1
|
Sallicandro L, Gliozheni E, Feudi D, Sabbatini P, Pellegrino RM, Alabed HBR, Baldini D, Gerli S, Alviggi C, Cascardi E, Cicinelli E, Malvasi A, Fioretti B. Increased Vasoactive Intestinal Peptide (VIP) in polycystic ovary syndrome patients undergoing IVF. Front Endocrinol (Lausanne) 2024; 15:1331282. [PMID: 38774232 PMCID: PMC11106456 DOI: 10.3389/fendo.2024.1331282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction Polycystic ovary syndrome (PCOS) is a common multifactorial and polygenic disorder of the endocrine system, affecting up to 20% of women in reproductive age with a still unknown etiology. Follicular fluid (FF) represents an environment for the normal development of follicles rich in metabolites, hormones and neurotransmitters, but in some instances of PCOS the composition can be different. Vasoactive intestinal peptide (VIP) is an endogenous autonomic neuropeptide involved in follicular atresia, granulosa cell physiology and steroidogenesis. Methods ELISA assays were performed to measure VIP and estradiol levels in human follicular fluids, while AMH, FSH, LH, estradiol and progesterone in the plasma were quantified by chemiluminescence. UHPLC/QTOF was used to perform the untargeted metabolomic analysis. Results Our ELISA and metabolomic results show: i) an increased concentration of VIP in follicular fluid of PCOS patients (n=9) of about 30% with respect to control group (n=10) (132 ± 28 pg/ml versus 103 ± 26 pg/ml, p=0,03) in women undergoing in vitro fertilization (IVF), ii) a linear positive correlation (p=0.05, r=0.45) between VIP concentration and serum Anti-Müllerian Hormone (AMH) concentration and iii) a linear negative correlation between VIP and noradrenaline metabolism. No correlation between VIP and estradiol (E2) concentration in follicular fluid was found. A negative correlation was found between VIP and noradrenaline metabolite 3,4-dihydroxyphenylglycolaldehyde (DOPGAL) in follicular fluids. Conclusion VIP concentration in follicular fluids was increased in PCOS patients and a correlation was found with noradrenaline metabolism indicating a possible dysregulation of the sympathetic reflex in the ovarian follicles. The functional role of VIP as noradrenergic modulator in ovarian physiology and PCOS pathophysiology was discussed.
Collapse
Affiliation(s)
- Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Elko Gliozheni
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Davide Feudi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Paola Sabbatini
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
| | | | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Domenico Baldini
- In Vitro Fertilization (IVF) Center, Momo Fertilife, Bisceglie, Italy
| | - Sandro Gerli
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Perugia, Italy
- Department of Obstetrics and Gynecology, Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Carlo Alviggi
- Department of Clinical Gynecological Emergency, Obstetrics and Reproductive Medicine, University Federico II, Naples, Italy
| | - Eliano Cascardi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ettore Cicinelli
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Bari, Italy
| | - Antonio Malvasi
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, Bari, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
- Department of Obstetrics and Gynecology, Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
2
|
Liaghatdar A, Mazaheri-Tehrani S, Fakhrolmobasheri M. Management of Hypertension in Patients With Polycystic Ovarian Syndrome: A Mini-Review. Cardiol Rev 2024:00045415-990000000-00199. [PMID: 38305409 DOI: 10.1097/crd.0000000000000635] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Polycystic ovarian syndrome (PCOS) is a common problem among young women. It is characterized mainly by hyperandrogenism features, such as hirsutism, menstrual problems, and anovulation. Diagnosis is based on the existence of 2 items out of, oligo-ovulation, hyperandrogenism features, and ultrasounds findings. Cardiovascular complications such as hypertension are a prevalent serious condition in these patients which is mainly predisposed by the high levels of androgens, and insulin resistance. High blood pressure should be controlled well to prevent the progression of other serious conditions. Various antihypertensive drugs could be prescribed. However, in selecting an antihypertensive medication, other therapeutic properties of the drug should also be considered. Up to now, many clinicians do not differ between PCOS patients with hypertension and other hypertensive patients. However, being aware of the potential effects of each hypertension drug could help to choose better options for the patient. Here is a brief review of how each antihypertensive drug could affect PCOS women and if they cause any improvement in the disorder progression.
Collapse
Affiliation(s)
- Amin Liaghatdar
- From the Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sadegh Mazaheri-Tehrani
- From the Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Fakhrolmobasheri
- From the Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
3
|
Robeva R, Elenkova A, Kirilov G, Zacharieva S. Plasma-free metanephrines, nerve growth factor, and renalase significance in patients with PCOS. Endocrine 2023; 81:602-612. [PMID: 37248367 PMCID: PMC10226715 DOI: 10.1007/s12020-023-03404-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023]
Abstract
PURPOSE Polycystic ovarian syndrome (PCOS) is a common heterogeneous condition with probably multifactorial genesis. Animal studies have proven the essential role of the sympathetic nervous system in the syndrome development, while human studies are still contradictory. The present study aims to investigate the possible influence of plasma-free metanephrine (MN), and normetanephrine (NMN), nerve growth factor (NGF), and renalase (RNL) on the hormonal and metabolic parameters in women with PCOS and healthy controls. METHODS Fifty patients with PCOS and 30 healthy women participated in the study. The plasma-free MN and NMN, NGF, RNL, anti-Mullerian hormone (AMH), gonadotropin, androgen levels, and metabolic parameters were investigated. RESULTS Plasma-free NMN and NGF concentrations were increased in PCOS individuals, while RNL levels were decreased compared to healthy volunteers. Increased plasma-free NMN (OR = 1.0213 [95%CI 1.0064-1.0364], p = 0.005) and NGF (OR = 1.0078 [95%CI 1.0001-1.0155], p = 0.046) but not MN or RNL levels were associated with a higher risk of PCOS after adjustment for age. Plasma-free NMN levels were positively associated with the LH (r = +0.253; p = 0.039). androstenedione (r = +0.265; p = 0.029), 17-OH progesterone (r = +0.285; p = 0.024), NGF (r = +0.320; p = 0.008), and AMH (r = +0.417; p < 0.001) concentrations of the investigated women. RNL levels were inversely related to the BMI (r = -0.245; p = 0.029), HOMA-IR (r = -0.250; p = 0.030), free testosterone (r = -0.303; p = 0.006) levels. systolic (r = -0.294; p = 0.008) and diastolic (r = -0.342; p = 0.002) blood pressure. CONCLUSIONS Increased sympathetic noradrenergic activity and NGF synthesis might be related to the increased AMH and delta-4 androgen levels in a subgroup of PCOS patients. RNL levels might influence the metabolic status of PCOS patients. Further studies are needed to explore the significance of adrenal medullar and autonomic dysfunction for developing different PCOS phenotypes and their subsequent cardiovascular complications.
Collapse
Affiliation(s)
- Ralitsa Robeva
- Department of Endocrinology, Faculty of Medicine, Medical University - Sofia, USHATE "Acad. Iv. Penchev", 2, Zdrave Str., 1431, Sofia, Bulgaria.
| | - Atanaska Elenkova
- Department of Endocrinology, Faculty of Medicine, Medical University - Sofia, USHATE "Acad. Iv. Penchev", 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Georgi Kirilov
- Department of Endocrinology, Faculty of Medicine, Medical University - Sofia, USHATE "Acad. Iv. Penchev", 2, Zdrave Str., 1431, Sofia, Bulgaria
| | - Sabina Zacharieva
- Department of Endocrinology, Faculty of Medicine, Medical University - Sofia, USHATE "Acad. Iv. Penchev", 2, Zdrave Str., 1431, Sofia, Bulgaria
| |
Collapse
|
4
|
Atiakshin D, Patsap O, Kostin A, Mikhalyova L, Buchwalow I, Tiemann M. Mast Cell Tryptase and Carboxypeptidase A3 in the Formation of Ovarian Endometrioid Cysts. Int J Mol Sci 2023; 24:ijms24076498. [PMID: 37047472 PMCID: PMC10095096 DOI: 10.3390/ijms24076498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
The mechanisms of ovarian endometrioid cyst formation, or cystic ovarian endometriosis, still remain to be elucidated. To address this issue, we analyzed the involvement of mast cell (MC) tryptase and carboxypeptidase A3 (CPA3) in the development of endometriomas. It was found that the formation of endometrioid cysts was accompanied by an increased MC population in the ovarian medulla, as well as by an MC appearance in the cortical substance. The formation of MC subpopulations was associated with endometrioma wall structures. An active, targeted secretion of tryptase and CPA3 to the epithelium of endometrioid cysts, immunocompetent cells, and the cells of the cytogenic ovarian stroma was detected. The identification of specific proteases in the cell nuclei of the ovarian local tissue microenvironment suggests new mechanisms for the regulatory effects of MCs. The cytoplasmic outgrowths of MCs propagate in the structures of the stroma over a considerable distance; they offer new potentials for MC effects on the structures of the ovarian-specific tissue microenvironment under pathological conditions. Our findings indicate the potential roles of MC tryptase and CPA3 in the development of ovarian endometriomas and infer new perspectives on their uses as pharmacological targets in personalized medicine.
Collapse
Affiliation(s)
- Dmitri Atiakshin
- Research and Educational Resource Centre for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
- Research Institute of Experimental Biology and Medicine, Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Olga Patsap
- Research and Educational Resource Centre for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - Andrey Kostin
- Research and Educational Resource Centre for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | | | - Igor Buchwalow
- Research and Educational Resource Centre for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
- Institute for Hematopathology, 22547 Hamburg, Germany
| | | |
Collapse
|
5
|
Szukiewicz D, Wojdasiewicz P, Watroba M, Szewczyk G. Mast Cell Activation Syndrome in COVID-19 and Female Reproductive Function: Theoretical Background vs. Accumulating Clinical Evidence. J Immunol Res 2022; 2022:9534163. [PMID: 35785029 PMCID: PMC9242765 DOI: 10.1155/2022/9534163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), a pandemic disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, can affect almost all systems and organs of the human body, including those responsible for reproductive function in women. The multisystem inflammatory response in COVID-19 shows many analogies with mast cell activation syndrome (MCAS), and MCAS may be an important component in the course of COVID-19. Of note, the female sex hormones estradiol (E2) and progesterone (P4) significantly influence mast cell (MC) behavior. This review presents the importance of MCs and the mediators from their granules in the female reproductive system, including pregnancy, and discusses the mechanism of potential disorders related to MCAS. Then, the available data on COVID-19 in the context of hormonal disorders, the course of endometriosis, female fertility, and the course of pregnancy were compiled to verify intuitively predicted threats. Surprisingly, although COVID-19 hyperinflammation and post-COVID-19 illness may be rooted in MCAS, the available clinical data do not provide grounds for treating this mechanism as significantly increasing the risk of abnormal female reproductive function, including pregnancy. Further studies in the context of post COVID-19 condition (long COVID), where inflammation and a procoagulative state resemble many aspects of MCAS, are needed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Wojdasiewicz
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Mateusz Watroba
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Szewczyk
- Department of Biophysics Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
Metanephrine and Normetanephrine Urine Excretion in Patients with PCOS. ACTA MEDICA BULGARICA 2022. [DOI: 10.2478/amb-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Aims: The present study aims to investigate the association between catecholamine metabolites (normetanephrine [NMN] and metanephrine [MN]) and polycystic ovarian syndrome (PCOS) and its clinical manifestations.
Materials and methods: A total of 60 female patients (18-37 years) were included in the study. Twenty-five healthy women and 35 patients with PCOS provided blood and urine samples. Anthropometric, biochemical, and hormonal parameters, e.g. immunoreactive insulin, total testosterone, gonadotropins, and dehydroepiandrosterone-sulfate (DHEAS) have been investigated. Twenty-four-hour urinary metanephrines (MN and NMN) were measured by radioimmunoassay technique.
Results: MN and NMN showed a strong positive correlation (r = +0.645, p < 0.001) in women with PCOS, but nephrines were not significantly associated with the hormonal, anthropometric, and metabolic parameters of the patients (p > 0.05 for all). No differences in urinary nephrines concentrations between women with PCOS and healthy individuals after adjustment for age and BMI were established. However, lower 24-hour urinary MN levels predicted the development of menstrual irregularity after adjustment for age and BMI (OR = 1,015 [95% CI 1,001-1,029], p = 0,036).
Conclusions: Adrenomedullary dysfunction might influence the development of chronic anovulation in young women.
Collapse
|
7
|
Frungieri MB, Calandra RS, Bartke A, Matzkin ME. Male and female gonadal ageing: its impact on health span and life span. Mech Ageing Dev 2021; 197:111519. [PMID: 34139215 DOI: 10.1016/j.mad.2021.111519] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Ageing is linked to changes in the hypothalamic-pituitary-gonadal axis and a progressive decline in gonadal function. While women become infertile when they enter menopause, fertility decline in ageing men does not necessarily involve a complete cessation of spermatogenesis. Gonadal dysfunction in elderly people is characterized by morphological, endocrine and metabolic alterations affecting the reproductive function and quality of life. With advancing age, sexuality turns into a critical emotional and physical factor actually defining the number of years that ageing people live a healthy life. Gonadal ageing correlates with comorbidities and an increased risk of age-related diseases including diabetes, kidney problems, cardiovascular failures and cancer. This article briefly summarizes the current state of knowledge on ovarian and testicular senescence, explores the experimental models used in the study of gonadal ageing, and describes the local pro-inflammatory, oxidative and apoptotic events and the associated signalling pathways that take place in the gonads while people get older. Overall, literature reports that ageing exacerbates a mutual crosstalk among oxidative stress, apoptosis and the inflammatory response in the gonads leading to detrimental effects on fertility. Data also highlight the clinical implications of novel therapeutic interventions using antioxidant, anti-apoptotic and anti-inflammatory drugs on health span and life span.
Collapse
Affiliation(s)
- Mónica B Frungieri
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina; Cátedra de Química, Ciclo Básico Común, Ciudad de Buenos Aires, C1405CAE, Argentina.
| | - Ricardo S Calandra
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina
| | - Andrzej Bartke
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University, School of Medicine, Springfield, IL 62702, USA
| | - María E Matzkin
- Instituto de Biología y Medicina Experimental, CONICET, Ciudad de Buenos Aires, C1428ADN, Argentina; Cátedra de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, C1121ABG, Argentina
| |
Collapse
|
8
|
Gu X, Li SY, DeFalco T. Immune and vascular contributions to organogenesis of the testis and ovary. FEBS J 2021; 289:2386-2408. [PMID: 33774913 PMCID: PMC8476657 DOI: 10.1111/febs.15848] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/07/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Gonad development is a highly regulated process that coordinates cell specification and morphogenesis to produce sex-specific organ structures that are required for fertility, such as testicular seminiferous tubules and ovarian follicles. While sex determination occurs within specialized gonadal supporting cells, sexual differentiation is evident throughout the entire organ, including within the interstitial compartment, which contains immune cells and vasculature. While immune and vascular cells have been traditionally appreciated for their supporting roles during tissue growth and homeostasis, an increasing body of evidence supports the idea that these cell types are critical drivers of sexually dimorphic morphogenesis of the gonad. Myeloid immune cells, such as macrophages, are essential for multiple aspects of gonadogenesis and fertility, including for forming and maintaining gonadal vasculature in both sexes at varying stages of life. While vasculature is long known for supporting organ growth and serving as an export mechanism for gonadal sex steroids in utero, it is also an important component of fetal testicular morphogenesis and differentiation; additionally, it is vital for ovarian corpus luteal function and maintenance of pregnancy. These findings point toward a new paradigm in which immune cells and blood vessels are integral components of sexual differentiation and organogenesis. In this review, we discuss the state of the field regarding the diverse roles of immune and vascular cells during organogenesis of the testis and ovary and highlight outstanding questions in the field that could stimulate new research into these previously underappreciated constituents of the gonad.
Collapse
Affiliation(s)
- Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, OH, USA
| |
Collapse
|
9
|
Manti M, Pui HP, Edström S, Risal S, Lu H, Lindgren E, Ohlsson C, Jerlhag E, Benrick A, Deng Q, Stener-Victorin E. Excess of ovarian nerve growth factor impairs embryonic development and causes reproductive and metabolic dysfunction in adult female mice. FASEB J 2020; 34:14440-14457. [PMID: 32892421 DOI: 10.1096/fj.202001060r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/05/2020] [Accepted: 08/13/2020] [Indexed: 12/30/2022]
Abstract
Nerve growth factor (NGF) is critical for the development and maintenance of the peripheral sympathetic neurons. NGF is also involved in the ovarian sympathetic innervation and in the development and maintenance of folliculogenesis. Women with the endocrine disorder, polycystic ovary syndrome (PCOS), have an increased sympathetic nerve activity and increased ovarian NGF levels. The role of ovarian NGF excess in the PCOS pathophysiology and in the PCOS-related features is unclear. Here, using transgenic mice overexpressesing NGF in the ovarian theca cells (17NF mice), we assessed the female embryonic development, and the reproductive and metabolic profile in adult females. Ovarian NGF excess caused growth restriction in the female fetuses, and a delayed gonocyte and primary oocyte maturation. In adulthood, the 17NF mice displayed irregular estrous cycles and altered ovarian expression of steroidogenic and epigenetic markers. They also exhibited an increased sympathetic output with increased circulating dopamine, and metabolic dysfunction reflected by aberrant adipose tissue morphology and function, impaired glucose metabolism, decreased energy expenditure, and hepatic steatosis. These findings indicate that ovarian NGF excess leads to adverse fetal development and to reproductive and metabolic complications in adulthood, mirroring common features of PCOS. This work provides evidence that NGF excess may be implicated in the PCOS pathophysiology.
Collapse
Affiliation(s)
- Maria Manti
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Han-Pin Pui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sonja Edström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sanjiv Risal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Haojiang Lu
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eva Lindgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Benrick
- Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,School of Health and Education, University of Skövde, Skövde, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
10
|
Zangeneh FZ, Bagheri M, Shoushtari MS, Naghizadeh MM. Expression of ADR-α1, 2 and ADR-β2 in cumulus cell culture of infertile women with polycystic ovary syndrome and poor responder who are a candidate for IVF: the novel strategic role of clonidine in this expression. J Recept Signal Transduct Res 2020; 41:263-272. [PMID: 32878560 DOI: 10.1080/10799893.2020.1806320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Alpha and beta-adrenoceptors (ADR-α1, 2, and β2) play a regulatory role in the folliculogenesis and steroidogenesis in the ovarian follicles. This study aimed to measure these adrenoceptors mRNA and its protein levels in cumulus cells (CCs) culture of poor ovarian reserve (POR) and polycystic ovarian syndrome (PCOS) infertile women (IVF candidate) and the effect of clonidine treatment at CCs culture. METHODS This case/control study was conducted in 2017 includes a control (donation oocytes) and two studies (PCO and POR) groups. The ovulation induction drugs were prescribed in all groups. After the oocyte puncture, the follicular fluid was collected and CCs were isolated were cultured. RNA was extracted and cDNA was synthesized and designed the primer for the ADR-α1, 2 and ADR-β2 gene expression. The protein levels were investigated by Western Blot. RESULTS The results showed a high level of three adrenergic expressions in PCO women compared to the control group (p-value <.001), which can be reduced by clonidine. POR group showed a significant decrease in the gene expression of ADR-α1 (p-value = .004) and ADR-α2 (p-value = .003) compared to the control group and clonidine treatment had no effect. CONCLUSION The significant increase of three adrenoceptors gene expression and protein levels in CCs culture indicate to the hyperactivity of the ovarian sympathetic nervous system at the receptor levels in women with PCOS, and clonidine confirmed it by reducing this expression. In POR women, the reduction of ADR-α1, 2 expressions maybe lead to the aging process in the ovary.
Collapse
Affiliation(s)
| | - Maryam Bagheri
- Department of Reproductive Health, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
11
|
Tong X, Liu Y, Xu X, Shi J, Hu W, Ma T, Cui P, Lu W, Pei Z, Xu M, Zhang F, Li X, Feng Y. Ovarian Innervation Coupling With Vascularity: The Role of Electro-Acupuncture in Follicular Maturation in a Rat Model of Polycystic Ovary Syndrome. Front Physiol 2020; 11:474. [PMID: 32547407 PMCID: PMC7273926 DOI: 10.3389/fphys.2020.00474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
Low-frequency electro-acupuncture (EA) has been shown to restore ovulation in patients with polycystic ovary syndrome (PCOS), and previous animal experiments showed that EA improves ovarian blood flow and angiogenesis. We performed EA for 4 weeks in dihydrotestosterone (DHT)-induced PCOS-like rats and investigated the three-dimensional (3D) ovarian innervation to determine the role of innervation in folliculogenesis and vascularity. Ovarian tissues were made transparent following the CUBIC 3D tissue-clearing protocol and were immunostained using antibodies against platelet endothelial cell adhesion molecule-1 and tyrosine hydroxylase to visualize the ovarian vasculature and innervation, respectively. This was followed by 3D imaging using lightsheet microscopy and analysis using the Imaris software. In control rats, ovarian innervation increased with age, and the neuronal branching started from the ovarian hilum and reached the individual follicles at different follicle stages. At the individual follicle level, each follicle was mainly innervated by one neuronal fiber. Compared with control rats, ovaries from DHT-treated PCOS-like rats had more antral follicles and fewer preovulatory follicles and corpora lutea. Furthermore, PCOS ovaries showed decreased innervation of blood vessels near the hilum and the surrounding individual antral follicles. EA in PCOS-like rats led to increased numbers of preovulatory follicles and corpora lutea together with increased innervation of blood vessels near the hilum. To determine the role of ovarian innervation, we further performed unilateral sectioning of the superior ovarian nerve (SON) in PCOS + EA rats and found that the left sectioned ovary had fewer preovulatory follicles and corpora lutea compared with those in the right non-sectioned ovary. In conclusion, ovarian innervation likely played an important role in folliculogenesis, and EA might restore PCOS pathophysiology by regulating ovarian innervation, at least partially mediated through the SON.
Collapse
Affiliation(s)
- Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yanjun Liu
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| | - Xiaoqing Xu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jiemei Shi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Tong Ma
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Peng Cui
- Department of Obstetrics and Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenhan Lu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhenle Pei
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mingzhen Xu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Feifei Zhang
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| | - Xin Li
- Department of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Ibrahim RO, Omer SH, Fattah CN. The Correlation between Hormonal Disturbance in PCOS Women and Serum Level of Kisspeptin. Int J Endocrinol 2020; 2020:6237141. [PMID: 32411228 PMCID: PMC7199587 DOI: 10.1155/2020/6237141] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/21/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Kisspeptin is a neuropeptide that upregulates gonadotropin-releasing hormone (GnRH) secretion. It is an essential element for the luteinizing hormone (LH) surge and ovulation. Women with polycystic ovary syndrome (PCOS) expose alteration in both GnRH and LH secretion levels. OBJECTIVE This paper aims to evaluate serum kisspeptin levels in healthy and polycystic ovarian syndrome women. Furthermore, it investigates the effect of obesity and age on circulating kisspeptin levels in both normal and PCOS women. Moreover, it points out the correlation between kisspeptin and other hormonal parameters. Methods and Patients. One hundred women (60 are with PCOS and 40 are normal) were enrolled in the study. Five milliliter samples of blood from all the patients and control women were obtained twice during the menstrual cycle. All the study samples were classified depending on the age factor for several subgroups. RESULTS Kisspeptin levels were higher in PCOS patients than those in the normal group. Kisspeptin correlated with serum free testosterone level (r=0.26). In healthy women, preovulatory kisspeptin levels were higher than follicular kisspeptin levels (P < 0.05), while this difference was insignificant in PCOS patients. The variation in serum kisspeptin levels between overweight/obese and normal-weight women was insignificant. In normal women, serum kisspeptin levels were higher in women >35 years than those <24 years at (P=0.03). CONCLUSION The serum kisspeptin level is higher in PCOS women. Its levels fluctuate during the menstrual cycle, but these fluctuations are disturbed in PCOS women. The effect of BMI on serum kisspeptin levels is insignificant, and kisspeptin serum levels increase with age.
Collapse
Affiliation(s)
- Razaw O. Ibrahim
- Department of Physiology, College of Medicine, University of Kirkuk, Kirkuk, Iraq
| | - Shirwan H. Omer
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
| | - Chro N. Fattah
- Department of Obstetrics and Gynecology, College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq
| |
Collapse
|
13
|
Venegas B, De León Gordillo LY, Rosas G, Espinoza JA, Morán C, Domínguez R, Morales-Ledesma L. In rats with estradiol valerate-induced polycystic ovary syndrome, the acute blockade of ovarian β-adrenoreceptors improve ovulation. Reprod Biol Endocrinol 2019; 17:95. [PMID: 31744506 PMCID: PMC6862835 DOI: 10.1186/s12958-019-0539-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome is characterized by hyperactivity of the ovarian sympathetic nervous system, increases in the content and release of norepinephrine, as well as decreases in the number of β-adrenoreceptors. In the present study, β-adrenoreceptors in the ovaries of rats with polycystic ovary syndrome were blocked and analyzed the resultant effects on ovulation, hormone secretion and the enzymes responsible for the synthesis of catecholamines. METHODS At 60 days of age, vehicle or estradiol valerate-treated rats were injected with propranolol [10- 4 M] into the ovarian bursas on oestrus day. The animals were sacrificed on the next day of oestrus, and the ovulation response, the steroid hormone levels in the serum and the immunoreactivity of tyrosine hydroxylase and dopamine β-hydroxylase in the ovaries were measured. RESULTS In animals with the induction of polycystic ovary syndrome and β-adrenoreceptor blocking, ovulation was restored in more than half of the animals and resulted in decreased hyperandrogenism with respect to the levels observed in the estradiol valerate-treated group. Tyrosine hydroxylase and dopamine β-hydroxylase were present in the theca cells of the growing follicles and the interstitial gland. Injection of propranolol restored the tyrosine hydroxylase and ovarian dopamine β-hydroxylase levels in rats with polycystic ovary syndrome induction. CONCLUSIONS The results suggest that a single injection into the ovarian bursas of propranolol, a nonselective antagonist of β-adrenoreceptor receptors, decreases the serum testosterone concentration and the formation of ovarian cysts, improving the ovulation rate that accompanies lower levels of tyrosine hydroxylase and dopamine β-hydroxylase in the ovary.
Collapse
Affiliation(s)
- Berenice Venegas
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico
- Área de Procesos Celulares Fundamentales, Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, 72570, Puebla, CP, Mexico
| | - Lizzbeth Yureli De León Gordillo
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico
| | - Gabriela Rosas
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico
| | - Julieta A Espinoza
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Roberto Domínguez
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico
| | - Leticia Morales-Ledesma
- Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP, 15000, México, DF, Mexico.
| |
Collapse
|
14
|
Moulana M. Immunophenotypic profile of leukocytes in hyperandrogenemic female rat an animal model of polycystic ovary syndrome. Life Sci 2019; 220:44-49. [PMID: 30708097 DOI: 10.1016/j.lfs.2019.01.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 11/17/2022]
Abstract
The immune etiology of polycystic ovary syndrome (PCOS) is an intriguing area. However, whether there is alteration in the leukocyte populations in different tissues remain ambiguous. AIM To characterize the leukocyte populations of hyperandrogenemic female (HAF) rat tissues. METHODS Female Sprague Dawley rats at 3 weeks of age were implanted subcutaneously with dihydrotestosterone (DHT) or placebo pellets. The rats were aged to 14-15 weeks and tissues were collected. RESULTS Peripheral blood (PB) and renal CD4+ (P < 0.03, P < 0.007), Th17 (P < 0.05, P < 0.002), and CD4+CD28null (P < 0.04, P < 0.001) were significantly increased in HAF rats compared to placebo, respectively, in spite of their lower percentage in the spleen. Although, the percentage of Treg T lymphocytes were significantly higher in the PB (P < 0.001) of HAF rats, the splenic (P < 0.01) and renal Treg cells (P < 0.03) were found to be significantly lower. Remarkably, HAF rats had higher renal mast cells (P < 0.00009) despite lower splenic (P < 0.002). The number of PB, renal, and splenic CD8+ T cells and IgM+-B cells in HAF rats remained unchanged. CONCLUSION Results from this study 1) provide the first evidence of significant alteration of T lymphocyte subsets and different leukocyte populations profile in a rat model of polycystic ovary syndrome, 2) demonstrate alteration of the immunological niche of blood, spleen, and kidney tissues in Hyperandrogenemia state in female rats, 3) imply potential immune system dysregulation in HAF rats which may suggest a link between excess androgen, chronic inflammation, and immune-mediated diseases in polycystic ovary syndrome patients.
Collapse
Affiliation(s)
- Mohadetheh Moulana
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, United States of America; Women's Health Research Center, University of Mississippi Medical Center, United States of America.
| |
Collapse
|
15
|
Kakoly NS, Moran LJ, Teede HJ, Joham AE. Cardiometabolic risks in PCOS: a review of the current state of knowledge. Expert Rev Endocrinol Metab 2019; 14:23-33. [PMID: 30556433 DOI: 10.1080/17446651.2019.1556094] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/03/2018] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Polycystic ovary syndrome (PCOS) is a common endocrine disorder affecting up to 18% women of reproductive age. It is associated with a range of metabolic, reproductive, and psychological features. Current evidence indicates a role of PCOS in the development of metabolic and increased cardiovascular risk factors (CVRF) with implications for compromised cardiovascular endpoint disease, which may have a considerable impact on health and health care costs. AREAS COVERED Existing studies examining long-term cardiometabolic health in PCOS are heterogeneous with inconsistent findings. In the current review, we aim to explore and critically review retrospective, prospective, meta-analysis and review articles relating to PCOS on cardiometabolic risk factors and clinical consequences to summarize the evidence, note evidence gaps, and suggest implications for future research. EXPERT COMMENTARY Although there is an established association between PCOS and metabolic health, implications on cardiac health are more uncertain with associations observed for CVRF and subclinical disease, yet limited and conflicting data on actual cardiovascular endpoints. There is a lack of population-based long-term studies examining cardiometabolic morbidity and mortality in PCOS with a need for further research to progress toward a better understanding of the long-term cardiometabolic impacts in women with PCOS.
Collapse
Affiliation(s)
- N S Kakoly
- a Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine , Monash University , Melbourne , Australia
| | - L J Moran
- a Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine , Monash University , Melbourne , Australia
| | - H J Teede
- a Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine , Monash University , Melbourne , Australia
- b Endocrine and Diabetes Units , Monash Health , Clayton , Australia
- c Monash Partners Academic Health Sciences Centre , Melbourne , Australia
| | - A E Joham
- a Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine , Monash University , Melbourne , Australia
- b Endocrine and Diabetes Units , Monash Health , Clayton , Australia
| |
Collapse
|
16
|
Álvarez D, Ceballo K, Olguín S, Martinez-Pinto J, Maliqueo M, Fernandois D, Sotomayor-Zárate R, Cruz G. Prenatal metformin treatment improves ovarian function in offspring of obese rats. J Endocrinol 2018; 239:325-338. [PMID: 30334444 DOI: 10.1530/joe-18-0352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023]
Abstract
Maternal obesity causes a wide range of impairment in offspring, such as metabolic and reproductive dysfunctions. We previously demonstrated that female offspring of obese rats have increased serum estradiol levels during early postnatal life, probably because of decreased hepatic cytochrome P450 3A2 levels, which could lead to early onset of puberty and polycystic ovary condition in adulthood. Using metformin during pregnancy and nursing to improve the metabolic status of obese mothers could prevent the sequence of events that lead to an increase in postnatal serum estradiol levels in female offspring and, hence, reproductive dysfunction. We found that metformin prevented an increase in serum estradiol levels at postnatal day 14 in female offspring of obese mothers, which was associated with a restoration of hepatic cytochrome P450 3A2 levels to control values. Treatment using metformin could not prevent advanced puberty, but we observed that the number of antral follicles, follicular cysts and multi-oocyte follicles returned to control values in the female offspring of obese mothers treated with metformin. We also observed an increase in the levels of norepinephrine and the norepinephrine metabolite 3-methoxy-4-hydroxyphenylglycol in the ovaries, indicating increased sympathetic activity in female offspring induced by an obesogenic uterine environment. We found that this effect was prevented by metformin administration. From the results of this study, we concluded that metformin administration to obese mothers during pregnancy and nursing partially prevents ovarian dysfunction in female offspring during adulthood.
Collapse
Affiliation(s)
- Daniela Álvarez
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karina Ceballo
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Sofía Olguín
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jonathan Martinez-Pinto
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Manuel Maliqueo
- Department of Medicine West Division, Endocrinology and Metabolism Laboratory, School of Medicine, University of Chile, Santiago, Chile
| | - Daniela Fernandois
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Gonzalo Cruz
- Laboratorio de Alteraciones Reproductivas y Metabólicas, Centro de Neurobiología y Plasticidad Cerebral (CNPC), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
17
|
Shorakae S, Ranasinha S, Abell S, Lambert G, Lambert E, de Courten B, Teede H. Inter-related effects of insulin resistance, hyperandrogenism, sympathetic dysfunction and chronic inflammation in PCOS. Clin Endocrinol (Oxf) 2018; 89:628-633. [PMID: 29992612 DOI: 10.1111/cen.13808] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/16/2018] [Accepted: 07/09/2018] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Insulin resistance, hyperandrogenism, sympathetic dysfunction and chronic low-grade inflammation may act together in a vicious cycle in the pathophysiology of PCOS. However, the inter-relationships of these components are not fully understood. We aimed to study these mechanisms in the pathophysiology of PCOS. PARTICIPANTS AND METHODS Premenopausal women with PCOS (Rotterdam diagnostic criteria) and without PCOS were recruited from a community setting into a cross-sectional substudy within a randomized control trial. Insulin resistance (fasting insulin and glucose), hyperandrogenism (testosterone, sex hormone-binding globulin [SHBG] and Free Androgen Index [FAI]), muscle sympathetic nerve activity (MSNA) and markers of chronic low-grade inflammation (high sensitivity C-reactive protein [hs-CRP] and high molecular weight adiponectin [HMW-adiponectin]) were measured. RESULTS Forty-nine women with PCOS (mean age 30 ± 6 mean BMI 29 ± 5) and 23 controls (mean age 29 ± 8 mean BMI 33 ± 7) with included in this analysis. MSNA and testosterone level were most significantly associated with PCOS status, after adjustment for age and BMI. In women with PCOS, markers of sympathetic activity correlated inversely with HMW-adiponectin and HMW-adiponectin correlated inversely with FAI. Testosterone and FAI both correlated positively with insulin resistance in women PCOS. CONCLUSION Sympathetic dysfunction and hyperandrogenism are significantly associated with PCOS. Chronic low-grade inflammation potentially mediates the effect of sympathetic dysfunction on hyperandrogenism and insulin resistance.
Collapse
Affiliation(s)
- Soulmaz Shorakae
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia
| | - Sanjeeva Ranasinha
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sally Abell
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia
| | - Gavin Lambert
- Human Neurotransmitters Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Faculty of Health, Arts and Design, Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Elisabeth Lambert
- Human Neurotransmitters Laboratory, Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Faculty of Health, Arts and Design, Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia
| | - Helena Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Shorakae S, Lambert EA, Jona E, Ika Sari C, de Courten B, Dixon JB, Lambert GW, Teede HJ. Effect of Central Sympathoinhibition With Moxonidine on Sympathetic Nervous Activity in Polycystic Ovary Syndrome-A Randomized Controlled Trial. Front Physiol 2018; 9:1486. [PMID: 30410448 PMCID: PMC6210452 DOI: 10.3389/fphys.2018.01486] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Sympathetic nervous system (SNS) activity is increased in polycystic ovary syndrome (PCOS). Moxonidine is a centrally acting sympatholytic drug with known beneficial effects on hypertension, insulin sensitivity, dyslipidemia and inflammation. In this double-blind placebo controlled randomized clinical trial we examined the effect of moxonidine on modulating sympathetic activity and downstream metabolic abnormalities in 48 pre-menopausal women with PCOS (Rotterdam diagnostic criteria), recruited from the community (January 2013-August 2015). Participants received moxonidine (0.2 mg daily initially, up titrated to 0.4 mg daily in 2 weeks) (n = 23) or placebo (n = 25) for 12 weeks. Multiunit muscle sympathetic activity (by microneurography) and plasma noradrenaline levels were measured (primary outcomes). Fasting lipids, insulin resistance, serum androgens, and inflammatory markers were measured as secondary outcomes. Forty three women completed the trial (19 moxonidine, 24 placebo). Mean change in burst frequency (-3 ± 7 vs. -3 ± 8 per minute) and burst incidence (-3 ± 10 vs. -4 ± 12 per 100 heartbeat) did not differ significantly between moxonidine and placebo groups. Women on moxonidine had a significant reduction in hs-CRP compared to placebo group (-0.92 ± 2.3 vs. -0.04 ± 1.5) which did not persist post Bonferroni correction. There was a significant association between markers of insulin resistance at baseline and reduction in sympathetic activity with moxonidine. Moxonidine was not effective in modulating sympathetic activity in PCOS. Anti-inflammatory effects of moxonidine and a relationship between insulin resistance and sympathetic response to moxonidine are suggested which need to be further explored. Clinical Trial Registration Number: (NCT01504321).
Collapse
Affiliation(s)
- Soulmaz Shorakae
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, VIC, Australia
| | - Elisabeth A Lambert
- Faculty of Health, Arts and Design, Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Eveline Jona
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Carolina Ika Sari
- Human Neurotransmitters Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Barbora de Courten
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, VIC, Australia
| | - John B Dixon
- Human Neurotransmitters Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia.,Clinical Obesity Research Laboratories, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Gavin W Lambert
- Faculty of Health, Arts and Design, Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, VIC, Australia.,Human Neurotransmitters Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Helena J Teede
- Monash Centre for Health Research and Implementation, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Diabetes and Vascular Medicine Unit, Monash Health, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Ibáñez L, Oberfield SE, Witchel S, Auchus RJ, Chang RJ, Codner E, Dabadghao P, Darendeliler F, Elbarbary NS, Gambineri A, Garcia Rudaz C, Hoeger KM, López-Bermejo A, Ong K, Peña AS, Reinehr T, Santoro N, Tena-Sempere M, Tao R, Yildiz BO, Alkhayyat H, Deeb A, Joel D, Horikawa R, de Zegher F, Lee PA. An International Consortium Update: Pathophysiology, Diagnosis, and Treatment of Polycystic Ovarian Syndrome in Adolescence. Horm Res Paediatr 2018; 88:371-395. [PMID: 29156452 DOI: 10.1159/000479371] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022] Open
Abstract
This paper represents an international collaboration of paediatric endocrine and other societies (listed in the Appendix) under the International Consortium of Paediatric Endocrinology (ICPE) aiming to improve worldwide care of adolescent girls with polycystic ovary syndrome (PCOS)1. The manuscript examines pathophysiology and guidelines for the diagnosis and management of PCOS during adolescence. The complex pathophysiology of PCOS involves the interaction of genetic and epigenetic changes, primary ovarian abnormalities, neuroendocrine alterations, and endocrine and metabolic modifiers such as anti-Müllerian hormone, hyperinsulinemia, insulin resistance, adiposity, and adiponectin levels. Appropriate diagnosis of adolescent PCOS should include adequate and careful evaluation of symptoms, such as hirsutism, severe acne, and menstrual irregularities 2 years beyond menarche, and elevated androgen levels. Polycystic ovarian morphology on ultrasound without hyperandrogenism or menstrual irregularities should not be used to diagnose adolescent PCOS. Hyperinsulinemia, insulin resistance, and obesity may be present in adolescents with PCOS, but are not considered to be diagnostic criteria. Treatment of adolescent PCOS should include lifestyle intervention, local therapies, and medications. Insulin sensitizers like metformin and oral contraceptive pills provide short-term benefits on PCOS symptoms. There are limited data on anti-androgens and combined therapies showing additive/synergistic actions for adolescents. Reproductive aspects and transition should be taken into account when managing adolescents.
Collapse
Affiliation(s)
- Lourdes Ibáñez
- Endocrinology, Hospital Sant Joan de Deu, Esplugues, Barcelona, Spain.,CIBERDEM, ISCIII, Madrid, Spain
| | - Sharon E Oberfield
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Selma Witchel
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | | | - R Jeffrey Chang
- Department of Reproductive Medicine, UCSD School of Medicine, La Jolla, California, USA
| | - Ethel Codner
- Institute of Maternal and Child Research, University of Chile, School of Medicine, Santiago, Chile
| | - Preeti Dabadghao
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | - Alessandra Gambineri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Cecilia Garcia Rudaz
- Division of Women, Youth and Children, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Kathleen M Hoeger
- Department of OBGYN, University of Rochester Medical Center, Rochester, New York, USA
| | - Abel López-Bermejo
- Pediatric Endocrinology, Hospital de Girona Dr. Josep Trueta, Girona, Spain
| | - Ken Ong
- MRC Epidemiology Unit, University of Cambridge, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Alexia S Peña
- The University of Adelaide and Robinson Research Institute, Adelaide, South Australia, Australia
| | - Thomas Reinehr
- University of Witten/Herdecke, Vestische Kinder- und Jugendklinik, Pediatric Endocrinology, Diabetes, and Nutrition Medicine, Datteln, Germany
| | - Nicola Santoro
- Pediatrics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Rachel Tao
- Division of Pediatric Endocrinology, CUMC, New York-Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| | - Bulent O Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey
| | - Haya Alkhayyat
- Medical University of Bahrain, BDF Hospital, Riffa, Bahrein
| | - Asma Deeb
- Mafraq Hospital, Abu Dhabi, United Arab Emirates
| | - Dipesalema Joel
- Department of Paediatrics and Adolescent Health, University of Botswana Teaching Hospital, Gaborone, Botswana
| | - Reiko Horikawa
- Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Francis de Zegher
- Department Pediatrics, University Hospital Gasthuisberg, Leuven, Belgium
| | - Peter A Lee
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
20
|
Squicciarini V, Riquelme R, Wilsterman K, Bentley GE, Lara HE. Role of RFRP-3 in the development of cold stress-induced polycystic ovary phenotype in rats. J Endocrinol 2018; 239:81–91. [PMID: 30307156 DOI: 10.1530/joe-18-0357] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RFamide-related peptide (RFRP-3) is a regulator of GnRH secretion from the brain, but it can also act in human ovary to influence steroidogenesis. We aimed to study the putative local role of RFRP-3 in the ovary and its potential participation in the development of a polycystic ovary phenotype induced by chronic sympathetic stress (cold stress). We used adult Sprague–Dawley rats divided into control and stressed groups. In both groups, we studied the effect of intraovarian exposure to RFRP-3 on follicular development and plasma ovarian steroid concentrations. We also tested the effect of RFRP-3 on ovarian steroid production in vitro. Chronic in vivo intraovarian exposure to RFRP-3 decreased basal testosterone concentrations and cold stress-induced progesterone production by the ovary. In vitro, RFRP-3 decreased hCG-induced ovarian progesterone and testosterone secretion. Immunohistochemistry and mRNA expression analysis showed a decrease in Rfrp and expression of its receptor in the ovary of stressed rats, a result which is in line with the increased testosterone levels found in stressed rats. In vivo application of RFRP-3 recovered the low levels of secondary and healthy antral follicles found in stressed rats. Taken together, our data indicate a previously unknown response of hypothalamic and ovarian RFRP-3 to chronic cold stress, influencing ovarian steroidogenesis and follicular dynamics. Thus, it is likely that RFRP-3 modulation in the ovary is a key component of development of the polycystic ovary phenotype.
Collapse
Affiliation(s)
- V Squicciarini
- Center for Neurobiochemical Studies in Endocrine Diseases, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Laboratory of Neurobiochemistry, Universidad de Chile, Santiago, Chile
| | - R Riquelme
- Center for Neurobiochemical Studies in Endocrine Diseases, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Laboratory of Neurobiochemistry, Universidad de Chile, Santiago, Chile
| | - K Wilsterman
- Department of Integrative Biology, UC Berkeley, Berkeley, California, USA
| | - G E Bentley
- Department of Integrative Biology, UC Berkeley, Berkeley, California, USA
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, USA
| | - H E Lara
- Center for Neurobiochemical Studies in Endocrine Diseases, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Laboratory of Neurobiochemistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
21
|
Espinoza JA, Alvarado W, Venegas B, Domínguez R, Morales-Ledesma L. Pharmacological sympathetic denervation prevents the development of polycystic ovarian syndrome in rats injected with estradiol valerate. Reprod Biol Endocrinol 2018; 16:86. [PMID: 30193590 PMCID: PMC6128994 DOI: 10.1186/s12958-018-0400-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The injection of estradiol valerate in female rats induces polycystic ovary syndrome, which is characterized by polycystic ovaries, anovulation, and hyperandrogenism. These characteristics have been associated with an increase in the ovarian concentration of norepinephrine, which occurs before establishing the polycystic ovary syndrome. The bilateral section of the superior ovarian nerve restores ovarian functions in animals with polycystic ovary syndrome. The superior ovarian nerve provides norepinephrine and vasoactive intestinal peptide to the ovary. An increase in the activity of both neurotransmitters has been associated with the development of polycystic ovary syndrome. The purpose of the present study was analyzed the participation of the noradrenergic nervous system in the development of polycystic ovary syndrome using guanethidine as a pharmacological tool that destroys peripheral noradrenergic nerve fibers. METHODS Fourteen-day old female rats of the CIIZ-V strain were injected with estradiol valerate or vehicle solution. Rats were randomly allotted to one of three guanethidine treatment groups for denervation: 1) guanethidine treatment at age 7 to 27-days, 2) guanethidine treatment at age 14 to 34- days, and 3) guanethidine treatment at age 70 to 90- days. All animals were sacrificed when presenting vaginal oestrus at age 90 to 94-days. The parameters analyzed were the number of ova shed by ovulating animals, the ovulation rate (i.e., the numbers of ovulating animals/the numbers of used animals), the serum concentration of progesterone, testosterone, oestradiol and the immunoreactivity for tyrosine hydroxylase enzyme. All data were analyzed statistically. A p-value of less than 0.05 was considered significant. RESULTS Our results show that the elimination of noradrenergic fibers before the establishment of polycystic ovary syndrome prevents two characteristics of the syndrome, blocking of ovulation and hyperandrogenism. We also found that in animals that have already developed polycystic ovary syndrome, sympathetic denervation restores ovulatory capacity, but it was not as efficient in reducing hyperandrogenism. CONCLUSION The results of the present study suggest that the noradrenergic fibers play a stimulant role in the establishment of polycystic ovary syndrome.
Collapse
Affiliation(s)
- Julieta A. Espinoza
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM AP 9-020, CP 15000 México, DF Mexico
| | - Wendy Alvarado
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM AP 9-020, CP 15000 México, DF Mexico
| | - Berenice Venegas
- 0000 0001 2112 2750grid.411659.eFacultad de Ciencias Biológicas de la Benemérita Universidad Autónoma de Puebla, Blvd. Valsequillo, Av. San Claudio, Edificio 112-A, Cd Universitaria, Col. Jardines de San Manuel, Puebla, Mexico
| | - Roberto Domínguez
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM AP 9-020, CP 15000 México, DF Mexico
| | - Leticia Morales-Ledesma
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM AP 9-020, CP 15000 México, DF Mexico
| |
Collapse
|
22
|
El-Shamy FF, El-Kholy SS, El-Rahman MMA. Effectiveness of Laser Acupoints on Women With Polycystic Ovarian Syndrome: A Randomized Controlled Trial. J Lasers Med Sci 2018; 9:113-120. [PMID: 30026896 DOI: 10.15171/jlms.2018.22] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: Polycystic ovarian syndrome (PCOS) is one of the most widely recognized reasons for infertility. The aim of this study was to examine the impact of laser acupuncture on PCOS women. Methods: Twenty-five PCOS women were randomly allocated to either the study group (SG; n=13), treated by laser acupuncture, or the control group (CG; n = 12). Blood hormonal levels and insulin resistance were measured at baseline and after 12 weeks of intervention. Results: The pre-intervention levels showed no statistically significant differences between SG and CG for baseline characteristics (P>0.05). After 12 weeks of intervention, within-group analyses showed that body mass index (BMI), blood hormonal levels, and insulin resistance were significantly decreased (P<0.05), while no significant changes in follicle-stimulating hormone (P>0.05) were recorded in the 2 groups. Between-groups analyses showed that most outcomes measures were significantly decreased (P<0.05) in SG compared with CG, with no significant changes in FSH and BMI (P>0.05). Conclusion: Laser acupuncture can be suggested as an effective management for PCOS women.
Collapse
Affiliation(s)
- Fayiz F El-Shamy
- Department of Physical Therapy for Women's Health, Faculty of Physical Therapy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Sand S El-Kholy
- Department of Physiology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Marwa M Abd El-Rahman
- Department of Physical Therapy for Gynaecology and Obstetrics, Faculty of Physical Therapy, Cairo University, Cairo, Egypt
| |
Collapse
|
23
|
Shorakae S, Abell SK, Hiam DS, Lambert EA, Eikelis N, Jona E, Sari CI, Stepto NK, Lambert GW, de Courten B, Teede HJ. High-molecular-weight adiponectin is inversely associated with sympathetic activity in polycystic ovary syndrome. Fertil Steril 2018; 109:532-539. [DOI: 10.1016/j.fertnstert.2017.11.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 01/22/2023]
|
24
|
Wang BQ, Chen YY, Lan XX, Zhou ZY, Xu XX, Wu XQ. The effect of neonatal immune challenge on reproduction by altering intraovarian kisspeptin/GPR54 system in the rat. Reprod Toxicol 2017; 74:40-47. [DOI: 10.1016/j.reprotox.2017.08.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/12/2017] [Accepted: 08/30/2017] [Indexed: 01/20/2023]
|
25
|
Sominsky L, Hodgson DM, McLaughlin EA, Smith R, Wall HM, Spencer SJ. Linking Stress and Infertility: A Novel Role for Ghrelin. Endocr Rev 2017; 38:432-467. [PMID: 28938425 DOI: 10.1210/er.2016-1133] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/24/2017] [Indexed: 12/23/2022]
Abstract
Infertility affects a remarkable one in four couples in developing countries. Psychological stress is a ubiquitous facet of life, and although stress affects us all at some point, prolonged or unmanageable stress may become harmful for some individuals, negatively impacting on their health, including fertility. For instance, women who struggle to conceive are twice as likely to suffer from emotional distress than fertile women. Assisted reproductive technology treatments place an additional physical, emotional, and financial burden of stress, particularly on women, who are often exposed to invasive techniques associated with treatment. Stress-reduction interventions can reduce negative affect and in some cases to improve in vitro fertilization outcomes. Although it has been well-established that stress negatively affects fertility in animal models, human research remains inconsistent due to individual differences and methodological flaws. Attempts to isolate single causal links between stress and infertility have not yet been successful due to their multifaceted etiologies. In this review, we will discuss the current literature in the field of stress-induced reproductive dysfunction based on animal and human models, and introduce a recently unexplored link between stress and infertility, the gut-derived hormone, ghrelin. We also present evidence from recent seminal studies demonstrating that ghrelin has a principal role in the stress response and reward processing, as well as in regulating reproductive function, and that these roles are tightly interlinked. Collectively, these data support the hypothesis that stress may negatively impact upon fertility at least in part by stimulating a dysregulation in ghrelin signaling.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Eileen A McLaughlin
- School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland 1010, New Zealand.,School of Environmental & Life Sciences, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Lookout Road, New Lambton Heights, New South Wales 2305, Australia.,Priority Research Centre in Reproductive Science, The University of Newcastle, New South Wales 2308, Australia
| | - Hannah M Wall
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| |
Collapse
|
26
|
Zangeneh FZ, Naghizadeh MM, Bagheri M, Jafarabadi M. Are CRH & NGF as psychoneuroimmune regulators in women with polycystic ovary syndrome? Gynecol Endocrinol 2017; 33:227-233. [PMID: 27908212 DOI: 10.1080/09513590.2016.1250152] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) affects quality of life and can worsen anxiety and depression either due to the features of PCOS or due to the diagnosis of a chronic disease. Corticotrophin-releasing hormone (CRH) and nerves growth factor (NGF) are the modulator for the actions of the sympathetic nervous and immune systems. METHODS In total, 171 women divided into two groups: study and control groups. Serum CRH, NGF, and interleukins: IL-1α. IL-1β, 17A, and TNFα were determined by ELISA Kits in both groups. RESULTS The results showed that IL-1α (p < 0.001) and β (p = 0.017) significantly increased in PCO group. CRH, NGF, and IL-17α in serum of patients with PCO significantly lower than the control group (p < 0.001). The results of this study indicate: (1) destruction of three cytokines pattern, (2) Reduction of CRH, NGF, and IL-17α in serum of PCO patients can be under the direct influence of the sympathetic nervous system (SAS), and (3) reduction of CRH and NGFα can be reason of psych/emotional distress in women with PCOS. CONCLUSIONS The results of this study confirm (1) low-grade chronic inflammation in PCOS. This impaired cytokine pattern can play a major role in the immune-pathogenesis of PCOS; (2) hyponeurotrophinemia and reduction of CRH in women with PCOS could reflect deficit of neuronal stress-adaptation in these patients.
Collapse
Affiliation(s)
- F Z Zangeneh
- a Reproductive Health Research Center, Tehran University of Medical Sciences , Tehran , Iran and
| | - M M Naghizadeh
- b Department of Community Medicine , Medical Faculty, Fasa University of Medical Sciences , Fasa , Iran
| | - M Bagheri
- a Reproductive Health Research Center, Tehran University of Medical Sciences , Tehran , Iran and
| | - M Jafarabadi
- a Reproductive Health Research Center, Tehran University of Medical Sciences , Tehran , Iran and
| |
Collapse
|
27
|
Cruz G, Fernandois D, Paredes AH. Ovarian function and reproductive senescence in the rat: role of ovarian sympathetic innervation. Reproduction 2017; 153:R59-R68. [DOI: 10.1530/rep-16-0117] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 10/21/2016] [Accepted: 10/31/2016] [Indexed: 01/13/2023]
Abstract
Successful reproduction is the result of a myriad interactions in which the ovary and the ovarian follicular reserve play a fundamental role. At present, women who delay maternity until after 30 years of age have a decreased fertility rate due to various causes, including damaged follicles and a reduction in the reserve pool of follicles. Therefore, the period just prior to menopause, also known as the subfertile period, is important. The possibility of modulating the follicular pool and the health of follicles during this period to improve fertility is worth exploring. We have developed an animal model to study the ovarian ageing process during this subfertile period to understand the mechanisms responsible for reproductive senescence. In the rat model, we have shown that the sympathetic nervous system participates in regulating the follicular development during ovarian ageing. This article reviews the existing evidence on the presence and functional role of sympathetic nerve activity in regulating the follicular development during ovarian ageing, with a focus on the subfertile period.Free Spanish abstract: A Spanish translation of this abstract is freely available athttp://www.reproduction-online.org/content/153/2/R59/suppl/DC1.
Collapse
|
28
|
Rosenfield RL, Ehrmann DA. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr Rev 2016; 37:467-520. [PMID: 27459230 PMCID: PMC5045492 DOI: 10.1210/er.2015-1104] [Citation(s) in RCA: 833] [Impact Index Per Article: 92.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 07/20/2016] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) was hypothesized to result from functional ovarian hyperandrogenism (FOH) due to dysregulation of androgen secretion in 1989-1995. Subsequent studies have supported and amplified this hypothesis. When defined as otherwise unexplained hyperandrogenic oligoanovulation, two-thirds of PCOS cases have functionally typical FOH, characterized by 17-hydroxyprogesterone hyperresponsiveness to gonadotropin stimulation. Two-thirds of the remaining PCOS have FOH detectable by testosterone elevation after suppression of adrenal androgen production. About 3% of PCOS have a related isolated functional adrenal hyperandrogenism. The remaining PCOS cases are mild and lack evidence of steroid secretory abnormalities; most of these are obese, which we postulate to account for their atypical PCOS. Approximately half of normal women with polycystic ovarian morphology (PCOM) have subclinical FOH-related steroidogenic defects. Theca cells from polycystic ovaries of classic PCOS patients in long-term culture have an intrinsic steroidogenic dysregulation that can account for the steroidogenic abnormalities typical of FOH. These cells overexpress most steroidogenic enzymes, particularly cytochrome P450c17. Overexpression of a protein identified by genome-wide association screening, differentially expressed in normal and neoplastic development 1A.V2, in normal theca cells has reproduced this PCOS phenotype in vitro. A metabolic syndrome of obesity-related and/or intrinsic insulin resistance occurs in about half of PCOS patients, and the compensatory hyperinsulinism has tissue-selective effects, which include aggravation of hyperandrogenism. PCOS seems to arise as a complex trait that results from the interaction of diverse genetic and environmental factors. Heritable factors include PCOM, hyperandrogenemia, insulin resistance, and insulin secretory defects. Environmental factors include prenatal androgen exposure and poor fetal growth, whereas acquired obesity is a major postnatal factor. The variety of pathways involved and lack of a common thread attests to the multifactorial nature and heterogeneity of the syndrome. Further research into the fundamental basis of the disorder will be necessary to optimally correct androgen levels, ovulation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Robert L Rosenfield
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637
| | - David A Ehrmann
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637
| |
Collapse
|
29
|
Rudolph LM, Bentley GE, Calandra RS, Paredes AH, Tesone M, Wu TJ, Micevych PE. Peripheral and Central Mechanisms Involved in the Hormonal Control of Male and Female Reproduction. J Neuroendocrinol 2016; 28:10.1111/jne.12405. [PMID: 27329133 PMCID: PMC5146987 DOI: 10.1111/jne.12405] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 05/25/2016] [Accepted: 06/20/2016] [Indexed: 12/18/2022]
Abstract
Reproduction involves the integration of hormonal signals acting across multiple systems to generate a synchronised physiological output. A critical component of reproduction is the luteinising hormone (LH) surge, which is mediated by oestradiol (E2 ) and neuroprogesterone interacting to stimulate kisspeptin release in the rostral periventricular nucleus of the third ventricle in rats. Recent evidence indicates the involvement of both classical and membrane E2 and progesterone signalling in this pathway. A metabolite of gonadotrophin-releasing hormone (GnRH), GnRH-(1-5), has been shown to stimulate GnRH expression and secretion, and has a role in the regulation of lordosis. Additionally, gonadotrophin release-inhibitory hormone (GnIH) projects to and influences the activity of GnRH neurones in birds. Stress-induced changes in GnIH have been shown to alter breeding behaviour in birds, demonstrating another mechanism for the molecular control of reproduction. Peripherally, paracrine and autocrine actions within the gonad have been suggested as therapeutic targets for infertility in both males and females. Dysfunction of testicular prostaglandin synthesis is a possible cause of idiopathic male infertility. Indeed, local production of melatonin and corticotrophin-releasing hormone could influence spermatogenesis via immune pathways in the gonad. In females, vascular endothelial growth factor A has been implicated in an angiogenic process that mediates development of the corpus luteum and thus fertility via the Notch signalling pathway. Age-induced decreases in fertility involve ovarian kisspeptin and its regulation of ovarian sympathetic innervation. Finally, morphological changes in the arcuate nucleus of the hypothalamus influence female sexual receptivity in rats. The processes mediating these morphological changes have been shown to involve the rapid effects of E2 controlling synaptogenesis in this hypothalamic nucleus. In summary, this review highlights new research in these areas, focusing on recent findings concerning the molecular mechanisms involved in the central and peripheral hormonal control of reproduction.
Collapse
Affiliation(s)
- L M Rudolph
- Department of Neurobiology, Laboratory of Neuroendocrinology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - G E Bentley
- Department of Integrative Biology, and Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - R S Calandra
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - A H Paredes
- Laboratory of Neurobiochemistry, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Independencia, Santiago, Chile
| | - M Tesone
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - T J Wu
- Department of Obstetrics and Gynecology, Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, MD, USA
| | - P E Micevych
- Department of Neurobiology, Laboratory of Neuroendocrinology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
30
|
Wu XQ, Li XF, Xia WT, Ye B, O’Byrne KT. The effects of small litter rearing on ovarian function at puberty and adulthood in the rat. Reprod Biol 2016; 16:130-7. [DOI: 10.1016/j.repbio.2016.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/24/2016] [Accepted: 02/05/2016] [Indexed: 01/14/2023]
|
31
|
Fernandois D, Na E, Cuevas F, Cruz G, Lara HE, Paredes AH. Kisspeptin is involved in ovarian follicular development during aging in rats. J Endocrinol 2016; 228:161-70. [PMID: 26698566 DOI: 10.1530/joe-15-0429] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 11/08/2022]
Abstract
We have previously reported that kisspeptin (KP) may be under the control of the sympathetic innervation of the ovary. Considering that the sympathetic activity of the ovary increases with aging, it is possible that ovarian KP also increases during this period and participates in follicular development. To evaluate this possibility, we determined ovarian KP expression and its action on follicular development during reproductive aging in rats. We measured ovarian KP mRNA and protein levels in 6-, 8-, 10- and 12-month-old rats. To evaluate follicular developmental changes, intraovarian administration of KP or its antagonist, peptide 234 (P234), was performed using a mini-osmotic pump, and to evaluate FSH receptor (FSHR) changes in the senescent ovary, we stimulated cultured ovaries with KP, P234 and isoproterenol (ISO). Our results shows that KP expression in the ovary was increased in 10- and 12-month-old rats compared with 6-month-old rats, and this increase in KP was strongly correlated with the increase in ovarian norepinephrine observed with aging. The administration of KP produced an increase in corpora lutea and type III follicles in 6- and 10-month-old rats, which was reversed by P234 administration at 10 months. In addition, KP decreased the number and size of antral follicles in 6- and 10-month-old rats, while P234 administration produced an increase in these structures at the same ages. In ovarian cultures KP prevented the induction of FSHR by ISO. These results suggest that intraovarian KP negatively participates in the acquisition of FSHR, indicating a local role in the regulation of follicular development and ovulation during reproductive aging.
Collapse
Affiliation(s)
- D Fernandois
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - E Na
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - F Cuevas
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - G Cruz
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - H E Lara
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - A H Paredes
- Laboratory of NeurobiochemistryDepartment of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago 8380492, ChileLaboratorio de alteraciones Reproductivas y MetabólicasFacultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
32
|
Ovarian kisspeptin expression is related to age and to monocyte chemoattractant protein-1. J Assist Reprod Genet 2016; 33:535-43. [PMID: 26879207 DOI: 10.1007/s10815-016-0672-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/26/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE The objective of this study was to test the hypothesis that ovarian kisspeptin (kiss1) and its receptor (kiss1r) expression are affected by age, obesity, and the age- and obesity-related chemokine monocyte chemoattractant protein-1 (MCP-1). METHODS Ovaries from reproductive-aged and older C57BL/6J mice fed normal chow (NC) or high-fat (HF) diet, ovaries from age-matched young MCP-1 knockout and young control mice on NC, and finally, cumulus and mural granulosa cells (GCs) from women who underwent in vitro fertilization (IVF) were collected. Kiss1, kiss1r, anti-Mullerian hormone (AMH), and AMH receptor (AMHR-II) messenger RNA (mRNA) expression levels were quantified using real-time polymerase chain reaction (RT-PCR). RESULTS In mouse ovaries, kiss1 and kiss1r mRNA levels were significantly higher in old compared to reproductive-aged mice, and diet-induced obesity did not alter kiss1 or kiss1r mRNA levels. Compared to young control mice, young MCP-1 knockout mice had significantly lower ovarian kiss1 mRNA but significantly higher AMH and AMHR-II mRNA levels. In human cumulus GCs, kiss1r mRNA levels were positively correlated with age but not with BMI. There was no expression of kiss1 mRNA in either cumulus or mural GCs. CONCLUSION These data suggest a possible age-related physiologic role for the kisspeptinergic system in ovarian physiology. Additionally, the inflammatory MCP-1 may be associated with kiss1 and AMH genes, which are important in ovulation and folliculogenesis, respectively.
Collapse
|
33
|
Dumesic DA, Oberfield SE, Stener-Victorin E, Marshall JC, Laven JS, Legro RS. Scientific Statement on the Diagnostic Criteria, Epidemiology, Pathophysiology, and Molecular Genetics of Polycystic Ovary Syndrome. Endocr Rev 2015; 36:487-525. [PMID: 26426951 PMCID: PMC4591526 DOI: 10.1210/er.2015-1018] [Citation(s) in RCA: 609] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous and complex disorder that has both adverse reproductive and metabolic implications for affected women. However, there is generally poor understanding of its etiology. Varying expert-based diagnostic criteria utilize some combination of oligo-ovulation, hyperandrogenism, and the presence of polycystic ovaries. Criteria that require hyperandrogenism tend to identify a more severe reproductive and metabolic phenotype. The phenotype can vary by race and ethnicity, is difficult to define in the perimenarchal and perimenopausal period, and is exacerbated by obesity. The pathophysiology involves abnormal gonadotropin secretion from a reduced hypothalamic feedback response to circulating sex steroids, altered ovarian morphology and functional changes, and disordered insulin action in a variety of target tissues. PCOS clusters in families and both female and male relatives can show stigmata of the syndrome, including metabolic abnormalities. Genome-wide association studies have identified a number of candidate regions, although their role in contributing to PCOS is still largely unknown.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Sharon E Oberfield
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Elisabet Stener-Victorin
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - John C Marshall
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Joop S Laven
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Richard S Legro
- Department of Obstetrics and Gynecology (D.A.D.), David Geffen School of Medicine at UCLA, Los Angeles, California 90095; Division of Pediatric Endocrinology (S.E.O.), Children's Hospital of New York-Presbyterian, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Physiology (E.S.-V.), Karolinska Institutet, 171 77 Stockholm, Sweden; Center for Research in Reproduction and Division of Endocrinology (J.C.M.), Department of Internal Medicine, University of Virginia Health System, Charlottesville, Virginia 22903; Division of Reproductive Medicine (J.S.L.), Department of Obstetrics and Gynecology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands; and Department of Obstetrics and Gynecology (R.S.L.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| |
Collapse
|
34
|
Anesetti G, Chávez-Genaro R. Neonatal testosterone exposure induces early development of follicular cysts followed by sympathetic ovarian hyperinnervation. Reprod Fertil Dev 2015; 28:RD14460. [PMID: 25989716 DOI: 10.1071/rd14460] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/05/2015] [Indexed: 01/22/2023] Open
Abstract
This study analysed the temporal association between ovarian cyst development induced by neonatal androgenisation and sympathetic innervation. Neonatal rats (postnatal Days 1 to 5) were treated with testosterone or dihydrotestosterone and the effects were evaluated at postnatal Days 20, 40, 90 or 180. Ovulation rate, number of cystic follicles and density of sympathetic fibres were analysed. The effects of surgical denervation or gonadotrophin stimulation were also assessed. Rats exposed to testosterone showed no oestrous cycle activity and did not ovulate, maintaining a polycystic ovarian morphology at all ages studied. Also, a significant increase in ovarian density of noradrenergic fibres was detected at postnatal Days 90 and 180. Sympathectomy was unable to re-establish ovarian activity; however, human chorionic gonadotrophin stimulation was enough to induce ovulation. The impact of dihydrotestosterone on ovarian function was less noticeable, showing the coexistence of corpora lutea and cystic structures without changes in sympathetic innervation. Our findings suggest that a remodelling of ovarian sympathetic innervation occurs as a response to modifications in the pattern of follicular growth induced by testosterone. A role of sympathetic innervation in the maintenance of the polycystic condition is suggested.
Collapse
|
35
|
Merz C, Saller S, Kunz L, Xu J, Yeoman RR, Ting AY, Lawson MS, Stouffer RL, Hennebold JD, Pau F, Dissen GA, Ojeda SR, Zelinski MB, Mayerhofer A. Expression of the beta-2 adrenergic receptor (ADRB-2) in human and monkey ovarian follicles: a marker of growing follicles? J Ovarian Res 2015; 8:8. [PMID: 25824473 PMCID: PMC4356150 DOI: 10.1186/s13048-015-0136-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/23/2015] [Indexed: 12/03/2022] Open
Abstract
Background ADRB-2 was implicated in rodent ovarian functions, including initial follicular growth. In contrast, ADRB-2 expression and function in nonhuman primate and human ovary were not fully known but innervation and significant levels of norepinephrine (NE), which is a ligand at the ADRB-2, were reported in the ovary. Methods We studied expression of ADRB-2 in human and rhesus monkey ovary (RT-PCR, immunohistochemistry; laser micro dissection) and measured levels of norepinephrine (NE; ELISA) in monkey follicular fluid (FF). 3D cultures of monkey follicles (4 animals) were exposed to NE or the ADRB-2 agonist isoproterenol (ISO), and follicular development (size) was monitored. Upon termination expression of ADRB-2, FSH receptor and aromatase genes were examined. Results Immunohistochemistry and RT-PCR of either human follicular granulosa cells (GCs) obtained by laser micro dissection or isolated monkey follicles revealed ADRB-2 in GCs of primordial, primary, secondary and tertiary follicles. Staining of GCs in primordial and primary follicles was intense. In large preantral and antral follicles the staining was heterogeneous, with positive and negative GCs present but GCs lining the antrum of large follicles were generally strongly immunopositive. Theca, interstitial, and ovarian surface epithelial cells were also positive. NE was detected in FF of preovulatory antral monkey follicles (0.37 + 0.05 ng/ml; n = 7; ELISA) but not in serum. We examined preantral follicles ranging from 152 to 366 μm in diameter in a 3D culture in media supplemented with follicle stimulating hormone (FSH). Under these conditions, neither NE, nor ISO, influenced growth rate in a period lasting up to one month. Upon termination of the cultures, all surviving follicles expressed aromatase and FSH receptors, but only about half of them also co-expressed ADRB-2. The ADRB-2 expression was not correlated with the treatment but was positively correlated with the follicular size at the beginning and at the end of the culture period. Hence, expression of ADRB-2 was found in the largest and fastest-in vitro growing follicles. Conclusions The results imply ADRB-2-mediated actions in the development of primate follicles. Drugs interfering with ADRB-2 are used to treat medical conditions and may have unexplored effects in the human ovary.
Collapse
|
36
|
Wojtkiewicz J, Jana B, Kozłowska A, Crayton R, Majewski M, Zalecki M, Baranowski W, Radziszewski P. Innervation pattern of polycystic ovaries in the women. J Chem Neuroanat 2014; 61-62:147-52. [DOI: 10.1016/j.jchemneu.2014.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 01/19/2023]
|
37
|
Wilson JL, Chen W, Dissen GA, Ojeda SR, Cowley MA, Garcia-Rudaz C, Enriori PJ. Excess of nerve growth factor in the ovary causes a polycystic ovary-like syndrome in mice, which closely resembles both reproductive and metabolic aspects of the human syndrome. Endocrinology 2014; 155:4494-506. [PMID: 25211588 PMCID: PMC4197978 DOI: 10.1210/en.2014-1368] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polycystic ovarian syndrome (PCOS), the most common female endocrine disorder of unknown etiology, is characterized by reproductive abnormalities and associated metabolic conditions comprising insulin resistance, type 2 diabetes mellitus, and dyslipidemia. We previously reported that transgenic overexpression of nerve growth factor (NGF), a marker of sympathetic hyperactivity, directed to the ovary by the mouse 17α-hydroxylase/C17-20 lyase promoter (17NF mice), results in ovarian abnormalities similar to those seen in PCOS women. To investigate whether ovarian overproduction of NGF also induces common metabolic alterations of PCOS, we assessed glucose homeostasis by glucose tolerance test, plasma insulin levels, and body composition by dual-energy x-ray absorptiometry scan in young female 17NF mice and wild-type mice. 17NF mice exhibited increased body weight and alterations in body fat distribution with a greater accumulation of visceral fat compared with sc fat (P < .01). 17NF mice also displayed glucose intolerance (P < .01), decreased insulin-mediated glucose disposal (P < .01), and hyperinsulinemia (P < .05), which, similar to PCOS patients, occurred independently of body weight. Additionally, 17NF mice exhibited increased sympathetic outflow observed as increased interscapular brown adipose tissue temperature. This change was evident during the dark period (7 pm to 7 am) and occurred concomitant with increased interscapular brown adipose tissue uncoupling protein 1 expression. These findings suggest that overexpression of NGF in the ovary may suffice to cause both reproductive and metabolic alterations characteristic of PCOS and support the hypothesis that sympathetic hyperactivity may contribute to the development and/or progression of PCOS.
Collapse
Affiliation(s)
- Jenny L Wilson
- Monash Obesity and Diabetes Institute/Department of Physiology (J.L.W., W.C., M.A.C., P.J.E.), Monash University, Clayton, 3800 Victoria, Australia; Division of Neuroscience (G.A.D., S.R.O.), Oregon Health and Science University, Portland, Oregon 97239; and Department of Paediatrics (C.G.-R.), Faculty of Medicine, Nursing and Health Sciences, Monash University and Monash Children's Hospital, Monash Health, Clayton, 3168 Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
38
|
Kampfer C, Saller S, Windschüttl S, Berg D, Berg U, Mayerhofer A. Pigment-Epithelium Derived Factor (PEDF) and the human ovary: A role in the generation of ROS in granulosa cells. Life Sci 2014; 97:129-36. [DOI: 10.1016/j.lfs.2013.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 12/04/2013] [Accepted: 12/07/2013] [Indexed: 11/16/2022]
|
39
|
Saller S, Kunz L, Berg D, Berg U, Lara H, Urra J, Hecht S, Pavlik R, Thaler CJ, Mayerhofer A. Dopamine in human follicular fluid is associated with cellular uptake and metabolism-dependent generation of reactive oxygen species in granulosa cells: implications for physiology and pathology. Hum Reprod 2013; 29:555-67. [PMID: 24287819 DOI: 10.1093/humrep/det422] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION Is the neurotransmitter dopamine (DA) in the human ovary involved in the generation of reactive oxygen species (ROS)? SUMMARY ANSWER Human ovarian follicular fluid contains DA, which causes the generation of ROS in cultured human granulosa cells (GCs), and alterations of DA levels in follicular fluid and DA uptake/metabolism in GCs in patients with polycystic ovary syndrome (PCOS) are linked to increased levels of ROS. WHAT IS KNOWN ALREADY DA is an important neurotransmitter in the brain, and the metabolism of DA results in the generation of ROS. DA was detected in human ovarian homogenates, but whether it is present in follicular fluid and plays a role in the follicle is not known. STUDY DESIGN, SIZE AND DURATION We used human follicular fluid from patients undergoing in vitro fertilization (IVF), GCs from patients with or without PCOS and also employed mathematical modeling to investigate the presence of DA and its effects on ROS. PARTICIPANTS/MATERIALS, SETTING AND METHODS DA in follicular fluid and GCs was determined by enzyme-linked immunosorbent assay. GC viability, apoptosis and generation of ROS were monitored in GCs upon addition of DA. Inhibitors of DA uptake and metabolism, an antioxidant and DA receptor agonists, were used to study cellular uptake and the mechanism of DA-induced ROS generation. Human GCs were examined for the presence and abundance of transcripts of the DA transporter (DAT; SLC6A3), the DA-metabolizing enzymes monoamine oxidases A/B (MAO-A/B) and catechol-O-methyltransferase and the vesicular monoamine transporter. A computational model was developed to describe and predict DA-induced ROS generation in human GCs. MAIN RESULTS AND ROLE OF CHANCE We found DA in follicular fluid of ovulatory follicles of the human ovary and in GCs. DAT and MAO-A/B, which are expressed by GCs, are prerequisites for a DA receptor-independent generation of ROS in GCs. Blockers of DAT and MAO-A/B, as well as an antioxidant, prevented the generation of ROS (P < 0.05). Agonists of DA receptors (D1 and D2) did not induce ROS. DA, in the concentration range found in follicular fluid, did not induce apoptosis of cultured GCs. Computational modeling suggested, however, that ROS levels in GCs depend on the concentrations of DA and on the cellular uptake and metabolism. In PCOS-derived follicular fluid, the levels of DA were higher (P < 0.05) in GCs, the transcript levels of DAT and MAO-A/B in GCs were 2-fold higher (P < 0.05) and the DA-induced ROS levels were found to be more than 4-fold increased (P < 0.05) compared with non-PCOS cells. Furthermore, DA at a high concentration induced apoptosis in PCOS-derived GCs. LIMITATIONS, REASONS FOR CAUTION While the results in IVF-derived follicular fluid and in GCs reveal for the first time the presence of DA in the human follicular compartment, functions of DA could only be studied in IVF-derived GCs, which can be viewed as a cellular model for the periovulatory follicular phase. The full functional importance of DA-induced ROS in small follicles and other compartments of the ovary, especially in PCOS samples, remains to be shown. WIDER IMPLICATIONS OF THE FINDINGS The results identify DA as a factor in the human ovary, which, via ROS generation, could play a role in ovarian physiology and pathology. The results obtained in samples from women with PCOS suggest the involvement of DA, acting via ROS, in this condition. STUDY FUNDING/COMPETING INTERESTS This work was supported by a grant from DFG MA1080/17-3 and in part MA1080/19-1. There are no competing interests.
Collapse
Affiliation(s)
- S Saller
- Anatomy III-Cell Biology, Ludwig Maximilian University Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hendriks ML, König T, Soleman RS, Korsen T, Schats R, Hompes PGA, Homburg R, Lambalk CB. Influence of ovarian manipulation on reproductive endocrinology in polycystic ovarian syndrome and regularly cycling women. Eur J Endocrinol 2013; 169:503-10. [PMID: 23904283 DOI: 10.1530/eje-13-0334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Little is known about the function of the ovarian neuronal network in humans. In many species, copulation influences endocrinology through this network. As a first step, the possible influence of ovarian mechanical manipulation on pituitary and ovarian hormones was evaluated in polycystic ovarian syndrome (PCOS) and regularly cycling women. DESIGN Prospective case-control study (2008-2010). METHODS Ten PCOS women (Rotterdam criteria) undergoing ovulation induction with recombinant-FSH and ten normal ovulatory controls were included in an academic fertility clinic. In the late follicular phase blood was drawn every 10 min for 6 h. After 3 h the ovaries were mechanically manipulated by moving a transvaginal ultrasound probe firmly over each ovary ten times. Main outcome measures were LH and FSH pulsatility and ovarian hormones before and after ovarian manipulation. RESULTS All PCOS patients showed an LH decline after the ovarian manipulation (before 13.0 U/l and after 10.4 U/l, P<0.01), probably based on a combination of a longer LH pulse interval and smaller amplitude (P=0.07). The controls showed no LH change (before 9.6 U/l and after 9.3 U/l, P=0.67). None of the ovarian hormones (estradiol, progesterone, anti-Müllerian hormone, inhibin B, androstenedione and testosterone) changed in either group. CONCLUSIONS Ovarian mechanical manipulation lowers LH secretion immediately and typically only in preovulatory PCOS patients. The immediate LH change after the ovarian manipulation without any accompanying ovarian hormonal changes point to nonhormonal communication from the ovaries to the pituitary. A neuronal pathway from the ovaries communicating to the hypothalamic-pituitary system is the most reasonable explanation.
Collapse
Affiliation(s)
- M L Hendriks
- Division of Reproductive Medicine, Department of Obstetrics and Gynecology
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Johansson J, Stener-Victorin E. Polycystic ovary syndrome: effect and mechanisms of acupuncture for ovulation induction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:762615. [PMID: 24073009 PMCID: PMC3773899 DOI: 10.1155/2013/762615] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/23/2013] [Indexed: 12/22/2022]
Abstract
Polycystic ovary syndrome (PCOS), the most common endocrine disorder among women of reproductive age, is characterized by the coexistence of hyperandrogenism, ovulatory dysfunction, and polycystic ovaries (PCO). PCOS also represents the largest part of female oligoovulatory infertility, and the management of ovulatory and menstrual dysfunction, comprises a third of the high costs of PCOS treatment. Current pharmacological and surgical treatments for reproductive symptoms are effective, however, associated with negative side effects, such as cardiovascular complications and multiple pregnancies. For menstrual irregularities and ovulation induction in women with PCOS, acupuncture has indicated beneficial effects. This review will focus on the results from randomized controlled acupuncture trials for regulation of menstrual dysfunction and for inducing ovulation in women with PCOS although there are uncontrolled trials with nonetheless interesting results. Animal experimental studies will be further discussed when they can provide a more mechanistic explanatory view.
Collapse
Affiliation(s)
- Julia Johansson
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 434, 405 30 Gothenburg, Sweden
| | - Elisabet Stener-Victorin
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 434, 405 30 Gothenburg, Sweden
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
42
|
Zheng YH, Wang XH, Lai MH, Yao H, Liu H, Ma HX. Effectiveness of Abdominal Acupuncture for Patients with Obesity-Type Polycystic Ovary Syndrome: A Randomized Controlled Trial. J Altern Complement Med 2013; 19:740-5. [PMID: 23676106 DOI: 10.1089/acm.2012.0429] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Yan-Hua Zheng
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Xin-Hua Wang
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Mao-Hua Lai
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Hong Yao
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Hua Liu
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Hong-Xia Ma
- First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| |
Collapse
|
43
|
KOZŁOWSKA A, WOJTKIEWICZ J, MAJEWSKI M, JANA B. The Noradrenergic Innervation and Steroidogenic Activity of Porcine Cystic Ovaries. Physiol Res 2013; 62:421-33. [DOI: 10.33549/physiolres.932471] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The aim of the present study was to investigate the distribution and density of noradrenergic nerve fibres (NNFs), content of catecholamines (CATs) and steroids in the cystic ovaries of gilts receiving DXM from middle luteal phase. Cystic status of ovaries was induced by i.m. DXM injections on days 7-21 of the estrous cycle. During the same time, gilts in the control group received saline. The ovaries were collected on predicted day 11 of the second studied estrous cycle. The cystic ovaries were supplied by more numerous NNFs than the control gonads. Moreover after DXM injections, the content of CATs and progesterone and androstendione (A4) in the cystic wall were elevated, while the levels of A4, testosterone and estradiol-17β in the cystic fluid were lowered. Our results show that in the porcine cystic ovaries, induced by DXM injections from middle phase of estrous cycle, increased the density of NNFs and level of CATs, and that it was accompanied by changes in the content of steroids. Moreover, this study is a further confirmation that the morphological and functional changes of cystic ovaries are partly dependent on phase of the estrous cycle in which the induction of the ovarian cysts was initiated.
Collapse
Affiliation(s)
- A. KOZŁOWSKA
- Department of Human Physiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland
| | | | | | | |
Collapse
|
44
|
Stener-Victorin E. Hypothetical physiological and molecular basis for the effect of acupuncture in the treatment of polycystic ovary syndrome. Mol Cell Endocrinol 2013; 373:83-90. [PMID: 23416841 DOI: 10.1016/j.mce.2013.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 01/10/2013] [Accepted: 01/10/2013] [Indexed: 10/27/2022]
Abstract
Clinical and experimental evidence indicates that acupuncture may be a safe alternative or complement in the treatment of endocrine and reproductive function in women with polycystic ovary syndrome (PCOS). This review describes potential etiological factors of PCOS with the aim to support potential mechanism of action of acupuncture to relieve PCOS related symptoms. The theory that increased sympathetic activity contributes to the development and maintenance of PCOS is presented, and that the effects of acupuncture are, at least in part, mediated by modulation of sympathetic outflow. While there are no relevant randomized controlled studies on the use of acupuncture to treat metabolic abnormalities in women with PCOS, a number of experimental studies indicate that acupuncture may improve metabolic dysfunction. For each aspect of PCOS, it is important to pursue new treatment strategies that have fewer negative side effects than drug treatments, as women with PCOS often require prolonged treatment.
Collapse
Affiliation(s)
- Elisabet Stener-Victorin
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Box 434, SE-405 30 Gothenburg, Sweden.
| |
Collapse
|
45
|
Johansson J, Redman L, Veldhuis PP, Sazonova A, Labrie F, Holm G, Johannsson G, Stener-Victorin E. Acupuncture for ovulation induction in polycystic ovary syndrome: a randomized controlled trial. Am J Physiol Endocrinol Metab 2013; 304:E934-43. [PMID: 23482444 PMCID: PMC4116535 DOI: 10.1152/ajpendo.00039.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Acupuncture has been demonstrated to improve menstrual frequency and to decrease circulating testosterone in women with polycystic ovary syndrome (PCOS). Our aim was to investigate whether acupuncture affects ovulation frequency and to understand the underlying mechanisms of any such effect by analyzing LH and sex steroid secretion in women with PCOS. This prospective, randomized, controlled clinical trial was conducted between June 2009 and September 2010. Thirty-two women with PCOS were randomized to receive either acupuncture with manual and low-frequency electrical stimulation or to meetings with a physical therapist twice a week for 10-13 wk. Main outcome measures were changes in LH secretion patterns from baseline to after 10-13 wk of treatment and ovulation frequency during the treatment period. Secondary outcomes were changes in the secretion of sex steroids, anti-Müllerian hormone, inhibin B, and serum cortisol. Ovulation frequency during treatment was higher in the acupuncture group than in the control group. After 10-13 wk of intervention, circulating levels of estrone, estrone sulfate, estradiol, dehydroepiandrosterone, dehydroepiandrosterone sulfate, androstenedione, testosterone, free testosterone, dihydrotestosterone, androsterone glucuronide, androstane-3α,17β-diol-3-glucuronide, and androstane-3α,17β-diol-17-glucuronide decreased within the acupuncture group and were significantly lower than in the control group for all of these except androstenedione. We conclude that repeated acupuncture treatments resulted in higher ovulation frequency in lean/overweight women with PCOS and were more effective than just meeting with the therapist. Ovarian and adrenal sex steroid serum levels were reduced with no effect on LH secretion.
Collapse
Affiliation(s)
- Julia Johansson
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Göteborg, Gothenburg, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Rashidi BH, Tehrani ES, Hamedani NA, Pirzadeh L. Effects of acupuncture on the outcome of in vitro fertilisation and intracytoplasmic sperm injection in women with polycystic ovarian syndrome. Acupunct Med 2013; 31:151-6. [PMID: 23376852 DOI: 10.1136/acupmed-2012-010198] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Different mechanisms have been described for the effects of acupuncture on fertility. Acupuncture may mediate neurotransmitter release, which results in the stimulation of gonadotrophin-releasing hormone secretion. The production of neurotransmitters such as endogenous opioids may also inhibit central nervous system outflow and biological stress response. Acupuncture may also stimulate blood flow to the uterus by inhibiting uterine central sympathetic nerve activity. Despite some evidence suggesting beneficial effects of acupuncture on in vitro fertilisation (IVF) success rates, recent clinical trials could not duplicate these effects, especially in patients who are infertile with polycystic ovarian syndrome (PCOS) who are undergoing IVF. METHODS A total of 62 women with PCOS undergoing IVF/intracytoplasmic sperm injection (ICSI) at Shayamehr IVF Clinic in Tehran (Iran) participated in a randomised, controlled trial. Participants were randomly allocated to one of two groups: there were 31 women each in the acupuncture (group I) and control groups (group II). Acupuncture based on traditional Chinese medicine was performed in group I versus no adjunctive treatment in group II. All acupuncture procedures were performed in five sessions: start of downregulation, start of stimulation, 2 days before ovum pick-up, and immediately before and after embryo transfer. We measured the rate of oocytes in metaphase II, the fertilisation rate and the quality of embryos. RESULTS Our results showed a statistically higher mean of good quality embryos in group I compared with group II (p=0.044). However, the mean number of metaphase II oocytes retrieved and fertilisation rate were similar between the two groups. We also could not find any significant differences among clinical, biochemical or ongoing pregnancy rates. CONCLUSIONS Acupuncture at an early stage of oocyte recruitment may have a beneficial effect on embryo quality. However, it seems to have no significant effect on other IVF/ICSI outcomes of women with PCOS. TRIAL REGISTRATION NUMBER (IRCT ID: 201011275181N4).
Collapse
Affiliation(s)
- Batool Hossein Rashidi
- Department of Obstetrics and Gynecology, Vali-e-Asr Reproductive Health Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | |
Collapse
|
47
|
Bernuci MP, Leite CM, Barros P, Kalil B, Leoni GB, Del Bianco-Borges B, Franci CR, Szawka RE, Lara HE, Anselmo-Franci JA. Transitory activation of the central and ovarian norepinephrine systems during cold stress-induced polycystic ovary in rats. J Neuroendocrinol 2013; 25:23-33. [PMID: 22882492 DOI: 10.1111/j.1365-2826.2012.02373.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 07/02/2012] [Accepted: 08/02/2012] [Indexed: 11/30/2022]
Abstract
Cold stress-induced ovarian sympathetic activation is associated with the development of ovarian cysts in rats. Although we have hypothesised that polycystic ovary (PCO) features induced by cold stress, as prevented by lesion of the noradrenergic nucleus locus coeruleus (LC), were a result of the increased activity of the ovarian norepinephrine (NE) system, this was not evident after 8 weeks of stress. In the present study, we investigated the temporal changes in LC and ovarian NE activities and steroid secretion in rats exposed to single (SS) or repeated (RS) cold stress. SS and 4 week (4W)-RS but not 8 week (8W)-RS increased c-Fos expression in the LC and ovarian NE release. Plasma oestradiol, testosterone and progesterone levels tended to increase in 4W-RS and were elevated in 8W-RS rats, which displayed PCO morphology. β-adrenergic receptor agonist increased steroid hormone release from the ovary of unstressed (US) but not from 8W-RS rats. To determine whether increased activity of noradrenergic system during the initial 4 weeks of RS would be sufficient to promote PCO, rats were exposed to 4 weeks of cold stress and kept in ambient temperature for the next 4 weeks (4W-RS/4W-US). Accordingly, PCO morphology, increased steroid secretion and decreased ovulation rate were found in 4W-RS/4W-US rats, strengthening the hypothesis that the initial increase in NE release triggers the development of PCO. The correlated activity of LC neurones and ovarian noradrenergic terminals and the induction of PCO in 4W-RS/4W-US rats provide functional evidence for a major role of NE in disrupting follicular development and causing the long-lasting endocrine abnormalities found in stress-induced PCO.
Collapse
Affiliation(s)
- M P Bernuci
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lansdown A, Rees DA. The sympathetic nervous system in polycystic ovary syndrome: a novel therapeutic target? Clin Endocrinol (Oxf) 2012; 77:791-801. [PMID: 22882204 DOI: 10.1111/cen.12003] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine condition associated with long-term health risks, including type 2 diabetes and vascular dysfunction in addition to reproductive sequelae. Many of the common features of PCOS, such as central obesity, hyperinsulinaemia and obstructive sleep apnoea (OSA), are associated with chronic sympathetic overactivity, suggesting that sympathoexcitation may be involved in the pathogenesis of this condition. Rodent models of polycystic ovaries have shown that ovarian sympathetic outflow may be increased, accompanied by elevated intra-ovarian synthesis of nerve growth factor (NGF) which may be involved in initiation of ovarian pathology. Patients with PCOS have evidence of increased muscle sympathetic nerve activity (MSNA), altered heart rate variability and attenuated heart rate recovery postexercise, compared with age- and BMI-matched controls, suggesting a generalized increase in sympathetic nerve activity. Active weight loss can reduce MSNA and whole body noradrenaline spillover, whereas low-frequency electroacupuncture decreased MSNA in overweight women with PCOS. Treatment of OSA with continuous positive airways pressure may reduce plasma noradrenaline levels and diastolic blood pressure and improve cardiac sympathovagal balance. Renal sympathetic denervation also reduced MSNA, noradrenaline spillover and blood pressure in two PCOS subjects with hypertension, accompanied by improved insulin sensitivity. The sympathetic nervous system may thus offer a new therapeutic target in PCOS but larger and longer-term studies are needed before these treatments can be considered in clinical practice.
Collapse
Affiliation(s)
- Andrew Lansdown
- Institute of Experimental and Molecular Medicine, Cardiff University, Cardiff, UK
| | | |
Collapse
|
49
|
Temporal window in which exposure to estradiol permanently modifies ovarian function causing polycystic ovary morphology in rats. Fertil Steril 2012; 98:1283-90. [PMID: 22854013 DOI: 10.1016/j.fertnstert.2012.07.1060] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 06/18/2012] [Accepted: 07/05/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the developmental window in which E(2) exposure produces irreversible changes in ovarian function resulting in polycystic ovary. DESIGN Basic experimental study. SETTING University animal laboratory. ANIMAL(S) Thirty Sprague-Dawley rats were administered a single E(2) valerate dose (10 mg/kg of weight) at 1, 7, 14, 21, or 30 days of age. Control rats were injected with the vehicle at 1 day of age. All rats were sacrificed at 6 months of age. INTERVENTION(S) Observation of vaginal opening, estrous cyclicity by vaginal smears, and ovarian morphometry in the 6-month-old rat. MAIN OUTCOME MEASURE(S) Measurement of ovarian noradrenaline by high-performance liquid chromatography coupled with electrochemical detection, serum levels of LH by enzyme-linked immunoassay, P, androstenedione, and E(2) by enzyme immunoassay. RESULT(S) Rats exposed to E(2) at 1, 7, or 14 days of life did not show estrual cycling activity and maintained a polycystic ovary (PCO) condition throughout the entirety of the study. However, if the exposure to E(2) occurred after postnatal day 21, the PCO-induced condition was reversible. In rats that developed a permanent PCO condition, we observed significant effects of E(2) on ovarian morphology if exposure occurred on postnatal day 1 and a presumable effect on the hypothalamus if the exposure occurred between postnatal days 1 and 14. CONCLUSION(S) Our findings suggest that in rats, the most sensitive period for the promotion of an irreversible PCO morphology by estrogenic compounds is during neonatal early follicular development.
Collapse
|
50
|
Abstract
Steroid hormones modulate a wide array of physiological processes including development, metabolism, and reproduction in various species. It is generally believed that these biological effects are predominantly mediated by their binding to specific intracellular receptors resulting in conformational change, dimerization, and recruitment of coregulators for transcription-dependent genomic actions (classical mechanism). In addition, to their cognate ligands, intracellular steroid receptors can also be activated in a "ligand-independent" manner by other factors including neurotransmitters. Recent studies indicate that rapid, nonclassical steroid effects involve extranuclear steroid receptors located at the membrane, which interact with cytoplasmic kinase signaling molecules and G-proteins. The current review deals with various mechanisms that function together in an integrated manner to promote hormone-dependent actions on the central and sympathetic nervous systems.
Collapse
Affiliation(s)
- S K Mani
- Department of Molecular & Cellular Biology and Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | |
Collapse
|