha Tsai
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic 3168, Australia.,Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Vic 3168, Australia
| | - Sonika Tyagi
- Monash Bioinformatics Platform, Monash University, Clayton, Vic 3168, Australia
| | - Justin C St John
- Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, Vic 3168, Australia.,Department of Molecular and Translational Science, Monash University, 27-31 Wright Street, Clayton, Vic 3168, Australia
| |
Collapse
|
17
|
Siristatidis CS, Maheshwari A, Vaidakis D, Bhattacharya S. In vitro maturation in subfertile women with polycystic ovarian syndrome undergoing assisted reproduction. Cochrane Database Syst Rev 2018; 11:CD006606. [PMID: 30480769 PMCID: PMC6517219 DOI: 10.1002/14651858.cd006606.pub4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) occurs in 4% to 7% of all women of reproductive age and 50% of women presenting with subfertility. Subfertility affects 15% to 20% of couples trying to conceive. A significant proportion of these women ultimately need assisted reproductive technology (ART). In vitro fertilisation (IVF) is one of the assisted reproduction techniques employed to raise the chances of achieving a pregnancy. For the standard IVF technique, stimulating follicle development and growth before oocyte retrieval is essential, for which a large number of different methods combining gonadotrophins with a gonadotrophin-releasing hormone (GnRH) agonist or antagonist are used. In women with PCOS, the supra-physiological doses of gonadotrophins used for controlled ovarian hyperstimulation (COH) often result in an exaggerated ovarian response, characterised by the development of a large cohort of follicles of uneven quality, retrieval of immature oocytes, and increased risk of ovarian hyperstimulation syndrome (OHSS). A potentially effective intervention for women with PCOS-related subfertility involves earlier retrieval of immature oocytes at the germinal-vesicle stage followed by in vitro maturation (IVM). So far, the only data available have derived from observational studies and non-randomised clinical trials. OBJECTIVES To assess the effectiveness and safety of IVM followed by IVF or ICSI versus conventional IVF or ICSI among women with PCOS undergoing assisted reproduction. SEARCH METHODS This is the second update of this review. We performed the search on 17 April 2018.The search was designed with the help of the Cochrane Gynaecology and Fertility Group Information Specialist, for all published and unpublished randomised controlled trials (RCTs).We searched the the Cochrane Gynaecology and Fertility Group Specialised Register of controlled trials, CENTRAL via the Cochrane Central Register of Studies Online, MEDLINE, Embase, CINAHL, and the trial registers for ongoing and registered trials and the Open Grey database for grey literature from Europe. We made further searches in the National Institute for Health and Care Excellence (NICE) fertility assessment and treatment guidelines. We handsearched reference lists of relevant systematic reviews and RCTs, together with PubMed and Google for any recent trials that have not yet been indexed in the major databases. SELECTION CRITERIA All RCTs on the intention to perform IVM before IVF or ICSI compared with conventional IVF or ICSI for subfertile women with PCOS, irrespective of language and country of origin. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, assessed risk of bias, extracted data from studies, and attempted to contact the authors of studies for which data were missing. Our primary outcomes were live birth per woman randomised and miscarriage. We performed statistical analysis using Review Manager 5. We assessed evidence quality using GRADE methods. MAIN RESULTS We found two RCTs suitable for inclusion in the review and six ongoing trials that have not yet reported results. Both included studies were published as abstracts in international conferences.Both studies were at unclear or high risk of bias for most of the seven domains assessed. Common problems were unclear reporting of study methods and lack of blinding. The main limitations in the overall quality of the evidence were high risk of bias and serious imprecision.There were no data on the primary outcomes of this review, namely live birth per woman randomised and miscarriage.Both studies reported clinical pregnancy rate: there was evidence of an effect between IVM and IVF, favouring the former (odds ratio 3.10, 95% confidence interval 1.06 to 9.00; 71 participants; 2 studies; I2 = 0%; very low-quality evidence). The incidence of OHSS was zero in both studies in both groups.There were no data for the other outcomes specified in this review. AUTHORS' CONCLUSIONS Though promising data on the in vitro maturation (IVM) technique have been published, unfortunately there is still no evidence from properly conducted randomised controlled trials upon which to base any practice recommendations regarding IVM before in vitro fertilisation (IVF) or intracytoplasmic sperm injection for women with polycystic ovarian syndrome. Regarding our secondary outcomes, very low-quality evidence showed that clinical pregnancy was higher with IVM when compared to IVF, whereas the incidence of ovarian hyperstimulation syndrome was zero in both studies in both groups. We are awaiting the results of six ongoing trials and eagerly anticipate further evidence from good-quality trials in the field.
Collapse
Affiliation(s)
- Charalampos S Siristatidis
- Medical School, National and Kapodistrian University of AthensAssisted Reproduction Unit, 3rd Department of Obstetrics and GynaecologyAttikon University HospitalRimini 1AthensChaidariGreece12462
| | - Abha Maheshwari
- University of AberdeenDivision of Applied Health SciencesAberdeenUKAB25 2ZL
| | - Dennis Vaidakis
- University of Athens3rd Department of Obstetrics and Gynecology'Attikon' Hospital, ChaidariAthensGreece
| | | | | |
Collapse
|
18
|
Lu C, Chi H, Wang Y, Feng X, Wang L, Huang S, Yan L, Lin S, Liu P, Qiao J. Transcriptome analysis of PCOS arrested 2-cell embryos. Cell Cycle 2018; 17:1007-1013. [PMID: 29912628 DOI: 10.1080/15384101.2018.1467678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In an attempt to explore the early developmental arrest in embryos from polycystic ovarian syndrome (PCOS) patients, we sequenced the transcriptome profiles of PCOS arrested 2-cell embryos, non-PCOS arrested 2-cell embryos and non-arrested 2-cell embryos using single-cell RNA-Seq technique. Differential expression analysis was performed using the DEGSeq R package. Gene Ontology (GO) enrichment was analyzed using the GOseq R package. Data revealed 62 differentially expressed genes between non-PCOS arrested and PCOS arrested embryos and 2217 differentially expressed genes between PCOS arrested and non-arrested 2-cell embryos. A total of 49 differently expressed genes (DEGs) were annotated with GO terms in the up-regulated genes between PCOS arrested and non-PCOS arrested embryos after GO enrichment. A total of 29 DEGs were annotated with GO terms in the down-regulated genes between PCOS arrested and non-arrested 2-cell embryos after GO enrichment. These data can provide a reference for screening specific genes involved in the arrest of PCOS embryos.
Collapse
Affiliation(s)
- Cuiling Lu
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Hongbin Chi
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Yapeng Wang
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Xue Feng
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Lina Wang
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Shuo Huang
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Liying Yan
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Shengli Lin
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Ping Liu
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| | - Jie Qiao
- a Center for Reproductive Medicine, Department of Obstetrics and Gynecology , Peking University Third Hospital , Beijing , China
| |
Collapse
|
19
|
Cecchino GN, Seli E, Alves da Motta EL, García-Velasco JA. The role of mitochondrial activity in female fertility and assisted reproductive technologies: overview and current insights. Reprod Biomed Online 2018; 36:686-697. [DOI: 10.1016/j.rbmo.2018.02.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 02/18/2018] [Accepted: 02/28/2018] [Indexed: 12/21/2022]
|
20
|
Effects of methotrexate on the quality of oocyte maturation in vitro. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 47:249-260. [DOI: 10.1007/s00249-017-1254-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 09/02/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
|
21
|
Sampo AV, Palena C, Ganzer L, Maccari V, Estofán G, Hernández M. The adverse effect of overweight in assisted reproduction treatment outcomes. JBRA Assist Reprod 2017; 21:212-216. [PMID: 28837030 PMCID: PMC5574643 DOI: 10.5935/1518-0557.20170041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/17/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To assess Body Mass Index (BMI) effects on the results obtained from ICSI cycles. METHODS We studied 266 ICSI cycles performed between January 2014 and December 2016. The patients were grouped according to their BMI in: Normal (18.5-24.9), Overweight (25.0-29.9) and Obese (>30). We compared the following variables between the groups: number of antral follicles, ovarian stimulation length, gonadotropin dose used, maximum estradiol level, follicles developed/antral follicles, retrieved oocytes/developed follicles and mature/retrieved oocytes, normal fertilization rate, embryo achieved/normal fertilized oocytes, clinical pregnancy and implantation rates. We used the Kruskal-Wallis and the Chi square tests. p<0.05 was considered significant. RESULTS Normal, Overweight and Obese patients presented comparable values for number of antral follicles (11.6±5.4, 12.5±5.5, 12.2±5.7), ovarian stimulation length (7.5±1.4, 7.6±1.1, 7.8±1.3) and gonadotropin dose used (2043±489, 1940±536, 2109±605). Obese patients had lower values of estradiol (1560±610, 1511±635, 1190±466; p=0.018), developed follicles (81%, 76%, 70%; p<0.0001), and retrieved oocytes (91%, 90%, 84%; p=0.0017); and not significantly lower values of mature oocytes (82%, 82%, 77%; p=0.26). The groups had comparable fertilization rates (72%, 73%, 69%) and embryo achieved rates (67%, 63%, 72%). The normal group had higher, but not significantly higher pregnancy and implantation rates (43%, 40%, 38%, p=0.53; and 33%, 26%, 23%; p=0.11), and significantly higher ongoing pregnancy rates (37%, 33%, 33%, p=0.042). CONCLUSION Increased BMI patients had impaired ovarian response and lower pregnancy rates in ICSI cycles.
Collapse
Affiliation(s)
| | - Celina Palena
- CIGOR - Centro Integral de Ginecología, Obstetricia y
Reproducción. Córdoba, Argentina
| | - Luciano Ganzer
- CIGOR - Centro Integral de Ginecología, Obstetricia y
Reproducción. Córdoba, Argentina
| | - Virginia Maccari
- CIGOR - Centro Integral de Ginecología, Obstetricia y
Reproducción. Córdoba, Argentina
| | - Gustavo Estofán
- CIGOR - Centro Integral de Ginecología, Obstetricia y
Reproducción. Córdoba, Argentina
| | - Mariana Hernández
- CIGOR - Centro Integral de Ginecología, Obstetricia y
Reproducción. Córdoba, Argentina
| |
Collapse
|
22
|
Fournier A, Torre A, Delaroche L, Gala A, Mullet T, Ferrières A, Hamamah S. [Quality of oocytes and embryos from women with polycystic ovaries syndrome: State of the art]. ACTA ACUST UNITED AC 2017; 45:429-438. [PMID: 28757106 DOI: 10.1016/j.gofs.2017.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Indexed: 12/21/2022]
Abstract
The frequency of polycystic ovary syndrome (PCOS) and the consequent fertility disorders cause many difficulties in the management of the assisted reproductive technics. Some studies are focused on different additional treatments, stimulation protocols or techniques that could optimize the in vitro fertilization cycles. The quality of the oocytes and embryos of these patients is also an outstanding issue. They remain difficult to actually evaluate during management, and none of the few published studies on this subject demonstrated any inferiority, compared to control patients. However, many differences have been highlighted, studying intra- and extra-ovarian factors. The advent of new genetic techniques could allow a better understanding of the pathophysiological mechanisms of the syndrome, as well as refining the evaluation of oocytes and embryos, in order to better predict the results of in vitro fertilization attempts. Pregnancy and birth rates, however, appear to be comparable to those of the general population.
Collapse
Affiliation(s)
- A Fournier
- Département de biologie de la reproduction et du diagnostic pré-implantatoire, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - A Torre
- Département gynécologie-obstétrique, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - L Delaroche
- Clinique P. Cherest, centre de fécondation in vitro, 5, rue Pierre-Cherest, 92200 Neuilly-sur-Seine, France
| | - A Gala
- Département de biologie de la reproduction et du diagnostic pré-implantatoire, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France; Inserm U1203, institut de médecine régénérative et biothérapies (IRMB), hôpital Saint-Éloi, CHRU de Montpellier, 80, rue Augustin-Fliche, 34295 Montpellier, France
| | - T Mullet
- Département de biologie de la reproduction et du diagnostic pré-implantatoire, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France; Inserm U1203, institut de médecine régénérative et biothérapies (IRMB), hôpital Saint-Éloi, CHRU de Montpellier, 80, rue Augustin-Fliche, 34295 Montpellier, France
| | - A Ferrières
- Département de biologie de la reproduction et du diagnostic pré-implantatoire, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France; Inserm U1203, institut de médecine régénérative et biothérapies (IRMB), hôpital Saint-Éloi, CHRU de Montpellier, 80, rue Augustin-Fliche, 34295 Montpellier, France
| | - S Hamamah
- Département de biologie de la reproduction et du diagnostic pré-implantatoire, hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France; Inserm U1203, institut de médecine régénérative et biothérapies (IRMB), hôpital Saint-Éloi, CHRU de Montpellier, 80, rue Augustin-Fliche, 34295 Montpellier, France.
| |
Collapse
|
23
|
Abstract
This review attempts to summarize the known literature on high responders to ovarian stimulation during assisted reproductive techniques (ART). Response to gonadotrophins is subject to significant interindividual and intercycle variation, thus carrying a risk of high response or poor response to ovarian stimulation regimens. The main risk for high responders is the development of ovarian hyperstimulation syndrome (OHSS) which is associated with significant morbidity. Hence, the definition of high responders in the literature has primarily focussed on risk factors for OHSS. Strategies to reduce OHSS including tailoring of the ovarian stimulation regimens and adjusting gonadotrophin doses according to patient characteristics and findings during the cycle of stimulation. In addition, modifying the type of ovulation trigger used and adjuvant therapies, such as metformin, intravenous colloids and vascular endothelial growth factor blockers, have also been studied as options to reduce OHSS. Apart from the risk of OHSS, high response also appears to have an adverse impact on the oocyte and endometrium, though there is a paucity of data regarding the extent and mechanisms behind this impact.
Collapse
Affiliation(s)
- Mariano Mascarenhas
- a Seacroft Hospital , Leeds Centre for Reproductive Medicine , Leeds , United Kingdom
| | - Adam H Balen
- a Seacroft Hospital , Leeds Centre for Reproductive Medicine , Leeds , United Kingdom
| |
Collapse
|
24
|
Tsai T, St John JC. The role of mitochondrial DNA copy number, variants, and haplotypes in farm animal developmental outcome. Domest Anim Endocrinol 2016; 56 Suppl:S133-46. [PMID: 27345311 DOI: 10.1016/j.domaniend.2016.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 03/11/2016] [Accepted: 03/15/2016] [Indexed: 01/20/2023]
Abstract
The vast majority of cellular energy is generated through the process of oxidative phosphorylation, which takes place in the electron transport chain in the mitochondria. The electron transport chain is encoded by 2 genomes, the chromosomal and the mitochondrial genomes. Mitochondrial DNA is associated with a number of traits, which include tolerance to heat, growth and physical performance, meat and milk quality, and fertility. Mitochondrial genomes can be clustered into groups known as mtDNA haplotypes. Mitochondrial DNA haplotypes are a potential genetic source for manipulating phenotypes in farm animals. The use of assisted reproductive technologies, such as nuclear transfer, allows favorable chromosomal genetic traits to be mixed and matched with sought after mtDNA haplotype traits. As a result super breeds can be generated.
Collapse
Affiliation(s)
- Tesha Tsai
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Vic, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Vic, 3168, Australia
| | - Justin C St John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Vic, 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Vic, 3168, Australia.
| |
Collapse
|
25
|
Pruksananonda K, Wasinarom A, Sereepapong W, Sirayapiwat P, Rattanatanyong P, Mutirangura A. Epigenetic modification of long interspersed elements-1 in cumulus cells of mature and immature oocytes from patients with polycystic ovary syndrome. Clin Exp Reprod Med 2016; 43:82-9. [PMID: 27358825 PMCID: PMC4925871 DOI: 10.5653/cerm.2016.43.2.82] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/24/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The long interspersed elements (LINE-1, L1s) are a group of genetic elements found in large numbers in the human genome that can translate into phenotype by controlling genes. Growing evidence supports the role of epigenetic in polycystic ovary syndrome (PCOS). The purpose of this study is to evaluate the DNA methylation levels in LINE-1 in a tissue-specific manner using cumulus cells from patients with PCOS compared with normal controls. METHODS The study included 19 patients with PCOS and 22 control patients who were undergoing controlled ovarian hyperstimulation. After oocyte retrieval, cumulus cells were extracted. LINE-1 DNA methylation levels were analysed by bisulfite treatment, polymerase chain reaction, and restriction enzyme digestion. The Connection Up- and Down-Regulation Expression Analysis of Microarrays software package was used to compare the gene regulatory functions of intragenic LINE-1. RESULTS The results showed higher LINE-1 DNA methylation levels in the cumulus cells of mature oocytes in PCOS patients, 79.14 (±2.66) vs. 75.40 (±4.92); p=0.004, but no difference in the methylation of cumulus cells in immature oocytes between PCOS and control patients, 70.33 (±4.79) vs. 67.79 (±5.17); p=0.155. However, LINE-1 DNA methylation levels were found to be higher in the cumulus cells of mature oocytes than in those of immature oocytes in both PCOS and control patients. CONCLUSION These findings suggest that the epigenetic modification of LINE-1 DNA may play a role in regulating multiple gene expression that affects the pathophysiology and development of mature oocytes in PCOS.
Collapse
Affiliation(s)
- Kamthorn Pruksananonda
- Reproductive Medicine Division, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Artisa Wasinarom
- Reproductive Medicine Division, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wisan Sereepapong
- Reproductive Medicine Division, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Porntip Sirayapiwat
- Reproductive Medicine Division, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prakasit Rattanatanyong
- Center of Excellence in Molecular Genetics of Cancer and Human Disease, Department of Anatomy, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Apiwat Mutirangura
- Center of Excellence in Molecular Genetics of Cancer and Human Disease, Department of Anatomy, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
26
|
St John JC, Tsai TS, Cagnone GL. Mitochondrial DNA supplementation as an enhancer of female reproductive capacity. Curr Opin Obstet Gynecol 2016; 28:211-6. [DOI: 10.1097/gco.0000000000000265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Wang Q, Luo L, Lei Q, Lin MM, Huang X, Chen MH, Zeng YH, Zhou CQ. Low aneuploidy rate in early pregnancy loss abortuses from patients with polycystic ovary syndrome. Reprod Biomed Online 2016; 33:85-92. [PMID: 27157933 DOI: 10.1016/j.rbmo.2016.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/15/2022]
Abstract
A prospective cohort study was conducted to determine whether chromosome aneuploidy increases the risk of early spontaneous abortions in patients with polycystic ovary syndrome (PCOS). A total of 1461 patients who conceived after IVF and embryo transfer were followed; 100 patients who had experienced clinical spontaneous abortion were recruited, 32 with PCOS and 68 without PCOS. Before 2013, genetic analysis comprised conventional cultured villus chromosome karyotyping and a multiplex ligation-dependent probe amplification subtelomere assay combined with fluorescence in-situ hybridization; since 2013, array-based comparative genomic hybridization technique combined with chromosome karyotyping has been used. Age, BMI, pregnancy history, gestational age and total gonadotrophin dosage did not differ significantly between the PCOS and non-PCOS groups. In the PCOS group, 28.1% of abortuses demonstrated aneuploidy, which was significantly lower (P = 0.001) than in the non-PCOS group (72.1%). Further statistical analyses controlling for maternal age demonstrated that abortuses of women with PCOS were significantly less (P = 0.001) likely to have chromosome aneuploidy. Embryonic aneuploidy does not play a vital role in early spontaneous abortion in women with PCOS. Maternal factors resulting in endometrial disorders are more likely to be responsible for the increased risk of early spontaneous abortion in patients with PCOS.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
| | - Lu Luo
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
| | - Qiong Lei
- Department of Obstetrics, Guangdong Women and Children Hospital, Guangzhou, China
| | - Ming-Mei Lin
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China; Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Xuan Huang
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ming-Hui Chen
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
| | - Yan-Hong Zeng
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
| | - Can-Quan Zhou
- Department of Obstetrics and Gynecology, The Center of Reproductive Medicine, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China.
| |
Collapse
|
28
|
Restoration of normal embryogenesis by mitochondrial supplementation in pig oocytes exhibiting mitochondrial DNA deficiency. Sci Rep 2016; 6:23229. [PMID: 26987907 PMCID: PMC4796791 DOI: 10.1038/srep23229] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/03/2016] [Indexed: 11/08/2022] Open
Abstract
An increasing number of women fail to achieve pregnancy due to either failed fertilization or embryo arrest during preimplantation development. This often results from decreased oocyte quality. Indeed, reduced mitochondrial DNA copy number (mitochondrial DNA deficiency) may disrupt oocyte quality in some women. To overcome mitochondrial DNA deficiency, whilst maintaining genetic identity, we supplemented pig oocytes selected for mitochondrial DNA deficiency, reduced cytoplasmic maturation and lower developmental competence, with autologous populations of mitochondrial isolate at fertilization. Supplementation increased development to blastocyst, the final stage of preimplantation development, and promoted mitochondrial DNA replication prior to embryonic genome activation in mitochondrial DNA deficient oocytes but not in oocytes with normal levels of mitochondrial DNA. Blastocysts exhibited transcriptome profiles more closely resembling those of blastocysts from developmentally competent oocytes. Furthermore, mitochondrial supplementation reduced gene expression patterns associated with metabolic disorders that were identified in blastocysts from mitochondrial DNA deficient oocytes. These results demonstrate the importance of the oocyte’s mitochondrial DNA investment in fertilization outcome and subsequent embryo development to mitochondrial DNA deficient oocytes.
Collapse
|
29
|
Huang Y, Yu Y, Gao J, Li R, Zhang C, Zhao H, Zhao Y, Qiao J. Impaired oocyte quality induced by dehydroepiandrosterone is partially rescued by metformin treatment. PLoS One 2015; 10:e0122370. [PMID: 25811995 PMCID: PMC4374838 DOI: 10.1371/journal.pone.0122370] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/15/2015] [Indexed: 12/25/2022] Open
Abstract
The present study evaluated the influence of hyperandrogenism on oocyte quality using a murine PCOS model induced by dehydroepiandrosterone (DHEA) and further explored the effect of metformin treatment. Female BALB/c mice were treated with a vehicle control or DHEA (6 mg /100 g body weight) or DHEA plus metformin (50 mg /100 g body weight) for 20 consecutive days. DHEA-induced mice resembled some characters of human PCOS, such as irregular sexual cycles and polycystic ovaries. After the model validation was completed, metaphase II (MII) oocytes were retrieved and subsequent analyses of oocyte quality were performed. DHEA-treated mice yielded fewer MII oocytes, which displayed decreased mtDNA copy number, ATP content, inner mitochondrial membrane potential, excessive oxidative stress and impaired embryo development competence compared with those in control mice. Metformin treatment partially attenuated those damages, as evidenced by the increased fertilization and blastocyst rate, ATP content, GSH concentration and GSH/GSSG ratio, and decreased reactive oxygen species levels. No significant difference in normal spindle assembly was observed among the three groups. During in vitro maturation (IVM), the periods of germinal vesicle breakdown (GVBD) and the first polar body (PB1) extrusion were extended and the maturation rate of GVBD oocytes was decreased in DHEA mice compared with controls. Metformin treatment decreased the time elapsed of GVBD while had no effect on PB1 extrusion. These results indicated that excessive androgen is detrimental to oocyte quality while metformin treatment is, directly or indirectly, beneficial for oocyte quality improvement.
Collapse
Affiliation(s)
- Ying Huang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yang Yu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Jiangman Gao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rong Li
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chunmei Zhang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Hongcui Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yue Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- * E-mail: (JQ); (YZ)
| | - Jie Qiao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- * E-mail: (JQ); (YZ)
| |
Collapse
|
30
|
Uppangala S, Dhiman S, Salian SR, Singh VJ, Kalthur G, Adiga SK. In vitro matured oocytes are more susceptible than in vivo matured oocytes to mock ICSI induced functional and genetic changes. PLoS One 2015; 10:e0119735. [PMID: 25786120 PMCID: PMC4364773 DOI: 10.1371/journal.pone.0119735] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/13/2015] [Indexed: 11/19/2022] Open
Abstract
Background Concerns regarding the safety of ICSI have been intensified recently due to increased risk of birth defects in ICSI born children. Although fertilization rate is significantly higher in ICSI cycles, studies have failed to demonstrate the benefits of ICSI in improving the pregnancy rate. Poor technical skill, and suboptimal in vitro conditions may account for the ICSI results however, there is no report on the effects of oocyte manipulations on the ICSI outcome. Objective The present study elucidates the influence of mock ICSI on the functional and genetic integrity of the mouse oocytes. Methods Reactive Oxygen Species (ROS) level, mitochondrial status, and phosphorylation of H2AX were assessed in the in vivo matured and IVM oocytes subjected to mock ICSI. Results A significant increase in ROS level was observed in both in vivo matured and IVM oocytes subjected to mock ICSI (P<0.05-0.001) whereas unique mitochondrial distribution pattern was found only in IVM oocytes (P<0.01-0.001). Importantly, differential H2AX phosphorylation was observed in both in vivo matured and IVM oocytes subjected to mock ICSI (P <0.001). Conclusion The data from this study suggests that mock ICSI can alter genetic and functional integrity in oocytes and IVM oocytes are more vulnerable to mock ICSI induced changes.
Collapse
Affiliation(s)
- Shubhashree Uppangala
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
| | - Shilly Dhiman
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
| | - Sujit Raj Salian
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
| | - Vikram Jeet Singh
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
| | - Guruprasad Kalthur
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
| | - Satish Kumar Adiga
- Division of Clinical Embryology, Department of Obstetrics & Gynecology, Kasturba Medical College, Manipal University, Manipal-576 104, India
- * E-mail:
| |
Collapse
|
31
|
Katulski K, Czyzyk A, Podfigurna-Stopa A, Genazzani AR, Meczekalski B. Pregnancy complications in polycystic ovary syndrome patients. Gynecol Endocrinol 2015; 31:87-91. [PMID: 25356655 DOI: 10.3109/09513590.2014.974535] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [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
Infertility is a widely disputed problem affecting patients suffering from polycystic ovary syndrome (PCOS). As a serious dysfunction, it frequently occurs in PCOS patients. It is, therefore, important to devote more attention to pregnancy in PCOS sufferers. According to various data, the risk of miscarriage in PCOS women is three times higher than the risk of miscarriage in healthy women. Unfortunately, the risk of most frequent pregnancy pathologies is also higher for PCOS patients, as gestational diabetes (GD), pregnancy-induced hypertension and pre-eclampsia, and small for gestational age (SGA) children. Impaired glucose tolerance and GD in pregnant PCOS patients occur more frequently than in healthy women. A quadruple increase in the risk of pregnancy-induced hypertension linked to arterial wall stiffness has also been observed in PCOS patients. The risk of pre-eclampsia, the most severe of all complications, is also four times higher in those suffering from PCOS. Pre-eclampsia is also more frequent in patients presenting additional risk factors accompanying PCOS, such as obesity or GD. At that point, it should be mentioned that PCOS patients are under 2.5 higher risk of giving birth to SGA children than healthy women. It appears that SGA can be linked to insulin resistance and insulin-dependent growth dysfunction. Therefore, PCOS pregnant women are patients of special obstetrical care.
Collapse
Affiliation(s)
- Krzysztof Katulski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences , Poznan , Poland and
| | | | | | | | | |
Collapse
|
32
|
Gu L, Liu H, Gu X, Boots C, Moley KH, Wang Q. Metabolic control of oocyte development: linking maternal nutrition and reproductive outcomes. Cell Mol Life Sci 2014; 72:251-71. [PMID: 25280482 DOI: 10.1007/s00018-014-1739-4] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 02/01/2023]
Abstract
Obesity, diabetes, and related metabolic disorders are major health issues worldwide. As the epidemic of metabolic disorders continues, the associated medical co-morbidities, including the detrimental impact on reproduction, increase as well. Emerging evidence suggests that the effects of maternal nutrition on reproductive outcomes are likely to be mediated, at least in part, by oocyte metabolism. Well-balanced and timed energy metabolism is critical for optimal development of oocytes. To date, much of our understanding of oocyte metabolism comes from the effects of extrinsic nutrients on oocyte maturation. In contrast, intrinsic regulation of oocyte development by metabolic enzymes, intracellular mediators, and transport systems is less characterized. Specifically, decreased acid transport proteins levels, increased glucose/lipid content and elevated reactive oxygen species in oocytes have been implicated in meiotic defects, organelle dysfunction and epigenetic alteration. Therefore, metabolic disturbances in oocytes may contribute to the diminished reproductive potential experienced by women with metabolic disorders. In-depth research is needed to further explore the underlying mechanisms. This review also discusses several approaches for metabolic analysis. Metabolomic profiling of oocytes, the surrounding granulosa cells, and follicular fluid will uncover the metabolic networks regulating oocyte development, potentially leading to the identification of oocyte quality markers and prevention of reproductive disease and poor outcomes in offspring.
Collapse
Affiliation(s)
- Ling Gu
- College of Animal Science and Technology, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, Jiangsu, China,
| | | | | | | | | | | |
Collapse
|
33
|
Maruthini D, Harris SE, Barth JH, Balen AH, Campbell BK, Picton HM. The effect of metformin treatment in vivo on acute and long-term energy metabolism and progesterone production in vitro by granulosa cells from women with polycystic ovary syndrome. Hum Reprod 2014; 29:2302-16. [PMID: 25139174 PMCID: PMC4164147 DOI: 10.1093/humrep/deu187] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION What are the consequences of polycystic ovary syndrome (PCOS) pathology and metformin-pretreatment in vivo in women with PCOS on the metabolism and steroid production of follicular phenotype- and long-term cultured-granulosa cells (GC)? SUMMARY ANSWER PCOS pathology significantly compromised glucose metabolism and the progesterone synthetic capacity of follicular- and long-term cultured-GCs and the metabolic impact of PCOS on GC function was alleviated by metformin-pretreatment in vivo. WHAT IS KNOWN ALREADY Granulosa cells from women with PCOS have been shown to have an impaired insulin-stimulated glucose uptake and lactate production in vitro. However, these results were obtained by placing GCs in unphysiological conditions in culture medium containing high glucose and insulin concentrations. Moreover, existing data on insulin-responsive steroid production in vitro by PCOS GCs vary. STUDY DESIGN, SIZE AND DURATION Case-control experimental research comparing glucose uptake, pyruvate and lactate production and progesterone production in vitro by GCs from three aetiological groups, all undergoing IVF; healthy control women (Control, n = 12), women with PCOS treated with metformin in vivo (Metformin, n = 8) and women with PCOS not exposed to metformin (PCOS, n = 8). The study was conducted over a period of 3 years between 2007 and 2010. PARTICIPANTS/MATERIALS, SETTING, METHODS Rotterdam criteria were used for the diagnosis of PCOS; all subjects were matched for age, BMI and baseline FSH. Individual patient cultures were undertaken with cells incubated in a validated, physiological, serum-free culture medium containing doses of 0–6 mM glucose and 0–100 ng/ml insulin for 6 h and 144 h to quantify the impact of treatments on acute and long-term metabolism, respectively, and progesterone production. The metabolite content of spent media was measured using spectrophotometric plate reader assay. The progesterone content of spent media was measured by enzyme-linked immunosorbent assay. Viable GC number was quantified after 144 h of culture by the vital dye Neutral Red uptake assay. MAIN RESULTS AND THE ROLE OF CHANCE Granulosa cells from women with PCOS pathology revealed reduced pyruvate production and preferential lactate production in addition to their reduced glucose uptake during cultures (P < 0.05). Metformin pretreatment alleviated this metabolic lesion (P < 0.05) and enhanced cell proliferation in vitro (P < 0.05), but cells retained a significantly reduced capacity for progesterone synthesis compared with controls (P < 0.05). LIMITATIONS, REASONS FOR CAUTION Although significant treatment effects were detected in this small cohort, further studies are required to underpin the molecular mechanisms of the effect of metformin on GCs. WIDER IMPLICATIONS OF THE FINDINGS The individual patient culture strategy combined with multifactorial experimental design strengthens the biological interpretation of the data. Collectively, these results support the notion that there is an inherent impairment in progesterone biosynthetic capacity of the GCs from women with PCOS. The positive, acute metabolic effect and the negative long-term steroidogenic effect on GCs following metformin exposure in vivo may have important implications for follicular development and luteinized GC function when the drug is used in clinical practice. STUDY FUNDING/COMPETING INTEREST(S) No competing interests. This work was supported by the UK Medical Research Council Grant Reference number G0800250.
Collapse
Affiliation(s)
- D Maruthini
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - S E Harris
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| | - J H Barth
- Department of Clinical Biochemistry, Leeds Teaching Hospitals NHS Trust, Leeds General Infirmary, Great George Street, Leeds LS1 3EX, UK
| | - A H Balen
- The Leeds Centre for Reproductive Medicine, Seacroft Hospital, York Road, Leeds LS14 6UH, UK
| | - B K Campbell
- Division of Human Development at Nottingham University Hospitals NHS Trust, Department of Obstetrics and Gynaecology, Queen's Medical Centre Campus, Nottingham NG7 2UH, UK
| | - H M Picton
- Division of Reproduction and Early Development, Leeds Institute for Genetics, Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
| |
Collapse
|
34
|
Liu HY, Liu JQ, Mai ZX, Zeng YT. A subpathway-based method of drug reposition for polycystic ovary syndrome. Reprod Sci 2014; 22:423-30. [PMID: 25015903 DOI: 10.1177/1933719114542025] [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] [Indexed: 11/17/2022]
Abstract
The need for development of new therapeutic agents for polycystic ovary syndrome (PCOS) is urgent due to general lack of efficient and specialized drugs currently available. We aimed to explore the metabolic mechanism of PCOS and inferred drug reposition for PCOS by a subpathway-based method. Using the GSE34526 microarray data from the Gene Expression Omnibus database, we first identified the differentially expressed genes (DEGs) between PCOS and normal samples. Then, we identified 13 significantly enriched metabolic subpathways that may be involved in the development of PCOS. Finally, by an integrated analysis of PCOS-involved subpathways and drug-affected subpathways, we identified 54 novel small molecular drugs capable to target the PCOS-involved subpathways. We also mapped the DEGs of PCOS and a potential novel drug (alprostadil) into purine metabolism pathway to illustrate the potentially active mechanism of alprostadil on PCOS. Candidate agents identified by our approach may provide insights into a novel therapy approach for PCOS.
Collapse
Affiliation(s)
- Hai-Ying Liu
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Jian-Qiao Liu
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Zi-Xin Mai
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| | - Yan-Ting Zeng
- Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, PR China
| |
Collapse
|
35
|
Impact of PCOS on early embryo cleavage kinetics. Reprod Biomed Online 2014; 28:508-14. [DOI: 10.1016/j.rbmo.2013.11.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 12/19/2022]
|
36
|
Murri M, Insenser M, Escobar-Morreale HF. Metabolomics in polycystic ovary syndrome. Clin Chim Acta 2014; 429:181-8. [DOI: 10.1016/j.cca.2013.12.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/12/2013] [Accepted: 12/14/2013] [Indexed: 10/25/2022]
|
37
|
Siristatidis CS, Vrachnis N, Creatsa M, Maheshwari A, Bhattacharya S. In vitro maturation in subfertile women with polycystic ovarian syndrome undergoing assisted reproduction. Cochrane Database Syst Rev 2013:CD006606. [PMID: 24101529 DOI: 10.1002/14651858.cd006606.pub3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) occurs in 4% to 7% of all women of reproductive age and 50% of women presenting with subfertility. Subfertility affects 15% to 20% of couples trying to conceive. A significant proportion of these women ultimately need assisted reproductive technology (ART). In vitro fertilisation (IVF) is one of the assisted reproduction techniques employed to raise the chances of achieving a pregnancy. For the standard IVF technique, stimulating follicle development and growth before oocyte retrieval is essential, for which a large number of different methods combining gonadotrophins with a gonadotrophin-releasing hormone (GnRH) agonist or antagonist are used. In women with PCOS, the supra-physiological doses of gonadotrophins used for controlled ovarian hyperstimulation (COH) often result in an exaggerated ovarian response, characterised by the development of a large cohort of follicles of uneven quality, retrieval of immature oocytes, and increased risk of ovarian hyperstimulation syndrome. A potentially effective intervention for women with PCOS-related subfertility involves earlier retrieval of immature oocytes at the germinal-vesicle stage followed by in vitro maturation (IVM). So far, the only data available have derived from observational studies and non-randomised clinical trials. OBJECTIVES To compare outcomes associated with in vitro maturation (IVM) followed by vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI) versus conventional IVF or ICSI, among women with polycystic ovarian syndrome (PCOS) undergoing assisted reproductive technologies (ART). SEARCH METHODS We searched the Menstrual Disorders and Subfertility Group (MDSG) Specialised Register of controlled trials to May 2013 for any relevant trials identified from the title, abstract, or keyword sections. This was followed by a search of the electronic database MEDLINE, EMBASE, LILACS and CINAHL, without language restriction. We also performed a manual search of the references of all retrieved articles; sought unpublished papers and abstracts submitted to international conferences, searched the clinicaltrials.gov and WHO portal registries for submitted protocols of clinical trials, and contacted experts. In addition, we examined the National Institute of Clinical Excellence (NICE) fertility assessment and treatment guidelines and handsearched reference lists of relevant articles (from 1970 to May 2013). SELECTION CRITERIA All randomised trials (RCTs) on the intention to perform IVM before IVF or ICSI compared with conventional IVF or ICSI for subfertile women with PCOS. DATA COLLECTION AND ANALYSIS Three review authors (CS, MK and NV) independently assessed eligibility and quality of trials. Primary outcome measure was live birth rate per randomised woman. MAIN RESULTS There were no RCTs suitable for inclusion in the review, although there are currently three ongoing trials that have not yet reported results. AUTHORS' CONCLUSIONS Though promising data on the IVM technique have been published, unfortunately there is still no evidence from RCTs upon which to base any practice recommendations regarding IVM before IVF or ICSI for women with PCOS. Meanwhile, the results of the above-mentioned ongoing trials are awaited and, of course, further evidence from good quality trials in the field is eagerly anticipated.
Collapse
Affiliation(s)
- Charalampos S Siristatidis
- Assisted Reproduction Unit, 3rd Department of Obstetrics and Gynaecology, University of Athens, Attikon University Hospital,, Rimini 1, Athens, Chaidari, Greece, 12462
| | | | | | | | | |
Collapse
|
38
|
Cotterill M, Harris SE, Collado Fernandez E, Lu J, Huntriss JD, Campbell BK, Picton HM. The activity and copy number of mitochondrial DNA in ovine oocytes throughout oogenesis in vivo and during oocyte maturation in vitro. Mol Hum Reprod 2013; 19:444-50. [PMID: 23468533 PMCID: PMC3690804 DOI: 10.1093/molehr/gat013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria are responsible for the production of ATP, which drives cellular metabolic and biosynthetic processes. This is the first study to quantify the mtDNA copy number across all stages of oogenesis in a large monovulatory species, it includes assessment of the activity of mitochondria in germinal vesicle (GV) and metaphase II (MII) oocytes through JC1 staining. Primordial to early antral follicles (n = 249) were isolated from the sheep ovarian cortex following digestion at 37°C for 1 h and all oocytes were disaggregated from their somatic cells. Germinal vesicle oocytes (n = 133) were aspirated from 3- to 5-mm diameter antral follicles, and mature MII oocytes (n = 71) were generated following in vitro maturation (IVM). The mtDNA copy number in each oocyte was quantified using real-time PCR and showed a progressive, but variable increase in the amount of mtDNA in oocytes from primordial follicles (605 ± 205, n = 8) to mature MII oocytes (744 633 ± 115 799, n = 13; P < 0.05). Mitochondrial activity (P > 0.05) was not altered during meiotic progression from GV to MII during IVM. The observed increase in the mtDNA copy number across oogenesis reflects the changing ATP demands needed to orchestrate cytoskeletal and cytoplasmic reorganization during oocyte growth and maturation and the need to fuel the resumption of meiosis in mature oocytes following the pre-ovulatory gonadotrophin surge.
Collapse
Affiliation(s)
- Matthew Cotterill
- Division of Reproduction and Early Development, Leeds Institute of Genetics Health and Therapeutics, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK.
| | | | | | | | | | | | | |
Collapse
|
39
|
Lindenberg S. New approach in patients with polycystic ovaries, lessons for everyone. Fertil Steril 2013; 99:1170-2. [PMID: 23465705 DOI: 10.1016/j.fertnstert.2013.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/01/2013] [Accepted: 02/06/2013] [Indexed: 11/25/2022]
Abstract
In vitro maturation is a safe and simple procedure for both the clinician and the patient. The discrepancy between the early clinical results and today's pregnancy rates (PR) of 30% is primarily due to the following modification of the original protocol recently reported by several groups: Priming with FSH and hCG, timing oocyte collection from follicles larger than 12 mm in diameter, and using blastocyst transfer. Furthermore, the endometrial priming should start 2 days before oocyte pick-up. This provides a PR of 40% per ET.
Collapse
|
40
|
Hemmings KE, Maruthini D, Vyjayanthi S, Hogg JE, Balen AH, Campbell BK, Leese HJ, Picton HM. Amino acid turnover by human oocytes is influenced by gamete developmental competence, patient characteristics and gonadotrophin treatment. Hum Reprod 2013; 28:1031-44. [PMID: 23335609 PMCID: PMC3600837 DOI: 10.1093/humrep/des458] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
STUDY QUESTION Can amino acid profiling differentiate between human oocytes with differing competence to mature to metaphase II (MII) in vitro? SUMMARY ANSWER Oocytes which remained arrested at the germinal vesicle (GV) stage after 24 h of in vitro maturation (IVM) displayed differences in the depletion/appearance of amino acids compared with oocytes which progressed to MII and patient age, infertile diagnosis and ovarian stimulation regime significantly affected oocyte amino acid turnover during IVM. WHAT IS KNOWN ALREADY Amino acid profiling has been proposed as a technique which can distinguish between human pronucleate zygotes and cleavage stage embryos with the potential to develop to the blastocyst stage and implant to produce a pregnancy and those that arrest. Most recently, the amino acid turnover by individual bovine oocytes has been shown to be predictive of oocyte developmental competence as indicated by the gamete's capacity to undergo fertilization and early cleavage divisions in vitro. STUDY DESIGN, SIZE, DURATION The study was conducted between March 2005 and March 2010. A total of 216 oocytes which were at the GV or metaphase I (MI) stages at the time of ICSI were donated by 67 patients. PARTICIPANTS/MATERIALS, SETTINGS, METHODS The research was conducted in university research laboratories affiliated to a hospital-based infertility clinic. Oocytes were cultured for 24 h and the depletion/appearance of amino acids was measured during the final 6 h of IVM. Amino acid turnover was analysed in relation to oocyte meiotic progression, patient age, disease aetiology and controlled ovarian stimulation regime. MAIN RESULTS AND THE ROLE OF CHANCE The depletion/appearance of key amino acids was linked to the maturation potential of human oocytes in vitro. Oocytes which arrested at the GV stage (n = 9) depleted significantly more valine and isoleucine than those which progressed to MI (n = 32) or MII (n = 107) (P < 0.05). Glutamate, glutamine, arginine and valine depletion or appearance differed in MII versus degenerating oocytes (n = 20) (P < 0.05). Glutamine, arginine, methionine, phenylalanine, total depletion and total turnover all differed in oocytes from patients aged < 35 years versus patients ≥35 years (P < 0.05). MII oocytes obtained following ovarian stimulation with recombinant FSH depleted more isoleucine (P < 0.05) and more alanine and lysine (P < 0.05) appeared than oocytes from hMG-stimulated cycles. MII oocytes from patients with a polycystic ovary (PCO) morphology (n = 33) depleted more serine (P < 0.05) than oocytes from women with normal ovaries (n = 61). LIMITATIONS, REASONS FOR CAUTION Immature oocytes collected at the time of ICSI were used as the model for human oocyte maturation. These oocytes have therefore failed to respond to the ovulatory hCG trigger in vivo (they are meiotically incompetent), and have limited capacity to support embryo development in vitro. The lack of cumulus cells and stress of the conditions in vitro may have influenced turnover of amino acids, and owing to the small sample sizes further studies are required to confirm these findings. WIDER IMPLICATIONS OF THE FINDINGS The findings provide support for the hypothesis that oocyte metabolism reflects oocyte quality. Longitudinal studies are required to link these functional metabolic indices of human oocyte quality with embryo developmental competence. Oocyte amino acid profiling may be a useful tool to quantify the impact of new assisted reproduction technologies (ART) on oocyte quality. STUDY FUNDING/COMPETING INTERESTS This project was funded by the UK Biology and Biotechnology Research Council (BB/C007395/1) and the Medical Research Council (G 0800250). K.E.H was in receipt of a British Fertility Society/Merck Serono studentship. H.J.L. is a shareholder in Novocellus Ltd, a company which seeks to devise a non-invasive biochemical test of embryo health.
Collapse
Affiliation(s)
- K E Hemmings
- Division of Reproduction and Early Development, University of Leeds, Leeds LS2 9JT, UK.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
McRae C, Sharma V, Fisher J. Metabolite Profiling in the Pursuit of Biomarkers for IVF Outcome: The Case for Metabolomics Studies. Int J Reprod Med 2013; 2013:603167. [PMID: 25763388 PMCID: PMC4334075 DOI: 10.1155/2013/603167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/02/2013] [Indexed: 01/24/2023] Open
Abstract
Background. This paper presents the literature on biomarkers of in vitro fertilisation (IVF) outcome, demonstrating the progression of these studies towards metabolite profiling, specifically metabolomics. The need for more, and improved, metabolomics studies in the field of assisted conception is discussed. Methods. Searches were performed on ISI Web of Knowledge SM for literature associated with biomarkers of oocyte and embryo quality, and biomarkers of IVF outcome in embryo culture medium, follicular fluid (FF), and blood plasma in female mammals. Results. Metabolomics in the field of female reproduction is still in its infancy. Metabolomics investigations of embryo culture medium for embryo selection have been the most common, but only within the last five years. Only in 2012 has the first metabolomics investigation of FF for biomarkers of oocyte quality been reported. The only metabolomics studies of human blood plasma in this context have been aimed at identifying women with polycystic ovary syndrome (PCOS). Conclusions. Metabolomics is becoming more established in the field of assisted conception, but the studies performed so far have been preliminary and not all potential applications have yet been explored. With further improved metabolomics studies, the possibility of identifying a method for predicting IVF outcome may become a reality.
Collapse
Affiliation(s)
- C. McRae
- School of Chemistry, University of Leeds, Leeds LS2 9JT, UK
| | - V. Sharma
- The Leeds Centre for Reproductive Medicine, Leeds Teaching Hospitals NHS Trust, Seacroft Hospital, Leeds LS14 6UH, UK
| | - J. Fisher
- School of Chemistry, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
42
|
Kim MK, Park EA, Kim HJ, Choi WY, Cho JH, Lee WS, Cha KY, Kim YS, Lee DR, Yoon TK. Does supplementation of in-vitro culture medium with melatonin improve IVF outcome in PCOS? Reprod Biomed Online 2012. [PMID: 23177415 DOI: 10.1016/j.rbmo.2012.10.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human pre-ovulatory follicular fluid (FF) contains a higher concentration of melatonin than serum. The aim of this study was to evaluate the effect of melatonin supplementation of culture medium on the clinical outcomes of an in-vitro maturation (IVM) IVF-embryo transfer programme for patients with polycystic ovarian syndrome (PCOS). Melatonin concentrations in the culture media of granulosa cells (GC) or cumulus-oocyte-complexes (COC) were measured and the clinical outcomes after using IVM media with or without melatonin were analysed. In the culture media of GC or COC, melatonin concentrations gradually increased. When human chorionic gonadotrophin priming protocols were used, implantation rates in the melatonin-supplemented group were higher than those of the non-supplemented control group (P<0.05). Pregnancy rates were also higher, although not significantly. The findings suggest that the addition of melatonin to IVM media may improve the cytoplasmic maturation of human immature oocytes and subsequent clinical outcomes. It is speculated that follicular melatonin may be released from luteinizing GC during late folliculogenesis and that melatonin supplementation may be used to improve the clinical outcomes of IVM IVF-embryo transfer. Melatonin is primarily produced by the pineal gland and regulates a variety of important central and peripheral actions related to circadian rhythms and reproduction. Interestingly, human pre-ovulatory follicular fluid contains a higher concentration of melatonin than serum. However, in contrast to animal studies, the direct role of melatonin on oocyte maturation in the human system has not yet been investigated. So, the aim of the study was to evaluate the effect of melatonin supplementation of culture medium on the clinical outcome of an in-vitro maturation (IVM) IVF-embryo transfer programme for PCOS patients. The melatonin concentrations in culture medium of granulosa cells (GC) or cumulus-oocyte-complexes (COC) were measured and the clinical outcomes of IVM IVF-embryo transfer using IVM medium alone or supplemented with melatonin were analysed. In the culture media of GC or COC, the melatonin concentration gradually increased. With human chorionic gonadotrophin priming, the pregnancy and implantation rates in the melatonin-supplemented group were higher than those of the non-supplemented control (P<0.05). Our findings suggest that follicular melatonin is released from luteinizing GC during late folliculogenesis and plays a positive role in oocyte maturation. Therefore, addition of melatonin into IVM medium may improve cytoplasmic maturation of human immature oocytes and subsequent clinical outcomes.
Collapse
Affiliation(s)
- Mi Kyoung Kim
- Fertility Center of CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul 135-081, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Escobar-Morreale HF, Samino S, Insenser M, Vinaixa M, Luque-Ramírez M, Lasunción MA, Correig X. Metabolic Heterogeneity in Polycystic Ovary Syndrome Is Determined by Obesity: Plasma Metabolomic Approach Using GC-MS. Clin Chem 2012; 58:999-1009. [DOI: 10.1373/clinchem.2011.176396] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract
BACKGROUND
Abdominal adiposity and obesity influence the association of polycystic ovary syndrome (PCOS) with insulin resistance and diabetes. We aimed to characterize the intermediate metabolism phenotypes associated with PCOS and obesity.
METHODS
We applied a nontargeted GC-MS metabolomic approach to plasma samples from 36 patients with PCOS and 39 control women without androgen excess, matched for age, body mass index, and frequency of obesity.
RESULTS
Patients with PCOS were hyperinsulinemic and insulin resistant compared with the controls. The increase in plasma long-chain fatty acids, such as linoleic and oleic acid, and glycerol in the obese patients with PCOS suggests increased lipolysis, possibly secondary to impaired insulin action at adipose tissue. Conversely, nonobese patients with PCOS showed a metabolic profile consisting of suppression of lipolysis and increased glucose utilization (increased lactic acid concentrations) in peripheral tissues, and PCOS patients as a whole showed decreased 2-ketoisocaproic and alanine concentrations, suggesting utilization of branched-chain amino acids for protein synthesis and not for gluconeogenesis. These metabolic processes required effective insulin signaling; therefore, insulin resistance was not universal in all tissues of these women, and different mechanisms possibly contributed to their hyperinsulinemia. PCOS was also associated with decreased α-tocopherol and cholesterol concentrations irrespective of obesity.
CONCLUSIONS
Substantial metabolic heterogeneity, strongly influenced by obesity, underlies PCOS. The possibility that hyperinsulinemia may occur in the absence of universal insulin resistance in nonobese women with PCOS should be considered when designing diagnostic and therapeutic strategies for the management of this prevalent disorder.
Collapse
Affiliation(s)
- Héctor F Escobar-Morreale
- Diabetes, Obesity and Human Reproduction Research Group, Hospital Universitario Ramón y Cajal & Universidad de Alcalá, Madrid, Spain
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Sara Samino
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
- Metabolomics Platform, Universitat Rovira i Virgili & Institut d'Investigació Sanitaria Pere Virgili, Tarragona, Spain
| | - María Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Hospital Universitario Ramón y Cajal & Universidad de Alcalá, Madrid, Spain
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - María Vinaixa
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
- Metabolomics Platform, Universitat Rovira i Virgili & Institut d'Investigació Sanitaria Pere Virgili, Tarragona, Spain
| | - Manuel Luque-Ramírez
- Diabetes, Obesity and Human Reproduction Research Group, Hospital Universitario Ramón y Cajal & Universidad de Alcalá, Madrid, Spain
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
| | - Miguel A Lasunción
- Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Department of Biochemistry-Research, Hospital Universitario Ramón y Cajal & Universidad de Alcalá, Madrid, Spain, and CIBER Fisiopatología de la Obesidad y la Nutrición, Spain
| | - Xavier Correig
- CIBER Diabetes y Enfermedades Metabólicas Asociadas, Spain
- Metabolomics Platform, Universitat Rovira i Virgili & Institut d'Investigació Sanitaria Pere Virgili, Tarragona, Spain
| |
Collapse
|
44
|
Baird DT, Balen A, Escobar-Morreale HF, Evers JLH, Fauser BCJM, Franks S, Glasier A, Homburg R, La Vecchia C, Devroey P, Diedrich K, Fraser L, Gianaroli L, Liebaers I, Sunde A, Tapanainen JS, Tarlatzis B, Van Steirteghem A, Veiga A, Crosignani PG, Evers JLH. Health and fertility in World Health Organization group 2 anovulatory women. Hum Reprod Update 2012; 18:586-99. [DOI: 10.1093/humupd/dms019] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
45
|
Hemmings KE, Leese HJ, Picton HM. Amino Acid Turnover by Bovine Oocytes Provides an Index of Oocyte Developmental Competence In Vitro1. Biol Reprod 2012; 86:165, 1-12. [DOI: 10.1095/biolreprod.111.092585] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
46
|
Consensus on women's health aspects of polycystic ovary syndrome (PCOS): the Amsterdam ESHRE/ASRM-Sponsored 3rd PCOS Consensus Workshop Group. Fertil Steril 2011; 97:28-38.e25. [PMID: 22153789 DOI: 10.1016/j.fertnstert.2011.09.024] [Citation(s) in RCA: 1034] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/13/2011] [Indexed: 12/11/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in females, with a high prevalence. The etiology of this heterogeneous condition remains obscure, and its phenotype expression varies. Two widely cited previous ESHRE/ASRM sponsored PCOS consensus workshops focused on diagnosis (published in 2004) and infertility management (published in 2008), respectively. The present third PCOS consensus report summarizes current knowledge and identifies knowledge gaps regarding various women's health aspects of PCOS. Relevant topics addressed-all dealt with in a systematic fashion-include adolescence, hirsutism and acne, contraception, menstrual cycle abnormalities, quality of life, ethnicity, pregnancy complications, long-term metabolic and cardiovascular health, and finally cancer risk. Additional, comprehensive background information is provided separately in an extended online publication.
Collapse
|
47
|
Siristatidis CS, Maheshwari A, Bhattacharya S. In vitro maturation in sub fertile women with polycystic ovarian syndrome undergoing assisted reproduction. Cochrane Database Syst Rev 2009:CD006606. [PMID: 19160291 DOI: 10.1002/14651858.cd006606.pub2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) occurs in 4% to 7% of all women of reproductive age and 20% of women presenting with sub-fertility. A significant proportion of these women will ultimately need assisted reproductive techniques (ART). In women with PCOS, the supra-physiological doses of gonadotrophins used for controlled ovarian hyperstimulation (COH) often result in an exaggerated ovarian response characterised by the development of a large cohort of follicles of uneven quality, retrieval of immature oocytes, and increased risk of ovarian hyperstimulation syndrome. A potentially useful intervention for women with PCOS-related infertility involves earlier retrieval of immature oocytes followed by in vitro maturation (IVM). OBJECTIVES To compare live birth rates per woman following in vitro maturation (IVM) with conventional in vitro fertilisation (IVF) or intracytoplasmic sperm injection (ICSI) for women with PCOS undergoing ART. SEARCH STRATEGY We searched the Menstrual Disorders and Subfertility Group Specialised Register of controlled trials for any relevant trials from the title, abstract, or keyword sections. Following a search of electronic databases (MEDLINE in all languages) using Ovid software we also performed a manual search of the references of all retrieved articles, sought unpublished papers and abstracts submitted to international conferences, and contacted experts. In addition, we searched the National Institute of Clinical Excellence fertility assessment and treatment guidelines (NICE 2004) and handsearched reference lists of relevant systematic reviews and randomised trials. SELECTION CRITERIA All randomised trials on the intention to perform IVM before IVF or ICSI and conventional IVF or ICSI for sub-fertile women with PCOS. DATA COLLECTION AND ANALYSIS We identified no studies that met the inclusion criteria. MAIN RESULTS There were no trials suitable for inclusion in the review. AUTHORS' CONCLUSIONS There are no randomised controlled trials upon which to base any practice recommendations regarding IVM before IVF or ICSI for women with PCOS. There is an urgent need for randomised trials in this field.
Collapse
Affiliation(s)
- Charalambos S Siristatidis
- Department of Obstetrics & Gynaecology, University of Aberdeen, Assisted Reproduction Unit, Aberdeen Maternity Hospital, Forest Hill, Aberdeen, UK, AB25 2ZD.
| | | | | |
Collapse
|