51
|
Çolakoğlu HE, Küplülü S, Polat IM, Pekcan M, Özenç E, Baklacı C, Seyrek-İntaş K, Gümen A, Vural MR. Association among lipopolysaccharide, the transforming growth factor-beta superfamily, follicular growth, and transcription factors in spontaneous bovine ovarian cysts. Domest Anim Endocrinol 2020; 70:106398. [PMID: 31677486 DOI: 10.1016/j.domaniend.2019.106398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 01/16/2023]
Abstract
The aim of this study was to investigate some of the growth and transcriptional factors originating from oocytes and granulosa cells in follicular fluid and to identify the relationships between the basic blood metabolite-metabolic hormones and intrafollicular lipopolysaccharide (LPS) concentrations. Thirty cows included in the study were allocated into 2 groups comprising 15 cows with healthy preovulatory follicles (cyclic cows) and 15 cows with confirmed cystic follicles. The ovaries and uteri of all cows were assessed by transrectal ultrasonographic examination. Blood serum samples were collected at 15, 25, 35, 45, and 55 d after calving for analysis of nonesterified fatty acids, β-hydroxybutyrate, insulin, glucose, IGF-I, ACTH, and cortisol. Ovaries and uteri were examined using transrectal ultrasound. Vaginal discharge was evaluated on the same days. Follicular fluid was also aspirated on days 35-55 from the healthy preovulatory follicles and cystic follicles using a transvaginal ovum pickup method. The densitometric levels of inhibin-α, growth and differentiation factor (GDF-9), bone morphogenetic protein (BMP-6), and GATA-4 and GATA-6 proteins were analyzed by the Western blotting technique; the concentrations of antimullerian hormone (AMH), IGF-I, estradiol-17 beta (E2), and progesterone (P4) were determined by ELISA; and the concentrations of LPS in the follicular fluid were measured by the Limulus amebocyte lysate test. The serum insulin, ACTH, and cortisol concentrations were higher in cystic cows than cyclic cows, but serum IGF-I concentrations were lower in cystic cows. The IGF-I concentrations of cystic follicular fluids were lower, whereas AMH levels were significantly greater than those of healthy preovulatory follicular fluids. The cystic follicles had significantly lower expression levels of GDF-9, BMP-6, GATA-4, and GATA-6; in contrast, inhibin-α expression and LPS concentrations were significantly higher than in healthy preovulatory follicles. The proportion of pathologic vaginal discharge within 25 d postpartum in cystic cows were higher than in the cyclic group. In conclusion, it is suggested that intrafollicular dysregulation of the transforming growth factor-β superfamily, growth, and transcriptional factors is affected by high intrafollicular LPS concentrations and systemic metabolic changes and these disturbances may be responsible for the generation of ovarian cysts.
Collapse
Affiliation(s)
- H E Çolakoğlu
- Department of Obstetrics and Gynecology, Ankara University, Faculty of Veterinary Medicine, Ankara, Turkey
| | - S Küplülü
- Department of Obstetrics and Gynecology, Ankara University, Faculty of Veterinary Medicine, Ankara, Turkey
| | - I M Polat
- Department of Obstetrics and Gynecology, Kırıkkale University, Faculty of Veterinary Medicine, Kırıkkale, Turkey
| | - M Pekcan
- Department of Biochemistry, Ankara University, Faculty of Veterinary Medicine, Ankara, Turkey
| | - E Özenç
- Department of Obstetrics and Gynecology, Afyon Kocatepe University, Faculty of Veterinary Medicine, Afyonkarahisar, Turkey
| | - C Baklacı
- Field Veterinarian, Alaca Farm, Bursa, Turkey
| | - K Seyrek-İntaş
- Department of Obstetrics and Gynecology, Bursa Uludağ University, Faculty of Veterinary Medicine, Bursa, Turkey
| | - A Gümen
- Department of Obstetrics and Gynecology, Bursa Uludağ University, Faculty of Veterinary Medicine, Bursa, Turkey
| | - M R Vural
- Department of Obstetrics and Gynecology, Ankara University, Faculty of Veterinary Medicine, Ankara, Turkey.
| |
Collapse
|
52
|
Kalhori Z, Mehranjani MS, Azadbakht M, Shariatzadeh MA. L-Carnitine improves endocrine function and folliculogenesis by reducing inflammation, oxidative stress and apoptosis in mice following induction of polycystic ovary syndrome. Reprod Fertil Dev 2019; 31:282-293. [PMID: 31039949 DOI: 10.1071/rd18131] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 06/29/2018] [Indexed: 01/15/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is related to low levels of serum l-carnitine, which has antioxidant, anti-inflammatory and antiapoptotic properties. The aim of this study was to investigate the effect of l-carnitine on folliculogenesis in mice following induction of PCOS. PCOS was induced by daily injections of testosterone enanthate (1mg per 100g, s.c., for 35 days). NMRI mice (21 days old) were divided into four groups (n=6 per group): Control, Control+l-carnitine, PCOS and PCOS+l-carnitine. Mice were treated with 500mgkg-1, i.p., l-carnitine every second day for 28 days. Ovaries were studied stereologically and serum concentrations of FSH, LH, testosterone, interleukin (IL)-6 and tumour necrosis factor (TNF)-α were determined using ELISA kits. Serum concentrations of malondialdehyde (MDA) and the ferric ion reducing antioxidant power (FRAP) were also analysed. Apoptosis of follicles was evaluated by terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end-labelling (TUNEL). CD31 was assessed immunohistochemically. Data were analysed using one-way analysis of variance (ANOVA) and Tukey's test, differences considered significant at P<0.05.The total volume of the ovary, cortex volume, oocyte volume, zona pellucida thickness and the number of antral follicles increased significantly, whereas the number of primary and preantral follicles decreased significantly, in the PCOS+l-carnitine versus PCOS group. In the PCOS+l-carnitine group, serum concentrations of FSH and FRAP increased significantly, whereas there were significant decreases in serum concentrations of testosterone, LH, MDA, IL-6 and TNF-α, as well as in the percentage of TUNEL-positive apoptotic cells, compared with the PCOS group. l-Carnitine improves folliculogenesis and is therefore suggested as a therapeutic supplement in the treatment of PCOS.
Collapse
Affiliation(s)
- Zahra Kalhori
- Department of Biology, Faculty of Science, Arak University, PO Box 381-5688138, Arak, Iran
| | | | - Mehri Azadbakht
- Department of Biology, Faculty of Science, Razi University, PO Box 6714967346, Kermanshah, Iran
| | | |
Collapse
|
53
|
Yan YL, Batzel P, Titus T, Sydes J, Desvignes T, BreMiller R, Draper B, Postlethwait JH. A Hormone That Lost Its Receptor: Anti-Müllerian Hormone (AMH) in Zebrafish Gonad Development and Sex Determination. Genetics 2019; 213:529-553. [PMID: 31399485 PMCID: PMC6781894 DOI: 10.1534/genetics.119.302365] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/04/2019] [Indexed: 12/26/2022] Open
Abstract
Fetal mammalian testes secrete Anti-Müllerian hormone (Amh), which inhibits female reproductive tract (Müllerian duct) development. Amh also derives from mature mammalian ovarian follicles, which marks oocyte reserve and characterizes polycystic ovarian syndrome. Zebrafish (Danio rerio) lacks Müllerian ducts and the Amh receptor gene amhr2 but, curiously, retains amh To discover the roles of Amh in the absence of Müllerian ducts and the ancestral receptor gene, we made amh null alleles in zebrafish. Results showed that normal amh prevents female-biased sex ratios. Adult male amh mutants had enormous testes, half of which contained immature oocytes, demonstrating that Amh regulates male germ cell accumulation and inhibits oocyte development or survival. Mutant males formed sperm ducts and some produced a few offspring. Young female mutants laid a few fertile eggs, so they also had functional sex ducts. Older amh mutants accumulated nonvitellogenic follicles in exceedingly large but sterile ovaries, showing that Amh helps control ovarian follicle maturation and proliferation. RNA-sequencing data partitioned juveniles at 21 days postfertilization (dpf) into two groups that each contained mutant and wild-type fish. Group21-1 upregulated ovary genes compared to Group21-2, which were likely developing as males. By 35 dpf, transcriptomes distinguished males from females and, within each sex, mutants from wild types. In adult mutants, ovaries greatly underexpressed granulosa and theca genes, and testes underexpressed Leydig cell genes. These results show that ancestral Amh functions included development of the gonadal soma in ovaries and testes and regulation of gamete proliferation and maturation. A major gap in our understanding is the identity of the gene encoding a zebrafish Amh receptor; we show here that the loss of amhr2 is associated with the breakpoint of a chromosome rearrangement shared among cyprinid fishes.
Collapse
Affiliation(s)
- Yi-Lin Yan
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Peter Batzel
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Tom Titus
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Jason Sydes
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Ruth BreMiller
- Institute of Neuroscience, University of Oregon, Eugene, Oregon 97403
| | - Bruce Draper
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616
| | | |
Collapse
|
54
|
Reive C. The Biological Measurements of Mindfulness-based Stress Reduction: A Systematic Review. Explore (NY) 2019; 15:295-307. [DOI: 10.1016/j.explore.2019.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
|
55
|
Hosseini E, Shahhoseini M, Afsharian P, Karimian L, Ashrafi M, Mehraein F, Afatoonian R. Role of epigenetic modifications in the aberrant CYP19A1 gene expression in polycystic ovary syndrome. Arch Med Sci 2019; 15:887-895. [PMID: 31360184 PMCID: PMC6657255 DOI: 10.5114/aoms.2019.86060] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/23/2017] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION In this study, the global DNA methylation, histone acetylation and methylation levels of cumulus cells (CCs) in infertile polycystic ovary syndrome (PCOS) patients and the correlation of these epigenetic modifications with the expression of the ovarian aromatase gene (as an important marker in the etiology of PCOS) were investigated. MATERIAL AND METHODS A cross-sectional study was conducted on 24 patients (12 PCOS patients and 12 healthy women), who underwent ovarian stimulation. Nucleosome ELISA was performed, in order to identify the global occupancy level of Mecp2 (as a marker of DNA methylation) and H3K9me2/H3K9ac as histone modification markers in chromatin fractions obtained from CCs. The CYP19A1 gene expression was measured by qRT-PCR. The level of DNA incorporation of MeCP2, histone modification markers and binding of estrogen receptor β (ERβ) to CYP19A1 regulatory sequences were examined by ChIP-QPCR assay. RESULTS The data demonstrate a significant increase in global occupancy levels of MeCP2 and H3K9ac markers and a decrease of H3K9me2 to chromatin in CCs of PCOS patients vs. control group. Furthermore, CYP19A1 gene expression, and the incorporation of H3K9ac in PII, PI.3, and PI.4 promoters of CYP19A1 in PCOS, were higher than those of controls. Also, significant hypomethylation of H3K9 at PII and DNA hypomethylated at PII and PI.3 promoters and differential binding of ERβ to three promoters were observed in PCOS patients (p < 0.05). CONCLUSIONS Aromatase expression can be affected by epigenetic modifications and differential ERβ binding to the proximal CYP19A1 promoters. These mechanisms may be involved in the enhanced aromatase transcription during ovarian stimulation in PCOS patients.
Collapse
Affiliation(s)
- Elham Hosseini
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Obstetrics and Gynecology, IVF Clinic, Mousavi Hospital, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Shahhoseini
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Parvaneh Afsharian
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Leila Karimian
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mahnaz Ashrafi
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Fereshteh Mehraein
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Afatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
56
|
Fruzzetti F, Capozzi A, Canu A, Lello S. Treatment with d-chiro-inositol and alpha lipoic acid in the management of polycystic ovary syndrome. Gynecol Endocrinol 2019; 35:506-510. [PMID: 30612488 DOI: 10.1080/09513590.2018.1540573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To evaluate the effects of the combination of d-chiro inositol and alpha lipoic acid on menstrual cycles and insulin sensitivity in women with polycystic ovary syndrome (PCOS). Forty-one women with PCOS and 31 controls have been enrolled in the study. The menstrual cycle, BMI, homeostasis model assessment index (HOMA-I), and insulin secretion in response to an OGTT were evaluated before and after 6 months of treatment. During the observation period, the patients have been asked to not modify their diet and physical activity. The menstrual cycle length improved in 76.7% of the women. Ovulation was restored in 40%. During treatment, BMI significantly decreased (p<.002). The HOMA-I and insulin secretion were unchanged by treatment. However, when women were divided according to the presence of insulin resistance (IR; HOMA-I > 2.5), in those with IR the HOMA-I and the insulin secretion significantly decreased (p<.05 and p<.006). The association of d-chiro-inositol and alpha lipoic acid improves menstrual cycle length, restoring ovulation in the majority of women. Insulin sensitivity improved in women with IR only, confirming that in presence of IR the d-chiro-inositol has a role in restoring the insulin action overcoming the inactivity of epimerase in transforming myo-inositol to d-chiro inositol.
Collapse
Affiliation(s)
- Franca Fruzzetti
- a Department of Obstetrics and Gynecology , Santa Chiara Hospital , Pisa , Italy
| | - Anna Capozzi
- b Department of Woman and Child Health , Policlinico Gemelli Foundation IRCCS , Rome , Italy
| | - Alessandro Canu
- a Department of Obstetrics and Gynecology , Santa Chiara Hospital , Pisa , Italy
| | - Stefano Lello
- b Department of Woman and Child Health , Policlinico Gemelli Foundation IRCCS , Rome , Italy
| |
Collapse
|
57
|
Mohammadi M. Oxidative Stress and Polycystic Ovary Syndrome: A Brief Review. Int J Prev Med 2019; 10:86. [PMID: 31198521 PMCID: PMC6547785 DOI: 10.4103/ijpvm.ijpvm_576_17] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common hormonal disorders, occurring in 5-10% women in reproductive ages. Despite a long history of studies on PCOS, its etiology is still unknown. Oxidative stress is now recognized to play a central role in the pathophysiology of many different disorders, including PCOS. Although intracellular reactive oxygen species (ROS) production and propagation are controlled by highly complex antioxidant enzymatic and non-enzymatic systems, understanding of mechanisms that oxidative stress is important to develop strategies for prevention and therapy of PCOS. This article reviews the literature data related to the mechanisms of oxidative stress in PCOS.
Collapse
Affiliation(s)
- Masoumeh Mohammadi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
58
|
Rashad NM, Ateya MAM, Saraya YS, Elnagar WM, Helal KF, Lashin MEB, Abdelrhman AA, Alil AE, Yousef MS. Association of miRNA - 320 expression level and its target gene endothelin-1 with the susceptibility and clinical features of polycystic ovary syndrome. J Ovarian Res 2019; 12:39. [PMID: 31064393 PMCID: PMC6505291 DOI: 10.1186/s13048-019-0513-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/16/2019] [Indexed: 01/01/2023] Open
Abstract
Background Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder characterized by obesity, hyperandrogenism, and insulin resistance (IR). MicroRNAs (miRNAs) are small noncoding RNA associated with ovarian follicle development and female fertility. The objective of this study was to investigate the role of miRNA- 320 and its target gene endothelin-1 (ET-1) as a noninvasive biomarker of PCOS and to evaluate its possible relationship with IR as well as clinic-morphological features of PCOS. Methods Case-control study enrolled 60 patients with PCOS and 40 control group. We subdivided our PCOS women according to homeostasis model assessments of insulin resistance (HOMA-IR) to PCOS women with and without IR.ET-1 levels were measured by ELISA. We estimated the serum expression level of miRNA- 320 by real-time polymerase chain reaction. Results Our results revealed that serum miR-320 expression level was lower in PCOS patients compared to controls, in particular, PCOS women with IR. Moreover, it was negatively correlated to its target gene; ET-I as well as fasting serum insulin (FSI), HOMA-IR, PCOS phenotype; hirsutism score, ovarian volume and antral follicle count (AFC). In the PCOS group, linear regression analysis revealed that only hirsutism and HOMA-IR was the main predictor of expression levels of miRNA − 320 among other clinical and laboratory biomarkers of PCOS. The sensitivity and specificity of serum miR-320 expression levels in diagnosis PCOS was 80, and 97.5% respectively. Conclusion The Expression serum levels of miR-320 were lower in PCOS compared to control and it could be a noninvasive diagnostic biomarker of PCOS.
Collapse
Affiliation(s)
- Nearmeen M Rashad
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | | | - Yasser S Saraya
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Walid Mohamed Elnagar
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Khaled Fathy Helal
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamed El-Bakry Lashin
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amr Ahmed Abdelrhman
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ayman E Alil
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammed S Yousef
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
59
|
Merlo E, Schereider IRG, Simões MR, Vassallo DV, Graceli JB. Mercury leads to features of polycystic ovary syndrome in rats. Toxicol Lett 2019; 312:45-54. [PMID: 31071422 DOI: 10.1016/j.toxlet.2019.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 05/05/2019] [Indexed: 01/28/2023]
Abstract
Mercury (Hg) is a heavy metal and Hg exposure is associated with various neural, immune, and cardiovascular abnormalities. However, few studies have evaluated Hg's toxicologic effect on reproductive and metabolic functions. In this study, we assessed whether Hg exposure results in reproductive and metabolic abnormalities. Hg was administered to adult female Wistar rats, mimicking the Hg levels found in exposed human blood, and their reproductive and metabolic function was assessed. Rats exposed to Hg displayed abnormal estrous cyclicity and ovarian follicular development, with a reduction in ovarian antral follicles and an increase in atretic and cystic ovarian follicles. Uterine atrophy with the presence of inflammatory cells was observed in Hg-exposed rats. The presence of abnormal ovarian fat accumulation, as well as increased ovarian lipid drops accumulation, was observed in Hg-exposed rats. Ovarian oxidative stress was also present in the Hg-exposed rats. High fasting glucose levels, glucose, and insulin intolerance were observed in Hg-exposed rats. Thus, these data suggest that Hg exposure led to abnormal reproductive and metabolic features similar to those found in the polycystic ovary syndrome (PCOS) rat models.
Collapse
Affiliation(s)
- Eduardo Merlo
- Dept of Morphology, Federal University of Espírito Santo, Brazil
| | | | - Maylla R Simões
- Dept of Physiology, Federal University of Espírito Santo, Brazil
| | | | - Jones B Graceli
- Dept of Morphology, Federal University of Espírito Santo, Brazil.
| |
Collapse
|
60
|
Sagvekar P, Kumar P, Mangoli V, Desai S, Mukherjee S. DNA methylome profiling of granulosa cells reveals altered methylation in genes regulating vital ovarian functions in polycystic ovary syndrome. Clin Epigenetics 2019; 11:61. [PMID: 30975191 PMCID: PMC6458760 DOI: 10.1186/s13148-019-0657-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022] Open
Abstract
Background Women with polycystic ovary syndrome (PCOS) manifest a host of ovarian defects like impaired folliculogenesis, anovulation, and poor oocyte quality, which grossly affect their reproductive health. Addressing the putative epigenetic anomalies that tightly regulate these events is of foremost importance in this disorder. We therefore aimed to carry out DNA methylome profiling of cumulus granulosa cells and assess the methylation and transcript expression profiles of a few differentially methylated genes contributing to ovarian defects in PCOS. A total of 20 controls and 20 women with PCOS were selected from a larger cohort of women undergoing IVF, after carefully screening their sera and follicular fluids for hormonal and biochemical parameters. DNA extracted from cumulus granulosa cells of three women each, from control and PCOS groups was subjected to high-throughput, next generation bisulfite sequencing, using the Illumina HiSeq 2500® platform. Remaining samples were used for the validation of methylation status of some identified genes by pyrosequencing, and the transcript expression profiles of these genes were assessed by quantitative real-time PCR. Results In all, 6486 CpG sites representing 3840 genes associated with Wnt signaling, G protein receptor, endothelin/integrin signaling, angiogenesis, chemokine/cytokine-mediated inflammation, etc., showed differential methylation in PCOS. Hypomethylation was noted in 2977 CpGs representing 2063 genes while 2509 CpGs within 1777 genes showed hypermethylation. Methylation differences were also noted in noncoding RNAs regulating several ovarian functions that are dysregulated in PCOS. Few differentially methylated genes such as aldo-keto reductase family 1 member C3, calcium-sensing receptor, resistin, mastermind-like domain 1, growth hormone-releasing hormone receptor and tumor necrosis factor, which predominantly contribute to hyperandrogenism, premature luteolysis, and oocyte development defects, were explored as novel epigenetic candidates in mediating ovarian dysfunction. Methylation profiles of these genes matched with our NGS findings, and their transcript expression patterns correlated with the gene hypo- or hypermethylation status. Conclusion Our findings suggest that the epigenetic dysregulation of genes involved in important processes associated with follicular development may contribute to ovarian defects observed in women with PCOS. Electronic supplementary material The online version of this article (10.1186/s13148-019-0657-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pooja Sagvekar
- Department of Molecular Endocrinology, ICMR-National Institute for Research in Reproductive Health, J.M. Street, Parel, Mumbai, Maharashtra, 400012, India
| | - Pankaj Kumar
- Colin Jamura Lab, Institute for Stem Cell Biology and Regenerative Medicine (inStem), National Centre for Biological Sciences (NCBS), GKVK, Bellary Road, Bangalore, 560065, India
| | - Vijay Mangoli
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai, Maharashtra, 400007, India
| | - Sadhana Desai
- Fertility Clinic and IVF Center, 12-Springfield, 19-Vachha Gandhi Road, Gamdevi, Mumbai, Maharashtra, 400007, India
| | - Srabani Mukherjee
- Department of Molecular Endocrinology, ICMR-National Institute for Research in Reproductive Health, J.M. Street, Parel, Mumbai, Maharashtra, 400012, India.
| |
Collapse
|
61
|
Usselman CW, Yarovinsky TO, Steele FE, Leone CA, Taylor HS, Bender JR, Stachenfeld NS. Androgens drive microvascular endothelial dysfunction in women with polycystic ovary syndrome: role of the endothelin B receptor. J Physiol 2019; 597:2853-2865. [PMID: 30847930 DOI: 10.1113/jp277756] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/04/2019] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS Polycystic ovary syndrome (PCOS) is a complex syndrome with cardiovascular risk factors, including obesity and insulin resistance. PCOS is also associated with high androgens, increases the risk of cardiovascular dysfunction in women. Due to the complexity of PCOS, had it has been challenging to isolate specific causes of the cardiovascular dysfunction. Our measure of cardiovascular dysfunction (endothelial dysfunction) was most profound in lean women with PCOS. The endothelin-1-induced vasodilation in these PCOS subject, was dependent on the ETB R but was not NO-dependent. We also demonstrated oestrogen administration improved endothelial function in lean and obese women with PCOS likely because oestrogen increased NO availability. Our studies indicate a primary role for androgens in cardiovascular dysfunction in PCOS. ABSTRACT Endothelin-1 (ET-1) is an indicator of endothelial injury and dysfunction and is elevated in women with androgen excess polycystic ovary syndrome (AE-PCOS). The endothelin B receptor (ETB R) subtype mediates vasodilatation, but is blunted in women with PCOS. We hypothesized that androgen drives endothelial dysfunction in AE-PCOS women and oestradiol (EE) administration reverses these effects. We assessed microvascular endothelial function in women with (7 lean and 7 obese) and without AE-PCOS (controls, 6 lean, 7 obese). Only obese AE-PCOS women were insulin resistant (IR). We evaluated cutaneous vascular conductance (%CVCmax ) with laser Doppler flowmetry during low dose intradermal microdialysis ET-1 perfusions (1, 3, 4, 5 and 7 pmol) with either lactated Ringer solution alone, or with ETB R (BQ-788), or nitric oxide (NO) inhibition (l-NAME). Log[ET-1]-%maxCVC dose-response curves demonstrated reduced vasodilatory responses to ET-1 in lean AE-PCOS (logED50 , 0.59 ± 0.08) versus lean controls (logED50 , 0.49 ± 0.09, P < 0.05), but not compared to obese AE-PCOS (logED50 , 0.65 ± 0.09). ETB R inhibition decreased ET-1-induced vasodilatation in AE-PCOS women (logED50 , 0.64 ± 0. 22, P < 0.05). This was mechanistically observed at the cellular level, with ET-1-induced, DAF-FM-measurable endothelial cell NO production, which was abrogated by dihydrotestosterone in an androgen receptor-dependent manner. EE augmented the cutaneous vasodilating response to ET-1(logED50 0.29 ± 0.21, 0.47 ± 0.09, P < 0.05 for lean and obese, respectively). Androgens drive endothelial dysfunction in lean and obese AE-PCOS. We propose that the attenuated ET-1-induced vasodilatation in AE-PCOS is a consequence of androgen receptor-mediated, suppressed ETB R-stimulated NO production, and is reversed with EE.
Collapse
Affiliation(s)
- Charlotte W Usselman
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.,Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Timur O Yarovinsky
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Frances E Steele
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Cheryl A Leone
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey R Bender
- Departments of Internal Medicine (Cardiovascular Medicine) and Immunobiology, Yale School of Medicine, New Haven, CT, USA.,Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Nina S Stachenfeld
- John B. Pierce Laboratory, Yale School of Medicine, New Haven, CT, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
62
|
Reckelhoff JF. Androgens and Blood Pressure Control: Sex Differences and Mechanisms. Mayo Clin Proc 2019; 94:536-543. [PMID: 30713048 PMCID: PMC6546172 DOI: 10.1016/j.mayocp.2018.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/13/2023]
Abstract
The role that androgens play in mediating elevated blood pressure is unclear. Low levels of androgens in men and increased levels of androgens in women, as occurs with polycystic ovary syndrome (PCOS), are both associated with increased risk for cardiovascular disease and elevated blood pressure. We have used animal models to evaluate the potential mechanisms by which men and women have differential responses to androgens that affect regulation of blood pressure and the implications these may have for the health of men and women.
Collapse
Affiliation(s)
- Jane F Reckelhoff
- Department of Cell and Molecular Biology, Mississippi Center of Excellence in Perinatal Research and Women's Health Research Center, University of Mississippi Medical Center, Jackson.
| |
Collapse
|
63
|
Merhi Z, Kandaraki EA, Diamanti-Kandarakis E. Implications and Future Perspectives of AGEs in PCOS Pathophysiology. Trends Endocrinol Metab 2019; 30:150-162. [PMID: 30712978 DOI: 10.1016/j.tem.2019.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 01/12/2023]
Abstract
Human, animal, and in vitro studies provide evidence that advanced glycation end-products (AGEs) may contribute to the pathogenesis of polycystic ovary syndrome (PCOS) and its metabolic and reproductive consequences. AGEs are able to induce, via activation of key intracellular signaling pathways, the generation of oxidative stress and proinflammatory cytokines, thus contributing to the adverse health impact of PCOS. This review presents the implications of AGEs in several disease pathophysiologies, including PCOS, as well as the cellular and systemic effects of AGEs on insulin resistance (IR), hyperandrogenemia, endoplasmic reticulum (ER) stress, hypoxia, and ovarian function. The gaps in our knowledge will serve as launching pad for future developments ranging from dietary and lifestyle changes to pharmaceutical interventions aiming at potential applications in women with PCOS.
Collapse
Affiliation(s)
- Zaher Merhi
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Eleni A Kandaraki
- St Bartholomew's Hospital, Department of Endocrinology, London EC1A 7BE, UK
| | - Evanthia Diamanti-Kandarakis
- Medical School, University of Athens, 11527 Goudi, Athens, Greece; Endocrinology and Diabetes Department, Hygeia Hospital, 15123 Marousi, Athens, Greece
| |
Collapse
|
64
|
Jia C, Xu H, Xu Y, Xu Y, Shi Q. Serum metabolomics analysis of patients with polycystic ovary syndrome by mass spectrometry. Mol Reprod Dev 2019; 86:292-297. [PMID: 30624822 DOI: 10.1002/mrd.23104] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a set of symptoms caused by elevated androgens (male hormones) in females. PCOS is the most common endocrine disorder among women between 18 and 44 years. Currently, the pathogenesis of PCOS remains unclear. Liquid chromatography-mass spectrometry (LC/MS)-based metabolomics is becoming more and more useful for medical research, especially in revealing the mechanism of the disease. The aim of this study was to investigate the difference of serum metabolic profiles in patients with PCOS and healthy control to better understand the mechanism of this disease. Ten patients with PCOS and 10 healthy people were recruited for this study. The serum samples were collected for LC/MS analysis. Multivariate statistical analysis was performed to discover and identify the potential biomarkers. Six biomarkers were found and identified. The biomarkers belonged to different metabolic pathway including lipid metabolism, carnitine metabolism, androgen metabolism, and bile acid metabolism. Those biomarkers also played different roles in disease progression. Metabolomics is a powerful tool used in research of the mechanism involved in this disease to provide useful information for better understanding of PCOS.
Collapse
Affiliation(s)
- Chunshu Jia
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital of Jilin University, Changchun, China
| | - Hongmei Xu
- Department of Obstetrics, First Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Department of Nephrology, First Hospital of Jilin University, China
| | - Qingyang Shi
- Centre for Reproductive Medicine, Centre for Prenatal Diagnosis, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
65
|
Kupreeva M, Diane A, Lehner R, Watts R, Ghosh M, Proctor S, Vine D. Effect of metformin and flutamide on insulin, lipogenic and androgen-estrogen signaling, and cardiometabolic risk in a PCOS-prone metabolic syndrome rodent model. Am J Physiol Endocrinol Metab 2019; 316:E16-E33. [PMID: 30153063 PMCID: PMC6417686 DOI: 10.1152/ajpendo.00018.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is highly associated with cardiometabolic risk and the metabolic syndrome (MetS), predisposing women to increased risk of developing type 2 diabetes and cardiovascular disease. Metformin is commonly used to treat insulin resistance-glucose intolerance, and flutamide, an androgen receptor (AR) antagonist, is used to target hyperandrogenemia and dyslipidemia. Currently, the physiological mechanism of action of these treatments on androgen, lipidogenic, and insulin signaling pathways remains unclear in PCOS. The aim of this study was to investigate the effects and mechanisms of action of metformin and flutamide on plasma lipid-apolipoprotein (Apo)B-lipoprotein and insulin-glucose metabolism, and endocrine-reproductive indices in a PCOS-prone MetS rodent model. PCOS-prone rodents were treated with metformin (300 mg/kg body wt), flutamide (30 mg/kg body wt), or metformin + flutamide combination treatment for 6 wk. Metformin was shown to improve fasting insulin and HOMA-IR, whereas flutamide and combination treatment were shown to reduce plasma triglycerides, ApoB48, and ApoB100, and this was associated with decreased intestinal secretion of ApoB48/triglyceride. Flutamide and metformin were shown to reduce plasma androgen indices and to improve ovarian primary and preovulatory follicle frequency. Metformin treatment increased hepatic estrogen receptor (ER)α, and metformin-flutamide decreased intestinal AR and increased ERα mRNA expression. Metformin-flutamide treatment upregulated hepatic and intestinal insulin signaling, including insulin receptor, MAPK1, and AKT2. In conclusion, cardiometabolic risk factors, in particular ApoB-hypertriglyceridemia, are independently modulated via the AR, and understanding the contribution of AR and insulin-signaling pathways further may facilitate the development of targeted interventions in high-risk women with PCOS and MetS.
Collapse
Affiliation(s)
- M. Kupreeva
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - A. Diane
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - R. Lehner
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - R. Watts
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - M. Ghosh
- Division of Endocrinology and Metabolism, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - S. Proctor
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - D. Vine
- Metabolic and Cardiovascular Disease Laboratory, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
66
|
Stanhewicz AE, Wenner MM, Stachenfeld NS. Sex differences in endothelial function important to vascular health and overall cardiovascular disease risk across the lifespan. Am J Physiol Heart Circ Physiol 2018; 315:H1569-H1588. [PMID: 30216121 PMCID: PMC6734083 DOI: 10.1152/ajpheart.00396.2018] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/22/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
Diseases of the cardiovascular system are the leading cause of morbidity and mortality in men and women in developed countries, and cardiovascular disease (CVD) is becoming more prevalent in developing countries. The prevalence of atherosclerotic CVD in men is greater than in women until menopause, when the prevalence of CVD increases in women until it exceeds that of men. Endothelial function is a barometer of vascular health and a predictor of atherosclerosis that may provide insights into sex differences in CVD as well as how and why the CVD risk drastically changes with menopause. Studies of sex differences in endothelial function are conflicting, with some studies showing earlier decrements in endothelial function in men compared with women, whereas others show similar age-related declines between the sexes. Because the increase in CVD risk coincides with menopause, it is generally thought that female hormones, estrogens in particular, are cardioprotective. Moreover, it is often proposed that androgens are detrimental. In truth, the relationships are more complex. This review first addresses female and male sex hormones and their receptors and how these interact with the cardiovascular system, particularly the endothelium, in healthy young women and men. Second, we address sex differences in sex steroid receptor-independent mechanisms controlling endothelial function, focusing on vascular endothelin and the renin-angiotensin systems, in healthy young women and men. Finally, we discuss sex differences in age-associated endothelial dysfunction, focusing on the role of attenuated circulating sex hormones in these effects.
Collapse
Affiliation(s)
- Anna E Stanhewicz
- Department of Kinesiology, Pennsylvania State University , University Park, Pennsylvania
| | - Megan M Wenner
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| | - Nina S Stachenfeld
- The John B. Pierce Laboratory, New Haven, Connecticut
- Department of Obstetrics, Gynecology and Reproductive Sciences and Yale School of Public Health, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
67
|
Tonellotto Dos Santos J, Escarião da Nóbrega J, Serrano Mujica LK, Dos Santos Amaral C, Machado FA, Manta MW, Rizzetti TM, Zanella R, Fighera R, Antoniazzi AQ, Gonçalves PBD, Comim FV. Prenatal Androgenization of Ewes as a Model of Hirsutism in Polycystic Ovary Syndrome. Endocrinology 2018; 159:4056-4064. [PMID: 30376052 DOI: 10.1210/en.2018-00781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/24/2018] [Indexed: 02/04/2023]
Abstract
The main clinical feature associated with hyperandrogenism in polycystic ovary syndrome (PCOS) in humans is hirsutism, where hair increases its length, pigmentation, and particularly its diameter. Currently, it is not known whether PCOS animal models also exhibit changes in the hair. Therefore, the aim of this study was to explore the wool characteristics in sheep prenatally androgenized (PA) with testosterone propionate. After 4 and 13 months of life, wool was collected from the top of the shoulder of both females and males (both androgenized and controls). The offspring sheep were followed for up to 19 months of life to evaluate testosterone and androstenedione serum levels by ultra-high-performance liquid chromatography-tandem mass spectrometry, determine insulin and glucose response to intravenous glucose tolerance test, and address estrus cyclicity during the second breeding season. PA male animals showed a reduction in wool fiber diameter at 4 months of age compared with controls (P = 0.02) but not at 13 months, whereas PA females showed increased hair diameter at 13 months (P = 0.002), with no difference at 4 months. No substantial changes in other hair parameters (length, color, and medullation) were identified. In addition, increased levels of serum testosterone were observed in PA female sheep compared with controls at 12 months (P = 0.03). Our results indicate for the first time, to our knowledge, that changes in wool fiber diameter observed in PA ewes replicate, at the translational level, the increase in hair diameter in hirsute women with PCOS.
Collapse
Affiliation(s)
- Joabel Tonellotto Dos Santos
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Jandui Escarião da Nóbrega
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Lady Katerine Serrano Mujica
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Carolina Dos Santos Amaral
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Fabrício Amadori Machado
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Manuela W Manta
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Tiele Medianeira Rizzetti
- Laboratory of Pesticide Analysis (LARP), Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Renato Zanella
- Laboratory of Pesticide Analysis (LARP), Department of Chemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rafael Fighera
- Department of Pathology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Alfredo Quites Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
| | - Fabio Vasconcellos Comim
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria, Santa Maria, Brazil
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
68
|
Amelia D, Santoso B, Purwanto B, Miftahussurur M, Joewono HT. Effects of Moringa oleifera on Insulin Levels and Folliculogenesis in Polycystic Ovary Syndrome Model with Insulin Resistance. ACTA ACUST UNITED AC 2018; 18:22-30. [PMID: 30369967 PMCID: PMC6174639 DOI: 10.2174/1871522218666180426100754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/14/2022]
Abstract
Background: Insulin resistance is a frequent metabolic disorder in Polycystic Ovary Syndrome (PCOS). Moringa oleifera has been shown to increase insulin expres-sion and decrease the degree of insulin in diabetes mellitus, therefore it is expected that Moringa oleifera could decrease insulin levels and increase folliculogenesis in PCOS. Objective: To prove the effect of Moringa oleifera leaf extract in various doses might decrease the insulin levels and increase folliculogenesis in female PCOS-insulin resistant rats. Methods: The three month old white rat of Wistar strain (Rattus norvegicus) 100-130 grams were divided into five groups (n=8) including normal control, PCOS-insulin re-sistance, PCOS-insulin resistance given metformin and PCOS-resistance insulin were giv-en Moringa oleifera leaf extract in two doses. Then, the PCOS model-insulin resistance by injection of testosterone propionate for 28 days. After 14 days treatment, we analysed insulin levels and folliculogenesis. Results: The PCOS control group showed a significant increase in insulin levels compared to the normal control group. The insulin levels from group treatment with Moringa oleifera leaf extract of 250 mg/kgBW was significantly lower than the PCOS control group. Ovarian histology analysis found that the number and diameter of follicle of PCOS control group showed a significant decrease compared to normal control group. In addition, the treatment with metformin and leaf Moringa oleifera dose 250 mg/kgBW and 500 mg/kgBW showed significant increase of folliculogenesis compared to PCOS control group. Conclusions: Moringa oleifera could lowering the blood insulin levels, subsequently decreasing the androgen thus allowed the increasing of folliculogenesis in PCOS.
Collapse
Affiliation(s)
- Dessy Amelia
- School of Reproduction Health, Faculty of Medicine of Universitas Airlangga, Mayjen Prof. Dr. Moestopo 47, Pacar Kembang, Surabaya, Jawa Timur 60132, Indonesia
| | - Budi Santoso
- Department of Obstetrics and Gynaecology, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Mayjend Prof. Dr. Moestopo No. 6 - 8, Airlangga, Gubeng, Kota Surabaya, Jawa Timur 60285, Indonesia
| | - Bambang Purwanto
- Department of Physiology, Faculty of Medicine of Universitas Airlangga, Mayjen Prof. Dr. Moestopo 47, Pacar Kembang, Surabaya, Jawa Timur 60132, Indonesia
| | - Muhammad Miftahussurur
- Gastroentero-Hepatology Division, Department of Internal Medicine, Faculty of Medicine-Dr. Soetomo Teaching Hospital-Institute of Tropical Disease, Universitas Airlangga, Mayjend Prof. Dr. Moestopo No. 6 - 8, Airlangga, Gubeng, Kota Surabaya, Jawa Timur 60285, Indonesia
| | - Hermanto Tri Joewono
- Department of Obstetrics and Gynaecology, Faculty of Medicine-Dr. Soetomo Teaching Hospital, Universitas Airlangga, Mayjend Prof. Dr. Moestopo No. 6 - 8, Airlangga, Gubeng, Kota Surabaya, Jawa Timur 60285, Indonesia
| | | |
Collapse
|
69
|
Patil CN, Racusen LC, Reckelhoff JF. Consequences of advanced aging on renal function in chronic hyperandrogenemic female rat model: implications for aging women with polycystic ovary syndrome. Physiol Rep 2018; 5:5/20/e13461. [PMID: 29051304 PMCID: PMC5661229 DOI: 10.14814/phy2.13461] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 01/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine and reproductive disorder in premenopausal women, characterized by hyperandrogenemia, metabolic syndrome, and inflammation. Women who had PCOS during their reproductive years remain hyperandrogenemic after menopause. The consequence of chronic hyperandrogenemia with advanced aging has not been studied to our knowledge. We have characterized a model of hyperandrogenemia in female rats and have aged them to 22–25 months to mimic advanced aging in hyperandrogenemic women, and tested the hypothesis that chronic exposure to hyperandrogenemia with aging has a deleterious effect on renal function. Female rats were chronically implanted with dihydrotestosterone pellets (DHT 7.5 mg/90 days) that were changed every 85 days or placebo pellets, and renal function was measured by clearance methods. Aging DHT‐treated females had a threefold higher level of DHT with significantly higher body weight, mean arterial pressure, left kidney weight, proteinuria, and kidney injury molecule‐1 (KIM‐1), than did age‐matched controls. In addition, DHT‐treated‐old females had a 60% reduction in glomerular filtration rate, 40% reduction in renal plasma flow, and significant reduction in urinary nitrate and nitrite excretion (UNOxV), an index of nitric oxide production. Morphological examination of kidneys showed that old DHT‐treated females had significant focal segmental glomerulosclerosis, global sclerosis, and interstitial fibrosis compared to controls. Thus chronic hyperandrogenemia that persists into old age in females is associated with renal injury. These data suggest that women with chronic hyperandrogenemia such as in PCOS may be at increased risk for development of chronic kidney disease with advanced age.
Collapse
Affiliation(s)
- Chetan N Patil
- Department of Physiology, The Women's Health Research Center University of Mississippi Medical Center, Jackson, Mississippi.,Department of Biophysics, The Women's Health Research Center University of Mississippi Medical Center, Jackson, Mississippi
| | - Lorraine C Racusen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jane F Reckelhoff
- Department of Physiology, The Women's Health Research Center University of Mississippi Medical Center, Jackson, Mississippi .,Department of Biophysics, The Women's Health Research Center University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
70
|
Bakhshalizadeh S, Amidi F, Shirazi R, Shabani Nashtaei M. Vitamin D3 regulates steroidogenesis in granulosa cells through AMP-activated protein kinase (AMPK) activation in a mouse model of polycystic ovary syndrome. Cell Biochem Funct 2018; 36:183-193. [PMID: 29676471 DOI: 10.1002/cbf.3330] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/20/2018] [Accepted: 03/04/2018] [Indexed: 01/11/2023]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic disorder in reproductive-aged women. Hormonal abnormality caused by steroidogenesis disturbances appears to be the main culprit of the clinical picture in PCOS. Vitamin D3 could regulate steroidogenesis in granulosa cells, but the mechanism of action of vitamin D3 on steroidogenesis remains unknown. AMP-activated protein kinase (AMPK) has a modulating role in steroid hormone production. We investigated the effect of vitamin D3 on steroidogenesis in cultured granulosa cells of dehydroepiandrosterone-induced PCOS mice and studied the involvement of AMPK signalling pathway in the current process. Immunoblotting assay showed that vitamin D3 could increase phosphorylation of AMPK alpha and acetyl-CoA carboxylase, main substrate of AMPK. Vitamin D3 and 5-aminoimidazole-4-carboxamide-1-β-D-riboside or Aicar (AMPK activator) not only reduced gene expression of steroidogenic enzymes (P450scc or Cyp11a1, StAR, Cyp19a1 and 3B-HSD), but also reduced production of progesterone and 17B-estradiol assessed by radioimmunoassay. Pretreatment with compound C (AMPK inhibitor) decreased APMK phosphorylation and eliminated the effects of vitamin D3 and Aicar on steroidogenic enzymes expression and estradiol and progesterone production. This study showed that vitamin D3 has the main role in regulating of steroidogenesis in granulosa cells of mouse polycystic ovary through activation of the AMPK signalling pathway. SIGNIFICANCE OF THE STUDY Polycystic ovarian syndrome (PCOS) is an endocrine disorder of women in reproductive age. This disorder is partly related to disruption in steroidogenesis pathway and dysregulation of estradiol and progesterone production in granulosa cells of polycystic ovaries. Previously, we have shown that vitamin D3 could modulate steroidogenesis pathway in PCOS granulosa cells. In this study, we investigate the molecular mechanism of vitamin D3 in regulation of steroidogenesis pathway. We have shown that vitamin D3 has a modulating role in steroidogenesis pathway of granulosa cells by regulation of AMP-activated protein kinase (AMPK) as an underlying molecular mechanism in mouse polycystic ovary.
Collapse
Affiliation(s)
- Shabnam Bakhshalizadeh
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Fardin Amidi
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirazi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
71
|
Chaudhari N, Dawalbhakta M, Nampoothiri L. GnRH dysregulation in polycystic ovarian syndrome (PCOS) is a manifestation of an altered neurotransmitter profile. Reprod Biol Endocrinol 2018; 16:37. [PMID: 29642911 PMCID: PMC5896071 DOI: 10.1186/s12958-018-0354-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/02/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND GnRH is the master molecule of reproduction that is influenced by several intrinsic and extrinsic factors such as neurotransmitters and neuropeptides. Any alteration in these regulatory loops may result in reproductive-endocrine dysfunction such as the polycystic ovarian syndrome (PCOS). Although low dopaminergic tone has been associated with PCOS, the role of neurotransmitters in PCOS remains unknown. The present study was therefore aimed at understanding the status of GnRH regulatory neurotransmitters to decipher the neuroendocrine pathology in PCOS. METHODS PCOS was induced in rats by oral administration of letrozole (aromatase inhibitor). Following PCOS validation, animals were assessed for gonadotropin levels and their mRNA expression. Neurotrasnmitter status was evaluated by estimating their levels, their metabolism and their receptor expression in hypothalamus, pituitary, hippocampus and frontal cortex of PCOS rat model. RESULTS We demonstrate that GnRH and LH inhibitory neurotransmitters - serotonin, dopamine, GABA and acetylcholine - are reduced while glutamate, a major stimulator of GnRH and LH release, is increased in the PCOS condition. Concomitant changes were observed for neurotransmitter metabolising enzymes and their receptors as well. CONCLUSION Our results reveal that increased GnRH and LH pulsatility in PCOS condition likely result from the cumulative effect of altered GnRH stimulatory and inhibitory neurotransmitters in hypothalamic-pituitary centre. This, we hypothesise, is responsible for the depression and anxiety-like mood disorders commonly seen in PCOS women.
Collapse
Affiliation(s)
- Nirja Chaudhari
- 0000 0001 2154 7601grid.411494.dReproductive-Neuro-Endocrinology Lab, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Mitali Dawalbhakta
- 0000 0001 2154 7601grid.411494.dReproductive-Neuro-Endocrinology Lab, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Laxmipriya Nampoothiri
- 0000 0001 2154 7601grid.411494.dReproductive-Neuro-Endocrinology Lab, Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| |
Collapse
|
72
|
Kalhori Z, Soleimani Mehranjani M, Azadbakht M, Shariaatzadeh MA. Ovary stereological features and serum biochemical factors following induction of polycystic ovary syndrome with testosterone enanthate in mice: An experimental study. Int J Reprod Biomed 2018. [DOI: 10.29252/ijrm.16.4.267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
73
|
Patel R, Shah G. High-fat diet exposure from pre-pubertal age induces polycystic ovary syndrome (PCOS) in rats. Reproduction 2018; 155:141-151. [DOI: 10.1530/rep-17-0584] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 10/30/2017] [Accepted: 11/09/2017] [Indexed: 01/17/2023]
Abstract
Polycystic ovary syndrome (PCOS) is associated with hyperandrogenism, oligo-anovulation, polycystic ovaries and metabolic syndrome. Many researchers reported that PCOS often starts with menarche in adolescents. Presently available animal model focuses on ovarian but not metabolic features of PCOS. Therefore, we hypothesized that high-fat diet feeding to pre-pubertal female rats results in both reproductive and metabolic features of PCOS. Pre-pubertal female rats were divided into two groups: group I received normal pellet diet and group II received high-fat diet (HFD). In the letrozole study, adult female rats were divided into two groups: group I received 1% carboxy methyl cellulose and group II received 1 mg/kg letrozole orally. Oral glucose tolerance test, lipid profile, fasting glucose, insulin, estrus cycle, hormonal profile, ovary weight, luteinizing hormone (LH) receptor and follicle-stimulating hormone receptor expression were measured. Polycystic ovarian morphology was assessed through histopathological changes of ovary. Feeding of HFD gradually increase glucose intolerance and fasting insulin levels. Triglyceride level was higher in HFD study while total cholesterol level was higher in the letrozole study. Alteration in testosterone and estrogen levels was observed in both studies. LH receptor expression was upregulated only in HFD study. Histopathological changes like increase cystic follicle, diminished granulosa cell layer and thickened theca cell layer were observed in letrozole as well as HFD study. High-fat diet initiated at pre-puberty age in rats produces both metabolic disturbances and ovarian changes similar to that observed clinically in PCOS patients. Letrozole on the other hand induces change in ovarian structure and function.
Collapse
Affiliation(s)
- Roshni Patel
- 1Department of Pharmacology and Pharmacy Practice, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar, Gujarat, India
| | - Gaurang Shah
- 2Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| |
Collapse
|
74
|
Richards JS, Ren YA, Candelaria N, Adams JE, Rajkovic A. Ovarian Follicular Theca Cell Recruitment, Differentiation, and Impact on Fertility: 2017 Update. Endocr Rev 2018; 39:1-20. [PMID: 29028960 PMCID: PMC5807095 DOI: 10.1210/er.2017-00164] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/12/2017] [Indexed: 12/24/2022]
Abstract
The major goal of this review is to summarize recent exciting findings that have been published within the past 10 years that, to our knowledge, have not been presented in detail in previous reviews and that may impact altered follicular development in polycystic ovarian syndrome (PCOS) and premature ovarian failure in women. Specifically, we will cover the following: (1) mouse models that have led to discovery of the derivation of two precursor populations of theca cells in the embryonic gonad; (2) the key roles of the oocyte-derived factor growth differentiation factor 9 on the hedgehog (HH) signaling pathway and theca cell functions; and (3) the impact of the HH pathway on both the specification of theca endocrine cells and theca fibroblast and smooth muscle cells in developing follicles. We will also discuss the following: (1) other signaling pathways that impact the differentiation of theca cells, not only luteinizing hormone but also insulinlike 3, bone morphogenic proteins, the circadian clock genes, androgens, and estrogens; and (2) theca-associated vascular, immune, and fibroblast cells, as well as the cytokines and matrix factors that play key roles in follicle growth. Lastly, we will integrate what is known about theca cells from mouse models, human-derived theca cell lines from patients who have PCOS and patients who do not have PCOS, and microarray analyses of human and bovine theca to understand what pathways and factors contribute to follicle growth as well as to the abnormal function of theca.
Collapse
Affiliation(s)
- JoAnne S. Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Yi A. Ren
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Nicholes Candelaria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jaye E. Adams
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology and Reproductive Medicine, Magee-Women’s Research Institute, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
75
|
Bellver J, Rodríguez-Tabernero L, Robles A, Muñoz E, Martínez F, Landeras J, García-Velasco J, Fontes J, Álvarez M, Álvarez C, Acevedo B. Polycystic ovary syndrome throughout a woman's life. J Assist Reprod Genet 2018; 35:25-39. [PMID: 28951977 PMCID: PMC5758469 DOI: 10.1007/s10815-017-1047-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/12/2017] [Indexed: 01/22/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-aged women and the main cause of infertility due to anovulation. However, this syndrome spans the lives of women affecting them from in-utero life until death, leading to several health risks that can impair quality of life and increase morbidity and mortality rates. Fetal programming may represent the beginning of the condition characterized by hyperandrogenism and insulin resistance which leads to a series of medical consequences in adolescence, adulthood, and old age. Menstrual and fertility problems evolve into metabolic complications as age advances. An early and precise diagnosis is important for an adequate management of PCOS, especially at the extreme ends of the reproductive lifespan. However, many different phenotypes are included under the same condition, being important to look at these different phenotypes separately, as they may require different treatments and have different consequences. In this way, PCOS exhibits a great metabolic complexity and its diagnosis needs to be revised once again and adapted to recent data obtained by new technologies. According to the current medical literature, lifestyle therapy constitutes the first step in the management, especially when excess body weight is associated. Pharmacotherapy is frequently used to treat the most predominant manifestations in each age group, such as irregular menses and hirsutism in adolescence, fertility problems in adulthood, and metabolic problems and risk of cancer in old age. Close surveillance is mandatory in each stage of life to avoid health risks which may also affect the offspring, since fetal and post-natal complications seem to be increased in PCOS women.
Collapse
Affiliation(s)
- José Bellver
- IVI-Valencia, University of Valencia, Valencia, Spain.
| | | | | | | | | | | | | | - Juan Fontes
- Hospital Virgen de las Nieves, Granada, Spain
| | | | | | | |
Collapse
|
76
|
Reckelhoff JF. Sex Differences in Regulation of Blood Pressure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1065:139-151. [PMID: 30051382 DOI: 10.1007/978-3-319-77932-4_9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hypertension is one of the leading risk factors for cardiovascular disease, myocardial infarction, and stroke. There are gender differences in the prevalence of hypertension and in the mechanisms responsible for hypertension in humans. This review will discuss the mechanisms for regulation of blood pressure, sex differences that have been identified in animal studies, and the gender differences that have been identified in humans.
Collapse
Affiliation(s)
- Jane F Reckelhoff
- Department of Cell and Molecular Biology and Women's Health Research Center and The Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
77
|
Hormone-induced rat model of polycystic ovary syndrome: A systematic review. Life Sci 2017; 191:259-272. [DOI: 10.1016/j.lfs.2017.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/04/2017] [Accepted: 10/17/2017] [Indexed: 12/16/2022]
|
78
|
Yan YL, Desvignes T, Bremiller R, Wilson C, Dillon D, High S, Draper B, Buck CL, Postlethwait J. Gonadal soma controls ovarian follicle proliferation through Gsdf in zebrafish. Dev Dyn 2017; 246:925-945. [PMID: 28856758 PMCID: PMC5761338 DOI: 10.1002/dvdy.24579] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/20/2017] [Accepted: 08/01/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Aberrant signaling between germ cells and somatic cells can lead to reproductive disease and depends on diffusible signals, including transforming growth factor-beta (TGFB) -family proteins. The TGFB-family protein Gsdf (gonadal soma derived factor) controls sex determination in some fish and is a candidate for mediating germ cell/soma signaling. RESULTS Zebrafish expressed gsdf in somatic cells of bipotential gonads and expression continued in ovarian granulosa cells and testicular Sertoli cells. Homozygous gsdf knockout mutants delayed leaving the bipotential gonad state, but then became a male or a female. Mutant females ovulated a few oocytes, then became sterile, accumulating immature follicles. Female mutants stored excess lipid and down-regulated aromatase, gata4, insulin receptor, estrogen receptor, and genes for lipid metabolism, vitellogenin, and steroid biosynthesis. Mutant females contained less estrogen and more androgen than wild-types. Mutant males were fertile. Genomic analysis suggests that Gsdf, Bmp15, and Gdf9, originated as paralogs in vertebrate genome duplication events. CONCLUSIONS In zebrafish, gsdf regulates ovarian follicle maturation and expression of genes for steroid biosynthesis, obesity, diabetes, and female fertility, leading to ovarian and extra-ovarian phenotypes that mimic human polycystic ovarian syndrome (PCOS), suggesting a role for a related TGFB signaling molecule in the etiology of PCOS. Developmental Dynamics 246:925-945, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yi-Lin Yan
- Institute of Neuroscience, University of Oregon, Eugene, Oregon
| | | | - Ruth Bremiller
- Institute of Neuroscience, University of Oregon, Eugene, Oregon
| | | | - Danielle Dillon
- Center for Bioengineering Innovation, Northern Arizona University, Flagstaff, Arizona
| | - Samantha High
- Institute of Neuroscience, University of Oregon, Eugene, Oregon
| | - Bruce Draper
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California
| | - Charles Loren Buck
- Center for Bioengineering Innovation, Northern Arizona University, Flagstaff, Arizona
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona
| | | |
Collapse
|
79
|
Unfer V, Facchinetti F, Orrù B, Giordani B, Nestler J. Myo-inositol effects in women with PCOS: a meta-analysis of randomized controlled trials. Endocr Connect 2017; 6:647-658. [PMID: 29042448 PMCID: PMC5655679 DOI: 10.1530/ec-17-0243] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Myo-inositol (MI) supplementation in women with polycystic ovary syndrome (PCOS) has been evaluated over the last years. Many hormonal and reproductive impairments associated with this disorder seem relieved by the supplement. The objective of the meta-analysis was to assess the effects of MI alone or combined with d-chiro-inositol (DCI) on the endocrine and metabolic abnormalities of women with PCOS. Literature was retrieved from selected databases, MEDLINE, EMBASE, PubMed and Research Gate (up to November 2016). Only randomized controlled trials (RCTs) investigating the effects of MI alone or combined with DCI were reviewed. Nine RCTs involving 247 cases and 249 controls were included. Significant decreases in fasting insulin (SMD = -1.021 µU/mL, 95% CI: -1.791 to -0.251, P = 0.009) and homeostasis model assessment (HOMA) index (SMD = -0.585, 95% CI: -1.145 to -0.025, P = 0.041) were identified after MI supplementation. The trial sequential analysis of insulin meta-analysis illustrates that the cumulative z-curve crossed the monitoring boundary, providing firm evidence of the intervention effect. A slight trend toward a reduction of testosterone concentration by MI with respect to controls was found (SMD = -0.49, 95% CI: -1.072 to 0.092, P = 0.099), whereas androstenedione levels remained unaffected. Throughout a subgroup's meta-analysis, a significant increase in serum SHBG was observed only in those studies where MI was administered for at least 24 weeks (SMD = 0.425 nmol/L, 95% CI: 0.050-0.801, P = 0.026). These results highlight the beneficial effect of MI in improving the metabolic profile of women with PCOS, concomitantly reducing their hyperandrogenism.
Collapse
Affiliation(s)
- Vittorio Unfer
- Health DepartmentUniPoliSi - Institut des Etudes Universitaires, Disentis, Switzerland
| | - Fabio Facchinetti
- Mother-Infant DepartmentUniversity of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Orrù
- Medical Affairs DepartmentLo.Li. Pharma, Rome, Italy
| | | | - John Nestler
- Departments of Medicine and Obstetrics and GynecologyVirginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
80
|
Delitala AP, Capobianco G, Delitala G, Cherchi PL, Dessole S. Polycystic ovary syndrome, adipose tissue and metabolic syndrome. Arch Gynecol Obstet 2017. [DOI: 10.1007/s00404-017-4429-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
81
|
Sagvekar P, Mangoli V, Desai S, Patil A, Mukherjee S. LINE1 CpG-DNA Hypomethylation in Granulosa Cells and Blood Leukocytes Is Associated With PCOS and Related Traits. J Clin Endocrinol Metab 2017; 102:1396-1405. [PMID: 28324041 DOI: 10.1210/jc.2016-2645] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
CONTEXT Altered global DNA methylation is indicative of epigenomic instability concerning chronic diseases. Investigating its incidence and association with polycystic ovary syndrome (PCOS) is essential to understand the etiopathogenesis of this disorder. OBJECTIVES We assessed global DNA methylation differences in peripheral blood leukocytes (PBLs) and cumulus granulosa cells (CGCs) of controls and women with PCOS; and their association with PCOS and its traits. DESIGN, SETTING, PARTICIPANTS, MAIN OUTCOME MEASURE This study included a total of 102 controls and women with PCOS. Forty-one women undergoing controlled ovarian hyperstimulation (COH) and 61 women not undergoing COH were recruited from in vitro fertilization (IVF) and infertility clinics. DNA methylation was measured by ELISA for 5'-methyl-cytosine content and bisulfite sequencing of 5'-untranslated region (5'-UTR) of long interspersed nucleotide element-1 (LINE1/L1). RESULTS Total 5'-methyl-cytosine and L1 methylation levels in PBLs and CGCs were similar between controls and women with PCOS. Methylation assessed at CpG sites of L1 5'-UTR revealed a single CpG-site (CpG-4) to be consistently hypomethylated in PBLs of both PCOS groups and CGCs of stimulated PCOS group. In unstimulated women, hypomethylation at CpG-4 was strongly associated with PCOS susceptibility, whereas in stimulated group it showed strong associations with PCOS and its hormonal traits. Furthermore, CGCs demonstrated consistent global and CpG-DNA hypomethylation relative to PBLs, irrespective of normal or disease states. CONCLUSION Our study revealed strong association of single hypomethylated CpG-site with PCOS. Identification and characterization of more such methyl-CpG signatures in repetitive elements in larger study populations would provide valuable epigenetic insights into PCOS.
Collapse
Affiliation(s)
| | - Vijay Mangoli
- Fertility Clinic and IVF Center, Gamdevi, Mumbai 400007, Maharashtra, India
| | - Sadhana Desai
- Fertility Clinic and IVF Center, Gamdevi, Mumbai 400007, Maharashtra, India
| | - Anushree Patil
- Department of Infertility and Endocrinology, National Institute for Research in Reproductive Health, Parel, Mumbai 400012, India
| | | |
Collapse
|
82
|
Bakhshalizadeh S, Amidi F, Alleyassin A, Soleimani M, Shirazi R, Shabani Nashtaei M. Modulation of steroidogenesis by vitamin D3 in granulosa cells of the mouse model of polycystic ovarian syndrome. Syst Biol Reprod Med 2017; 63:150-161. [DOI: 10.1080/19396368.2017.1296046] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Shabnam Bakhshalizadeh
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Alleyassin
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Shirazi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
83
|
Fruzzetti F, Perini D, Russo M, Bucci F, Gadducci A. Comparison of two insulin sensitizers, metformin and myo-inositol, in women with polycystic ovary syndrome (PCOS). Gynecol Endocrinol 2017; 33:39-42. [PMID: 27808588 DOI: 10.1080/09513590.2016.1236078] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Insulin resistance (IR) plays a pivotal role in PCOS. Insulin-sensitizer agents such as metformin and inositols have been shown to improve the endocrine and metabolic aspects of PCOS. The purpose of this study is to compare their effects on the clinical and metabolic features of the women with PCOS. Fifty PCOS women with IR and/or hyperinsulinemia were randomized to treatment with metformin (1500 mg/day) or myo-inositol (4 g/day). IR was defined as HOMA-IR >2.5, while hyperinsulinemia was defined as a value of AUC for insulin after a glucose load over the cutoff of our laboratory obtained in normal women. The Matsusa Index has been calculated. The women have been evaluated for insulin secretion, BMI, menstrual cycle length, acne and hirsutism, at baseline and after 6 months of therapy. The results obtained in both groups were similar. The insulin sensitivity improved in both treatment groups. The BMI significantly decreased and the menstrual cycle was normalized in about 50% of the women. No significant changes in acne and hirsutism were observed. The two insulin-sensitizers, metformin and myo-inositol, show to be useful in PCOS women in lowering BMI and ameliorating insulin sensitivity, and improving menstrual cycle without significant differences between the two treatments.
Collapse
Affiliation(s)
- Franca Fruzzetti
- a Department of Obstetrics and Gynecology , University of Pisa , Pisa , Italy
| | - Daria Perini
- a Department of Obstetrics and Gynecology , University of Pisa , Pisa , Italy
| | - Marinella Russo
- a Department of Obstetrics and Gynecology , University of Pisa , Pisa , Italy
| | - Fiorella Bucci
- a Department of Obstetrics and Gynecology , University of Pisa , Pisa , Italy
| | - Angiolo Gadducci
- a Department of Obstetrics and Gynecology , University of Pisa , Pisa , Italy
| |
Collapse
|
84
|
Geronikolou SA, Bacopoulou F, Cokkinos D. Bioimpedance Measurements in Adolescents with Polycystic Ovary Syndrome: A Pilot Study. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 987:291-299. [PMID: 28971467 DOI: 10.1007/978-3-319-57379-3_26] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Limited data are available on the body composition of adolescent women with polycystic ovary syndrome (PCOS). The aim of this study was to examine differences in body composition indices of metabolism, homeostasis and inflammation, between Greek adolescent females suffering from PCOS and age- and body mass index (BMI)-matched non-PCOS controls. Thirteen PCOS patients and nine non-PCOS controls, aged 13-24 years participated in this cross-sectional study. Study participants underwent assessment by a novel dual frequency bioimpedance device (BIA-ACC). The following body composition indices were measured in each adolescent: extra cellular water (ECW) as inflammation marker, total body water (TBW) as homeostasis marker, extracellular mass to body cell mass ratio (ECM/BCM), fat mass (FM), fat-free mass (FFM) and intracellular water (ICW) as markers of body mass composition and metabolism. Non-linear analysis showed no statistically significant differences in the body composition characteristics between PCOS patients and controls. Further studies with larger sample sizes are needed to confirm whether adolescents with PCOS actually have similar body composition profile with their non-PCOS peers.
Collapse
Affiliation(s)
- Styliani A Geronikolou
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation of Academy of Athens, 4 Soranou Ephessiou Street, 11527, Athens, Greece.
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, 3 Thivon Street, 11527, Athens, Greece
| | - Dennis Cokkinos
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation of Academy of Athens, 4 Soranou Ephessiou Street, 11527, Athens, Greece
| |
Collapse
|
85
|
Sun Y, Yuan Y, Yang H, Li J, Feng T, Ouyang Y, Jin T, Liu M. Association between Common Genetic Variants and Polycystic Ovary Syndrome Risk in a Chinese Han Population. J Clin Res Pediatr Endocrinol 2016; 8:405-410. [PMID: 27217259 PMCID: PMC5197998 DOI: 10.4274/jcrpe.2784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is one of the most common endocrinopathies affecting 5-7% of reproductive age women worldwide. The aim of our study was to explore the PCOS-related single nucleotide polymorphism (SNP) associations between common genetic variants and PCOS risk in a Han Chinese women population. METHODS In this case-control study, 285 Chinese Han women aged 28.50±6.858 years with PCOS and 299 controls of a mean age of 32.66±7.018 years were compared. We selected recently published genome-wide association studies (GWAS) which identified several genetic loci in PCOS. All the SNPs were genotyped by Sequenom Mass-ARRAY technology. Associations between the gene and the risk of PCOS were tested using various genetic models by Statistical Package for the Social Sciences and Plink. RESULTS We found that rs705702 in the RAB5B/SUOX was associated with PCOS (odds ratio=1.42; 95% confidence interval=1.08-1.87, p=0.011) and increased the PCOS risk. The genotypic model analysis also showed that rs705702 was associated with PCOS risk. CONCLUSION Our results suggest that SNPs rs705702 in gene RAB5B/SUOX was associated with PCOS in Han Chinese women.
Collapse
Affiliation(s)
- Ying Sun
- Xi’an Jiaotong University School of Medicine, Department of Pathology, Xi’an, China
| | - Yi Yuan
- Han Zhong Central Hospital, Clinic of Obstetrics and Gynecology, Han Zhong, China
| | - Hua Yang
- Northwest University School of Life Sciences, Xi’an, China
| | - Jingjie Li
- Northwest University School of Life Sciences, Xi’an, China
| | - Tian Feng
- Northwest University School of Life Sciences, Xi’an, China
| | - Yongri Ouyang
- Northwest University School of Life Sciences, Xi’an, China
| | - Tianbo Jin
- Northwest University School of Life Sciences, Xi'an, China, Phone: +86-29-88302831 E-mail:
| | - Ming Liu
- Xi’an Jiatong University Second Affiliated Hospital, Department of Obstetrics and Gynecology, Xi’an, China
| |
Collapse
|
86
|
Ożegowska K, Bogacz A, Bartkowiak-Wieczorek J, Seremak-Mrozikiewicz A, Pawelczyk L. Association between the angiotensin converting enzyme gene insertion/deletion polymorphism and metabolic disturbances in women with polycystic ovary syndrome. Mol Med Rep 2016; 14:5401-5407. [DOI: 10.3892/mmr.2016.5910] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/29/2016] [Indexed: 11/06/2022] Open
|
87
|
Garg D, Merhi Z. Relationship between Advanced Glycation End Products and Steroidogenesis in PCOS. Reprod Biol Endocrinol 2016; 14:71. [PMID: 27769286 PMCID: PMC5073880 DOI: 10.1186/s12958-016-0205-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/14/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Women with PCOS have elevated levels of the harmful Advanced Glycation End Products (AGEs), which are highly reactive molecules formed after glycation of lipids and proteins. Additionally, AGEs accumulate in the ovaries of women with PCOS potentially contributing to the well-documented abnormal steroidogenesis and folliculogenesis. MAIN BODY A systematic review of articles and abstracts available in PubMed was conducted and presented in a systemic manner. This article reports changes in steroidogenic enzyme activity in granulosa and theca cells in PCOS and PCOS-models. It also described the changes in AGEs and their receptors in the ovaries of women with PCOS and presents the underlying mechanism(s) whereby AGEs could be responsible for the PCOS-related changes in granulosa and theca cell function thus adversely impacting steroidogenesis and follicular development. AGEs are associated with hyperandrogenism in PCOS possibly by altering the activity of various enzymes such as cholesterol side-chain cleavage enzyme cytochrome P450, steroidogenic acute regulatory protein, 17α-hydroxylase, and 3β-hydroxysteroid dehydrogenase. AGEs also affect luteinizing hormone receptor and anti-Mullerian hormone receptor expression as well as their signaling pathways in granulosa cells. CONCLUSIONS A better understanding of how AGEs alter granulosa and theca cell function is likely to contribute meaningfully to a conceptual framework whereby new interventions to prevent and/or treat ovarian dysfunction in PCOS can ultimately be developed.
Collapse
Affiliation(s)
- Deepika Garg
- Department of Obstetrics and Gynecology, Maimonides Medical Center, Brooklyn, NY 11219 USA
| | - Zaher Merhi
- Division of Reproductive Biology, Department of Obstetrics and Gynecology, NYU School of Medicine, 180 Varick Street, sixth floor, New York City, NY 11014 USA
| |
Collapse
|
88
|
Diane A, Pierce WD, Kelly SE, Sokolik S, Borthwick F, Jacome-Sosa M, Mangat R, Pradillo JM, Allan SM, Ruth MR, Field CJ, Hutcheson R, Rocic P, Russell JC, Vine DF, Proctor SD. Mechanisms of Comorbidities Associated With the Metabolic Syndrome: Insights from the JCR:LA-cp Corpulent Rat Strain. Front Nutr 2016; 3:44. [PMID: 27777929 PMCID: PMC5056323 DOI: 10.3389/fnut.2016.00044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 01/08/2023] Open
Abstract
Obesity and its metabolic complications have emerged as the epidemic of the new millennia. The use of obese rodent models continues to be a productive component of efforts to understand the concomitant metabolic complications of this disease. In 1978, the JCR:LA-cp rat model was developed with an autosomal recessive corpulent (cp) trait resulting from a premature stop codon in the extracellular domain of the leptin receptor. Rats that are heterozygous for the cp trait are lean-prone, while those that are homozygous (cp/cp) spontaneously display the pathophysiology of obesity as well as a metabolic syndrome (MetS)-like phenotype. Over the years, there have been formidable scientific contributions that have originated from this rat model, much of which has been reviewed extensively up to 2008. The premise of these earlier studies focused on characterizing the pathophysiology of MetS-like phenotype that was spontaneously apparent in this model. The purpose of this review is to highlight areas of recent advancement made possible by this model including; emerging appreciation of the "thrifty gene" hypothesis in the context of obesity, the concept of how chronic inflammation may drive obesogenesis, the impact of acute forms of inflammation to the brain and periphery during chronic obesity, the role of dysfunctional insulin metabolism on lipid metabolism and vascular damage, and the mechanistic basis for altered vascular function as well as novel parallels between the human condition and the female JCR:LA-cp rat as a model for polycystic ovary disease (PCOS).
Collapse
Affiliation(s)
- Abdoulaye Diane
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - W. David Pierce
- Department of Sociology, University of Alberta, Edmonton, AB, Canada
| | - Sandra E. Kelly
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Sharon Sokolik
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Faye Borthwick
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Miriam Jacome-Sosa
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | | | - Stuart McRae Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Megan R. Ruth
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Catherine J. Field
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | | | | | - James C. Russell
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Donna F. Vine
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
89
|
Rutkowska AZ, Diamanti-Kandarakis E. Polycystic ovary syndrome and environmental toxins. Fertil Steril 2016; 106:948-58. [PMID: 27559705 DOI: 10.1016/j.fertnstert.2016.08.031] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 08/15/2016] [Accepted: 08/15/2016] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common, heterogeneous, and multifactorial endocrine disorder in premenopausal women. The pathophysiology of this endocrinopathy is still unclear; however, the heterogeneity of its features within ethnic races, geographic location, and families suggests that environment and lifestyle are of prime importance. This work is mainly focused on the possible role of the most common and studied environmental toxins for this syndrome in the pathogenesis of PCOS. Plasticizers, such as bisphenol A (BPA) or phthalates, which belong to the categories of endocrine disrupting chemicals (EDCs) and advanced glycation end products (AGEs), affect humans' health in everyday, industrialized life; therefore special attention should be paid to such exposure. Timing of exposure to EDCs is crucial for the intensity of adverse health effects. It is now evident that fetuses, infants, and/or young children are the most susceptible groups, especially in the early development periods. Prenatal exposure to EDCs that mimic endogenous hormones may contribute to the altered fetal programming and in consequence lead to PCOS and other adverse health effects, potentially transgenerationally. Acute or prolonged exposure to EDCs and AGEs through different life cycle stages may result in destabilization of the hormonal homeostasis and lead to disruption of reproductive functions. They may also interfere with metabolic alterations such as obesity, insulin resistance, and compensatory hyperinsulinemia that can exacerbate the PCOS phenotype and contribute to PCOS consequences such as type 2 diabetes and cardiovascular disease. Since wide exposure to environmental toxins and their role in the pathophysiology of PCOS are supported by extensive data derived from diverse scientific models, protective strategies and strong recommendations should be considered to reduce human exposure to protect present and future generations from their adverse health effects.
Collapse
Affiliation(s)
| | - Evanthia Diamanti-Kandarakis
- Department of Endocrinology and Diabetes Center of Excellence, Medical School University of Athens, EUROCLINIC, Athens, Greece.
| |
Collapse
|
90
|
De Leo V, Musacchio MC, Cappelli V, Massaro MG, Morgante G, Petraglia F. Genetic, hormonal and metabolic aspects of PCOS: an update. Reprod Biol Endocrinol 2016; 14:38. [PMID: 27423183 PMCID: PMC4947298 DOI: 10.1186/s12958-016-0173-x] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/08/2016] [Indexed: 12/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrine disorder affecting 5-10 % of women of reproductive age. It generally manifests with oligo/anovulatory cycles, hirsutism and polycystic ovaries, together with a considerable prevalence of insulin resistance. Although the aetiology of the syndrome is not completely understood yet, PCOS is considered a multifactorial disorder with various genetic, endocrine and environmental abnormalities. Moreover, PCOS patients have a higher risk of metabolic and cardiovascular diseases and their related morbidity, if compared to the general population.
Collapse
Affiliation(s)
- V. De Leo
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| | - M. C. Musacchio
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| | - V. Cappelli
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| | - M. G. Massaro
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| | - G. Morgante
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| | - F. Petraglia
- Department Molecular Medicine and Development, University of Siena, Policlinico Le Scotte, Viale Bracci, 53100 Siena, Italy
| |
Collapse
|
91
|
Dalmasso C, Maranon R, Patil C, Bui E, Moulana M, Zhang H, Smith A, Yanes Cardozo LL, Reckelhoff JF. Cardiometabolic Effects of Chronic Hyperandrogenemia in a New Model of Postmenopausal Polycystic Ovary Syndrome. Endocrinology 2016; 157:2920-7. [PMID: 27145003 PMCID: PMC4929551 DOI: 10.1210/en.2015-1617] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Postmenopausal women who have had polycystic ovary syndrome (PCOS) and chronic hyperandrogenemia may be at a greater risk for cardiovascular disease than normoandrogenemic postmenopausal women. The cardiometabolic effect of chronic hyperandrogenemia in women with PCOS after menopause is unclear. The present study was performed to test the hypothesis that chronic hyperandrogenemia in aging female rats would have more deleterious effects on metabolic function, blood pressure, and renal function than in normoandrogenemic age-matched females. Female Sprague Dawley were implanted continuously, beginning at 4-5 weeks, with dihydrotestosterone (postmenopausal hyperandrogenemic female [PMHAF]) or placebo pellets (controls), and were studied at 13 months of age. Plasma DHT was 3-fold higher, and estradiol was 90% lower in PMHAF than controls. Body weights were higher; EchoMRI showed greater fat and lean mass; and computed tomography showed more sc and visceral adiposity in PMHAF, but with similar femur length compared with controls. Insulin resistance was present in PMHAF with higher plasma insulin, normal fasting blood glucose, abnormal oral glucose tolerance test, and higher nonfasting blood glucose. Blood pressure (radiotelemetry) was significantly higher and heart rate was lower, and renal function (glomerular filtration rate) was reduced by 40% in PMHAF. Thus the aging chronically hyperandrogenemic female rat is a new model of postmenopausal PCOS, which exhibits insulin resistance and visceral obesity, hypertension, and impairment in renal function. This new model provides a unique tool to study the deleterious effects of chronic androgen excess in postmenopausal females rats.
Collapse
Affiliation(s)
- Carolina Dalmasso
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Rodrigo Maranon
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Chetan Patil
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Elizabeth Bui
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Mohadetheh Moulana
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Howei Zhang
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Andrew Smith
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Licy L Yanes Cardozo
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Jane F Reckelhoff
- Departments of Physiology and Biophysics (C.D., R.M., C.P., L.L.Y.C., J.F.R.), Medicine (E.B., R.M., L.L.Y.C.), Psychiatry (M.M.), Radiology (H.Z., A.S.), Endocrinology (L.L.Y.C.), and Women's Health Research Center (C.D., R.M., C.P., L.L.Y.C., J.F.R.), University of Mississippi Medical Center, Jackson, Mississippi 39216
| |
Collapse
|
92
|
Comim FV, Marchesan LQ, Copes RM, de Vieira AR, Moresco RN, Compston JE, Premaor MO. Increased risk of humerus and lower leg fractures in postmenopausal women with self-reported premenopausal hirsutism and/or oligomenorrhea. Eur J Obstet Gynecol Reprod Biol 2016; 203:162-6. [PMID: 27318183 DOI: 10.1016/j.ejogrb.2016.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 05/05/2016] [Accepted: 05/13/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the prevalence of fracture in women aged >55 years with self-reported premenopausal hirsutism and/or oligomenorrhea. STUDY DESIGN A cross-sectional study including 1057 post-menopausal women aged >55 years who were treated in a primary care facility in Santa Maria, South Brazil, from March 2013 through August 2013. Data were collected using a standardized questionnaire for characteristics including fracture history, medication use, and reproductive history (oligomenorrhea, hirsutism, miscarriage, the diagnosis or treatment of hypothyroidism, hyperprolactinemia, or infertility). RESULTS A non-significant trend towards an increased prevalence of all fragility fractures was observed in women with premenopausal hirsutism and/or oligomenorrhea when compared to those without (20.4% vs. 15.7%). After correction for age, falls, and comorbidities, fracture prevalence was significantly higher in the lower leg (OR 3.1 [CI 1.1-8.6]; P=0.029), and humerus (OR 2.6 [CI 1.2-5.4]; P=0.015) in the women with hirsutism and/or oligomenorrhea. CONCLUSION This is a hypothesis-generating study which evaluated the association between hirsutism and/or oligomenorrhea and fractures in postmenopausal women. Our results suggest that premenopausal hirsutism and/or oligomenorrhea may be associated with an increased risk of fracture postmenopause, particularly in the humerus and lower leg.
Collapse
Affiliation(s)
- Fabio V Comim
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil.
| | - Luana Q Marchesan
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rafaela M Copes
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Adhan R de Vieira
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Rafael N Moresco
- Pharmaceutical Science, Postgraduate Course, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Melissa O Premaor
- Department of Clinical Medicine, Health Science Center, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
93
|
Dalmasso C, Maranon R, Patil C, Moulana M, Romero DG, Reckelhoff JF. 20-HETE and CYP4A2 ω-hydroxylase contribute to the elevated blood pressure in hyperandrogenemic female rats. Am J Physiol Renal Physiol 2016; 311:F71-7. [PMID: 27194719 DOI: 10.1152/ajprenal.00458.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/17/2016] [Indexed: 01/08/2023] Open
Abstract
In male rats, androgen supplements increase 20-hydroxyeicosatetraenoic acid (20-HETE) via cytochrome P-450 (CYP)4A ω-hydroxylase and cause an increase in blood pressure (BP). In the present study, we determined the roles of 20-HETE and CYP4A2 on the elevated BP in hyperandrogenemic female rats. Chronic dihydrotestosterone (DHT) increased mean arterial pressure (MAP) in female Sprague-Dawley rats (96 ± 2 vs. 108 ± 2 mmHg, P < 0.05) and was associated with increased renal microvascular CYP4A2 mRNA expression (15-fold), endogenous renal 20-HETE (5-fold), and ω-hydroxylase activity (3-fold). Chronic DHT also increased MAP in low salt-fed Dahl salt-resistant female rats (81 ± 4 vs. 95 ± 1 mmHg, P < 0.05) but had no effect on MAP in Dahl salt-sensitive female rats (154 ± 3 vs. 153 ± 3 mmHg), which are known to be 20-HETE deficient. To test the role of CYP4A2, female CYP4A2(-/-) and SS.5(Bn) (wild type) rats were treated with DHT. DHT increased MAP in SS.5(Bn) female rats (104 ± 1 vs. 128 ± 1 mmHg, P < 0.05) but had no effect in CYP4A2(-/-) female rats (118 ± 1 vs. 120 ± 1 mmHg). Renal microvascular 20-HETE was reduced in control CYP4A2(-/-) female rats and was increased with DHT in SS.5(Bn) female rats (6-fold) but not CYP4A2(-/-) female rats. ω-Hydroxylase activity was 40% lower in control CYP4A2(-/-) female rats than in SS.5(Bn) female rats, and DHT decreased ω-hydroxylase activity in SS.5(Bn) female rats (by 50%) but significantly increased ω-hydroxylase activity in CYP4A2(-/-) female rats (3-fold). These data suggest that 20-HETE via CYP4A2 contributes to the elevation in BP in hyperandrogenemic female rats. The data also suggest that 20-HETE synthesis inhibition may be effective in treating the elevated BP in women with hyperandrogenemia, such as women with polycystic ovary syndrome.
Collapse
Affiliation(s)
- Carolina Dalmasso
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rodrigo Maranon
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Department of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Chetan Patil
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mohadetheh Moulana
- Department of Psychiatry, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Damian G Romero
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi; and Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jane F Reckelhoff
- Department of Physiology, University of Mississippi Medical Center, Jackson, Mississippi; Women's Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
94
|
Comim FV, Gutierrez K, Bridi A, Bochi G, Chemeris R, Rigo ML, Dau AMP, Cezar AS, Moresco RN, Gonçalves PBD. Effects of Adiponectin Including Reduction of Androstenedione Secretion and Ovarian Oxidative Stress Parameters In Vivo. PLoS One 2016; 11:e0154453. [PMID: 27158926 PMCID: PMC4861339 DOI: 10.1371/journal.pone.0154453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 04/13/2016] [Indexed: 12/19/2022] Open
Abstract
Adiponectin is the most abundantly produced human adipokine with anti-inflammatory, anti-oxidative, and insulin-sensitizing properties. Evidence from in vitro studies has indicated that adiponectin has a potential role in reproduction because it reduces the production of androstenedione in bovine theca cells in vitro. However, this effect on androgen production has not yet been observed in vivo. The current study evaluated the effect of adiponectin on androstenedione secretion and oxidative stress parameters in a rodent model. Seven-week-old female Balb/c mice (n = 33), previously treated with equine gonadotropin chorionic, were assigned to one of four different treatments: Group 1, control (phosphate-buffered saline); Group 2, adiponectin 0.1 μg/mL; Group 3, adiponectin 1.0 μg/mL; Group 4, adiponectin 5.0 μg/mL. After 24 h, all animals were euthanized and androstenedione levels were measured in the serum while oxidative stress markers were quantified in whole ovary tissue. Female mice treated with adiponectin exhibited a significant reduction (about 60%) in serum androstenedione levels in comparison to controls. Androstenedione levels decreased from 0.78 ± 0.4 ng/mL (mean ± SD) in controls to 0.28 ± 0.06 ng/mL after adiponectin (5 μg/mL) treatment (P = 0.01). This change in androgen secretion after 24 hours of treatment was associated with a significant reduction in the expression of CYP11A1 and STAR (but not CYP17A1). In addition, ovarian AOPP product levels, a direct product of protein oxidation, decreased significantly in adiponectin-treated mice (5 μg/mL); AOPP (mean ± SD) decreased to 4.3 ± 2.1 μmol/L in comparison with that of the controls (11.5 ± 1.7 μmol/L; P = 0.0003). Our results demonstrated for the first time that acute treatment with adiponectin reduced the levels of a direct oxidative stress marker in the ovary as well as decreased androstenedione serum levels in vivo after 24 h.
Collapse
Affiliation(s)
- Fabio V. Comim
- Department of Clinical Medicine, Federal University of Santa Maria (UFSM), Santa Maria, Brazil
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
- * E-mail:
| | - Karina Gutierrez
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Alessandra Bridi
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Guilherme Bochi
- Laboratory of Clinical Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Raisa Chemeris
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Melânia L. Rigo
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Andressa Minussi P. Dau
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Alfredo S. Cezar
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Rafael Noal Moresco
- Laboratory of Clinical Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Federal University of Santa Maria (UFSM), Santa Maria, Brazil
| |
Collapse
|
95
|
Association between Polycystic Ovary Syndrome and Gut Microbiota. PLoS One 2016; 11:e0153196. [PMID: 27093642 PMCID: PMC4836746 DOI: 10.1371/journal.pone.0153196] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 03/24/2016] [Indexed: 11/19/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most frequent endocrinopathy in women of reproductive age. It is difficult to treat PCOS because of its complex etiology and pathogenesis. Here, we characterized the roles of gut microbiota on the pathogenesis and treatments in letrozole (a nonsteroidal aromatase inhibitor) induced PCOS rat model. Changes in estrous cycles, hormonal levels, ovarian morphology and gut microbiota by PCR-DGGE and real-time PCR were determined. The results showed that PCOS rats displayed abnormal estrous cycles with increasing androgen biosynthesis and exhibited multiple large cysts with diminished granulosa layers in ovarian tissues. Meanwhile, the composition of gut microbiota in letrozole-treated rats was different from that in the controls. Lactobacillus, Ruminococcus and Clostridium were lower while Prevotella was higher in PCOS rats when compared with control rats. After treating PCOS rats with Lactobacillus and fecal microbiota transplantation (FMT) from healthy rats, it was found that the estrous cycles were improved in all 8 rats in FMT group, and in 6 of the 8 rats in Lactobacillus transplantation group with decreasing androgen biosynthesis. Their ovarian morphologies normalized. The composition of gut microbiota restored in both FMT and Lactobacillus treated groups with increasing of Lactobacillus and Clostridium, and decreasing of Prevotella. These results indicated that dysbiosis of gut microbiota was associated with the pathogenesis of PCOS. Microbiota interventions through FMT and Lactobacillus transplantation were beneficial for the treatments of PCOS rats.
Collapse
|
96
|
Does myo-inositol effect on PCOS follicles involve cytoskeleton regulation? Med Hypotheses 2016; 91:1-5. [PMID: 27142131 DOI: 10.1016/j.mehy.2016.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/25/2016] [Indexed: 11/24/2022]
Abstract
Inositol metabolism is severely impaired in follicles obtained from cystic ovaries, leading to deregulated insulin transduction and steroid synthesis. On the contrary, inositol administration to women suffering from polycystic ovary syndrome (PCOS) has been proven to efficiently counteract most of the clinical hallmarks displayed by PCOS patients, including insulin resistance, hyperandrogenism and oligo-amenorrhea. We have recently observed that myo-inositol induces significant changes in cytoskeletal architecture of breast cancer cells, by modulating different biochemical pathways, eventually modulating the epithelial-mesenchymal transition. We hypothesize that inositol and its monophosphate derivatives, besides their effects on insulin transduction, may efficiently revert histological and functional features of cystic ovary by inducing cytoskeleton rearrangements. We propose an experimental model that could address not only whether inositol modulates cytoskeleton dynamics in both normal and cystic ovary cells, but also whether this effect may interfere with ovarian steroidogenesis. A more compelling understanding of the mechanisms of action of inositol (and its derivatives) would greatly improve its therapeutic utilization, by conferring to current treatments a well-grounded scientific rationale.
Collapse
|
97
|
Rahaman SNA, Mat Yusop J, Mohamed-Hussein ZA, Ho KL, Teh AH, Waterman J, Ng CL. Cloning, expression, purification, crystallization and X-ray crystallographic analysis of recombinant human C1ORF123 protein. Acta Crystallogr F Struct Biol Commun 2016; 72:207-13. [PMID: 26919524 PMCID: PMC4774879 DOI: 10.1107/s2053230x16002016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/02/2016] [Indexed: 01/17/2023] Open
Abstract
C1ORF123 is a human hypothetical protein found in open reading frame 123 of chromosome 1. The protein belongs to the DUF866 protein family comprising eukaryote-conserved proteins with unknown function. Recent proteomic and bioinformatic analyses identified the presence of C1ORF123 in brain, frontal cortex and synapses, as well as its involvement in endocrine function and polycystic ovary syndrome (PCOS), indicating the importance of its biological role. In order to provide a better understanding of the biological function of the human C1ORF123 protein, the characterization and analysis of recombinant C1ORF123 (rC1ORF123), including overexpression and purification, verification by mass spectrometry and a Western blot using anti-C1ORF123 antibodies, crystallization and X-ray diffraction analysis of the protein crystals, are reported here. The rC1ORF123 protein was crystallized by the hanging-drop vapor-diffusion method with a reservoir solution comprised of 20% PEG 3350, 0.2 M magnesium chloride hexahydrate, 0.1 M sodium citrate pH 6.5. The crystals diffracted to 1.9 Å resolution and belonged to an orthorhombic space group with unit-cell parameters a = 59.32, b = 65.35, c = 95.05 Å. The calculated Matthews coefficient (VM) value of 2.27 Å(3) Da(-1) suggests that there are two molecules per asymmetric unit, with an estimated solvent content of 45.7%.
Collapse
Affiliation(s)
| | - Jastina Mat Yusop
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
| | - Zeti-Azura Mohamed-Hussein
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Aik-Hong Teh
- Centre for Chemical Biology, Universiti Sains Malaysia, 11900 Bayan Lepas, Penang, Malaysia
| | - Jitka Waterman
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, England
| | - Chyan Leong Ng
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor, Malaysia
| |
Collapse
|
98
|
Zahiri Z, Sharami SH, Milani F, Mohammadi F, Kazemnejad E, Ebrahimi H, Dalil Heirati SF. Metabolic Syndrome in Patients with Polycystic Ovary Syndrome in Iran. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2015; 9:490-6. [PMID: 26985336 PMCID: PMC4793169 DOI: 10.22074/ijfs.2015.4607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 07/27/2014] [Indexed: 11/04/2022]
Abstract
BACKGROUND The prevalence of metabolic syndrome (MetS) in polycystic ovary syndrome (PCOS) has been studied in different populations, but their results were so controversial regarding Iranian women. These controversial data indicated the need for more investigation of MetS characteristics in PCOS patients in our population. So this study aimed to evaluate the clinical and laboratory characteristics and metabolic features of patients with PCOS in Rasht. MATERIALS AND METHODS This prospective cross sectional study was conducted on 215 PCOS women who lived in Rasht, north of Iran, from March 2010 to July 2012. The participants were then divided into two groups of women with MetS (n=62) and women without MetS (n=153). The diagnosis of PCOS and MetS were based on the Rotterdam 2003 criteria and the Adult Treatment Panel III (ATP III) criteria, respectively. Demographic characteristics, fertility characteristics, family history and laboratory findings were assessed. RESULTS The prevalence of MetS in women with PCOS was 28.8%. In PCOS women of both groups, the waist circumference (WC) exceeded 88cm in 72.6%, hypertension [systolic blood pressure (SBP) and/or diastolic blood pressure (DBP) ≥130/85mm Hg] was prevalent in 9.3%, fasting blood sugar (FBS) level was ≥110 mg/dl in 6%, triglycerides (Tg) level were ≥150 mg/dl in 47%, and high-density lipoprotein (HDL) level was <50 mg/dl in 86%. The values of WC, SBP, DBP, body mass index (BMI), ovarian size, Tg, cholesterol, FBS, 2-hour blood sugar, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) were significantly greater in PCOS women with MetS than women without MetS. Also HDL and luteinizing hormone (LH) levels in women with MetS were significantly lower than women without MetS. CONCLUSION Prevalence of MetS in PCOS women was 28.8%, indicating that this value is higher than other studies conducted on PCOS women in Iran and other studies conducted on general population in Iran. PCOS women are considered as a high-risk population for MetS. The special strategies are required to prevent MetS and its associated complications in PCOS women.
Collapse
Affiliation(s)
- Ziba Zahiri
- Reproductive Health Research Center, Obstetrics and Gynecology Department, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyedeh Hajar Sharami
- Reproductive Health Research Center, Obstetrics and Gynecology Department, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Forozan Milani
- Reproductive Health Research Center, Obstetrics and Gynecology Department, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | - Hannan Ebrahimi
- Reproductive Health Research Center, Obstetrics and Gynecology Department, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyedeh Fatemeh Dalil Heirati
- Reproductive Health Research Center, Obstetrics and Gynecology Department, Alzahra Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
99
|
Palioura E, Diamanti-Kandarakis E. Polycystic ovary syndrome (PCOS) and endocrine disrupting chemicals (EDCs). Rev Endocr Metab Disord 2015; 16:365-71. [PMID: 26825073 DOI: 10.1007/s11154-016-9326-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a heterogeneous disorder of unclear etiopathogenesis that is likely to involve genetic and environmental components synergistically contributing to its phenotypic expression. Endocrine disrupting chemicals (EDCs) and in particular Bisphenol A (BPA) represent a group of widespread pollutants intensively investigated as possible environmental contributors to PCOS pathogenesis. Substantial evidence from in vitro and animal studies incriminates endocrine disruptors in the induction of reproductive and metabolic aberrations resembling PCOS characteristics. In humans, elevated BPA concentrations are observed in adolescents and adult PCOS women compared to reproductively healthy ones and are positively correlated with hyperandrogenemia, implying a potential role of the chemical in PCOS pathophysiology, although a causal interference cannot yet be established. It is plausible that developmental exposure to specific EDCs could permanently alter neuroendocrine, reproductive and metabolic regulation favoring PCOS development in genetically predisposed individuals or it could accelerate and/or exacerbate the natural course of the syndrome throughout life cycle exposure.
Collapse
Affiliation(s)
- Eleni Palioura
- Department of Endocrinology and Center of Excellence in Diabetes, Euroclinic Athens, Athens, Greece
| | | |
Collapse
|
100
|
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: 565] [Impact Index Per Article: 62.8] [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
|