1
|
Cakir C, Kuspinar G, Aslan K, Bozyigit C, Kasapoglu I, Dirican M, Uncu G, Avci B. Dehydroepiandrosterone modulates the PTEN/PI3K/AKT signaling pathway to alleviate 4-vinylcyclohexene diepoxide-induced premature ovarian insufficiency in rats. Exp Anim 2024; 73:319-335. [PMID: 38494723 PMCID: PMC11254495 DOI: 10.1538/expanim.23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Dehydroepiandrosterone (DHEA) is frequently integrated as an adjuvant in over a quarter of controlled ovarian hyperstimulation (COH) protocols, despite the ongoing debate regarding its impact. This study aimed to evaluate the efficacy and mechanism of action of DHEA on ovarian follicular development and ovarian response in rats with varying ovarian reserves. The study involved 75 rats categorized into 15 distinct groups. The ovarian tissues of rats in both the normal ovarian reserve group and the premature ovarian insufficiency (POI) group, induced by 4-vinylcyclohexene diepoxide (VCD) injection, were subjected to histomorphological and biochemical analyses following the administration of DHEA, either alone or in combination with COH. Follicle counting was performed on histological sections obtained from various tissues. Serum concentrations of anti-Müllerian hormone (AMH) and the quantification of specific proteins in ovarian tissue, including phosphatase and tensin homolog of chromosome 10 (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (pAKT), cyclooxygenase 2 (COX-2), caspase-3, as well as assessments of total antioxidant status and total oxidant status, were conducted employing the ELISA method. The impact of DHEA exhibited variability based on ovarian reserve. In the POI model, DHEA augmented follicular development and ovarian response to the COH protocol by upregulating the PTEN/PI3K/AKT signaling pathway, mitigating apoptosis, inflammation, and oxidative stress, contrary to its effects in the normal ovarian reserve group. In conclusion, it has been determined that DHEA may exert beneficial effects on ovarian stimulation response by enhancing the initiation of primordial follicles and supporting antral follicle populations.
Collapse
Affiliation(s)
- Cihan Cakir
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Goktan Kuspinar
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Kiper Aslan
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Cengiz Bozyigit
- Department of Medical Biochemistry, Bursa City Hospital, Doğanköy District, Nilüfer Bursa, 16110, Türkiye
| | - Isil Kasapoglu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Melahat Dirican
- Department of Medical Biochemistry, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Gurkan Uncu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Berrin Avci
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| |
Collapse
|
2
|
Naik S, Lepine S, Nagels HE, Siristatidis CS, Kroon B, McDowell S. Androgens (dehydroepiandrosterone or testosterone) for women undergoing assisted reproduction. Cochrane Database Syst Rev 2024; 6:CD009749. [PMID: 38837771 PMCID: PMC11152211 DOI: 10.1002/14651858.cd009749.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
BACKGROUND Practitioners in the field of assisted reproductive technology (ART) continually seek alternative or adjunct treatments to improve ART outcomes. This Cochrane review investigates the adjunct use of synthetic versions of two naturally produced hormones, dehydroepiandrosterone (DHEA) and testosterone (T), in assisted reproduction. Steroid hormones are proposed to increase conception rates by positively affecting follicular response to gonadotrophin stimulation. This may lead to a greater oocyte yield and, subsequently, an increased chance of pregnancy. OBJECTIVES To assess the effectiveness and safety of DHEA and T as pre- or co-treatments in infertile women undergoing assisted reproduction. SEARCH METHODS We searched the following electronic databases up to 8 January 2024: the Gynaecology and Fertility Group (CGF) Specialised Register, CENTRAL, MEDLINE, Embase, PsycINFO, and trial registries for ongoing trials. We also searched citation indexes, Web of Science, PubMed, and OpenGrey. We searched the reference lists of relevant studies and contacted experts in the field for any additional trials. There were no language restrictions. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing DHEA or T as an adjunct treatment to any other active intervention, placebo, or no treatment in women undergoing assisted reproduction. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, extracted relevant data, and assessed risk of bias. We pooled data from studies using fixed-effect models. We calculated odds ratios (ORs) for each dichotomous outcome. Analyses were stratified by type of treatment. We assessed the certainty of evidence for the main findings using GRADE methods. MAIN RESULTS We included 28 RCTs. There were 1533 women in the intervention groups and 1469 in the control groups. Apart from three trials, trial participants were women identified as 'poor responders' to standard in vitro fertilisation (IVF) protocols. The included trials compared either T or DHEA treatment with placebo or no treatment. Pre-treatment with DHEA versus placebo/no treatment: DHEA likely results in little to no difference in live birth/ongoing pregnancy rates (OR 1.30, 95% confidence interval (CI) 0.95 to 1.76; I² = 16%, 9 RCTs, N = 1433, moderate certainty evidence). This suggests that in women with a 12% chance of live birth/ongoing pregnancy with placebo or no treatment, the live birth/ongoing pregnancy rate in women using DHEA will be between 12% and 20%. DHEA likely does not decrease miscarriage rates (OR 0.85, 95% CI 0.53 to 1.37; I² = 0%, 10 RCTs, N =1601, moderate certainty evidence). DHEA likely results in little to no difference in clinical pregnancy rates (OR 1.18, 95% CI 0.93 to 1.49; I² = 0%, 13 RCTs, N = 1886, moderate certainty evidence). This suggests that in women with a 17% chance of clinical pregnancy with placebo or no treatment, the clinical pregnancy rate in women using DHEA will be between 16% and 24%. We are very uncertain about the effect of DHEA on multiple pregnancy (OR 3.05, 95% CI 0.47 to 19.66; 7 RCTs, N = 463, very low certainty evidence). Pre-treatment with T versus placebo/no treatment: T likely improves live birth rates (OR 2.53, 95% CI 1.61 to 3.99; I² = 0%, 8 RCTs, N = 716, moderate certainty evidence). This suggests that in women with a 10% chance of live birth with placebo or no treatment, the live birth rate in women using T will be between 15% and 30%. T likely does not decrease miscarriage rates (OR 1.63, 95% CI 0.76 to 3.51; I² = 0%, 9 RCTs, N = 755, moderate certainty evidence). T likely increases clinical pregnancy rates (OR 2.17, 95% CI 1.54 to 3.06; I² = 0%, 13 RCTs, N = 1152, moderate certainty evidence). This suggests that in women with a 12% chance of clinical pregnancy with placebo or no treatment, the clinical pregnancy rate in women using T will be between 17% and 29%. We are very uncertain about the effect of T on multiple pregnancy (OR 2.56, 95% CI 0.59 to 11.20; 5 RCTs, N = 449, very low certainty evidence). We are uncertain about the effect of T versus estradiol or T versus estradiol + oral contraceptive pills. The certainty of the evidence was moderate to very low, the main limitations being lack of blinding in the included trials, inadequate reporting of study methods, and low event and sample sizes in the trials. Data on adverse events were sparse; any reported events were minor. AUTHORS' CONCLUSIONS Pre-treatment with T likely improves, and pre-treatment with DHEA likely results in little to no difference, in live birth and clinical pregnancy rates in women undergoing IVF who have been identified as poor responders. DHEA and T probably do not decrease miscarriage rates in women under IVF treatment. The effects of DHEA and T on multiple pregnancy are uncertain. Research is needed to identify the optimal duration of treatment with T. Future studies should include data collection on adverse events and multiple pregnancy.
Collapse
Affiliation(s)
- Sandeep Naik
- Obstetrics and Gynaecology, Capital Coast District Heath Board, Wellington, New Zealand
| | - Sam Lepine
- Department of Obstetrics and Gynaecology, Capital and Coast District Health Board, Wellington, New Zealand
| | - Helen E Nagels
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Charalampos S Siristatidis
- Assisted Reproduction Unit, 2nd Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ben Kroon
- Queensland Fertility Group Research Foundation, Brisbane, Australia
- The University of Queensland, Brisbane, Australia
| | - Simon McDowell
- Obstetrics and Gynaecology, Capital Coast District Heath Board, Wellington, New Zealand
| |
Collapse
|
3
|
Aliakbar VH, Tanha FD, Asbagh FA, Ebrahimi M, Shahraki Z. The effect of methyltestosterone on in vitro fertilization outcomes: A randomized clinical trial on patients with low ovarian response. Clin Exp Reprod Med 2024; 51:158-162. [PMID: 38812245 PMCID: PMC11140261 DOI: 10.5653/cerm.2023.05946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 05/31/2024] Open
Abstract
OBJECTIVE The aim of this study was to compare the outcomes of in vitro fertilization (IVF) in patients with a poor ovarian response who used methyltestosterone, versus those using a placebo, in an infertility clinic setting. METHODS This clinical trial included 120 women who had undergone IVF with intracytoplasmic sperm injection due to poor ovarian reserve and infertility. The study took place at the Yas Infertility Center in Tehran, Iran, between January 1, 2018 and January 1, 2019. In the intervention group, 25 mg of methyltestosterone was administered daily for 2 months prior to the initiation of assisted reproductive treatment. The control group was given placebo tablets for the same duration before starting their cycle. Each group was randomly assigned 60 patients. All analyses were performed using SPSS ver. 23 (IBM Corp.). RESULTS The endometrial thickness in the intervention group was 7.57±1.22 mm, whereas in the control group, it was 7.11±1.02 (p=0.028). The gonadotropin number was significantly higher in the control group (64.7±13.48 vs. 57.9±9.25, p=0.001). However, there was no significant difference between the two groups in the antral follicular count. The chemical and clinical pregnancy rates in the intervention group were 18.33% and 15% respectively, compared to 8.33% and 6.67% in the control group. The rate of definitive pregnancy was marginally higher in the intervention group (13.3% vs. 3.3%, p=0.05). CONCLUSION The findings of this study suggest that pretreatment with methyltestosterone significantly increases endometrium thickness and is associated with an increase in the definitive pregnancy rate.
Collapse
Affiliation(s)
- Venus Haj Aliakbar
- Department of Obstetrics and Gynecology, Arash Women’s Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Davari Tanha
- Department of Obstetrics and Gynecology, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Firouzeh Akbari Asbagh
- Department of Obstetrics and Gynecology, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahbod Ebrahimi
- Department of Obstetrics and Gynecology, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shahraki
- Department of Obstetrics and Gynecology, Amir-Al-Momenin Hospital, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
4
|
Boyle P, Andralojc K, van der Velden S, Najmabadi S, de Groot T, Turczynski C, Stanford JB. Restoration of serum estradiol and reduced incidence of miscarriage in patients with low serum estradiol during pregnancy: a retrospective cohort study using a multifactorial protocol including DHEA. FRONTIERS IN REPRODUCTIVE HEALTH 2024; 5:1321284. [PMID: 38239818 PMCID: PMC10794495 DOI: 10.3389/frph.2023.1321284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
Background Low serum estradiol in early pregnancy is associated with an elevated risk of miscarriage. We sought to determine whether efforts to restore low blood estradiol via estradiol or dehydroepiandrosterone (DHEA) supplementation would reduce the risk of miscarriage as part of a multifactorial symptom-based treatment protocol. Methods This retrospective cohort study included women with low serum estradiol levels in early pregnancy, defined as ≤50% of reference levels by gestational age. Estradiol or DHEA were administered orally, and the primary outcome measure was serum estradiol level, in reference to gestational age. The secondary outcome measures included miscarriage, birth weight, and gestational age at birth. Results We found no significant effect of estradiol supplementation on serum estradiol levels referenced to gestational age, while DHEA supplementation strongly increased estradiol levels. For pregnancies with low estradiol, the miscarriage rate in the non-supplemented group was 45.5%, while miscarriage rate in the estradiol and DHEA supplemented groups were 21.2% (p = 0.067) and 17.5% (p = 0.038), respectively. Birth weight, size, gestational age, and preterm deliveries were not significantly different. No sexual abnormalities were reported in children (n = 29) of DHEA-supplemented patients after 5-7 years follow-up. Conclusions In conclusion, DHEA supplementation restored serum estradiol levels, and when included in the treatment protocol, there was a statistically significant reduction in miscarriage.
Collapse
Affiliation(s)
- Phil Boyle
- International Institute for Restorative Reproductive Medicine, London, United Kingdom
- NeoFertility Clinic, Dublin, Ireland
| | - Karolina Andralojc
- NeoFertility Clinic, Dublin, Ireland
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Shahpar Najmabadi
- Office of Cooperative Reproductive Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, United States
| | - Theun de Groot
- NeoFertility Clinic, Dublin, Ireland
- Department of Medical Microbiology and Infectious Diseases, Canisius Wilhelmina Hospital, Nijmegen, Netherlands
| | - Craig Turczynski
- NeoFertility Clinic, Dublin, Ireland
- Billings Ovulation Method Association-USA, Saint Cloud, MN, United States
| | - Joseph B. Stanford
- International Institute for Restorative Reproductive Medicine, London, United Kingdom
- Office of Cooperative Reproductive Health, Department of Family and Preventive Medicine, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
5
|
La Marca A, Longo M, Sighinolfi G, Grisendi V, Imbrogno MG, Giulini S. New insights into the role of LH in early ovarian follicular growth: a possible tool to optimize follicular recruitment. Reprod Biomed Online 2023; 47:103369. [PMID: 37918055 DOI: 10.1016/j.rbmo.2023.103369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 11/04/2023]
Abstract
Evidence shows that LH participates in enhancing transition from the early stage to the antral stage of folliculogenesis. It has been demonstrated that functional LH receptors are expressed, albeit at a very low level and even in smaller follicles, during the phase that was traditionally considered to be gonadotrophin independent, suggesting a role for LH in accelerating the rate of progression of non-growing and primary follicles to the preantral/antral stage. Hypogonadotropic hypogonadism, together with other clinical conditions of pituitary suppression, has been associated with reduced functional ovarian reserve. The reduction in LH serum concentration is associated with a low concentration of anti-Müllerian hormone. This is the case in hypothalamic amenorrhoea, pregnancy, long-term GnRH-analogue therapy and hormonal contraception. The effect seems to be reversible, such that after pregnancy and after discontinuation of drugs, the functional ovarian reserve returns to the baseline level. Evidence suggests that women with similar primordial follicle reserves could present with different numbers of antral follicles, and that gonadotrophins may play a fundamental role in permitting a normal rate of progression of follicles through non-cyclic folliculogenesis. The precise role of gonadotrophins in early folliculogenesis, as well as their use to modify the functional ovarian reserve, must be investigated.
Collapse
Affiliation(s)
- Antonio La Marca
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy.
| | - Maria Longo
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy
| | - Giovanna Sighinolfi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy
| | - Valentina Grisendi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy
| | - Maria Giovanna Imbrogno
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy
| | - Simone Giulini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Policlinico di Modena, via del Pozzo, 41124 Modena, Italy
| |
Collapse
|
6
|
Glachant S, Salle B, Langlois-Jacques C, Labrune E, Renault L, Roy P, Benchaib M, Fraison E. [Predictive factors of spontaneous pregnancies among women with diminished ovarian reserve patients treated with DHEA]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2023; 51:400-407. [PMID: 37331511 DOI: 10.1016/j.gofs.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/17/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
INTRODUCTION Diminished ovarian reserve remains a challenge in the reproductive medicine field. Treatment options for these patients are limited and there is no consensus to make any recommendations. Regarding adjuvant supplements, DHEA could play a role in follicular recruitment and, therefore, may increase spontaneous pregnancy rate. MATERIALS AND METHODS This study was a monocentric historical and observational cohort study carried out in the reproductive medicine department at the University Hospital, Femme-Mère-Enfant in Lyon. All women presenting with a diminished ovarian reserve treated with 75mg/day of DHEA were consecutively included. The main objective was to evaluate the spontaneous pregnancy rate. The secondary objectives were to identify predictive factors for pregnancy and the evaluation of treatment side effects. RESULTS Four hundred and thirty-nine women were included. In all, 277 were analyzed, 59 had a spontaneous pregnancy (21.3%). The probability of being pregnant was respectively 13.2% (IC95 9-17.2%), 21.3% (IC95 15.1-27%) and 38.8% (IC95 29.3-48.4%) at 6, 12 and 24 months. Only 20.6% of patients complained of side effects. CONCLUSION DHEA may improve spontaneous pregnancies in women with diminished ovarian reserve without any stimulation.
Collapse
Affiliation(s)
- S Glachant
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Laennec, université Claude-Bernard, 7, rue Guillaume-Paradin, Lyon, France
| | - B Salle
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Lyon Sud, université Claude-Bernard, 165, chemin du Petit-Revoyet, Oullins, France; Inserm unité 1208, 18, avenue Doyen-Lépine, Bron, France
| | - C Langlois-Jacques
- Service de biostatistique et bioinformatique, hospices civils de Lyon, 69003 Lyon, France; CNRS, UMR 5558, laboratoire de biométrie et biologie évolutive, équipe biostatistique-santé, 69100 Villeurbanne, France
| | - E Labrune
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Laennec, université Claude-Bernard, 7, rue Guillaume-Paradin, Lyon, France; Inserm unité 1208, 18, avenue Doyen-Lépine, Bron, France
| | - L Renault
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Laennec, université Claude-Bernard, 7, rue Guillaume-Paradin, Lyon, France; Inserm unité 1208, 18, avenue Doyen-Lépine, Bron, France
| | - P Roy
- Service de biostatistique et bioinformatique, hospices civils de Lyon, 69003 Lyon, France; CNRS, UMR 5558, laboratoire de biométrie et biologie évolutive, équipe biostatistique-santé, 69100 Villeurbanne, France
| | - M Benchaib
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Laennec, université Claude-Bernard, 7, rue Guillaume-Paradin, Lyon, France; Inserm unité 1208, 18, avenue Doyen-Lépine, Bron, France
| | - E Fraison
- Service de médecine de la reproduction, hospices civils de Lyon, hôpital Mère-Enfant, 59, boulevard Pinel, Bron, France; Faculté de médecine Laennec, université Claude-Bernard, 7, rue Guillaume-Paradin, Lyon, France.
| |
Collapse
|
7
|
Zhang J, Jia H, Diao F, Ma X, Liu J, Cui Y. Efficacy of dehydroepiandrosterone priming in women with poor ovarian response undergoing IVF/ICSI: a meta-analysis. Front Endocrinol (Lausanne) 2023; 14:1156280. [PMID: 37361534 PMCID: PMC10288189 DOI: 10.3389/fendo.2023.1156280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Background Dehydroepiandrosterone (DHEA) may improve the outcomes of patients with poor ovarian response (POR) or diminished ovarian reserve (DOR) undergoing IVF/ICSI. However, the evidence remains inconsistent. This study aimed to investigate the efficacy of DHEA supplementation in patients with POR/DOR undergoing IVF/ICSI. Methods PubMed, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI) were searched up to October 2022. Results A total of 32 studies were retrieved, including 14 RCTs, 11 self-controlled studies and 7 case-controlled studies. In the subgroup analysis of only RCTs, DHEA treatment significantly increased the number of antral follicle count (AFC) (weighted mean difference : WMD 1.18, 95% confidence interval(CI): 0.17 to 2.19, P=0.022), while reduced the level of bFSH (WMD -1.99, 95% CI: -2.52 to -1.46, P<0.001), the need of gonadotropin (Gn) doses (WMD -382.29, 95% CI: -644.82 to -119.76, P=0.004), the days of stimulation (WMD -0.90, 95% CI: -1.34 to -0.47, P <0.001) and miscarriage rate (relative risk : RR 0.46, 95% CI: 0.29 to 0.73, P=0.001). The higher clinical pregnancy and live birth rates were found in the analysis of non-RCTs. However, there were no significant differences in the number of retrieved oocytes, the number of transferred embryos, and the clinical pregnancy and live birth rates in the subgroup analysis of only RCTs. Moreover, meta-regression analyses showed that women with lower basal FSH had more increase in serum FSH levels (b=-0.94, 95% CI: -1.62 to -0.25, P=0.014), and women with higher baseline AMH levels had more increase in serum AMH levels (b=-0.60, 95% CI: -1.15 to -0.06, P=0.035) after DHEA supplementation. In addition, the number of retrieved oocytes was higher in the studies on relatively younger women (b=-0.21, 95% CI: -0.39 to -0.03, P=0.023) and small sample sizes (b=-0.003, 95% CI: -0.006 to -0.0003, P=0.032). Conclusions DHEA treatment didn't significantly improve the live birth rate of women with DOR or POR undergoing IVF/ICSI in the subgroup analysis of only RCTs. The higher clinical pregnancy and live birth rates in those non-RCTs should be interpreted with caution because of potential bias. Further studies using more explicit criteria to subjects are needed. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD 42022384393.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongyan Jia
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Clinical Centre of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital Kangda College of Nanjing Medical University, Lianyungang, China
| | - Feiyang Diao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Ma
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Viardot-Foucault V, Zhou J, Bi D, Takinami Y, Chan JKY, Lee YH. Dehydroepiandrosterone supplementation and the impact of follicular fluid metabolome and cytokinome profiles in poor ovarian responders. J Ovarian Res 2023; 16:107. [PMID: 37268990 PMCID: PMC10239139 DOI: 10.1186/s13048-023-01166-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/25/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Poor ovarian responders (POR) are women undergoing in-vitro fertilization who respond poorly to ovarian stimulation, resulting in the retrieval of lower number of oocytes, and subsequently lower pregnancy rates. The follicular fluid (FF) provides a crucial microenvironment for the proper development of follicles and oocytes through tightly controlled metabolism and cell signaling. Androgens such as dehydroepiandrosterone (DHEA) have been proposed to alter the POR follicular microenvironment, but the impact DHEA imposes on the FF metabolome and cytokine profiles is unknown. Therefore, the objective of this study is to profile and identify metabolomic changes in the FF with DHEA supplementation in POR patients. METHODS FF samples collected from 52 POR patients who underwent IVF with DHEA supplementation (DHEA +) and without (DHEA-; controls) were analyzed using untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics and a large-scale multiplex suspension immunoassay covering 65 cytokines, chemokines and growth factors. Multivariate statistical modelling by partial least squares-discriminant regression (PLSR) analysis was performed for revealing metabolome-scale differences. Further, differential metabolite analysis between the two groups was performed by PLSR β-coefficient regression analysis and Student's t-test. RESULTS Untargeted metabolomics identified 118 FF metabolites of diverse chemistries and concentrations which spanned three orders of magnitude. They include metabolic products highly associated with ovarian function - amino acids for regulating pH and osmolarity, lipids such fatty acids and cholesterols for oocyte maturation, and glucocorticoids for ovarian steroidogenesis. Four metabolites, namely, glycerophosphocholine, linoleic acid, progesterone, and valine were significantly lower in DHEA + relative to DHEA- (p < 0.05-0.005). The area under the curves of progesterone glycerophosphocholine, linoleic acid and valine are 0.711, 0.730, 0.785 and 0.818 (p < 0.05-0.01). In DHEA + patients, progesterone positively correlated with IGF-1 (Pearson r: 0.6757, p < 0.01); glycerophosphocholine negatively correlated with AMH (Pearson r: -0.5815; p < 0.05); linoleic acid correlated with estradiol and IGF-1 (Pearson r: 0.7016 and 0.8203, respectively; p < 0.01 for both). In DHEA- patients, valine negatively correlated with serum-free testosterone (Pearson r: -0.8774; p < 0.0001). Using the large-scale immunoassay of 45 cytokines, we observed significantly lower MCP1, IFNγ, LIF and VEGF-D levels in DHEA + relative to DHEA. CONCLUSIONS In POR patients, DHEA supplementation altered the FF metabolome and cytokine profile. The identified four FF metabolites that significantly changed with DHEA may provide information for titrating and monitoring individual DHEA supplementation.
Collapse
Affiliation(s)
- Veronique Viardot-Foucault
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
| | - Jieliang Zhou
- Translational ‘Omics and Biomarkers Group, KK Research Centre, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
| | - Dexi Bi
- Department of Pathology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yoshihiko Takinami
- Bruker Japan, 3-9 Yokohama City, Kanagawa, 220-0022 Japan
- Present Address: Kanomax Analytical Incorportated, Shimizu Suita City, Osaka Japan
| | - Jerry. K. Y. Chan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
- Obstetrics and Gynaecology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
| | - Yie Hou Lee
- Translational ‘Omics and Biomarkers Group, KK Research Centre, KK Women’s and Children’s Hospital, 100 Bukit Timah Road, Singapore, 229899 Singapore
- Obstetrics and Gynaecology Academic Clinical Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857 Singapore
- Singapore-MIT Alliance for Research and Technoology, 1 CREATE Way, Singapore, 138602 Singapore
| |
Collapse
|
9
|
Tao S, Yang M, Pan B, Wang Y, Tian F, Han D, Shao W, Yang W, Xie Y, Fang X, Xia M, Hu J, Kan H, Li W, Xu Y. Maternal exposure to ambient PM 2.5 perturbs the metabolic homeostasis of maternal serum and placenta in mice. ENVIRONMENTAL RESEARCH 2023; 216:114648. [PMID: 36341790 DOI: 10.1016/j.envres.2022.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/02/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Epidemiological and animal studies have shown that maternal fine particulate matters (PM2.5) exposure correlates with various adverse pregnancy outcomes such as low birth weight (LBW) of offspring. However, the underlying biological mechanisms have not been fully understood. In this study, female C57Bl/6 J mice were exposed to filtered air (FA) or concentrated ambient PM2.5 (CAP) during pregestational and gestational periods, and metabolomics was performed to analyze the metabolic features in maternal serum and placenta by liquid chromatography-mass spectrometry (LC-MS). The partial least squares discriminate analysis (PLS-DA) displayed evident clustering of FA- and CAP-exposed samples for both maternal serum and placenta. In addition, pathway analysis identified that vitamin digestion and absorption was perturbed in maternal serum, while metabolic pathways including arachidonic acid metabolism, serotonergic synapse, 2-oxocarboxylic acid metabolism and cAMP signaling pathway were perturbed in placenta. Further analysis indicated that CAP exposure influenced the nutrient transportation capacity of placenta, by not only changing the ratios of some critical metabolites in placenta to maternal serum but also significantly altering the expressions of nutrition transporters in placenta. These findings reaffirm the importance of protecting women from PM2.5 exposure, and also advance our understanding of the toxic actions of ambient PM2.5.
Collapse
Affiliation(s)
- Shimin Tao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China; NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Mingjun Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Bin Pan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China; NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Yuzhu Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Fang Tian
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Dongyang Han
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Wenpu Shao
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Wenhui Yang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Yuanting Xie
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Xinyi Fang
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Minjie Xia
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Jingying Hu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Haidong Kan
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Weihua Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), School of Pharmacy, Fudan University, Shanghai, 200032, China.
| | - Yanyi Xu
- Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Jiang Y, Cheng Y, Xia M, Zhang B, Ding Q, Lu L, Wang JR, Mei X. Dehydroepiandrosterone Cocrystals with Improved Solubility and Bioavailability. Pharmaceutics 2022; 14:pharmaceutics14112478. [PMID: 36432669 PMCID: PMC9699216 DOI: 10.3390/pharmaceutics14112478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Dehydroepiandrosterone (DHEA) is an FDA-approved food supplement used as an assisted reproductive sex hormone. The bioavailability is severely limited by its poor solubility (23 µg/mL). Herein, we aimed to modulate its solubility through cocrystallization. Eight cocrystals of DHEA with pyrocatechol (CAT), hydroquinone (HQ), resorcinol (RES), phloroglucinol (PG), 1,5-dihydroxy naphthalene (DHN), p-hydroxybenzoic acid (PHBA), gallic acid (GA), and 5-hydroxyisophthalic acid (5HIPA) were designed and synthesized. Some basic characterization tools, including powder X-ray diffraction, thermogravimetric analysis, differential scanning calorimetry, and Fourier transform infrared spectroscopy, were also applied in our work for basic analyses of cocrystals. It is indicated that DHEA-GA exhibits its superiority in dissolution and pharmacokinetic behaviors. While the area under the curve values of DHEA-GA is improved at the ratio of 2.2, the corresponding bioavailability of DHEA is expected to be accordingly increased.
Collapse
Affiliation(s)
- Yihua Jiang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yinxiang Cheng
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Mengyuan Xia
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Bingrui Zhang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Qiaoce Ding
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liye Lu
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian-Rong Wang
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Correspondence: (J.-R.W.); (X.M.)
| | - Xuefeng Mei
- Pharmaceutical Analytical & Solid-State Chemistry Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Correspondence: (J.-R.W.); (X.M.)
| |
Collapse
|
11
|
Lv M, Yu J, Huang Y, Ma J, Xiang J, Wang Y, Li L, Zhang Z, Liao H. Androgen Signaling in Uterine Diseases: New Insights and New Targets. Biomolecules 2022; 12:1624. [PMID: 36358974 PMCID: PMC9687413 DOI: 10.3390/biom12111624] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 12/06/2023] Open
Abstract
Common uterine diseases include endometriosis, uterine fibroids, endometrial polyps, endometrial hyperplasia, endometrial cancer, and endometrial dysfunction causing infertility. Patients with uterine diseases often suffer from abdominal pain, menorrhagia, infertility and other symptoms, which seriously impair their health and disturb their lives. Androgens play important roles in the normal physiological functions of the uterus and pathological progress of uterine diseases. Androgens in women are synthesized in the ovaries and adrenal glands. The action of androgens in the uterus is mainly mediated by its ligand androgen receptor (AR) that regulates transcription of the target genes. However, much less is known about the signaling pathways through which androgen functions in uterine diseases, and contradictory findings have been reported. This review summarizes and discusses the progress of research on androgens and the involvement of AR in uterine diseases. Future studies should focus on developing new therapeutic strategies that precisely target specific AR and their related signaling pathways in uterine diseases.
Collapse
Affiliation(s)
- Mu Lv
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Juanjuan Yu
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yan Huang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Jie Ma
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jun Xiang
- Department of Urology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yanqiu Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Linxia Li
- Department of Obstetrics and Gynecology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Shanghai 200137, China
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Hong Liao
- Department of Clinical Laboratory Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| |
Collapse
|
12
|
La Marca A, Longo M. Extended LH administration as a strategy to increase the pool of recruitable antral follicles in hypothalamic amenorrhea: evidence from a case series. Hum Reprod 2022; 37:2655-2661. [DOI: 10.1093/humrep/deac195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/31/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
New evidence is indicating a growing role of LH in promoting ovarian follicular growth and maturation, even at the early stages. LH seems to enhance the transition of follicles to the antral stage hence leading to an increase in the so-called functional ovarian reserve (recruitable antral follicles). Hypogonadotropic hypogonadism is characterized by low, and sometimes undetectable, serum LH and FSH levels, which may indeed explain the low anti-Müllerian hormone (AMH) levels and antral follicle count (AFC) found in patients affected by this condition. We report here the cases of two young women affected by hypothalamic amenorrhea (HA) that presented for fertility treatment with very low functional ovarian reserve. The two patients were treated with exogenous LH for 1 and 2 months (extended LH administration: ELHA) at the dose of 187.5 IU LH every day and 150 IU LH every other day, respectively. In both the cases there was an increase in serum AMH levels and in the AFC. In one patient, the AMH and AFC increased from a baseline 1.3 ng/ml and 8 to 2.3 ng/ml and 14 at end of treatment, respectively. In the second case, serum AMH and AFC increased from 0.4 ng/ml and 6 to 1.6 ng/ml and 13, respectively. One patient underwent ovarian stimulation before and after ELHA, showing an increase in the number of mature oocytes recruited (3 versus 8 metaphase II (MII) oocytes before and after, respectively). The second patient underwent an IVF cycle after ELHA resulting in the retrieval of six MII oocytes and an ongoing pregnancy following transfer of a single blastocyst. Women with HA are characterized by chronic, low levels of gonadotrophins, which may impact not only on the cyclic recruitment of follicles but also the progression of small growing follicles through the first stages of folliculogenesis. Some women with HA may in fact show very low serum AMH and AFC. Our case series shows that the administration of LH at a dose of at least 150–187.5 IU every day or every other day may contribute to a clinically evident increase in the functional ovarian reserve (AFC), and probably accounts for a positive effect of LH on the progression of follicles throughout the early stages of folliculogenesis.
Collapse
Affiliation(s)
- Antonio La Marca
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia , Modena, Italy
- Clinica Eugin Modena , Modena, Italy
| | - Maria Longo
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia , Modena, Italy
| |
Collapse
|
13
|
Garzia E, Galiano V, Guarnaccia L, Marfia G, Murru G, Guermandi E, Riparini J, Sulpizio P, Marconi AM. Basal serum level of Δ4-androstenedione reflects the ovaries' ability to respond to stimulation in IVF cycles: setting up a new reliable index of both ovarian reserve and response. J Assist Reprod Genet 2022; 39:1917-1926. [PMID: 35759063 PMCID: PMC9428103 DOI: 10.1007/s10815-022-02546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/09/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Adequate androgen levels are necessary for regular follicular growth, progression beyond the pre-antral stage, and prevention of follicular atresia. The main purpose of this study was to investigate whether baseline androgen levels had a predictive value on stimulation outcomes in IVF cycles. The secondary purpose was to compare the possible predictive value of androgens with that of already known markers. METHODS The study included 91 infertile patients aged 30-45 years awaiting the first IVF cycle. All women underwent the same stimulation protocol and the same starting dose of recombinant FSH. As stimulation outcomes, the number of follicles recruited, estradiol and progesterone levels on the day of trigger, the total dose of gonadotropins administered, and the number of oocytes collected were recorded. Multiple linear regression and multivariate logistic regression were used to evaluate the significant predictive value of the variables for response to controlled ovarian stimulation (COS). By studying the reliability of different markers, an attempt was made to develop a single index with the highest predictive value. RESULTS Pearson's correlation revealed a statistically significant inverse correlation between oocytes collected and age (r = - 0.333, p < 0.001) and a positive correlation with AMH (anti-müllerian hormone) (r = 0.360, p < 0.001), antral follicle count (AFC) (r = 0.639, p < 0.001), and androstenedione (Δ4-A) (r = 0.359, p < 0.001). No significant correlation was reported with FSH (r = - 0.133, p = 0.207) and total testosterone (r = 0.180, p = 0.088). In COS good responders, the G-index (= AMH ng/mL*AFC/Δ4-A ng/dL) revealed a significantly higher level (p < 0.001) than AMH, AFC, and Δ4-A alone. CONCLUSION Baseline serum Δ4-A, presumably crucial for ensuring a regular follicular growth, is a reliable marker of ovarian response to stimulation. Since the ovarian capacity to respond to gonadotropins does not depend exclusively on the presence of follicles, we suggest a new index, the G-index, able to contemplate both the ovarian reserve and the Δ4-A level.
Collapse
Affiliation(s)
- Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
| | - Valentina Galiano
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
- Gynecology and Obstetrics Unit, San Carlo Borromeo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giovanni Marfia
- Istituto Di Medicina Aerospaziale “A. Mosso,” Aeronautica Militare, Milan, Italy
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Aldo Ravelli Research Center, Milan, Italy
| | - Giulia Murru
- Gynecology and Obstetrics Unit, ASST Rhodense, Ospedale G. Salvini, Garbagnate Milanese, Italy
| | - Ellade Guermandi
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Jennifer Riparini
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Patrizia Sulpizio
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 via di Rudinì, 8, Milan, Italy
| | - Anna Maria Marconi
- Gynecology and Obstetrics Unit, Department of Mother and Child, Department of Health Sciences, San Paolo Hospital Medical School, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Barad DH, Albertini DF, Molinari E, Gleicher N. Preliminary report of intraovarian injections of autologous platelet-rich plasma (PRP) in extremely poor prognosis patients with only oocyte donation as alternative: a prospective cohort study. Hum Reprod Open 2022; 2022:hoac027. [PMID: 35795849 PMCID: PMC9247703 DOI: 10.1093/hropen/hoac027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/29/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
STUDY QUESTION
Does intraovarian injection of platelet-rich plasma (PRP) change ovarian function in patients with extremely low functional ovarian reserve (LFOR) who, otherwise, would likely only have a chance of pregnancy through third-party oocyte donation?
SUMMARY ANSWER
No clinically significant effects of PRP treatment on ovarian function were observed over 1 year of follow-up.
WHAT IS KNOWN ALREADY
Several investigators have reported improved responses to ovulation induction after treatment with PRP. However, previous published reports have involved, at most, only small case series. Whether PRP actually improves ovarian performance is, therefore, still unknown. PRP is nevertheless widely offered as an ‘established’ fertility treatment, often under the term ‘ovarian rejuvenation’.
STUDY DESIGN, SIZE, DURATION
We are reporting a prospective cohort study of 80 consecutive patients at ages 28–54 with LFOR, defined by anti-Müllerian hormone <1.1 ng/ml, FSH >12 mIU/ml or at least one prior IVF cycle with ≤3 oocytes within 1 year. The women were followed for 1 year after an intraovarian PRP procedure.
PARTICIPANTS/MATERIALS, SETTING, METHODS
PRP (1.5 ml) was injected into the cortex of ovaries with an average of 12 injections per ovary. Study participants were followed every 3 days for 2 weeks after PRP treatment with estradiol and FSH measurements and vaginal ultrasound to observe follicle growth and thereafter followed weekly. Beginning 1 month after their PRP treatment, participants underwent one or more cycles of ovarian stimulation for IVF. Outcome measures were endocrine response, and numbers of oocytes and embryos produced in response to a maximal gonadotropin stimulation before and after PRP treatment.
MAIN RESULTS AND THE ROLE OF CHANCE
In this study, women failed to demonstrate statistically significant outcome benefits from intraovarian PRP. However, two 40-year-old very poor-prognosis patients, with prior failed IVF cycles that never reached embryo transfer at other centers, achieved pregnancy, resulting in an ongoing pregnancy rate of 4.7% among patients who, following PRP, produced at least one oocyte (n = 42).
LIMITATIONS, REASONS FOR CAUTION
As an observational study of patients who performed poorly in past ovarian stimulation cycles, the improvement may be accounted for by regression to the mean. Similar considerations may also explain the occurrence of the two pregnancies.
WIDER IMPLICATIONS OF THE FINDINGS
This study demonstrates that, even in extremely poor prognosis patients due to LFOR, sporadic pregnancies are possible. The study, however, does not allow for the conclusion that those pregnancies were the consequence of PRP treatments. A case series, indeed, does not allow for such conclusions, even if results are more suggestive than here. This registered study, therefore, must be viewed as a preliminary report, with further data expected from this study but also from two other prospectively randomized ongoing registered studies with more controlled patient selection.
STUDY FUNDING/COMPETING INTEREST(S)
This work was supported by intramural funds from The Center for Human Reproduction and the not-for-profit research Foundation for Reproductive Medicine, both in New York, NY, USA. N.G. and D.H.B. are listed as co-inventors on several US patents. Some of these patents relate to pre-supplementation of hypo-androgenic infertile women with androgens, such as dehydroepiandrosterone and testosterone and, therefore, at least peripherally relate to the subject of this manuscript. They, as well as D.F.A., have also received research support, travel funds and speaker honoraria from several pharmaceutical and medical device companies, though none related to the here presented subject and manuscript. N.G. is a shareholder in Fertility Nutraceuticals and he and D.H.B. receive royalty payments from Fertility Nutraceuticals LLC. E.M. has no conflicts of interest to declare.
TRIAL REGISTRATION NUMBER
NCT04275700
Collapse
Affiliation(s)
- D H Barad
- The Center for Human Reproduction , New York, NY, USA
- The Foundation for Reproductive Medicine , New York, NY, USA
| | - D F Albertini
- The Center for Human Reproduction , New York, NY, USA
- Department of Developmental Cell Biology, Bedford Research Foundation , Bedford, MA, USA
| | - E Molinari
- The Center for Human Reproduction , New York, NY, USA
| | - N Gleicher
- The Center for Human Reproduction , New York, NY, USA
- The Foundation for Reproductive Medicine , New York, NY, USA
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University , New York, NY, USA
- Department of Obstetrics and Gynecology, Medical University of Vienna , Vienna, Austria
| |
Collapse
|
15
|
Yin WW, Huang CC, Chen YR, Yu DQ, Jin M, Feng C. The effect of medication on serum anti-müllerian hormone (AMH) levels in women of reproductive age: a meta-analysis. BMC Endocr Disord 2022; 22:158. [PMID: 35698127 PMCID: PMC9195431 DOI: 10.1186/s12902-022-01065-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE The study aims to address whether serum anti-müllerian hormone (AMH) levels fluctuate in the short term after medication application, including oral contraceptives (OCs), metformin (MET), Gonadotropin-releasing hormone agonist (GnRH-a), dehydroepiandrosterone (DHEA), vitamin D (VD), clomiphene citrate (CC), and letrozole (LET). METHODS Published literature from PubMed, Embase, and Cochrane central was retrieved up until 19 September 2021. A total of 51 self-control studies with an average Newcastle-Ottawa quality assessment scale (NOS) score of 6.90 were analyzed. The extracted data were entered into Stata software, and the weighted mean difference/standardized mean difference (WMD/SMD) and 95% confidence interval (CI) were used for data analysis. RESULTS After OCs treatment the AMH level showed a significant decline in women with normal ovarian function, which was significant within 3 months (WMD = -1.43, 95% CI: -2.05 to -0.80, P < 0.00001). After MET treatment, the serum AMH decreased in polycystic ovary syndrome (PCOS) patients (WMD = -1.79, 95% CI: -2.32 to -1.26, P < 0.00001), in both obese and non-obese patients. GnRH-a treatment in endometriosis patients led to dynamic changes in the serum AMH levels, that is, ascent at 1 month (P = 0.05), and descent at 3 months (P = 0.02). After DHEA treatment the serum AMH increased in diminished ovarian reserve (DOR) / poor ovarian response (POR) patients (WMD = 0.18, 95% CI: 0.09 to 0.27, P < 0.0001). After VD treatment the serum AMH increased, and it was obvious in non-PCOS patients (WMD = 0.78, 95% CI: 0.34 to 1.21, P = 0.0004). After CC treatment the serum AMH decreased significantly in PCOS patients, specifically in non-obese patients (WMD = -1.24, 95% CI: -1.87 to -0.61, P = 0.0001). CONCLUSIONS Serum AMH levels may be affected in the short term after drug application. Specifically, OC, MET and CC lead to decreased AMH level, DHEA and VD lead to increased AMH level, and GnRH-a leads to dynamic variation, which is correlated with PCOS, obesity, age, and duration of medication. The impacts of these medications should be taken into consideration when AMH is used as a marker of ovarian reserve.
Collapse
Affiliation(s)
- Wei-Wei Yin
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Chang-Chang Huang
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Yi-Ru Chen
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Dan-Qing Yu
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Min Jin
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China
| | - Chun Feng
- Department of Reproductive Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
16
|
Bennett G, Cussen L, O'Reilly MW. The role for long-term use of dehydroepiandrosterone in adrenal insufficiency. Curr Opin Endocrinol Diabetes Obes 2022; 29:284-293. [PMID: 35621180 DOI: 10.1097/med.0000000000000728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Dehydroepiandrosterone (DHEA) is an androgen produced by the zona reticularis of the adrenal gland. Patients with adrenal insufficiency will have a deficiency of DHEA. Unlike glucocorticoid and mineralocorticoid replacement, DHEA supplementation is not considered essential for life and is therefore not routinely replaced in adrenal failure. DHEA deficiency is associated with morbidity, including adverse impacts on metabolic function, quality of life and sexuality in multiple studies. The role for replacement, however, remains unclear. RECENT FINDINGS The benefits of DHEA supplementation have been definitively demonstrated in a number of historical studies of patients with primary and secondary adrenal insufficiency. Beneficial impacts on quality of life, body composition, bone health and metabolic markers have been demonstrated. However, published data are inconsistent; controversies persist around the exact role of DHEA replacement and around which patient cohorts are most likely to benefit. There is also a paucity of recent randomized controlled trials in the medical literature to inform on optimal dose and duration of DHEA replacement in adrenal failure. SUMMARY Here, we review the evidence for DHEA supplementation in patients with adrenal insufficiency. We highlight knowledge gaps in the medical literature and areas that should be prioritized for future research endeavours.
Collapse
Affiliation(s)
| | - Leanne Cussen
- Department of Endocrinology, Beaumont Hospital
- Department of Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Republic of Ireland
| | - Michael W O'Reilly
- Department of Endocrinology, Beaumont Hospital
- Department of Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, Republic of Ireland
| |
Collapse
|
17
|
Chansel-Debordeaux L, Rault E, Depuydt C, Soula V, Hocké C, Jimenez C, Creux H, Papaxanthos-Roche A. Successful live birth after in vitro maturation treatment in a patient with autoimmune premature ovarian failure: a case report and review of the literature. Gynecol Endocrinol 2021; 37:1138-1142. [PMID: 34008458 DOI: 10.1080/09513590.2021.1928065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE We report a successful live birth after oocytes in vitro maturation (IVM) and fresh embryo transfer in a patient with autoimmune premature ovarian failure (POF) and performed a review of the literature of livebirths obtained after oocytes IVM treatment in this indication. METHODS The patient was a 24-year-old woman with autoimmune POF diagnosed post-partum, who developed autoimmune polyglandular syndrome with serum anti-ovarian and anti-21-hydroxylase antibodies. The patient had typical symptoms of POF: secondary amenorrhea with hypoestrogenism, elevated gonadotropins and infertility; however, the serum anti-Müllerian hormone level and total antral follicle count remained normal. IVM of immature oocytes was performed after the administration of 150 IU highly purified human menopausal gonadotropin for three consecutive days and an injection of 10,000 IU human chorionic gonadotropin to trigger ovulation. RESULTS The six oocyte-cumulus complexes collected matured in vitro. After intracytoplasmic sperm injection (ICSI), five embryos were obtained. Pregnancy was achieved after the fresh transfer of two embryos and appropriate endometrial preparation. A normal female child was delivered following a 37-weeks pregnancy characterized by the onset of adrenal insufficiency and unstable diabetes. CONCLUSIONS We report a successful livebirth after IVM treatment in a patient with autoimmune premature ovarian failure (POF). Management of reproductive age women with autoimmune pathology requires fertility counseling. Early diagnosis of autoimmune POF is important for early conception and oocyte preservation, because the only other option at present is ovum donation.
Collapse
Affiliation(s)
| | - Elisabeth Rault
- Service de Gynécologie Chirurgicale et Médecine de la Reproduction, Centre Aliénor d'Aquitaine, Bordeaux, France
| | - Chloé Depuydt
- Service de Biologie De la Reproduction-CECOS, Centre Aliénor d'Aquitaine, Bordeaux, France
| | - Volcy Soula
- Service de Biologie De la Reproduction-CECOS, Centre Aliénor d'Aquitaine, Bordeaux, France
| | - Claude Hocké
- Service de Gynécologie Chirurgicale et Médecine de la Reproduction, Centre Aliénor d'Aquitaine, Bordeaux, France
| | - Clément Jimenez
- Service de Biologie De la Reproduction-CECOS, Centre Aliénor d'Aquitaine, Bordeaux, France
| | - Hélène Creux
- Service de Gynécologie Chirurgicale et Médecine de la Reproduction, Centre Aliénor d'Aquitaine, Bordeaux, France
- Centre D'assistance Médicale à la Procréation, Polyclinique Saint-Roch, Montpellier, France
| | | |
Collapse
|
18
|
The Use of Androgen Priming in Women with Reduced Ovarian Reserve Undergoing Assisted Reproductive Technology. Semin Reprod Med 2021; 39:207-219. [PMID: 34500477 DOI: 10.1055/s-0041-1735646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Androgen priming with either dehydroepiandrosterone (DHEA) or testosterone has been suggested as an adjunct to improve in vitro fertilization (IVF) outcomes in women with diminished ovarian reserve (DOR). Numerous studies have investigated the effects of both DHEA and testosterone on IVF outcome. The results were inconsistent, and the quality of most studies is substandard. Meta-analyses have consistently reported that DHEA does appear to significantly improve IVF outcome in women with predicted or proven poor ovarian response (POR), but these have included some normal responders and/or nonrandomized studies. Our meta-analyses including randomized controlled trials (RCTs) incorporating only women with DOR or POR suggest that DHEA confers no benefit. While meta-analyses of RCTs on the use of testosterone in women with DOR or POR showed an improved IVF outcome, most studies included are of low quality with high risk of bias. When analysis of data from studies of only low-risk bias was performed, such a benefit with testosterone was not observed. Although recruitment may well be a challenge, a large, well-designed RCT is, however, still warranted to investigate whether or not androgen priming with either DHEA or testosterone should be recommended as an adjuvant treatment for women with DOR or POR undergoing IVF.
Collapse
|
19
|
Neves AR, Montoya-Botero P, Polyzos NP. The Role of Androgen Supplementation in Women With Diminished Ovarian Reserve: Time to Randomize, Not Meta-Analyze. Front Endocrinol (Lausanne) 2021; 12:653857. [PMID: 34079524 PMCID: PMC8165260 DOI: 10.3389/fendo.2021.653857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/23/2021] [Indexed: 11/24/2022] Open
Abstract
The management of patients with diminished ovarian reserve (DOR) remains one of the most challenging tasks in IVF clinical practice. Despite the promising results obtained from animal studies regarding the importance of androgens on folliculogenesis, the evidence obtained from clinical studies remains inconclusive. This is mainly due to the lack of an evidence-based methodology applied in the available trials and to the heterogeneity in the inclusion criteria and IVF treatment protocols. In this review, we analyze the available evidence obtained from animal studies and highlight the pitfalls from the clinical studies that prevent us from closing the chapter of this line of research.
Collapse
Affiliation(s)
- Ana Raquel Neves
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine, Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Pedro Montoya-Botero
- Department of Reproductive Medicine, Conceptum – Unidad de Fertilidad del Country, Bogotá, Colombia
| | - Nikolaos P. Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), Gent, Belgium
- *Correspondence: Nikolaos P. Polyzos,
| |
Collapse
|
20
|
Hormonal Effects in Reproductive Technology with Focus on Diminished Ovarian Reserve. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020. [PMID: 32406026 DOI: 10.1007/978-3-030-38474-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2023]
Abstract
Modern use of reproductive technologies has revolutionized the treatment of infertile couples. Strategies to improve ovarian function in cases of diminished ovarian reserve are perhaps the least understood area in this field and will be the chief focus of this chapter.
Collapse
|
21
|
Jiang L, Cui J, Zhang C, Xie J, Zhang S, Fu D, Duo W. Sigma-1 receptor is involved in diminished ovarian reserve possibly by influencing endoplasmic reticulum stress-mediated granulosa cells apoptosis. Aging (Albany NY) 2020; 12:9041-9065. [PMID: 32409627 PMCID: PMC7288944 DOI: 10.18632/aging.103166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/31/2020] [Indexed: 12/25/2022]
Abstract
Sigma non-opioid intracellular receptor 1 (sigma-1 receptor), a non-opioid transmembrane protein, is located on cellular mitochondrial membranes and endoplasmic reticulum. Current research has demonstrated that sigma-1 receptor is related to human degenerative diseases. This study is focused on the effects of sigma-1 receptor on the pathophysiological process of diminished ovarian reserve (DOR) and granulosa cells (GCs) apoptosis. Sigma-1 receptor concentration in follicular fluid (FF) and serum were negatively correlated with basal follicle-stimulating hormone (FSH) and positively correlated with anti-mullerian hormone (AMH), antral follicle count (AFC). Sigma-1 receptor reduction in GCs was accompanied by endoplasmic reticulum stress (ERS)-mediated apoptosis in women with DOR. Plasmid transfection was used to establish SIGMAR1-overexpressed and SIGMAR1-knockdown human granulosa-like tumor (KGN) cell and thapsigargin (TG) was used to induce ERS KGN cells. We found that KGN cells treated with endogenous sigma-1 receptor ligand dehydroepiandrosterone (DHEA) and sigma-1 receptor agonist PRE-084 showed similar biological effects to SIGMAR1-overexpressed KGN cells and opposite effects to SIGMAR1-knockdown KGN cells. DHEA may improve DOR patients' pregnancy outcomes by upregulating sigma-1 receptor and downregulating ERS-mediated apoptotic genes in GCs. Thus, sigma-1 receptor may be a potential ovarian reserve biomarker, and ligand-mediated sigma-1 receptor activation could be a future approach for DOR therapy.
Collapse
Affiliation(s)
- Lile Jiang
- Reproductive Medical Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jinquan Cui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Cuilian Zhang
- Reproductive Medical Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Juanke Xie
- Reproductive Medical Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shaodi Zhang
- Reproductive Medical Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongjun Fu
- School of Pharmaceutical Sciences and Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Duo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
22
|
Anğın AD, Gün I, Sakin Ö, Çıkman MS, Şimşek EE, Karakuş R, Başak K, Kaptanağası AO. Investigation of the preventive effects of dehydroepiandrosterone (DHEA) and Caffeic acid phenethyl ester (CAPE) on cisplatin-induced ovarian damage in rats. Ultrastruct Pathol 2020; 44:71-80. [PMID: 31909696 DOI: 10.1080/01913123.2019.1711479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
To investigate whether Dehydroepiandrosterone (DHEA) and Caffeic acid phenethyl ester (CAPE) had any preventive effect against the ovarian damage caused by cisplatin (CP) (cis-diamminedichloroplatinum) in rats. On the first day ovaries were removed, Anti-Müllerian hormone (AMH) was measured (Group1, n:6), in the other groups 7.5 mg/kg cisplatin was administered intraperitoneally. In Group 2 (n = 6), 0.1 ml saline, in Group 3 (n = 5), 20 umol/kg CAPE, in Group 4 (n = 7), DHEA 6 mg/kg were administered every day. On the 10th day, ovaries were removed, AMH was measured. Ovary reserve (primordial/primary/secondary/tertiary/atretic follicles, AMH), ovarian damage scores (follicular degeneration, congestion, hemorrhage, edema, inflammation) were compared. The number of tertiary follicles were statistically high in the CAPE group (p = .015), the inflammation score in the DHEA group (p = .012), AMH level (p = .009) in the control group. The lowest number of atretic follicles (AF) was in the control group, while the highest number of AF was in the DHEA group (p = .002). Significant decreases in AF were the case in the cisplatin and DHEA groups compared to the control group (p < .008). The AMH values had the highest positive correlation with the number of primordial follicles and the highest negative correlation with the number of AF. The cut off point for AMH was 1.57 ng/ml as an indicator of low ovarian reserve. Cisplatin causes total damage and increased numbers of AF on the ovary. Depending on this, AMH levels fall. These negative effects of cisplatin are not obstructed by CAPE or DHEA, and may even be increased by DHEA.
Collapse
Affiliation(s)
- Ali Doğukan Anğın
- Department of Obstetrics and Gynecology, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| | - Ismet Gün
- Department of Obstetrics and Gynecology, University of Health Sciences, Sultan Abdülhamid Han Training and Research Hospital, İstanbul, Turkey
| | - Önder Sakin
- Department of Obstetrics and Gynecology, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| | - Muzaffer Seyhan Çıkman
- Department of Obstetrics and Gynecology, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| | - Engin Ersin Şimşek
- Department of Family Medicine, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| | - Resul Karakuş
- Department of Obstetrics and Gynecology, University of Health Sciences, Zeynep Kamil Women's and Children's Disease Training and Research Hospital, İstanbul, Turkey
| | - Kayhan Başak
- Department of Pathology, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| | - Asuman Orçun Kaptanağası
- Department of Biochemistry, University of Health Sciences, Dr Lütfi Kırdar Kartal Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
23
|
Blumenfeld Z. What Is the Best Regimen for Ovarian Stimulation of Poor Responders in ART/IVF? Front Endocrinol (Lausanne) 2020; 11:192. [PMID: 32362870 PMCID: PMC7180183 DOI: 10.3389/fendo.2020.00192] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
The infertile patients with aging ovaries-also sometimes referred to as impending premature ovarian insufficiency (POI), impending premature ovarian failure (POF), or poor ovarian responders (POR), constitute a significant and increasing bulk of the patients appealing to IVF/ART. Different causes have been cited in the literature, among the identified etiologies, including chromosomal and genetic etiology, metabolic, enzymatic, iatrogenic, toxic, autoimmune, and infectious causes. Although the most successful and ultimate treatment of POI/POF/POR patients is egg donation (ED), many, if not most, of these infertile women are reluctant to consent to ED upon the initial diagnostic interview, requesting alternative solutions despite the low odds for success. Despite anecdotal case reports, no unequivocal treatment proved to be successful for these patients in prospective randomized controlled trials. Nevertheless, the addition of growth hormone (GH) to ovarian stimulation in POR with GH deficiency may improve the results of controlled ovarian hyperstimulation (COH) and the IVF success. In patients with autoimmune etiology for POR/POI, the combination of glucocorticosteroids, pituitary-ovarian suppression, and COH may be successful in achieving the desired conception.
Collapse
|
24
|
Løssl K, Freiesleben NLC, Wissing ML, Birch Petersen K, Holt MD, Mamsen LS, Anderson RA, Andersen CY. Biological and Clinical Rationale for Androgen Priming in Ovarian Stimulation. Front Endocrinol (Lausanne) 2020; 11:627. [PMID: 33013703 PMCID: PMC7498541 DOI: 10.3389/fendo.2020.00627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/31/2020] [Indexed: 12/24/2022] Open
Abstract
Androgen receptors are expressed by all stages of growing follicles, and follicular fluid androgen levels are positively correlated to granulosa cell androgen receptor and follicle-stimulating hormone (FSH) receptor expression. Thus, androgens may promote follicular growth, accumulation and/or responsiveness to gonadotropins. This is explored therapeutically in the concept of androgen priming, to improve the ovarian response to stimulation in assisted reproduction. Androgen effects may be achieved in two different ways, either directly by providing exogenous androgen or by providing luteinizing hormone (LH) activity [i.e., LH or human chorionic gonadotropin (hCG)] to stimulate local ovarian production of androgen. The androgen concentrations in follicular fluid by far exceed the levels in female circulation and it has recently been shown that there was no correlation between serum testosterone levels and follicular fluid androgen levels. There is some evidence that administration of exogenous dehydroepiandrosterone or testosterone increases live birth rates, but an optimal protocol has not been established and such adjuvant treatment should be considered experimental. Furthermore, studies exploring long-term administration of LH activity, achieving LH levels comparable to those seen in women with polycystic ovary syndrome, are awaited. The aim of the present review is to discuss critically the most suitable approach for androgen priming from a biological and clinical standpoint, and to evaluate current approaches and results obtained in clinical trials.
Collapse
Affiliation(s)
- Kristine Løssl
- The Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Claus Yding Andersen
| |
Collapse
|
25
|
Monteiro CDS, Scheffer BB, Carvalho RFD, Scheffer JB. The impact of dehydroepiandrosterone in poor ovarian responders on assisted reproduction technology treatment. JBRA Assist Reprod 2019; 23:414-417. [PMID: 31251010 PMCID: PMC6798587 DOI: 10.5935/1518-0557.20190045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
One in six couples worldwide will experience at least one infertility problem
during their reproductive years. Between 5.6% and 35.1% of women will exhibit
poor ovarian response. A variety of methods have been applied to improve ovarian
response, including dehydroepiandrosterone. In the ovaries,
dehydroepiandrosterone promotes follicular development and granulosa cell
proliferation by increasing intraovarian androgen concentrations while
simultaneously enhancing the level of follicular insulin-like growth factor-1,
which promotes folliculogenesis. Dehydroepiandrosterone supplementation may
improve in vitro fertilization outcomes and ovarian response in
patients with poor ovarian response. However, a few questions still loom over
the effectiveness of dehydroepiandrosterone.
Collapse
Affiliation(s)
| | - Bruno Brum Scheffer
- Brazilian Institute of Assisted Reproduction (IBRRA), Belo Horizonte, MG, Brazil
| | | | | |
Collapse
|
26
|
Jiang L, Chen Y, Wang Q, Wang X, Luo X, Chen J, Han H, Sun Y, Shen H. A Chinese practice guideline of the assisted reproductive technology strategies for women with advanced age. J Evid Based Med 2019; 12:167-184. [PMID: 31144467 DOI: 10.1111/jebm.12346] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 01/31/2023]
Abstract
More women postpone childbearing nowadays while female fertility begins to decline with advancing age. Furthermore, with the rolling out of the two-child policy, there is a huge demand for a second child for Chinese aged women. There are various assisted reproductive technology (ART) strategies applied for age-related infertility without solid evidence. On behalf of the Society of Reproductive Medicine, Chinese Medical Association, we would like to develop a Chinese guideline of ART strategies for age-related infertility. This guideline was produced following the recommendations for standard guidelines described in the 2012 WHO Handbook for guideline development. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) framework was also followed. A protocol was formulated and a Guideline Development Group was formed with specialists of reproductive medicine, methodologists from Chinese GRADE working group, and patient representative. Questions regarding the ART strategies for aged infertility were formulated and 8 most important ones were chosen to be structured in PICO format (Population, Intervention, Comparison, Outcomes). Comprehensive search and review of the literature were performed and the quality of the evidence was assessed and rated based on certain criteria and be categorized as high, moderate, low, or very low. Twenty-five recommendations were formulated among members of the Guidelines Development Group (Delphi method) basing on the overall quality of the evidence, in addition to the balance between benefits and harms, values and preferences, and resource implications. The final recommendations were agreed on by consensus during face-to-face meetings. This is the first Chinese practice guideline in reproductive medicine developed following the standard and scientific method.
Collapse
Affiliation(s)
- Li Jiang
- Reproductive Medicine Center, Peking University People's Hospital, Beijing, China
| | - Yaolong Chen
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
- Chinese GRADE Center, Lanzhou, China
- WHO Collaborating Centre for Guideline Implementation and Knowledge Translation, Lanzhou, China
| | - Qi Wang
- Health Policy PhD Program, Department of Health Research Methods, Evidence and Impact, Faculty of Health Sciences, McMaster University, Hamilton, Canada
- McMaster Health Forum, McMaster University, Hamilton, Canada
| | - Xiaoqin Wang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
- Chinese GRADE Center, Lanzhou, China
- WHO Collaborating Centre for Guideline Implementation and Knowledge Translation, Lanzhou, China
| | - Xufei Luo
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
- Chinese GRADE Center, Lanzhou, China
- WHO Collaborating Centre for Guideline Implementation and Knowledge Translation, Lanzhou, China
| | - Junqiao Chen
- Faculty of Science, University of Lisbon, Lisbon, Portugal
| | - Hongjing Han
- Reproductive Medicine Center, Peking University People's Hospital, Beijing, China
| | - Yingpu Sun
- Reproductive Medicine Center, Zhengzhou University First Affiliated Hospital, Zhengzhou, Henan, China
| | - Huan Shen
- Reproductive Medicine Center, Peking University People's Hospital, Beijing, China
| |
Collapse
|
27
|
Kushnir VA, Darmon SK, Barad DH, Weghofer A, Gleicher N. Effects of dehydroepiandrosterone (DHEA) supplementation on sexual function in premenopausal infertile women. Endocrine 2019; 63:632-638. [PMID: 30311171 DOI: 10.1007/s12020-018-1781-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE To investigate the effects of dehydroepiandrosterone (DHEA) supplementation on female sexual function in premenopausal infertile women of advanced ages. METHODS This observational study was conducted in an academically affiliated private fertility center. Patients included 87 premenopausal infertile women, 50 of whom completed the study including the Female Sexual Function Index (FSFI) questionnaires and comprehensive endocrine evaluation before and 4-8 weeks after initiating 25 mg of oral micronized DHEA TID. RESULTS Age of patients was 41.1 ± 4.2 years, BMI 24.4 ± 6.1 kg/m2, 86% were married, and 42% were parous. Following supplementation with DHEA, all serum androgen levels increased (each P < 0.0001), while FSH levels decreased by 2.6 ± 4.4 from a baseline of 10.3 ± 5.4 mIU/mL (P = 0.009). The FSFI score for the whole study group increased by 7% (from 27.2 ± 6.9 to 29.2 ± 5.6; P = 0.0166). Domain scores for desire increased by 17% (P = 0.0004) and by 12% for arousal (P = 0.0122); lubrication demonstrated an 8% trend towards improvement (P = 0.0551), while no changes in domain scores for orgasm, satisfaction, or pain were observed. Women in the lowest starting FSFI score quartile (<25.7), experienced a 6.1 ± 8.0 (34%) increase in total FSFI score following DHEA supplementation. Among these women, improvements in domain categories were noted for desire (40%), arousal (46%), lubrication (33%), orgasm (54%), satisfaction (24%), and pain (25%). CONCLUSIONS This uncontrolled observational study implies that supplementation with DHEA improves sexual function in older premenopausal women with low baseline FSFI scores.
Collapse
Affiliation(s)
- Vitaly A Kushnir
- The Center for Human Reproduction, New York, NY, USA.
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | | | - David H Barad
- The Center for Human Reproduction, New York, NY, USA
- Foundation for Reproductive Medicine, New York, NY, USA
| | - Andrea Weghofer
- The Center for Human Reproduction, New York, NY, USA
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Norbert Gleicher
- The Center for Human Reproduction, New York, NY, USA
- Foundation for Reproductive Medicine, New York, NY, USA
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY, USA
| |
Collapse
|
28
|
Wang W, Liu H, Li J, Wei D, Zhang J, Wang J, Ma J, Shi Y, Chen ZJ. Effect of preconceptional DHEA treatment on in vitro fertilization outcome in poor ovarian respond women: study protocol for a randomized controlled trial. Trials 2019; 20:50. [PMID: 30646929 PMCID: PMC6334415 DOI: 10.1186/s13063-018-3146-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Background Women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) with poor ovarian respond (POR) always have very low clinical pregnancy rates. In previous data, dehydroepiandrosterone (DHEA) was suggested as a promising treatment and maybe has a good pregnancy outcome. But there is no sufficient evidence from randomized clinical trials evaluating the effect of DHEA preconceptional treatment on live birth in POR. Methods This trial is a multicenter active-placebo double-blind clinical trial (1:1 treatment ratio of active versus placebo). The infertile POR patients undergoing IVF or ICSI will be enrolled and randomly assigned to two parallel groups. Participants in these two groups will be given 4–12 weeks’ treatment of DHEA or placebo, respectively. The primary outcome is live birth rate. Discussion The results of this study will provide evidence for the effect of preconceptional DHEA treatment on IVF outcome in POR. Trial registration Chinese Clinical Trial Registry, ChiCTR-IPR-15006909. Registered on November 9, 2015. Electronic supplementary material The online version of this article (10.1186/s13063-018-3146-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei Wang
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China.,Center for Reproductive Medicine, The Second Clinical Medical College, Yangtze University, No. 2 People Street, Jingzhou district, Jingzhou, 434020, China
| | - Hong Liu
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Jing Li
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Daimin Wei
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Jiangtao Zhang
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Jianfeng Wang
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China
| | - Yuhua Shi
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China.
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Key Laboratory of Reproductive Endocrinology, Ministry of Education, and National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong Provincial Clinical medicine Research Center for reproductive health, Shandong Provincial Hospital Affiliated with Shandong University, Shandong University, No. 157 Jing Liu Street, Shizhong district, Jinan, 250012, China.,Center for Reproductive Medicine, School of Medicine, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shandong University, Shanghai Jiao Tong University, No. 845 Lingshan Road, Pudong new district, Shanghai, 310101, China
| |
Collapse
|
29
|
Chen SN, Tsui KH, Wang PH, Chern CU, Wen ZH, Lin LT. Dehydroepiandrosterone Supplementation Improves the Outcomes of in vitro Fertilization Cycles in Older Patients With Diminished Ovarian Reserve. Front Endocrinol (Lausanne) 2019; 10:800. [PMID: 31803144 PMCID: PMC6873389 DOI: 10.3389/fendo.2019.00800] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 11/01/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Dehydroepiandrosterone (DHEA) supplementation has been reported to have beneficial effects on the in vitro fertilization (IVF) outcomes of patients with poor ovarian response or diminished ovarian reserve. The Patient-Oriented Strategies Encompassing IndividualizeD Oocyte Number (POSEIDON) stratification is a set of newly established criteria for low prognosis patients. The aim of this study was to examine the potential effects of DHEA supplementation on the IVF outcomes of patients who fulfill the POSEIDON group 4 criteria. Methods: This retrospective cohort study investigated 297 cycles that fulfilled the POSEIDON group 4 criteria and underwent IVF treatment using the gonadotropin-releasing hormone antagonist protocol. The study group contained 159 cycles that received DHEA (30 mg three times per day) daily for 12 weeks before their IVF cycles. The control group included 138 cycles that underwent IVF cycles but did not receive DHEA. The baseline characteristics and cycle parameters as well as the IVF outcomes of both groups were compared. Results: In terms of baseline characteristics, more previous IVF attempts and lower AMH levels were found in the study group than in the control group. Regarding IVF outcomes, patients in the study group had significantly higher follicular oocyte index and higher numbers of retrieved oocytes, metaphase II oocytes, fertilized oocytes, day 3 embryos and top-quality day 3 embryos than those in the control group. Besides, a higher cumulative pregnancy rate and lower cancellation rate were observed in the study group than in the control group although clinical pregnancy rate, live birth rate, and cumulative live birth rate did not differ between the two groups. Whether patients are aged ≤ 40 years or aged > 40, higher numbers of oocytes and embryos were observed in the study group than in the control group. In patients aged > 40, cumulative pregnancy rate was significantly higher in the study group than in the control group. Conclusions: Our data suggest that DHEA supplementation might increase both oocyte and embryo yields, as well as cumulative pregnancy rates, in patients fulfilling the POSEIDON group 4 criteria.
Collapse
Affiliation(s)
- San-Nung Chen
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Yanpu Township, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei City, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung City, Taiwan
| | - Chyi-Uei Chern
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung City, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei City, Taiwan
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung City, Taiwan
- *Correspondence: Li-Te Lin
| |
Collapse
|
30
|
Ali AENAEG, Khodry MM. Role of Dehydroepiandrosterone Supplementation in Improving Intracytoplasmic Sperm Injection Outcome for Women with Expected Poor Ovarian Response. OPEN JOURNAL OF OBSTETRICS AND GYNECOLOGY 2019; 09:353-362. [DOI: 10.4236/ojog.2019.93036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
31
|
Artini PG, Obino ME, Vergine F, Sergiampietri C, Papini F, Cela V. Assisted reproductive technique in women of advanced fertility age. ACTA ACUST UNITED AC 2018; 70:738-749. [DOI: 10.23736/s0026-4784.18.04247-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
32
|
Schwarze JE, Canales J, Crosby J, Ortega-Hrepich C, Villa S, Pommer R. DHEA use to improve likelihood of IVF/ICSI success in patients with diminished ovarian reserve: A systematic review and meta-analysis. JBRA Assist Reprod 2018; 22:369-374. [PMID: 30125071 PMCID: PMC6210617 DOI: 10.5935/1518-0557.20180046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The aim of this review is to determine if the use of DHEA increases the
likelihood of success in patients with POR. We searched MEDLINE and EMBASE using
the terms "DHEA and diminished ovarian reserve", "DHEA and poor response", "DHEA
and premature ovarian aging". A fixed effects model was used and Peto's method
to get the odds ratio (OR) with 95% confidence intervals (CI 95%). For
quantitative variables, Cohen's method was used to present the standardized mean
differences (SMD) with their corresponding confidence intervals. Only five
studied fulfilled the selection criteria. DHEA was administered in 25 mg doses,
three times a day. In all studies, the authors corrected for the presence of
confounding variables such as partner's age, infertility diagnosis and number of
transferred embryos. The meta-analysis of the five selected studies assessed a
total of 910 patients, who underwent IVF/ICSI, of which 413 had received DHEA.
DHEA use was associated with a significant increase in pregnancy likelihood (OR
1.8, CI 95% 1.29 to 2.51, p=0.001). When analyzing the
association between DHEA use and the likelihood of abortion, we found low
heterogeneity between studies (I2=0.0%) and the use of DHEA to be
associated to a significant reduction in the likelihood of abortion (OR 0.25, CI
0.07 to 0.95; p=0.045). Analysis of the association of DHEA
with average oocyte retrieval showed high variability between studies
(I2=98.6%), as well as no association between DHEA use and the
number of oocytes retrieved (SMD -0.01, CI 95% -0.16 to 0.13;
p<0.05).
Collapse
Affiliation(s)
- Juan Enrique Schwarze
- Reproductive Medicine Unit, Las Condes Clinic. Santiago, Chile.,Clinical Department of Obstetrics and Gynecology, Universidad de Santiago de Chile
| | - Johana Canales
- Clinical Department of Obstetrics and Gynecology, Universidad de Santiago de Chile
| | - Javier Crosby
- Reproductive Medicine Unit, Monteblanco Clinic. Santiago, Chile
| | | | - Sonia Villa
- Reproductive Medicine Unit, Monteblanco Clinic. Santiago, Chile
| | - Ricardo Pommer
- Reproductive Medicine Unit, Monteblanco Clinic. Santiago, Chile
| |
Collapse
|
33
|
Payne P, Fiering S, Zava D, Gould TJ, Brown A, Hage P, Gaudet C, Crane-Godreau M. Digital Delivery of Meditative Movement Training Improved Health of Cigarette-Smoke-Exposed Subjects. Front Public Health 2018; 6:282. [PMID: 30406067 PMCID: PMC6202937 DOI: 10.3389/fpubh.2018.00282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Many FA who flew prior to the ban on smoking in commercial aircraft exhibit an unusual pattern of long-term pulmonary dysfunction. This randomized controlled study tested the hypothesis that digitally delivered meditative movement (MM) training improves chronic obstructive pulmonary disease (COPD)-related symptoms in flight attendants (FA) who were exposed to second-hand cigarette smoke (SHCS) while flying. Phase I of this two-phase clinical trial was a single-arm non-randomized pilot study that developed and tested methods for MM intervention; we now report on Phase II, a randomized controlled trial comparing MM to a control group of similar FA receiving health education (HE) videos. Primary outcomes were the 6-min walk test and blood levels of high sensitivity C-reactive protein (hs-CRP). Pulmonary, cardiovascular, autonomic and affective measures were also taken. There were significant improvements in the 6-min walk test, the Multidimensional Assessment of Interoceptive Awareness (MAIA) score, and the COPD Assessment Test. Non-significant trends were observed for increased dehydroepiandrosterone sulfate (DHEAS) levels, decreased anxiety scores and reduced blood hs-CRP levels, and increased peak expiratory flow (PEF). In a Survey Monkey questionnaire, 81% of participants who completed pre and post-testing expressed mild to strong positive opinions of the study contents, delivery, or impact, while 16% expressed mild negative opinions. Over the course of the year including the study, participant adoption of the MM practices showed a significant and moderately large correlation with overall health improvement; Pearson's R = 0.62, p < 0.005. These results support the hypothesized benefits of video-based MM training for this population. No adverse effects were reported. Clinical Trial Registration:www.ClinicalTrials.gov, identifier: NCT02612389
Collapse
Affiliation(s)
- Peter Payne
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, PA, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, PA, United States
| | - David Zava
- ZRT Laboratory, Beaverton, OR, United States
| | - Thomas J Gould
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, United States
| | - Anthony Brown
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Paul Hage
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Carole Gaudet
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, PA, United States
| | - Mardi Crane-Godreau
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, PA, United States
| |
Collapse
|
34
|
Chern CU, Tsui KH, Vitale SG, Chen SN, Wang PH, Cianci A, Tsai HW, Wen ZH, Lin LT. Dehydroepiandrosterone (DHEA) supplementation improves in vitro fertilization outcomes of poor ovarian responders, especially in women with low serum concentration of DHEA-S: a retrospective cohort study. Reprod Biol Endocrinol 2018; 16:90. [PMID: 30223902 PMCID: PMC6142344 DOI: 10.1186/s12958-018-0409-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Dehydroepiandrosterone (DHEA) is now widely used as an adjuvant for in vitro fertilization (IVF) cycles in poor ovarian responders (PORs). Several studies showed that DHEA supplementation could improve IVF outcomes of PORs. However, most of the PORs do not respond to DHEA clinically. Therefore, the aim of this study is to confirm the beneficial effects of DHEA on IVF outcomes of PORs and to investigate which subgroups of PORs can best benefit from DHEA supplementation. METHODS This retrospective cohort study was performed between January 2015 and December 2017. A total of 151 PORs who fulfilled the Bologna criteria and underwent IVF cycles with the gonadotropin-releasing hormone antagonist protocol were identified. The study group (n = 67) received 90 mg of DHEA daily for an average of 3 months before the IVF cycles. The control group (n = 84) underwent the IVF cycles without DHEA pretreatment. The basic and cycle characteristics and IVF outcomes between the two groups were compared using independent t-tests, Chi-Square tests and binary logistic regression. RESULTS The study and control groups did not show significant differences in terms of basic characteristics. The study group demonstrated a significantly greater number of retrieved oocytes, metaphase II oocytes, fertilized oocytes, day 3 embryos and top-quality embryos at day 3 and a higher clinical pregnancy rate, ongoing pregnancy rate and live birth rate than those measures in the control group. The multivariate analysis revealed that DHEA supplementation was positively associated with clinical pregnancy rate (OR = 4.93, 95% CI 1.68-14.43, p = 0.004). Additionally, in the study group, the multivariate analysis showed that serum dehydroepiandrosterone-sulfate (DHEA-S) levels < 180 μg/dl were significantly associated with a rate of retrieved oocytes > 3 (OR = 5.92, 95% CI 1.48-23.26, p = 0.012). CONCLUSIONS DHEA supplementation improves IVF outcomes of PORs. In PORs with DHEA pretreatment, women with lower DHEA-S level may have greater possibility of attaining more than 3 oocytes.
Collapse
Affiliation(s)
- Chyi-Uei Chern
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
| | - Kuan-Hao Tsui
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0639 0943grid.412902.cDepartment of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, No.20, Weixin Rd, Yanpu, Township, Pingtung County 90741 Taiwan
| | - Salvatore Giovanni Vitale
- 0000 0004 1757 1969grid.8158.4Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - San-Nung Chen
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
| | - Peng-Hui Wang
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0604 5314grid.278247.cDepartment of Obstetrics and Gynecology, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei, 112 Taiwan
- 0000 0004 0572 9415grid.411508.9Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, North District, Taichung City, 40447 Taiwan
| | - Antonio Cianci
- 0000 0004 1757 1969grid.8158.4Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Hsiao-Wen Tsai
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
| | - Zhi-Hong Wen
- 0000 0004 0531 9758grid.412036.2Department of Marine Biotechnology and Resources, National Sun Yat-sen University, 70 Lienhai Rd, Kaohsiung City, 80424 Taiwan
| | - Li-Te Lin
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0531 9758grid.412036.2Department of Biological Science, National Sun Yat-Sen University, 70 Lienhai Rd, Kaohsiung City, 80424 Taiwan
| |
Collapse
|
35
|
Mostajeran F, Tehrani H, Ghoreishi E. Effects of Dehydroepiandrosterone on In Vitro Fertilization Among Women Aging Over 35 Years and Normal Ovarian Reserve. J Family Reprod Health 2018; 12:129-133. [PMID: 31223318 PMCID: PMC6571446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective: The fertility potential of women declines with aging and the likelihood of infertility and stillbirth increase. Treatment protocols involving dehydroepiandrosterone (DHEA) provide pathways on improving fertility and live birth rate. This randomized clinical trial aimed to evaluate the effect of DHEA on In Vitro Fertilization (IVF) outcomes in women over 35 years and normal ovarian reserve. Materials and methods: One hundred and six consecutive women with advanced reproductive age undergoing IVF due to infertility were enrolled in the study. Participants in the intervention group received 75 mg/day of DHEA orally 8 weeks before starting the cycle of ovulation induction. Participants in placebo group received placebo tablets during the same period. After 8 weeks, routine procedure of IVF was initiated then Clinical pregnancy, Miscarriage, Endometrial thickness and Duration of stimulation were evaluated for all the participants in both groups. Results: There is no significant difference between groups in terms of age and BMI. Mean endometrial thickness was significantly lower in DHEA group (9.63 ± 0.70vs.8.05 ± 0.70; p < 0.001) while Duration of stimulation was higher (8.98 ± 1.29vs.10.59 ± 1.43; p < 0.001). There was no significant difference between pregnancy rate, and miscarriage rate of the groups. Conclusion: According to the result of this study, DHEA supplementation may improve IVF outcomes in infertile women. Although additional larger and placebo-controlled studies using different DHEA protocols are required to support our present findings.
Collapse
|
36
|
Dawood AS, El-Sharawy MA, Nada DW, El-Sheikh MF. Premature ovarian failure of autoimmune etiology in 46XX patients: is there a hope? JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2018; 15:jcim-2017-0072. [PMID: 29794258 DOI: 10.1515/jcim-2017-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 03/21/2018] [Indexed: 11/15/2022]
Abstract
OBJECTIVES To evaluate the efficacy of live bee stings at fertility points and acupuncture in treating symptoms and managing infertility in premature ovarian failure (POF) of autoimmune etiology. PATIENTS AND METHODS Patients with primary POF were allocated randomly into two groups: group I: subjected to acupuncture at specific fertility points and group II: subjected to live bee stings at sites of fertility points. RESULTS A total of 24 cases show significant reduction of Follicle stimulating hormone (FSH) level to normal range with gradual decline over the study duration: 13 cases in group I and 11 cases in group II. Eight cases got pregnant while the other 13 cases regained normal menses but still infertile. CONCLUSIONS Both bee sting therapy and acupuncture were effective in reduction of FSH levels with restoration of regular menstrual patterns and restoration of fertility. The bee sting therapy was superior in the pregnancy rate, while acupuncture was superior in alleviation of symptoms.
Collapse
Affiliation(s)
- Ayman Shehata Dawood
- MD of Obstetrics and Gynecology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Doaa Waseem Nada
- MD of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Fadel El-Sheikh
- MD of Economic Entomology, Department of Plant Protection, Faculty of Agriculture, Tanta University, Tanta, Egypt
| |
Collapse
|
37
|
Lin X, Du J, Du Y, Wu R, Fang X, Liao Y, Quan S. Effects of dehydroepiandrosterone supplementation on mice with diminished ovarian reserve. Gynecol Endocrinol 2018; 34:357-359. [PMID: 29221424 DOI: 10.1080/09513590.2017.1409712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Dehydroepiandrosterone (DHEA) has been used to improve the pregnancy rate in women with diminished ovarian reserve(DOR) during in vitro fertilization. We aimed to validate the effects of DHEA and identify the possible mechanisms. We constructed a mice model with DOR and analyzed the hormone parameters and follicle counts. In vivo experiment, FSH and LH concentrations in the serum were significantly elevated in the DOR group. However, the FSH and LH concentrations were partially reversed in the DOR + DHEA group. The E2, AMH and INHB were down-regulated in the DOR group and reversed in the DOR + DHEA group. Our study supported evidences that DHEA might modulate the hormone receptors in the ovary and hormone secretions to the peripheral circulation to regulate the ovary reserve functions.
Collapse
Affiliation(s)
- Xiufeng Lin
- a Reproductive Medical Center, Department of Obstetrics and Gynecology , Nanfang Hospital, Southern Medical University , Guangzhou , China
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Jing Du
- a Reproductive Medical Center, Department of Obstetrics and Gynecology , Nanfang Hospital, Southern Medical University , Guangzhou , China
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Yan Du
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Riran Wu
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Xiaowu Fang
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Yuechan Liao
- b Reproductive Medical Center , Boai Hospital of Zhongshan , Zhongshan , Guangdong Province , China
| | - Song Quan
- a Reproductive Medical Center, Department of Obstetrics and Gynecology , Nanfang Hospital, Southern Medical University , Guangzhou , China
| |
Collapse
|
38
|
Mahmoud YI, Mahmoud AA, Abo-Zeid FS, Fares NH. Effects of dehydroepiandrosterone on the ovarian reserve and pregnancy outcomes in perimenopausal rats (DHEA and fertility in perimenopausal rats). Life Sci 2018. [DOI: 10.1016/j.lfs.2018.03.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Liu Y, Hu L, Fan L, Wang F. Efficacy of dehydroepiandrosterone (DHEA) supplementation for in vitro fertilization and embryo transfer cycles: a systematic review and meta-analysis. Gynecol Endocrinol 2018; 34:178-183. [PMID: 29073790 DOI: 10.1080/09513590.2017.1391202] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Dehydroepiandrosterone (DHEA) supplementation might hold some promise in vitro fertilization and embryo transfer cycles. However, the results remain controversial. We conducted a systematic review and meta-analysis to evaluate the efficacy of DHEA in patients for in vitro fertilization. PubMed, EMbase, Web of science, EBSCO and Cochrane library databases were systematically searched. Randomized controlled trials (RCTs) assessing the effect of DHEA versus placebo on in vitro fertilization were included. Two investigators independently searched articles, extracted data and assessed the quality of included studies. The primary outcomes were clinical pregnancy and live birth rate. Meta-analysis was performed using random-effect model. Six RCTs involving 745 patients were included in the meta-analysis. Overall, compared with placebo, DHEA supplementation was associated with the significant increase in clinical pregnancy (OR = 1.45; 95% CI = 1.04-2.03; p = .03), live birth rate (OR = 2.70; 95% CI = 1.24-5.85; p = .01) and endometrial thickness (Std. mean difference = 0.67; 95% CI = 0.02-1.32; p = .04) but showed no influence on E2 on hCG day (Std. mean difference = 0.69; 95% CI = -0.46 to 1.85; p = .24), embryos transferred (Std. mean difference = 0.42; 95% CI = -0.04 to 0.88; p = .07) and miscarriage rate (OR = 0.43; 95% CI = 0.03-6.66; p = .55). DHEA supplementation could significantly improve clinical pregnancy, live birth rate, endometrial thickness and retrieved oocytes but failed to alter E2 on hCG day, embryos transferred and miscarriage rate.
Collapse
Affiliation(s)
- Yaofang Liu
- a Department of Gynecology and Obstetrics , The Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , China
| | - Lina Hu
- b Department of Gynecology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Lingye Fan
- a Department of Gynecology and Obstetrics , The Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , China
| | - Fang Wang
- a Department of Gynecology and Obstetrics , The Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , China
| |
Collapse
|
40
|
Gibson DA, Simitsidellis I, Kelepouri O, Critchley HOD, Saunders PTK. Dehydroepiandrosterone enhances decidualization in women of advanced reproductive age. Fertil Steril 2018; 109:728-734.e2. [PMID: 29397924 PMCID: PMC5908781 DOI: 10.1016/j.fertnstert.2017.12.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/23/2017] [Accepted: 12/20/2017] [Indexed: 11/17/2022]
Abstract
Objective To investigate the impact of the androgen precursor dehydroepiandrosterone (DHEA) on the decidualization of human endometrial stromal cells isolated from women of advanced reproductive age. Design In vitro study. Setting University research institute. Patient(s) Proliferative phase primary human endometrial stromal fibroblasts (hESFs) were isolated from women of advanced reproductive age (n = 16; mean age, 44.7 ± 2.3). None of the women were receiving hormone therapy or had endometriosis. Intervention(s) Isolated hESFs were decidualized in vitro by incubation with P (1 μM) and cAMP (0.1 mg/mL) in the presence, or absence, of DHEA (10 nM, 100 nM). Main Outcome Measure(s) Secretion of androgens was assessed by ELISA. Expression of decidualization markers and endometrial receptivity markers was assessed by quantitative polymerase chain reaction and ELISA. Result(s) Decidualization responses were retained in hESF isolated from women of advanced reproductive age. Supplementation with DHEA increased androgen biosynthesis and concentrations of T and dihydrotestosterone were ∼3× greater after coincubation with DHEA compared with hESF stimulated with decidualization alone. Addition of DHEA to decidualized hESF increased expression of the decidualization markers IGFBP1 and PRL and the endometrial receptivity marker SPP1. DHEA enhanced secretion of IGFBP1, PRL, and SPP1 proteins maximally by day 8 of the decidualization time course concomitant with peak androgen concentrations. Conclusion(s) These novel results demonstrate DHEA can enhance in vitro decidualization responses of hESF from women of advanced reproductive age. Supplementation with DHEA during the receptive phase may augment endometrial function and improve pregnancy rates in natural or assisted reproductive cycles.
Collapse
Affiliation(s)
- Douglas A Gibson
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.
| | - Ioannis Simitsidellis
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Olympia Kelepouri
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Hilary O D Critchley
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Philippa T K Saunders
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
41
|
Keane KN, Hinchliffe PM, Rowlands PK, Borude G, Srinivasan S, Dhaliwal SS, Yovich JL. DHEA Supplementation Confers No Additional Benefit to that of Growth Hormone on Pregnancy and Live Birth Rates in IVF Patients Categorized as Poor Prognosis. Front Endocrinol (Lausanne) 2018; 9:14. [PMID: 29445356 PMCID: PMC5797762 DOI: 10.3389/fendo.2018.00014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/12/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In vitro fertilization (IVF) patients receive various adjuvant therapies to enhance success rates, but the true benefit is actively debated. Growth hormone (GH) and dehydroepiandrosterone (DHEA) supplementation were assessed in women undergoing fresh IVF transfer cycles and categorized as poor prognosis from five criteria. METHODS Data were retrospectively analyzed from 626 women undergoing 626 IVF cycles, where they received no adjuvant, GH alone, or GH-DHEA in combination. A small group received DHEA alone. The utilization of adjuvants was decided between the attending clinician and the patient depending on various factors including cost. RESULTS Despite patients being significantly older with lower ovarian reserve, live birth rates were significantly greater with GH alone (18.6%) and with GH-DHEA (13.0%) in comparison to those with no adjuvant (p < 0.003). No significant difference was observed between the GH groups (p = 0.181). Overall, patient age, quality of the transferred embryo, and GH treatment were the only significant independent predictors of live birth chance. Following adjustment for patient age, antral follicle count, and quality of transferred embryo, GH alone and GH-DHEA led to a 7.1-fold and 5.6-fold increase in live birth chance, respectively (p < 0.000). CONCLUSION These data indicated that GH adjuvant may support more live births, particularly in younger women, and importantly, the positive effects of GH treatment were still observed even if DHEA was also used in combination. However, supplementation with DHEA did not indicate any potentiating benefit or modify the effects of GH treatment. Due to the retrospective design, and the risk of a selection bias, caution is advised in the interpretation of the data.
Collapse
Affiliation(s)
- Kevin N. Keane
- School of Pharmacy and Biomedical Science, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- PIVET Medical Centre, Perth, WA, Australia
| | | | | | | | | | - Satvinder S. Dhaliwal
- Faculty of Health Sciences, School of Public Health, Curtin University, Perth, WA, Australia
| | - John L. Yovich
- School of Pharmacy and Biomedical Science, Faculty of Health Sciences, Curtin University, Perth, WA, Australia
- PIVET Medical Centre, Perth, WA, Australia
- *Correspondence: John L. Yovich,
| |
Collapse
|
42
|
Çelik Ö, Acet M, İmren A, Çelik N, Erşahin A, Aktun LH, Otlu B, Çelik S, Çalışkan E, Ünlü C. DHEA supplementation improves endometrial HOXA-10 mRNA expression in poor responders. J Turk Ger Gynecol Assoc 2017; 18:160-166. [PMID: 29278227 PMCID: PMC5776153 DOI: 10.4274/jtgga.2017.0054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: The study was planned to investigate whether DHEA supplementation had an impact on endometrial receptivity in women who were poor responders (POR). Material and Methods: Twenty-eight POR women who were undergoing hysteroscopy and five fertile control subjects were included. The POR women were equally subdivided into two separate groups as patients who were currently using DHEA and those who were not. Endometrial samples of the subjects were obtained during hysteroscopy at the late follicular phase. Expression levels of endometrial HOXA-10, HOXA-11, and LIF mRNA were measured with the using real-time polymerase chain reaction. Spontaneous clinical pregnancy rates were also noted. Results: Compared with POR women who were not given DHEA, upregulated endometrial HOXA-10 (7.33-fold) and HOXA-11 (2.39-fold) mRNA expression were detected in POR women on DHEA. The increase in HOXA-10 mRNA was significant (p<0.03). The fold increase in HOXA-11 mRNA was found as 2.39, which indicated a positive upregulation. However, this fold increment was insignificant (p<0.45). An insignificant increase in spontaneous clinical pregnancy rates in POR women on DHEA (53.3%) was observed compared with POR women who were not given DHEA (43.8%). Conclusion: Oral DHEA supplementation in POR upregulates endometrial HOXA-10 mRNA expression, which is known to positively modulate endometrial receptivity.
Collapse
Affiliation(s)
- Önder Çelik
- Private Clinic, Obstetrics and Gynecology, Uşak, Turkey
| | - Mustafa Acet
- Department of Obstetrics and Gynecology, Medipol University Faculty of Medicine, İstanbul, Turkey
| | - Aytaç İmren
- Clinic of Obstetric and Gynecology, Medical Park Hospital, Uşak, Turkey
| | - Nilüfer Çelik
- Clinic of Biochemistry, Behçet Uz Children's Hospital, İzmir, Turkey
| | - Aynur Erşahin
- Department of Obstetrics and Gynecology, Bahçeşehir University Faculty of Medicine, İstanbul, Turkey
| | - Lebriz Hale Aktun
- Department of Obstetrics and Gynecology, Medipol University Faculty of Medicine, İstanbul, Turkey
| | - Barış Otlu
- Department of Medical Microbiology, İnönü University Faculty of Medicine, Malatya, Turkey
| | | | - Eray Çalışkan
- Department of Obstetrics and Gynecology, Bahçeşehir University Faculty of Medicine, Kocaeli, Turkey
| | - Cihat Ünlü
- Department of Obstetrics and Gynecology, Acıbadem University Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
43
|
Gonda KJ, Domar AD, Gleicher N, Marrs RP. Insights from clinical experience in treating IVF poor responders. Reprod Biomed Online 2017; 36:12-19. [PMID: 29223475 DOI: 10.1016/j.rbmo.2017.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/27/2017] [Accepted: 09/19/2017] [Indexed: 12/18/2022]
Abstract
'Poor responders' is a term used to describe a subpopulation of IVF patients who do not respond well to ovarian stimulation with gonadotrophins. While there is no standard definition of a poor responder, these patients tend to be of advanced maternal age (≥40 years), have a history of poor ovarian response with conventional stimulation protocols, and/or have low ovarian reserve. Despite the heterogeneity of this patient group, there are characteristics and needs common to many poor responders that can be addressed through a holistic approach. Stimulation during the earlier stages of follicle maturation may help synchronize follicle development for improved response to later gonadotrophin stimulation, and supplementation with dehydroepiandrosterone or human growth hormone may promote early follicle development in poor responders. IVF protocols should be specifically tailored to poor responders to complement the patient's natural cycle. Because poor responders tend to have high levels of stress and anxiety, patients should receive psychological counselling and support, both prior to and during IVF cycles, to ensure optimal outcomes and improve patients' experience. It is important to set realistic expectations with poor responders and their partners to help patients make informed decisions and better manage their distress and anxiety.
Collapse
Affiliation(s)
| | - Alice D Domar
- Domar Center for Mind/Body Health, Waltham, MA, USA; Boston IVF, Waltham, MA, USA; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Norbert Gleicher
- Center for Human Reproduction, New York, NY, USA; Foundation for Reproductive Medicine, New York, NY, USA; Rockefeller University, New York, NY, USA; Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
44
|
Chu N, Gui Y, Qiu X, Zhang N, Li L, Li D, Tang W, Gober HJ, Zhang B, Wang L. The effect of DHEA on apoptosis and cohesin levels in oocytes in aged mice. Biosci Trends 2017; 11:427-438. [PMID: 28717062 DOI: 10.5582/bst.2017.01108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Female fertility declines with age as the number of ovarian follicles decreases and aneuploidy increases. Degradation of the cohesin complex might be responsible for age-related aneuploidy. Dehydroepiandrosterone (DHEA) can improve the ovarian reserve and reduce the rate of aneuploidy, but the relationship between DHEA and cohesin levels in oocytes is still unknown. The aim of the current study was to evaluate the effect of the supplement DHEA on ovarian function, including the number of follicles and cohesin levels in oocytes. C57BL/6J mice at 3 weeks, 6 weeks, 12 weeks, 6 months, and 10 months of age were used to obtain a systematic view into follicle apoptosis and cohesin levels in oocytes. Nine-month-old C57BL/6J mice were administered saline (n = 5), 17β-estradiol (100 µg/kg per day, n = 5), or DHEA (5mg/Kg per day, n = 5). After 4 weeks, aged mice were weighed and sacrificed, and ovarian tissue samples were prepared. Anti-VASA staining and HE staining were used to count the number of follicles. Anti-γH2AX staining and TUNEL were used to measure follicle apoptosis and immunofluorescent staining was used to detect the levels of three oocyte cohesin subunits: REC8, SMC1β, and SMC3. Administration of the supplements 17β-estradiol and DHEA to aged mice increased the number of primordial and primary follicles and decreased the age-related apoptosis of follicles. Levels of the cohesin subunits REC8 and SMC1β declined with age, but DHEA and 17β-estradiol tended to delay that decline. The supplement DHEA increased the number of primordial and primary follicles in aged mice by inhibiting follicle apoptosis and tended to delay the decrease in cohesin levels in oocytes.
Collapse
Affiliation(s)
- Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Yuyan Gui
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Na Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Lisha Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo
| | | | - Bin Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Fudan University Shanghai Medical College
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
45
|
Narkwichean A, Maalouf W, Baumgarten M, Polanski L, Raine-Fenning N, Campbell B, Jayaprakasan K. Efficacy of Dehydroepiandrosterone (DHEA) to overcome the effect of ovarian ageing (DITTO): A proof of principle double blinded randomized placebo controlled trial. Eur J Obstet Gynecol Reprod Biol 2017; 218:39-48. [PMID: 28934714 DOI: 10.1016/j.ejogrb.2017.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 09/03/2017] [Accepted: 09/08/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To evaluate the effect of DHEA supplementation on In-Vitro Fertilisation (IVF) outcome as assessed by ovarian response, oocyte developmental competence and live birth rates in women predicted to have poor ovarian reserve (OR). The feasibility of conducting a large trial is also assessed by evaluating the recruitment rates and compliance of the recruited participants with DHEA/placebo intake and follow-up rates. STUDY DESIGN A single centre, double blinded, placebo controlled, randomized trial was performed over two years with 60 women undergoing in-vitro fertilisation (IVF). Subjects were randomized, based on a computer-generated pseudo-random code to receive either DHEA or placebo with both capsules having similar colour, size and appearance. 60 women with poor OR based on antral follicle count or anti-Mullerian hormone thresholds undergoing IVF were recruited. They were randomised to receive DHEA 75mg/day or placebo for at-least 12 weeks before starting ovarian stimulation. They had long protocol using hMG 300 IU/day. Data analysed by "intention to treat". Ovarian response, live birth rates and molecular markers of oocyte quality were compared between the study and control groups. RESULTS The recruitment rate was 39% (60/154). A total of 52 participants (27 versus 25 in the study and placebo groups) were included in the final analysis after excluding eight. While the mean (standard deviation) DHEA levels were similar at recruitment (9.4 (5) versus 7.5 (2.4) ng/ml; P=0.1), the DHEA levels at pre-stimulation were higher in the study group than in the controls (16.3 (5.8) versus 11.1 (4.5) ng/ml; P<0.01). The number (median, range) of oocytes retrieved (4, 0-18 versus 4, 0-15 respectively; P=0.54) and live birth rates (7/27, 26% versus 8/25, 32% respectively; RR (95% CI): 0.74 (0.22-2.48) and mRNA expression of developmental biomarkers in granulosa and cumulus cells were similar between the groups. CONCLUSION Pre-treatment DHEA supplementation, albeit statistical power in this study is low, did not improve the response to controlled ovarian hyperstimulation or oocyte quality or live birth rates during IVF treatment with long protocol in women predicted to have poor OR.
Collapse
Affiliation(s)
- Amarin Narkwichean
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom; Department of Obstetrics and Gynaecology, Faculty of Medicine, Srinakharinwirot University, Nakhon-Nayok, 26120, Thailand
| | - Walid Maalouf
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom
| | - Miriam Baumgarten
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom
| | - Lukasz Polanski
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom
| | - Nick Raine-Fenning
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom; NURTURE Fertility, Nottingham, NG10 5QG, United Kingdom
| | - Bruce Campbell
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom
| | - Kannamannadiar Jayaprakasan
- Division of Obstetrics and Gynaecology, Faculty of Medicine & Health Sciences, University of Nottingham, Nottingham, Nottinghamshire, NG7 2UH, United Kingdom; Derby Fertility Unit, Royal Derby Hospital, Derby, DE22 3NE, United Kingdom.
| |
Collapse
|
46
|
Hui G, Tian X, Ruihong M, Zhimei Z, Xueru S, Baojuan W, Lijing L, Kaimei H, Guoqing W, Yu F, Guimin H. Heyan Kuntai capsule versus dehydroepiandrosterone in treating Chinese patients with infertility caused by diminished ovarian reserve: a multicenter, randomized controlled trial. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30160-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
47
|
Al-Turki HA. Dehydroepiandrosterone supplementation in women undergoing assisted reproductive technology with poor ovarian response. A prospective case-control study. J Int Med Res 2017; 46:143-149. [PMID: 28758852 PMCID: PMC6011324 DOI: 10.1177/0300060517720005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective The effects of dehydroepiandrosterone (DHEA) supplementation in Saudi Arabian women with poor ovarian response (POR) is presently unknown. The present study aimed to assess the benefits of DHEA supplementation in women undergoing in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI). Methods This was a prospective case-control study involving 62 women who were diagnosed with POR and underwent IVF/ICSI between January 2012 and June 2016. The positive influence of DHEA in 34 women, compared with 28 women without supplementation, was defined as improvements in the number of oocytes retrieved, the fertilization rate, the number of grade I embryos generated and the pregnancy rate. Results Both groups were evenly matched for age, body mass index and laboratory test parameters. There were statistically significant differences between the groups with and without DHEA supplementation for oocyte yield (6.35 ± 2.41 versus 3.98 ± 3.2), Grade I embryos generated (55% versus 30%), positive pregnancy rate (21/34 versus 10/28), and live birth rate (18/34 versus 4/28). Conclusion DHEA supplementation in women with POR had a positive effect on hormonal profiles, the quality of the endometrium, the number of oocytes retrieved, the quality of embryos, and the pregnancy and live birth rates.
Collapse
Affiliation(s)
- Haifa A Al-Turki
- Associate Professor of Infertility and Reproductive Medicine and Consultant Obstetrician and Gynecologist, Department of Obstetrics and Gynecology, College of Medicine, Imam AbdulRahman Bin Faisal University, Dammam and King Fahd Hospital of the University, Al-Khobar, Saudi Arabia
| |
Collapse
|
48
|
Abide Yayla C, Ozkaya E, Kayatas Eser S, Sanverdi I, Devranoglu B, Kutlu T. Association of basal serum androgen levels with ovarian response and ICSI cycle outcome. Ir J Med Sci 2017; 187:409-415. [DOI: 10.1007/s11845-017-1665-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 01/05/2023]
|
49
|
Griffith OW, Brandley MC, Whittington CM, Belov K, Thompson MB. Comparative genomics of hormonal signaling in the chorioallantoic membrane of oviparous and viviparous amniotes. Gen Comp Endocrinol 2017; 244:19-29. [PMID: 27102939 DOI: 10.1016/j.ygcen.2016.04.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 01/17/2016] [Accepted: 04/16/2016] [Indexed: 01/13/2023]
Abstract
In oviparous amniotes (reptiles, birds, and mammals) the chorioallantoic membrane (CAM) lines the inside of the egg and acts as the living point of contact between the embryo and the outside world. In livebearing (viviparous) amniotes, communication during embryonic development occurs across placental tissues, which form between the uterine tissue of the mother and the CAM of the embryo. In both oviparous and viviparous taxa, the CAM is at the interface of the embryo and the external environment and can transfer signals from there to the embryo proper. To understand the evolution of placental hormone production in amniotes, we examined the expression of genes involved in hormone synthesis, metabolism, and hormone receptivity in the CAM of species across the amniote phylogeny. We collected transcriptome data for the chorioallantoic membranes of the chicken (oviparous), the lizards Lerista bougainvillii (both oviparous and viviparous populations) and Pseudemoia entrecasteauxii (viviparous), and the horse Equus caballus (viviparous). The viviparous taxa differ in their mechanisms of nutrient provisioning: L. bougainvillii is lecithotrophic (embryonic nourishment is provided via the yolk only), but P. entrecasteauxii and the horse are placentotrophic (embryos are nourished via placental transport). Of the 423 hormone-related genes that we examined, 91 genes are expressed in all studied species, suggesting that the chorioallantoic membrane ancestrally had an endocrine function. Therefore, the chorioallantoic membrane appears to be a highly hormonally active organ in all amniotes. No genes are expressed only in viviparous species, suggesting that the evolution of viviparity has not required the recruitment of any specific hormone-related genes. Our data suggest that the endocrine function of the CAM as a placental tissue evolved in part through co-option of ancestral gene expression patterns.
Collapse
Affiliation(s)
- Oliver W Griffith
- School of Life and Environmental Sciences, Heydon-Laurence Building, University of Sydney, Sydney, NSW 2006, Australia; Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States.
| | - Matthew C Brandley
- School of Life and Environmental Sciences, Heydon-Laurence Building, University of Sydney, Sydney, NSW 2006, Australia; New York University - Sydney, The Rocks, NSW 2000, Australia
| | - Camilla M Whittington
- School of Life and Environmental Sciences, Heydon-Laurence Building, University of Sydney, Sydney, NSW 2006, Australia; Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Katherine Belov
- Faculty of Veterinary Science, University of Sydney, Sydney, NSW 2006, Australia
| | - Michael B Thompson
- School of Life and Environmental Sciences, Heydon-Laurence Building, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
50
|
Lin LT, Wang PH, Chen SN, Li CJ, Wen ZH, Cheng JT, Tsui KH. Protection of cumulus cells following dehydroepiandrosterone supplementation. Gynecol Endocrinol 2017; 33:100-104. [PMID: 27684542 DOI: 10.1080/09513590.2016.1214262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Growing studies have demonstrated that dehydroepiandrosterone (DHEA) may improve fertility outcomes in poor ovarian responders (PORs). The aim of this study was to compare clinical outcomes and cumulus cell (CC) expression before and after DHEA treatment in PORs undergoing in vitro fertilization (IVF) cycles. METHODS Six patients with poor ovarian response were enrolled in the study according to Bologna criteria. DHEA was supplied at least 2 months before patients entered into the next IVF cycle. Expression of apoptosis-related genes in CCs was determined by quantitative real-time PCR. Mitochondrial dehydrogenase activity of CCs was assessed by cell counting kit-8 assay. RESULTS Metaphase II oocytes, maturation rate, embryos at Day 3, and fertilization rate significantly increased following DHEA treatment. Expression of cytochrome c, caspase 9, and caspase 3 genes in CCs were significantly reduced after DHEA therapy. Additionally, increased mitochondrial activity of CCs was observed following DHEA supplementation. CONCLUSIONS DHEA supplementation may protect CCs via improved mitochondrial activity and decreased apoptosis, leading to better clinical outcomes in PORs.
Collapse
Affiliation(s)
- Li-Te Lin
- a Department of Biological Science , National Sun Yat-sen University , Kaohsiung , Taiwan
- b Department of Obstetrics and Gynecology , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan
- c Department of Obstetrics and Gynecology , National Yang-Ming University School of Medicine , Taipei , Taiwan
| | - Peng-Hui Wang
- c Department of Obstetrics and Gynecology , National Yang-Ming University School of Medicine , Taipei , Taiwan
- d Division of Gynecology , Department of Obstetrics and Gynecology, Taipei Veterans General Hospital , Taipei , Taiwan
- e Department of Obstetrics and Gynecology , National Yang-Ming University Hospital , Ilan , Taiwan
- f Immunology Center, Taipei Veterans General Hospital , Taipei , Taiwan
- g Department of Medical Research , China Medical University Hospital , Taichung , Taiwan
| | - San-Nung Chen
- b Department of Obstetrics and Gynecology , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan
| | - Chia-Jung Li
- h Institute of Clinical Medicine, College of Medicine, National Cheng Kung University , Taiwan
| | - Zhi-Hong Wen
- i Department of Marine Biotechnology and Resources , National Sun Yat-sen University , Kaohsiung , Taiwan , and
| | - Jiin-Tsuey Cheng
- a Department of Biological Science , National Sun Yat-sen University , Kaohsiung , Taiwan
| | - Kuan-Hao Tsui
- b Department of Obstetrics and Gynecology , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan
- c Department of Obstetrics and Gynecology , National Yang-Ming University School of Medicine , Taipei , Taiwan
- j Department of Pharmacy and Graduate Institute of Pharmaceutical Technology , Tajen University , Pingtung County , Taiwan
| |
Collapse
|