1
|
Baddela VS, Michaelis M, Tao X, Koczan D, Brenmoehl J, Vanselow J. Comparative analysis of PI3K-AKT and MEK-ERK1/2 signaling-driven molecular changes in granulosa cells. Reproduction 2025; 169:e240317. [PMID: 39665647 DOI: 10.1530/rep-24-0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/12/2024] [Indexed: 12/13/2024]
Abstract
In brief PI3K-AKT signaling activates steroidogenesis by inducing estradiol and progesterone production, while MEK-ERK1/2 signaling regulates steroidogenesis by inhibiting estradiol and inducing progesterone production in granulosa cells (GCs). Both pathways are essential for glycolytic and mitochondrial metabolism in these cells. Abstract The PI3K-AKT and MEK-ERK1/2 signaling pathways are integral to fundamental cellular processes, such as proliferation, viability and differentiation. In GCs, these pathways are activated by follicle-stimulating hormone (FSH) and IGF1 through respective receptors. We investigated the comparative transcriptome changes induced by the AKT and ERK (ERK1/2) pathways using corresponding inhibitors in GCs. GCs isolated from antral follicles showed positive signals for phospho-AKT and phospho-ERK proteins. Treatment of cultured GCs with FSH and IGF1 induced phospho-AKT and phospho-ERK levels. Transcriptome analysis revealed 1436 genes regulated by AKT and 654 genes regulated by the ERK pathway. Among these, 94 genes were commonly downregulated and 11 genes were commonly upregulated in both datasets, while 110 genes were oppositely regulated. Bioinformatics analysis revealed that the inhibition of the PI3K-AKT and MEK-ERK pathways downregulates key reproductive processes and upstream molecules. Notably, AKT inhibition affected FSH, ESRRG and HIF1 pathways, while ERK inhibition impacted CG, FOS, TGFβ, EGR1 and LH pathways. Transcriptome data showed that genes related to estradiol production were inhibited by ERK and induced by the AKT pathway. This was verified by radioimmunoassays, and mRNA and protein analysis of CYP19A1 and STAR genes. In addition, transcriptome data suggested the downregulation of glucose metabolism in GCs. Using validation experiments, we confirm that both pathways are essential for glucose uptake, lactate production and mitochondrial activity in GCs. These data provide a resource for informing future research for analyzing various novel candidate genes regulated by the AKT and ERK pathways in GCs and other cell types.
Collapse
|
2
|
Bashiri Z, Sheibak N, Amjadi F, Zandieh Z. The role of myo-inositol supplement in assisted reproductive techniques. HUM FERTIL 2023; 26:1044-1060. [PMID: 35730666 DOI: 10.1080/14647273.2022.2073273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/20/2021] [Indexed: 11/04/2022]
Abstract
Assisted reproductive techniques can help many infertile couples conceive. Therefore, there is a need for an effective method to overcome the widespread problems of infertile men and women. Oocyte and sperm quality can increase the chances of successful in vitro fertilisation. The maturation environment in which gametes are present can affect their competency for fertilisation. It is well established that myo-inositol (MI) plays a pivotal role in reproductive physiology. It participates in cell membrane formation, lipid synthesis, cell proliferation, cardiac regulation, metabolic alterations, and fertility. This molecule also acts as a direct messenger of insulin and improves glucose uptake in various reproductive tissues. Evidence suggests that MI regulates events such as gamete maturation, fertilisation, and embryo growth through intracellular Ca2 + release and various signalling pathways. In addition to the in-vivo production of MI from glucose in the reproductive organs, its synthesis by in vitro-cultured sperm and follicles has also been reported. Therefore, MI is suggested as a therapeutic approach to maintain sperm and oocyte health in men and women with reproductive disorders and individuals of reproductive age.
Collapse
Affiliation(s)
- Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility and Infertility Clinic, Hamedan, Iran
| | - Nadia Sheibak
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemehsadat Amjadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandieh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Genazzani AD, Genazzani AR. Polycystic Ovary Syndrome as Metabolic Disease: New Insights on Insulin Resistance. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:71-77. [PMID: 37313240 PMCID: PMC10258623 DOI: 10.17925/ee.2023.19.1.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 06/15/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a very frequent disease that affects reproductive ability and menstrual regularity. Other than the criteria established at the Rotterdam consensus, in these last few years a new issue, insulin resistance, has been found frequently, and at a very high grade, in patients with PCOS. Insulin resistance occurs for several factors, such as overweight and obesity, but it is now clear that it occurs in patients with PCOS with normal weight, thus supporting the hypothesis that insulin resistance is independent of body weight. Evidence shows that a complex pathophysiological situation occurs that impairs post-receptor insulin signalling, especially in patients with PCOS and familial diabetes. In addition, patients with PCOS have a high incidence of non-alcoholic fatty liver disease related to the hyperinsulinaemia. This narrative review focuses on the recent new insights about insulin resistance in patients with PCOS, to better understand the metabolic impairment accounting for most of the clinical signs/symptoms of PCOS.
Collapse
Affiliation(s)
- Alessandro D Genazzani
- Gynaecological Endocrinology Center, Department of Obstetrics and Gynaecology, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea R Genazzani
- Department of Obstetrics and Gynaecology, University of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Nevelli F, Palmese A, Gleixner R, Peroglio F, D'Acunto CW, Dadone A, D'Hooghe T, Lispi M. Biological Assay to Determine Gonadotropin Potency: From In Vivo to In Vitro Sustainable Method. Int J Mol Sci 2023; 24:ijms24098040. [PMID: 37175746 PMCID: PMC10178553 DOI: 10.3390/ijms24098040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/04/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023] Open
Abstract
Various preparations of follicle-stimulating hormone (FSH) are commercially available; however, they differ in glycoforms composition and purity owing to their respective sources. Additional chemical/physical changes can also be introduced during manufacturing and can impact their biological activity (biopotency), which is routinely assessed using an in vivo bioassay (Steelman-Pohley). This study aimed to determine whether an in vitro bioassay could assess biopotency by distinguishing between r-hFSH chemical/physical variants with similar ability to the in vivo bioassay. The specific activity (units of biological activity per mg of product) of variants of r-hFSH generated through enrichment (acidic/basic), stress (oxidative/acidic pH) and enzymatic treatment (desialylation and desialylation/degalactosylation) was compared using the in vivo and in vitro bioassays. The in vitro bioassay reliably detected potential chemical/physical modifications in r-hFSH variants that may impact biopotency. Overall, the methods demonstrated a comparable ability to detect changes in specific activities due to chemical/physical differences in r-hFSH variants. These data indicate that the in vitro bioassay is suitable to replace the in vivo bioassay.
Collapse
Affiliation(s)
- Francesco Nevelli
- Analytical Development Biotech-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Istituto di Ricerche Biomediche "Antoine Marxer" RBM S.p.A., Colleretto Giacosa, Via Ribes, 1, 10010 Samone, Italy
| | - Angelo Palmese
- Analytical Development Biotech-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Merck Serono S.p.A., Piazza del Pigneto, 9, 00176 Rome, Italy
| | - Ralf Gleixner
- Ares Trading S.A., Rue de l'Ouriette 151, 1170 Aubonne, Switzerland
| | - Flavio Peroglio
- GHO Ivrea QC & Scientific Excellence-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Istituto di Ricerche Biomediche "Antoine Marxer" RBM S.p.A., Colleretto Giacosa, Via Ribes, 1, 10010 Samone, Italy
| | - Cosimo-Walter D'Acunto
- Analytical Development Biotech-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Istituto di Ricerche Biomediche "Antoine Marxer" RBM S.p.A., Colleretto Giacosa, Via Ribes, 1, 10010 Samone, Italy
- Analytical Development Biotech-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Merck Serono S.p.A., Piazza del Pigneto, 9, 00176 Rome, Italy
| | - Aurora Dadone
- Analytical Development Biotech-Global Analytical Development-Global Development & Launch-Global Healthcare Operation, Istituto di Ricerche Biomediche "Antoine Marxer" RBM S.p.A., Colleretto Giacosa, Via Ribes, 1, 10010 Samone, Italy
| | - Thomas D'Hooghe
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
- Department of Development and Regeneration, Laboratory of Endometrium, Endometriosis & Reproductive Medicine, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University Medical School, 333 Cedar St., New Haven, CT 06510, USA
| | - Monica Lispi
- Global Medical Affairs Fertility, Merck Healthcare KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
- PhD School of Clinical and Experimental Medicine, Unit of Endocrinology, University of Modena and Reggio Emilia, Viale A. Allegri 9, 42121 Reggio Emilia, Italy
| |
Collapse
|
5
|
Hua SS, Ding JJ, Sun TC, Guo C, Zhang Y, Yu ZH, Cao YQ, Zhong LH, Wu Y, Guo LY, Luo JH, Cui YH, Qiu S. NMDAR-dependent synaptic potentiation via APPL1 signaling is required for the accessibility of a prefrontal neuronal assembly in retrieving fear extinction. Biol Psychiatry 2023:S0006-3223(23)00087-2. [PMID: 36842495 DOI: 10.1016/j.biopsych.2023.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND The ventromedial prefrontal cortex (vmPFC) has been viewed as a locus to store and recall extinction memory. However, the synaptic and cellular mechanisms underlying this process remain elusive. METHODS We combined transgenic mice, electrophysiological recording, activity-dependent cell labeling, and chemogenetic manipulation to analyze the role of adaptor protein APPL1 in the vmPFC for fear extinction retrieval. RESULTS We found that both constitutive and conditional APPL1 knockout decreases NMDA receptor (NMDAR) function in the vmPFC and impairs fear extinction retrieval. Moreover, APPL1 undergoes nuclear translocation during extinction retrieval. Blocking APPL1 nucleocytoplasmic translocation reduces NMDAR currents and disrupts extinction retrieval. We further identified a prefrontal neuronal ensemble that is both necessary and sufficient for the storage of extinction memory. Inducible APPL1 knockout in this ensemble abolishes NMDAR-dependent synaptic potentiation and disrupts extinction retrieval, while simultaneously chemogenetic activation of this ensemble rescues the impaired behaviors. CONCLUSIONS Therefore, our results indicate that a prefrontal neuronal ensemble stores extinction memory, and APPL1 signaling supports these neurons to retrieve extinction memory via controlling NMDAR-dependent potentiation.
Collapse
Affiliation(s)
- Shu-Shan Hua
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jin-Jun Ding
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tian-Cheng Sun
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chen Guo
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ying Zhang
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zi-Hui Yu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yi-Qing Cao
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lin-Hong Zhong
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yu Wu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Lu-Ying Guo
- Kidney Disease Center of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jian-Hong Luo
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China
| | - Yi-Hui Cui
- Department of Neurobiology and Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Shuang Qiu
- Department of Neurobiology and Department of Anesthesiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology,ZhejiangUniversity ,Hangzhou ,310058 ,China.
| |
Collapse
|
6
|
Petrillo T, Semprini E, Tomatis V, Arnesano M, Ambrosetti F, Battipaglia C, Sponzilli A, Ricciardiello F, Genazzani AR, Genazzani AD. Putative Complementary Compounds to Counteract Insulin-Resistance in PCOS Patients. Biomedicines 2022; 10:biomedicines10081924. [PMID: 36009471 PMCID: PMC9406066 DOI: 10.3390/biomedicines10081924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/25/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most frequent endocrine-metabolic disorder among women at reproductive age. The diagnosis is based on the presence of at least two out of three criteria of the Rotterdam criteria (2003). In the last decades, the dysmetabolic aspect of insulin resistance and compensatory hyperinsulinemia have been taken into account as the additional key features in the etiopathology of PCOS, and they have been widely studied. Since PCOS is a complex and multifactorial syndrome with different clinical manifestations, it is difficult to find the gold standard treatment. Therefore, a great variety of integrative treatments have been reported to counteract insulin resistance. PCOS patients need a tailored therapeutic strategy, according to the patient’s BMI, the presence or absence of familiar predisposition to diabetes, and the patient’s desire to achieve pregnancy or not. The present review analyzes and discloses the main clinical insight of such complementary substances.
Collapse
Affiliation(s)
- Tabatha Petrillo
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Elisa Semprini
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Veronica Tomatis
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Melania Arnesano
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Fedora Ambrosetti
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Christian Battipaglia
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Alessandra Sponzilli
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Francesco Ricciardiello
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Andrea R. Genazzani
- Department of Obstetrics and Gynecology, University of Pisa, 56126 Pisa, Italy
| | - Alessandro D. Genazzani
- Gynecological Endocrinology Center, Department of Obstetrics and Gynecology, University of Modena and Reggio Emilia, 41121 Modena, Italy
- Correspondence:
| |
Collapse
|
7
|
Genazzani AD, Battipaglia C, Petrillo T, Piacquadio N, Ambrosetti F, Arnesano M, Semprini E, Sponzilli A, Tomatis V, Simoncini T. Familial diabetes predisposes PCOS patients to insulin resistance (IR), reproductive impairment and hepatic dysfunction: effects of d-chiro inositol (DCI) and alpha lipoic acid (ALA) administration on hepatic insulin extraction (HIE) index. Gynecol Endocrinol 2022; 38:681-688. [PMID: 35748584 DOI: 10.1080/09513590.2022.2089107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
ObjectivePCOS is a syndrome is characterized by 2 out of 3 of the criteria established during the Rotterdam Consensus Conference. Recently the issue of insulin resistance (IR) has caught attention.SubjectsA group of overweight/obese PCOS patients (n = 30) have been evaluated before and after 3 months of daily integrative administration of d-chiro inositol (DCI) (500 mg) and alpha lipoic acid (ALA) (300 mg).MethodsHormonal and metabolic profiles, oral glucose tolerance test (OGTT) for glucose, insulin and C-peptide response were performed in baseline conditions and after DCI plus ALA treatment. Hepatic Insulin Extraction (HIE) index was computed along the OGTT to evaluate the liver ability in degrading insulin.ResultsThe treatment decreased LH, Androstenedione (A), insulin plasma levels, BMI, HOMA index, AST and ALT. Considering patients for the presence (n = 17) or absence of familial diabetes (n = 13), the greatest improvements occurred in the former patients. Insulin response to OGTT was greatly reduced after the treatment interval in PCOS with familial diabetes. HIE computation disclosed that in presence of familial diabetes liver degradation of insulin is reduced thus leaving a higher amount of circulating insulin. DCI plus ALA administration decreased AST and ALT and restored hepatic insulin clearance since HIE profile was improved.ConclusionOur study demonstrates that in overweight/obese PCOS the predisposition to familial diabetes triggers IR not only through the endogenous impaired DCI and ALA synthesis but also through a reduced hepatic clearance of insulin. DCI plus ALA administration positively improved hormonal, metabolic profiles as well as liver function.
Collapse
Affiliation(s)
- Alessandro D Genazzani
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Christian Battipaglia
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Tabatha Petrillo
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicola Piacquadio
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Fedora Ambrosetti
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Melania Arnesano
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Semprini
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Sponzilli
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Veronica Tomatis
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Simoncini
- Department of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| |
Collapse
|
8
|
DiNicolantonio JJ, H O'Keefe J. Myo-inositol for insulin resistance, metabolic syndrome, polycystic ovary syndrome and gestational diabetes. Open Heart 2022; 9:openhrt-2022-001989. [PMID: 35236761 PMCID: PMC8896029 DOI: 10.1136/openhrt-2022-001989] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- James J DiNicolantonio
- Department of Preventive Cardiology, Saint Lukes Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - James H O'Keefe
- Department of Preventive Cardiology, Saint Lukes Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
9
|
Laganà AS, Forte G, Bizzarri M, Kamenov ZA, Bianco B, Kaya C, Gitas G, Alkatout I, Terzic M, Unfer V. Inositols in the ovaries: activities and potential therapeutic applications. Expert Opin Drug Metab Toxicol 2022; 18:123-133. [PMID: 35472446 DOI: 10.1080/17425255.2022.2071259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Myo-inositol (MI) and d-chiro-inositol (DCI) play a key role in ovarian physiology, as they are second messengers of insulin and gonadotropins. Ex-vivo and in-vitro experiments demonstrate that both isomers are deeply involved in steroid biosynthesis, and that reduced MI-to-DCI ratios are associated with pathological imbalance of sex hormones. AREAS COVERED This expert opinion provides an overview of the physiological distribution of MI and DCI in the ovarian tissues, and a thorough insight of their involvement into ovarian steroidogenesis. Insulin resistance and compensatory hyperinsulinemia dramatically reduce the MI-to-DCI ratio in the ovaries, leading to gynecological disorders characterized by hyperandrogenism, altered menstrual cycle and infertility. EXPERT OPINION Available evidence indicates that MI and DCI have very specific physiological roles and, seemingly, physiological MI-to-DCI ratios in the ovaries are crucial to maintain the correct homeostasis of steroids. Inositol treatments should be evaluated on the patients' specific conditions and needs, as long-term supplementation of high doses of DCI may cause detrimental effects on the ovarian functionality. In addition, the effects of inositol therapy on the different PCOS phenotypes should be further investigated in order to better tailor the supplementation.
Collapse
Affiliation(s)
- Antonio Simone Laganà
- The Experts Group on Inositols in Basic and Clinical Research (EGOI), Rome, Italy.,Unit of Gynecologic Oncology, ARNAS 'Civico - Di Cristina - Benfratelli', Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | | | - Mariano Bizzarri
- The Experts Group on Inositols in Basic and Clinical Research (EGOI), Rome, Italy.,Department of Experimental Medicine, Systems Biology Group Lab, Sapienza University of Rome, Rome, Italy
| | - Zdravko A Kamenov
- The Experts Group on Inositols in Basic and Clinical Research (EGOI), Rome, Italy.,Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Bianca Bianco
- Discipline of Sexual and Reproductive Health, and Populational Genetics - Department of Collective Health, Faculdade de Medicina do ABC/Centro Universitário FMABC, Santo André, Brazil
| | - Cihan Kaya
- Department of Obstetrics and Gynaecology, University of Health Sciences, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Georgios Gitas
- Department of Obstetrics and Gynecology, Charité Campus, Berlin, Germany
| | - Ibrahim Alkatout
- Department of Obstetrics and Gynecology, University Hospital Schleswig Holstein, Kiel, Germany
| | - Milan Terzic
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan.,National Research Center for Maternal and Child Health, Clinical Academic Department of Women's Health, University Medical Center, Nur-Sultan, Kazakhstan.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, 300 Halket Street, Pittsburgh, Pennsylvania, USA
| | - Vittorio Unfer
- The Experts Group on Inositols in Basic and Clinical Research (EGOI), Rome, Italy.,Systems Biology Group Lab, Rome, Italy
| |
Collapse
|
10
|
Haldar S, Agrawal H, Saha S, Straughn AR, Roy P, Kakar SS. Overview of follicle stimulating hormone and its receptors in reproduction and in stem cells and cancer stem cells. Int J Biol Sci 2022; 18:675-692. [PMID: 35002517 PMCID: PMC8741861 DOI: 10.7150/ijbs.63721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/21/2021] [Indexed: 11/05/2022] Open
Abstract
Follicle stimulating hormone (FSH) and its receptor (FSHR) have been reported to be responsible for several physiological functions and cancers. The responsiveness of stem cells and cancer stem cells towards the FSH-FSHR system make the function of FSH and its receptors more interesting in the context of cancer biology. This review is comprised of comprehensive information on FSH-FSHR signaling in normal physiology, gonadal stem cells, cancer cells, and potential options of utilizing FSH-FSHR system as an anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Swati Haldar
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India.,Current address: Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, Uttarakhand 249405
| | - Himanshu Agrawal
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India
| | - Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences Rishikesh, Uttarakhand 249203, India
| | - Alex R Straughn
- Department of Physiology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Uttarakhand 247667, India
| | - Sham S Kakar
- Department of Physiology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
11
|
Sirisin J, Kamnate A, Polsan Y, Somintara S, Chomphoo S, Sakagami H, Kondo H, Hipkaeo W. Localization of phosphatidylinositol 4-phosphate 5-kinase (PIP5K) α confined to the surface of lipid droplets and adjacent narrow cytoplasm in progesterone-producing cells of in situ ovaries of adult mice. Acta Histochem 2021; 123:151794. [PMID: 34624591 DOI: 10.1016/j.acthis.2021.151794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/15/2022]
Abstract
Phosphatidylinositol(4,5)bisphosphate (PI(4,5)P2) produced by phosphatidylinositol phosphate 5 kinase (PIP5K) plays not only as a precursor of second messengers in the phosphoinositide signal transduction, but also multiple roles influencing a variety of cellular activities. From this viewpoint, the present study attempted to localize PIP5Kα in the ovaries in situ of adult mice. PIP5Kα-immunoreactivity was confined to the surfaces of lipid droplets (LDs) and their adjacent cytoplasm in progesterone-producing cells of the interstitial glands, corpora lutea and theca interna. The LDs often contained membranous tubules/lamellae along their surfaces and within their interior whose membranes were continuous with those delineating LDs composed of a monolayer of phospholipids and were partially PIP5Kα-immunoreactive. Although granulosa cells of healthy-looking follicles were immunonegative, as the atresia progressed, PIP5Kα-immunoreactivity first appeared in sparsely dispersed dot forms in mural cells of the follicular epithelia, and then were dominant in almost all mural cells that remained after desquamation of the antral cells. The present study provides evidence suggesting that PI(4,5)P2 locally synthesized by PIP5K in LDs is involved in the lipid transfer between lipid droplets (LDs) and the endoplasmic reticulum, which eventually regulates ovarian progesterone production through control of multiple dynamic activities of LDs. It is also suggested that PIP5Kα and PI(4,5)P2 are implicated in the modulation of programmed cell death and/or acquiring the ability of progesterone production in some follicular cells surviving atresia.
Collapse
Affiliation(s)
- Juthathip Sirisin
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anussara Kamnate
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Yada Polsan
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Somsuda Somintara
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Surang Chomphoo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hiroyuki Sakagami
- Department of Anatomy, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Hisatake Kondo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Department of Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Wiphawi Hipkaeo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
12
|
Lee EB, Chakravarthi VP, Wolfe MW, Rumi MAK. ERβ Regulation of Gonadotropin Responses during Folliculogenesis. Int J Mol Sci 2021; 22:ijms221910348. [PMID: 34638689 PMCID: PMC8508937 DOI: 10.3390/ijms221910348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
Gonadotropins are essential for regulating ovarian development, steroidogenesis, and gametogenesis. While follicle stimulating hormone (FSH) promotes the development of ovarian follicles, luteinizing hormone (LH) regulates preovulatory maturation of oocytes, ovulation, and formation of corpus luteum. Cognate receptors of FSH and LH are G-protein coupled receptors that predominantly signal through cAMP-dependent and cAMP-independent mechanisms that activate protein kinases. Subsequent vital steps in response to gonadotropins are mediated through activation or inhibition of transcription factors required for follicular gene expression. Estrogen receptors, classical ligand-activated transcriptional regulators, play crucial roles in regulating gonadotropin secretion from the hypothalamic-pituitary axis as well as gonadotropin function in the target organs. In this review, we discuss the role of estrogen receptor β (ERβ) regulating gonadotropin response during folliculogenesis. Ovarian follicles in Erβ knockout (ErβKO) mutant female mice and rats cannot develop beyond the antral state, lack oocyte maturation, and fail to ovulate. Theca cells (TCs) in ovarian follicles express LH receptor, whereas granulosa cells (GCs) express both FSH receptor (FSHR) and LH receptor (LHCGR). As oocytes do not express the gonadotropin receptors, the somatic cells play a crucial role during gonadotropin induced oocyte maturation. Somatic cells also express high levels of estrogen receptors; while TCs express ERα and are involved in steroidogenesis, GCs express ERβ and are involved in both steroidogenesis and folliculogenesis. GCs are the primary site of ERβ-regulated gene expression. We observed that a subset of gonadotropin-induced genes in GCs, which are essential for ovarian follicle development, oocyte maturation and ovulation, are dependent on ERβ. Thus, ERβ plays a vital role in regulating the gonadotropin responses in ovary.
Collapse
Affiliation(s)
- Eun B. Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - V. Praveen Chakravarthi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: ; Tel.: +1-913-588-8059
| |
Collapse
|
13
|
Johnson GP, Jonas KC. Mechanistic insight into how gonadotropin hormone receptor complexes direct signaling†. Biol Reprod 2021; 102:773-783. [PMID: 31882999 DOI: 10.1093/biolre/ioz228] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/29/2022] Open
Abstract
Gonadotropin hormones and their receptors play a central role in the control of male and female reproduction. In recent years, there has been growing evidence surrounding the complexity of gonadotropin hormone/receptor signaling, with it increasingly apparent that the Gαs/cAMP/PKA pathway is not the sole signaling pathway that confers their biological actions. Here we review recent literature on the different receptor-receptor, receptor-scaffold, and receptor-signaling molecule complexes formed and how these modulate and direct gonadotropin hormone-dependent intracellular signal activation. We will touch upon the more controversial issue of extragonadal expression of FSHR and the differential signal pathways activated in these tissues, and lastly, highlight the open questions surrounding the role these gonadotropin hormone receptor complexes and how this will shape future research directions.
Collapse
Affiliation(s)
| | - Kim Carol Jonas
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, London, UK
| |
Collapse
|
14
|
Abstract
Gonadotropins are glycoprotein sex hormones regulating development and reproduction and bind to specific G protein–coupled receptors expressed in the gonads. Their effects on multiple signaling cascades and intracellular events have recently been characterized using novel technological and scientific tools. The impact of allosteric modulators on gonadotropin signaling, the role of sugars linked to the hormone backbone, the detection of endosomal compartments supporting signaling modules, and the dissection of different effects mediated by these molecules are areas that have advanced significantly in the last decade. The classic view providing the exclusive activation of the cAMP/protein kinase A (PKA) and the steroidogenic pathway by these hormones has been expanded with the addition of novel signaling cascades as determined by high-resolution imaging techniques. These new findings provided new potential therapeutic applications. Despite these improvements, unanswered issues of gonadotropin physiology, such as the intrinsic pro-apoptotic potential to these hormones, the existence of receptors assembled as heteromers, and their expression in extragonadal tissues, remain to be studied. Elucidating these issues is a challenge for future research.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Via P. Giardini 1355, 41126 Modena, Italy
| |
Collapse
|
15
|
Banerjee AA, Joseph S, Mahale SD. From cell surface to signalling and back: the life of the mammalian FSH receptor. FEBS J 2020; 288:2673-2696. [DOI: 10.1111/febs.15649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/17/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Antara A. Banerjee
- Division of Structural Biology National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| | - Shaini Joseph
- Genetic Research Center National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| | - Smita D. Mahale
- Division of Structural Biology National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
- ICMR Biomedical Informatics Centre National Institute for Research in Reproductive Health (Indian Council of Medical Research) Parel India
| |
Collapse
|
16
|
Affiliation(s)
- Alessandro Genazzani
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
17
|
Pani A, Giossi R, Menichelli D, Fittipaldo VA, Agnelli F, Inglese E, Romandini A, Roncato R, Pintaudi B, Del Sole F, Scaglione F. Inositol and Non-Alcoholic Fatty Liver Disease: A Systematic Review on Deficiencies and Supplementation. Nutrients 2020; 12:nu12113379. [PMID: 33153126 PMCID: PMC7694137 DOI: 10.3390/nu12113379] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Liver lipid accumulation is a hallmark of non-alcoholic fatty liver disease (NAFLD), broadly associated with insulin resistance. Inositols (INS) are ubiquitous polyols implied in many physiological functions. They are produced endogenously, are present in many foods and in dietary supplements. Alterations in INS metabolism seems to play a role in diseases involving insulin resistance such as diabetes and polycystic ovary syndrome. Given its role in other metabolic syndromes, the hypothesis of an INS role as a supplement in NAFLD is intriguing. We performed a systematic review of the literature to find preclinical and clinical evidence of INS supplementation efficacy in NAFLD patients. We retrieved 10 studies on animal models assessing Myoinosiol or Pinitol deficiency or supplementation and one human randomized controlled trial (RCT). Overall, INS deficiency was associated with increased fatty liver in animals. Conversely, INS supplementation in animal models of fatty liver reduced hepatic triglycerides and cholesterol accumulation and maintained a normal ultrastructural liver histopathology. In the one included RCT, Pinitol supplementation obtained similar results. Pinitol significantly reduced liver fat, post-prandial triglycerides, AST levels, lipid peroxidation increasing glutathione peroxidase activity. These results, despite being limited, indicate the need for further evaluation of INS in NAFLD in larger clinical trials.
Collapse
Affiliation(s)
- Arianna Pani
- Department of Oncology and Hemato-oncology, Postgraduate School of Clinical Pharmacology, University of Milan, 20129 Milan, Italy; (A.P.); (R.G.); (A.R.); (R.R.); (F.S.)
- Department of Neuroimmunology and Neuromuscular Diseases, Fondazione I.R.C.C.S., Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Riccardo Giossi
- Department of Oncology and Hemato-oncology, Postgraduate School of Clinical Pharmacology, University of Milan, 20129 Milan, Italy; (A.P.); (R.G.); (A.R.); (R.R.); (F.S.)
- Department of Neuroimmunology and Neuromuscular Diseases, Fondazione I.R.C.C.S., Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Danilo Menichelli
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Atherothrombosis Center, I Medical Clinic l, Sapienza University of Rome, 00161 Rome, Italy;
- Correspondence:
| | | | - Francesca Agnelli
- Internal Medicine Department, ASST Great Metropolitan Hospital Niguarda, 20162 Milan, Italy;
| | - Elvira Inglese
- Department of Laboratory Medicine, ASST Great Metropolitan Hospital Niguarda, 20162 Milan, Italy;
| | - Alessandra Romandini
- Department of Oncology and Hemato-oncology, Postgraduate School of Clinical Pharmacology, University of Milan, 20129 Milan, Italy; (A.P.); (R.G.); (A.R.); (R.R.); (F.S.)
| | - Rossana Roncato
- Department of Oncology and Hemato-oncology, Postgraduate School of Clinical Pharmacology, University of Milan, 20129 Milan, Italy; (A.P.); (R.G.); (A.R.); (R.R.); (F.S.)
- Experimental & Clinical Pharmacology Unit, Oncology Referral Center (CRO), IRCCS, 33081 Aviano, Italy
| | - Basilio Pintaudi
- SSD Diabetes Unit, ASST Great Metropolitan Hospital Niguarda, 20162 Milan, Italy;
| | - Francesco Del Sole
- Department of Clinical, Internal, Anesthesiologic and Cardiovascular Sciences, Atherothrombosis Center, I Medical Clinic l, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, Postgraduate School of Clinical Pharmacology, University of Milan, 20129 Milan, Italy; (A.P.); (R.G.); (A.R.); (R.R.); (F.S.)
- Department of Laboratory Medicine, ASST Great Metropolitan Hospital Niguarda, 20162 Milan, Italy;
| |
Collapse
|
18
|
Gupta D, Khan S, Islam M, Malik BH, Rutkofsky IH. Myo-Inositol's Role in Assisted Reproductive Technology: Evidence for Improving the Quality of Oocytes and Embryos in Patients With Polycystic Ovary Syndrome. Cureus 2020; 12:e8079. [PMID: 32542134 PMCID: PMC7292722 DOI: 10.7759/cureus.8079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is one of the most common causes of subfertility, and it is characterized by hormonal dysregulation like insulin resistance. Various measures have been taken in the past to overcome this insulin resistance to improve fertility treatment outcomes. The current paper aims to review and compare the existing studies and literature to assess the impact of myo-inositol (MI) on oocyte and embryo quality in assisted reproductive technology (ARTs). We thoroughly searched the PubMed and Google Scholar databases by using the keywords "PCOS, polycystic ovarian syndrome, inositol, oocyte quality, embryo quality, assisted conception, ART, IVF, and in vitro fertilization." Nine articles were finalized for review in this paper. Many of the reviewed studies have shown a trend toward the improvement of embryo quality in women with PCOS after MI supplementation; however, there is a lack of statistically significant evidence to support the use of MI in enhancing the quality of oocyte and/or embryo. Clear evidence regarding the role of MI in enhancing the quality of oocyte and embryo in PCOS is limited. A well-controlled, large, randomized controlled trial is required to definitively accept or refute its role.
Collapse
Affiliation(s)
- Deepti Gupta
- Reproductive Medicine, Saint Mary's Hospital, Manchester, GBR.,Obstetrics and Gynecology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Muhammad Islam
- Pediatrics, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Ian H Rutkofsky
- Psychiatry, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
19
|
Zariñán T, Butnev VY, Gutiérrez-Sagal R, Maravillas-Montero JL, Martínez-Luis I, Mejía-Domínguez NR, Juárez-Vega G, Bousfield GR, Ulloa-Aguirre A. In Vitro Impact of FSH Glycosylation Variants on FSH Receptor-stimulated Signal Transduction and Functional Selectivity. J Endocr Soc 2020; 4:bvaa019. [PMID: 32342021 PMCID: PMC7175721 DOI: 10.1210/jendso/bvaa019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
FSH exists as different glycoforms that differ in glycosylation of the hormone-specific β-subunit. Tetra-glycosylated FSH (FSH24) and hypo-glycosylated FSH (FSH18/21) are the most abundant glycoforms found in humans. Employing distinct readouts in HEK293 cells expressing the FSH receptor, we compared signaling triggered by human pituitary FSH preparations (FSH18/21 and FSH24) as well as by equine FSH (eFSH), and human recombinant FSH (recFSH), each exhibiting distinct glycosylation patterns. The potency in eliciting cAMP production was greater for eFSH than for FSH18/21, FSH24, and recFSH, whereas in the ERK1/2 activation readout, potency was highest for FSH18/21 followed by eFSH, recFSH, and FSH24. In β-arrestin1/2 CRISPR/Cas9 HEK293-KO cells, FSH18/21 exhibited a preference toward β-arrestin-mediated ERK1/2 activation as revealed by a drastic decrease in pERK during the first 15-minute exposure to this glycoform. Exposure of β-arrestin1/2 KO cells to H89 additionally decreased pERK1/2, albeit to a significantly lower extent in response to FSH18/21. Concurrent silencing of β-arrestin and PKA signaling, incompletely suppressed pERK response to FSH glycoforms, suggesting that pathways other than those dependent on Gs-protein and β-arrestins also contribute to FSH-stimulated pERK1/2. All FSH glycoforms stimulated intracellular Ca2+ (iCa2+) accumulation through both influx from Ca2+ channels and release from intracellular stores; however, iCa2+ in response to FSH18/21 depended more on the latter, suggesting differences in mechanisms through which glycoforms promote iCa2+ accumulation. These data indicate that FSH glycosylation plays an important role in defining not only the intensity but also the functional selectivity for the mechanisms leading to activation of distinct signaling cascades.
Collapse
Affiliation(s)
- Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Viktor Y Butnev
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Iván Martínez-Luis
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George R Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, Kansas, USA
| | - Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México (UNAM)-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
20
|
Caputo M, Bona E, Leone I, Samà MT, Nuzzo A, Ferrero A, Aimaretti G, Marzullo P, Prodam F. Inositols and metabolic disorders: From farm to bedside. J Tradit Complement Med 2020; 10:252-259. [PMID: 32670820 PMCID: PMC7340869 DOI: 10.1016/j.jtcme.2020.03.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 01/22/2023] Open
Abstract
Inositol and its derivates are catching interest in metabolism since taking part in several physiological processes, including endocrine modulation. Through several mechanisms mostly mediated by insulin signaling, these compounds regulate the activities of several hormones and are essential in oocytes maturation. It is interesting to point out the contribution of an inositol deficiency in the development of several diseases, mainly in the metabolic and endocrine setting. Inositols derive from both diet and endogenous production; among causes of inositol deficiency reduced dietary intake, increased catabolism and/or excretion, decreased biosynthesis, inhibition of gut and cellular uptake and altered microbiota could be considered. Mounting direct and indirect evidence suggests that the two main isoforms (Myo-inositol-inositol, D-chiro-inositol) are implied in glycemic and lipidic metabolism and supplementation yield a beneficial effect on these parameters without hazards for health. Moreover, they have a role in polycystic ovary syndrome, acting as insulin-sensitizing agents and free radical scavengers, helping to regulate metabolism and promoting ovulation. The aim of this narrative review is to discuss the role of inositols in metabolic function disorders paying attention to whether these compounds could be efficacious and safe as a therapeutic agent with a focus on dietary intake and the role of gut microbiota. Inositol deficiency is implicated in the development of metabolic and endocrine diseases. Inositol compounds could be safe food supplement to restore metabolic imbalance. Inositol compounds partly derive from microbiota phytases entering in bacterial metabolism. Diet inositol content has a role in shaping gut microbiota and the host metabolism.
Collapse
Affiliation(s)
- M Caputo
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - E Bona
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy.,Interdisciplinary Research Center of Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - I Leone
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - M T Samà
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - A Nuzzo
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - A Ferrero
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - G Aimaretti
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - P Marzullo
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Istituto Auxologico Italiano, I.R.C.C.S, Ospedale San Giuseppe, Piancavallo, Italy
| | - F Prodam
- Endocrinology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.,Interdisciplinary Research Center of Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy.,Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
21
|
Fruzzetti F, Benelli E, Fidecicchi T, Tonacchera M. Clinical and Metabolic Effects of Alpha-Lipoic Acid Associated with Two Different Doses of Myo-Inositol in Women with Polycystic Ovary Syndrome. Int J Endocrinol 2020; 2020:2901393. [PMID: 32256570 PMCID: PMC7106925 DOI: 10.1155/2020/2901393] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of this retrospective study was to evaluate the effects of a treatment with α-lipoic acid (ALA) associated with two different doses of myo-inositol (MI) on clinical and metabolic features of women with polycystic ovary syndrome (PCOS). Eighty-eight women received the treatment, and 71 among them had complete clinical charts and were considered eligible for this study. All women were treated with 800 mg of ALA per day: 43 patients received 2000 mg of MI and 28 received 1000 mg of MI per day. Menstrual cyclicity, BMI, FSH, LH, estradiol, testosterone, androstenedione, fasting insulin, HOMA-IR, and insulin response to a 2 h OGTT were evaluated before and after 6 months of treatment. The presence of diabetic relatives (DRs) was investigated. Cycle regularity was improved in 71.2% of women. The improvement of menstrual cyclicity occurred regardless of the state of IR and the presence of DRs of the patients. Women with IR mainly showed a significant improvement of metabolic parameters, while those without IR had significant changes of reproductive hormones. Patients with DRs did not show significant changes after the treatment. 85.7% of women taking 2000 mg of MI reported a higher improvement of menstrual regularity than those taking 1000 mg of MI (50%; p < 0.01). In conclusion, ALA + MI positively affects the menstrual regularity of women with PCOS, regardless of their metabolic phenotype, with a more evident effect with a higher dose of MI. This effect seems to be insulin independent. The presence of IR seems to be a predictor of responsivity to the treatment in terms of an improvement of the metabolic profile.
Collapse
Affiliation(s)
- Franca Fruzzetti
- Department of Obstetrics and Gynecology, Pisa University Hospital, Pisa, Italy
| | - Elena Benelli
- Department of Endocrinology, Pisa University Hospital, Pisa, Italy
| | - Tiziana Fidecicchi
- Department of Obstetrics and Gynecology, Pisa University Hospital, Pisa, Italy
| | | |
Collapse
|
22
|
Genazzani AD, Prati A, Marchini F, Petrillo T, Napolitano A, Simoncini T. Differential insulin response to oral glucose tolerance test (OGTT) in overweight/obese polycystic ovary syndrome patients undergoing to myo-inositol (MYO), alpha lipoic acid (ALA), or combination of both. Gynecol Endocrinol 2019; 35:1088-1093. [PMID: 31304823 DOI: 10.1080/09513590.2019.1640200] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Polycystic ovary syndrome is characterized by several endocrine impairments, insulin resistance and hyperinsulinemia. We aimed to evaluate the effects of myo-inositol (MYO), alpha-lipoic acid (ALA) and a combination of both. Setting: retrospective study. Ninety overweight/obese patients were considered. Presence or absence of first grade diabetic relatives was checked. Patients were administered MYO (1 g/die per os), ALA (400 mg/die per os), MYO (1 gr/die) + ALA (400 mg/die) per os. Only 76 out of 90 patients completed the 12 weeks of treatment. Patients were evaluated before and after the treatment interval for LH, FSH, E2 (estradiol), A (androstenedione), T (testosterone) plasma levels, oral glucose tolerance test (OGTT). All treatments demonstrated specific positive effects: MYO modulated more hormonal profiles and OGTT in polycystic ovary syndrome (PCOS) with no familial diabetes, ALA improved insulin response to OGTT and metabolic parameters in all patients with no effects on reproductive hormones, MYO + ALA improved hormonal and metabolic aspects and insulin response to OGTT in all patients. Presence of familial diabetes is a relevant clinical aspect. MYO is less effective when familial diabetes is present, ALA improved only metabolic aspects while MYO + ALA was effective on all PCOS patients independently from familial diabetes.
Collapse
Affiliation(s)
- Alessandro D Genazzani
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessia Prati
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Marchini
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Tabatha Petrillo
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonella Napolitano
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Modena, Italy
| | - Tommaso Simoncini
- Department of Gynecology and Obstetrics, University of Pisa, Pisa, Italy
| |
Collapse
|
23
|
Shokrpour M, Foroozanfard F, Afshar Ebrahimi F, Vahedpoor Z, Aghadavod E, Ghaderi A, Asemi Z. Comparison of myo-inositol and metformin on glycemic control, lipid profiles, and gene expression related to insulin and lipid metabolism in women with polycystic ovary syndrome: a randomized controlled clinical trial. Gynecol Endocrinol 2019; 35:406-411. [PMID: 30608001 DOI: 10.1080/09513590.2018.1540570] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
This investigation was conducted to evaluate comparison of myo-inositol and metformin on glycemic control, lipid profiles, and gene expression related to insulin and lipid metabolism in women with polycystic ovary syndrome (PCOS). This randomized controlled trial was conducted on 53 women with PCOS, aged 18-40 years old. Subjects were randomly allocated into two groups to take either myo-inositol (n = 26) or metformin (n = 27) for 12 weeks. Myo-inositol supplementation, compared with metformin, significantly reduced fasting plasma glucose (FPG) (β -5.12 mg/dL; 95% CI, -8.09, -2.16; p=.001), serum insulin levels (β -1.49 µIU/mL; 95% CI, -2.28, -0.70; p<.001), homeostasis model of assessment-insulin resistance (β -0.36; 95% CI, -0.55, -0.17; p<.001), serum triglycerides (β 12.42 mg/dL; 95% CI, -20.47, -4.37; p=.003) and VLDL-cholesterol levels (β -2.48 mg/dL; 95% CI, -4.09, -0.87; p=.003), and significantly increased the quantitative insulin sensitivity check index (β 0.006; 95% CI, 0.002, 0.01; p=.006) compared with metformin. Moreover, myo-inositol supplementation upregulated gene expression of peroxisome proliferator-activated receptor gamma (PPAR-γ) (p=.002) compared with metformin. Overall, taking myo-inositol, compared with metformin, for 12 weeks by women with PCOS had beneficial effects on glycemic control, triglycerides and VLDL-cholesterol levels, and gene expression of PPAR-γ.
Collapse
Affiliation(s)
- Maryam Shokrpour
- a Department of Gynecology and Obstetrics, School of Medicine , Endocrinology and Metabolism Research Center, Arak University of Medical Sciences , Arak , Iran
| | - Fatemeh Foroozanfard
- b Department of Gynecology and Obstetrics, School of Medicine , Kashan University of Medical Sciences , Kashan , Iran
| | - Faraneh Afshar Ebrahimi
- b Department of Gynecology and Obstetrics, School of Medicine , Kashan University of Medical Sciences , Kashan , Iran
| | - Zahra Vahedpoor
- b Department of Gynecology and Obstetrics, School of Medicine , Kashan University of Medical Sciences , Kashan , Iran
| | - Esmat Aghadavod
- c Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences , Kashan , Iran
| | - Amir Ghaderi
- d Department of Addiction Studies, School of Medicine , Kashan University of Medical Sciences , Kashan , Iran
| | - Zatollah Asemi
- c Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences , Kashan , Iran
| |
Collapse
|
24
|
Facchinetti F, Orrù B, Grandi G, Unfer V. Short-term effects of metformin and myo-inositol in women with polycystic ovarian syndrome (PCOS): a meta-analysis of randomized clinical trials. Gynecol Endocrinol 2019; 35:198-206. [PMID: 30614282 DOI: 10.1080/09513590.2018.1540578] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Metformin (MET), the most commonly used insulin sensitizer, is the reference off-label drug for the treatment of polycystic ovary syndrome (PCOS), worldwide. However, its use may be limited mainly by gastrointestinal adverse effects. Myo-inositol (MI), a well-recognized food supplement, also represents an evidence-based treatment for PCOS women, popular in many countries. Our aim is to provide a systematic review of the literature and a meta-analysis which compares these two treatments, for their short-term efficacy and safety in PCOS patients. Systematic review and meta-analysis of randomized clinical trials (RCTs). RCTs were identified from 1994 through 2017 using MEDLINE, Cochrane Library, PubMed, and ResearchGate. Included studies were limited to those one directly comparing MET to MI on several hormones changes. Standardized mean difference (SMD) or risk ratios (RRs) with 95% CIs were calculated. Changes in fasting insulin was the main outcome of measure. Six trials with a total of 355 patients were included. At the end of treatment, no difference between MET and MI was found on fasting insulin (SMD=0.08 µU/ml, 95% CI: -0.31-0.46, p=.697), HOMA index (SMD =0.17, 95% CI: -0.53-0.88, p=.635), testosterone (SMD= -0.01, 95% CI: -0.24-0.21, p=.922), SHBG levels (SMD= -0.50 nmol/l, 95% CI: -1.39-0.38, p=.263) and body mass index (BMI) (SMD= -0.22, 95% CI: -0.60-0.16, p=.265). There was strong evidence of an increased risk of adverse events among women receiving MET compared to those receiving MI (RR =5.17, 95% CI: 2.91-9.17, p<.001). No differences were found in the effect of MET and MI on short-term hormone changes. The better tolerability of MI makes it more acceptable for the recovery of androgenic and metabolic profile in PCOS women.
Collapse
Affiliation(s)
- Fabio Facchinetti
- a Department of Medical and Surgical Sciences for Mother, Child and Adult , Azienda Ospedaliero Universitaria Policlinico, University of Modena and Reggio Emilia , Modena , Italy
| | - Beatrice Orrù
- b Department of Medical Affairs , Lo.Li. Pharma , Rome , Italy
| | - Giovanni Grandi
- a Department of Medical and Surgical Sciences for Mother, Child and Adult , Azienda Ospedaliero Universitaria Policlinico, University of Modena and Reggio Emilia , Modena , Italy
| | - Vittorio Unfer
- c Department of Developmental and Social Psychology, Faculty of Medicine and Psychology, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
25
|
Pourghasem S, Bazarganipour F, Taghavi SA, Kutenaee MA. The effectiveness of inositol and metformin on infertile polycystic ovary syndrome women with resistant to letrozole. Arch Gynecol Obstet 2019; 299:1193-1199. [PMID: 30847561 DOI: 10.1007/s00404-019-05064-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 01/24/2019] [Indexed: 11/26/2022]
Abstract
PURPOSE The purpose is a comparison of effectiveness of myo-inositol and metformin in infertile women with polycystic ovary syndrome (PCOS) treated with letrozole. METHODS This study is a randomized single-blind controlled clinical trial undertaken in 150 infertile PCOS women. For all patients, letrozole is prescribed at a dose of 7.5 mg per day from the third day of menstruation for 5 days. Patients who did not ovulate were included and divided into three pretreatment groups: group I(control group), 200 µg of folic acid (as a placebo); group II, 1500 mg of metformin daily plus 200 µg of folic acid, and group III, inositol 2 g plus 200 µg of folic acid received twice daily for 3 months. In the last cycle, 7.5 mg letrozole was prescribed for the induction of ovulation. Primary outcomes were ovary function and pregnancy. RESULTS The ovarian function was not significantly different in those groups, whereas the ovarian function of inositol + folic acid group in normal BMI found significantly higher than other BMI spectra. In addition, the ovarian function is significantly higher in the inositol + folic acid group by increasing the infertility duration. The incidence of pregnancy is lower in letrozole + folic acid + inositol group than the other groups; however, it is not significant. CONCLUSION The addition of inositol and metformin to the treatment of infertile PCOS women with letrozole resistance improves the ovarian function; however, it is not significant. Of note, inositol was more effective than metformin in patients with normal BMI. IRCT REGISTRATION NUMBER IRCT2017070234845N1.
Collapse
Affiliation(s)
- Sajadeh Pourghasem
- Mother and Child Welfare Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Bazarganipour
- Midwifery Department, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Seyed Abdolvahab Taghavi
- Gynecologic and Obstetrics Department, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Maryam Azizi Kutenaee
- Fertility and Infertility Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
26
|
Landomiel F, De Pascali F, Raynaud P, Jean-Alphonse F, Yvinec R, Pellissier LP, Bozon V, Bruneau G, Crépieux P, Poupon A, Reiter E. Biased Signaling and Allosteric Modulation at the FSHR. Front Endocrinol (Lausanne) 2019; 10:148. [PMID: 30930853 PMCID: PMC6425863 DOI: 10.3389/fendo.2019.00148] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Knowledge on G protein-coupled receptor (GPCRs) structure and mechanism of activation has profoundly evolved over the past years. The way drugs targeting this family of receptors are discovered and used has also changed. Ligands appear to bind a growing number of GPCRs in a competitive or allosteric manner to elicit balanced signaling or biased signaling (i.e., differential efficacy in activating or inhibiting selective signaling pathway(s) compared to the reference ligand). These novel concepts and developments transform our understanding of the follicle-stimulating hormone (FSH) receptor (FSHR) biology and the way it could be pharmacologically modulated in the future. The FSHR is expressed in somatic cells of the gonads and plays a major role in reproduction. When compared to classical GPCRs, the FSHR exhibits intrinsic peculiarities, such as a very large NH2-terminal extracellular domain that binds a naturally heterogeneous, large heterodimeric glycoprotein, namely FSH. Once activated, the FSHR couples to Gαs and, in some instances, to other Gα subunits. G protein-coupled receptor kinases and β-arrestins are also recruited to this receptor and account for its desensitization, trafficking, and intracellular signaling. Different classes of pharmacological tools capable of biasing FSHR signaling have been reported and open promising prospects both in basic research and for therapeutic applications. Here we provide an updated review of the most salient peculiarities of FSHR signaling and its selective modulation.
Collapse
|
27
|
Abstract
The glycoprotein follicle-stimulating hormone (FSH) acts on gonadal target cells, hence regulating gametogenesis. The transduction of the hormone-induced signal is mediated by the FSH-specific G protein-coupled receptor (FSHR), of which the action relies on the interaction with a number of intracellular effectors. The stimulatory Gαs protein is a long-time known transducer of FSH signaling, mainly leading to intracellular cAMP increase and protein kinase A (PKA) activation, the latter acting as a master regulator of cell metabolism and sex steroid production. While in vivo data clearly demonstrate the relevance of PKA activation in mediating gametogenesis by triggering proliferative signals, some in vitro data suggest that pro-apoptotic pathways may be awakened as a "dark side" of cAMP/PKA-dependent steroidogenesis, in certain conditions. P38 mitogen-activated protein kinases (MAPK) are players of death signals in steroidogenic cells, involving downstream p53 and caspases. Although it could be hypothesized that pro-apoptotic signals, if relevant, may be required for regulating atresia of non-dominant ovarian follicles, they should be transient and counterbalanced by mitogenic signals upon FSHR interaction with opposing transducers, such as Gαi proteins and β-arrestins. These molecules modulate the steroidogenic pathway via extracellular-regulated kinases (ERK1/2), phosphatidylinositol-4,5-bisphosphate 3-kinases (PI3K)/protein kinase B (AKT), calcium signaling and other intracellular signaling effectors, resulting in a complex and dynamic signaling network characterizing sex- and stage-specific gamete maturation. Even if the FSH-mediated signaling network is not yet entirely deciphered, its full comprehension is of high physiological and clinical relevance due to the crucial role covered by the hormone in regulating human development and reproduction.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Department Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
- *Correspondence: Livio Casarini
| | - Pascale Crépieux
- PRC, UMR INRA0085, CNRS 7247, Centre INRA Val de Loire, Nouzilly, France
| |
Collapse
|
28
|
Lindgren I, Nenonen H, Henic E, Bungum L, Prahl A, Bungum M, Leijonhufvud I, Huhtaniemi I, Yding Andersen C, Lundberg Giwercman Y. Gonadotropin receptor variants are linked to cumulative live birth rate after in vitro fertilization. J Assist Reprod Genet 2018; 36:29-38. [PMID: 30232643 PMCID: PMC6338601 DOI: 10.1007/s10815-018-1318-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 01/06/2023] Open
Abstract
Purpose The objective was to investigate if the gonadotropin receptor variants N680S (N: asparagine, S: serine, rs6166) in the follicle-stimulating hormone receptor (FSHR) and N312S (rs2293275) in the luteinizing hormone/human chorionic gonadotropin receptor (LHCGR) predicted cumulative live birth rate after in vitro fertilization (IVF). Methods A total of 665 women were consecutively enrolled for IVF during the period 2007–2016. Inclusion criteria were < 40 years of age, body mass index < 30 kg/m2, non-smoking, regular menstruation cycle of 21–35 days, and bilateral ovaries. A blood sample was drawn for endocrine hormonal analysis and for DNA extraction with subsequent genotyping of the FSHR N680S and LHCGR N312S polymorphisms. Statistical analyses were done on all completed IVF cycles. Results Women homozygous for S in both receptors combined (4S) had significantly higher live birth rate compared to those with other receptor variants when combining the first three IVF cycles (OR = 2.00, 95% CI [1.02, 3.92], p = 0.043). Cumulatively higher chance of live birth rate, during all IVF cycles, was also evident (HR = 1.89, 95% CI [1.00, 3.57], p = 0.049). Conclusions Gonadotropin receptor variants are promising candidates for the prediction of the possibility to have a baby to take home after IVF treatment.
Collapse
Affiliation(s)
- I. Lindgren
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - H. Nenonen
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - E. Henic
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - L. Bungum
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - A. Prahl
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| | - M. Bungum
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - I. Leijonhufvud
- Reproductive Medicine Centre, Skåne University Hospital, JanWaldenströms gata 47, Plan 3, SE 21428 Malmö, Sweden
| | - I. Huhtaniemi
- Hammersmith Campus, Institute of Reproductive and Developmental Biology, Imperial College London, London, SW7 2AZ UK
| | - C. Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Y. Lundberg Giwercman
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 21428 Malmö, Sweden
| |
Collapse
|
29
|
Ulloa-Aguirre A, Reiter E, Crépieux P. FSH Receptor Signaling: Complexity of Interactions and Signal Diversity. Endocrinology 2018; 159:3020-3035. [PMID: 29982321 DOI: 10.1210/en.2018-00452] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
FSH is synthesized in the pituitary by gonadotrope cells. By binding to and interacting with its cognate receptor [FSH receptor (FSHR)] in the gonads, this gonadotropin plays a key role in the control of gonadal function and reproduction. Upon activation, the FSHR undergoes conformational changes leading to transduction of intracellular signals, including dissociation of G protein complexes into components and activation of several associated interacting partners, which concertedly regulate downstream effectors. The canonical Gs/cAMP/protein kinase A pathway, considered for a long time as the sole effector of FSHR-mediated signaling, is now viewed as one of several mechanisms employed by this receptor to transduce intracellular signals in response to the FSH stimulus. This complex network of signaling pathways allows for a fine-tuning regulation of the gonadotropic stimulus, where activation/inhibition of its multiple components vary depending on the cell context, cell developmental stage, and concentration of associated receptors and corresponding ligands. Activation of these multiple signaling modules eventually converge to the hormone-integrated biological response, including survival, proliferation and differentiation of target cells, synthesis and secretion of paracrine/autocrine regulators, and, at the molecular level, functional selectivity and differential gene expression. In this mini-review, we discuss the complexity of FSHR-mediated intracellular signals activated in response to ligand stimulation. A better understanding of the signaling pathways involved in FSH action might potentially influence the development of new therapeutic strategies for reproductive disorders.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eric Reiter
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| | - Pascale Crépieux
- Biology and Bioinformatics of Signaling Systems Group, Unité Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Institut National de la Recherche Agronomique, Nouzilly, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7247, Nouzilly, France
- Université François Rabelais, Nouzilly, France
| |
Collapse
|
30
|
Zhu D, Li X, Macrae VE, Simoncini T, Fu X. Extragonadal Effects of Follicle-Stimulating Hormone on Osteoporosis and Cardiovascular Disease in Women during Menopausal Transition. Trends Endocrinol Metab 2018; 29:571-580. [PMID: 29983231 DOI: 10.1016/j.tem.2018.06.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/03/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023]
Abstract
The risk of osteoporosis and cardiovascular disease increases significantly in postmenopausal women. Until recently, the underlying mechanisms have been primarily attributed to estrogen decline following menopause. However, follicle-stimulating hormone (FSH) levels rise sharply during menopausal transition and are maintained at elevated levels for many years. FSH receptor has been detected in various extragonadal sites, including osteoclasts and endothelial cells. Recent advances suggest FSH may contribute to postmenopausal osteoporosis and cardiovascular disease. Here, we review the key actions through which FSH contributes to the risk of osteoporosis and cardiovascular disease in women as they transition through menopause. Advancing our understanding of the precise mechanisms through which FSH promotes osteoporosis and cardiovascular disease may provide new opportunities for improving health-span for postmenopausal women.
Collapse
Affiliation(s)
- Dongxing Zhu
- Guangzhou Institute of Cardiovascular Diseases, The Second Affiliated Hospital; Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaosa Li
- Guangzhou Institute of Cardiovascular Diseases, The Second Affiliated Hospital; Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Vicky E Macrae
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK.
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Reproductive Medicine and Child Development, University of Pisa, Pisa 56100, Italy.
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Diseases, The Second Affiliated Hospital; Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
31
|
Integration of GPCR Signaling and Sorting from Very Early Endosomes via Opposing APPL1 Mechanisms. Cell Rep 2018; 21:2855-2867. [PMID: 29212031 PMCID: PMC5732320 DOI: 10.1016/j.celrep.2017.11.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/18/2017] [Accepted: 11/03/2017] [Indexed: 01/10/2023] Open
Abstract
Endocytic trafficking is a critical mechanism for cells to decode complex signaling pathways, including those activated by G-protein-coupled receptors (GPCRs). Heterogeneity in the endosomal network enables GPCR activity to be spatially restricted between early endosomes (EEs) and the recently discovered endosomal compartment, the very early endosome (VEE). However, the molecular machinery driving GPCR activity from the VEE is unknown. Using luteinizing hormone receptor (LHR) as a prototype GPCR for this compartment, along with additional VEE-localized GPCRs, we identify a role for the adaptor protein APPL1 in rapid recycling and endosomal cAMP signaling without impacting the EE-localized β2-adrenergic receptor. LHR recycling is driven by receptor-mediated Gαs/cAMP signaling from the VEE and PKA-dependent phosphorylation of APPL1 at serine 410. Receptor/Gαs endosomal signaling is localized to microdomains of heterogeneous VEE populations and regulated by APPL1 phosphorylation. Our study uncovers a highly integrated inter-endosomal communication system enabling cells to tightly regulate spatially encoded signaling. GPCRs that internalize to very early endosomes (VEEs) require APPL1 to recycle Receptor recycling is driven by cAMP/PKA to phosphorylate serine 410 on APPL1 cAMP signaling from GPCRs, such as LHR, occurs from distinct VEE microdomains APPL1 limits VEE cAMP signaling via opposing mechanisms required for GPCR sorting
Collapse
|
32
|
Nataraja S, Sriraman V, Palmer S. Allosteric Regulation of the Follicle-Stimulating Hormone Receptor. Endocrinology 2018; 159:2704-2716. [PMID: 29800292 DOI: 10.1210/en.2018-00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/16/2018] [Indexed: 01/08/2023]
Abstract
Follicle-stimulating hormone receptor (FSHR) belongs to the leucine-rich repeat family of the G protein-coupled receptor (LGR), which includes the glycoprotein hormone receptors luteinizing hormone receptor, thyrotropin receptor, and other LGRs 4, 5, 6, and 7. FSH is the key regulator of folliculogenesis in females and spermatogenesis in males. FSH elicits its physiological response through its cognate receptor on the cell surface. Binding of the hormone FSH to its receptor FSHR brings about conformational changes in the receptor that are transduced through the transmembrane domain to the intracellular region, where the downstream effector interaction takes place, leading to activation of the downstream signaling cascade. Identification of small molecules that could activate or antagonize FSHR provided interesting tools to study the signal transduction mechanism of the receptor. However, because of the nature of the ligand-receptor interaction of FSH-FSHR, which contains multiple sites in the extracellular binding domain, most of the small-molecule modulators of FSHR are unable to bind to the orthosteric site of the receptors. Rather they modulate receptor activation through allosteric sites in the transmembrane region. This review will discuss allosteric modulation of FSHR primarily through the discovery of small-molecule modulators, focusing on current data on the status of development and the utility of these as tools to better understand signaling mechanisms.
Collapse
|
33
|
English EJ, Mahn SA, Marchese A. Endocytosis is required for C XC chemokine receptor type 4 (CXCR4)-mediated Akt activation and antiapoptotic signaling. J Biol Chem 2018; 293:11470-11480. [PMID: 29899118 DOI: 10.1074/jbc.ra118.001872] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/07/2018] [Indexed: 11/06/2022] Open
Abstract
Signaling activated by binding of the CXC motif chemokine ligand 12 (CXCL12) to its cognate G protein-coupled receptor (GPCR), chemokine CXC motif receptor 4 (CXCR4), is linked to metastatic disease. However, the mechanisms governing CXCR4 signaling remain poorly understood. Here, we show that endocytosis and early endosome antigen 1 (EEA1), which is part of the endosome fusion machinery, are required for CXCL12-mediated AKT Ser/Thr kinase (Akt) signaling selective for certain Akt substrates. Pharmacological inhibition of endocytosis partially attenuated CXCL12-induced phosphorylation of Akt, but not phosphorylation of ERK-1/2. Similarly, phosphorylation of Akt, but not ERK-1/2, stimulated by CXCL13, the cognate ligand for the chemokine receptor CXCR5, was also attenuated by inhibited endocytosis. Furthermore, siRNA-mediated depletion of the Rab5-effector EEA1, but not of adaptor protein, phosphotyrosine-interacting with PH domain and leucine zipper 1 (APPL1), partially attenuated Akt, but not ERK-1/2, phosphorylation promoted by CXCR4. Attenuation of Akt phosphorylation through inhibition of endocytosis or EEA1 depletion was associated with reduced signaling to Akt substrate forkhead box O1/3a but not the Akt substrates TSC complex subunit 2 or glycogen synthase kinase 3β. This suggested that endocytosis and endosomes govern discrete aspects of CXCR4- or CXCR5-mediated Akt signaling. Consistent with this hypothesis, depletion of EEA1 reduced the ability of CXCL12 to attenuate apoptosis in suspended, but not adherent, HeLa cells. Our results suggest a mechanism whereby compartmentalized chemokine-mediated Akt signaling from endosomes suppresses the cancer-related process known as anoikis. Targeting this signaling pathway may help inhibit metastatic cancer involving receptors such as CXCR4.
Collapse
Affiliation(s)
- Elizabeth J English
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Sarah A Mahn
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|
34
|
Ben-Menahem D. Preparation, characterization and application of long-acting FSH analogs for assisted reproduction. Theriogenology 2018; 112:11-17. [DOI: 10.1016/j.theriogenology.2017.08.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 08/02/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022]
|
35
|
Genazzani AD, Shefer K, Della Casa D, Prati A, Napolitano A, Manzo A, Despini G, Simoncini T. Modulatory effects of alpha-lipoic acid (ALA) administration on insulin sensitivity in obese PCOS patients. J Endocrinol Invest 2018; 41:583-590. [PMID: 29090431 DOI: 10.1007/s40618-017-0782-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/22/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE To evaluate the efficacy of alpha-lipoic acid (ALA) administration on hormonal and metabolic parameters of obese PCOS patients. METHODS A group of 32 obese PCOS patients were selected after informed consent. 20 patients referred to have first grade relatives with diabetes type I or II. Hormonal and metabolic parameters as well as OGTT were evaluated before and after 12 weeks of ALA integrative administration (400 mg per os every day). RESULTS ALA administration significantly decreased insulin, glucose, BMI and HOMA index. Hyperinsulinemia and insulin response to OGTT decreased both as maximal response (Δmax) and as AUC. PCOS with diabetes relatives showed the decrease also of triglyceride and GOT. Interestingly in all PCOS no changes occurred on all hormonal parameters involved in reproduction such as LH, FSH, and androstenedione. CONCLUSIONS ALA integrative administration at a low dosage as 400 mg daily improved the metabolic impairment of all PCOS patients especially in those PCOS with familiar diabetes who have a higher grade of risk of NAFLD and predisposition to diabetes.
Collapse
Affiliation(s)
- A D Genazzani
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy.
| | - K Shefer
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - D Della Casa
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - A Prati
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - A Napolitano
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - A Manzo
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - G Despini
- Department of Obstetrics and Gynecology, Gynecological Endocrinology Center, University of Modena and Reggio Emilia, Via del Pozzo 71, 41100, Modena, Italy
| | - T Simoncini
- Department of Obstetrics and Gynecology, University of Pisa, Pisa, Italy
| |
Collapse
|
36
|
Das N, Kumar TR. Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol 2018; 60:R131-R155. [PMID: 29437880 PMCID: PMC5851872 DOI: 10.1530/jme-17-0308] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022]
Abstract
Follicle-stimulating hormone (FSH) plays fundamental roles in male and female fertility. FSH is a heterodimeric glycoprotein expressed by gonadotrophs in the anterior pituitary. The hormone-specific FSHβ-subunit is non-covalently associated with the common α-subunit that is also present in the luteinizing hormone (LH), another gonadotrophic hormone secreted by gonadotrophs and thyroid-stimulating hormone (TSH) secreted by thyrotrophs. Several decades of research led to the purification, structural characterization and physiological regulation of FSH in a variety of species including humans. With the advent of molecular tools, availability of immortalized gonadotroph cell lines and genetically modified mouse models, our knowledge on molecular mechanisms of FSH regulation has tremendously expanded. Several key players that regulate FSH synthesis, sorting, secretion and action in gonads and extragonadal tissues have been identified in a physiological setting. Novel post-transcriptional and post-translational regulatory mechanisms have also been identified that provide additional layers of regulation mediating FSH homeostasis. Recombinant human FSH analogs hold promise for a variety of clinical applications, whereas blocking antibodies against FSH may prove efficacious for preventing age-dependent bone loss and adiposity. It is anticipated that several exciting new discoveries uncovering all aspects of FSH biology will soon be forthcoming.
Collapse
Affiliation(s)
- Nandana Das
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
| | - T. Rajendra Kumar
- Division of Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Division of Reproductive Endocrinology and Infertility, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, U.S.A
- Author for Correspondence: T. Rajendra Kumar, PhD, Edgar L. and Patricia M. Makowski Professor, Associate Vice-Chair of Research, Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Mail Stop 8613, Research Complex 2, Room # 15-3000B, 12700 E. 19th Avenue, Aurora, CO 80045, USA, Tel: 303-724-8689,
| |
Collapse
|
37
|
Sayers N, Hanyaloglu AC. Intracellular Follicle-Stimulating Hormone Receptor Trafficking and Signaling. Front Endocrinol (Lausanne) 2018; 9:653. [PMID: 30450081 PMCID: PMC6225286 DOI: 10.3389/fendo.2018.00653] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/17/2018] [Indexed: 12/18/2022] Open
Abstract
Models of G protein-coupled receptor (GPCR) signaling have dramatically altered over the past two decades. Indeed, GPCRs such as the follicle-stimulating hormone receptor (FSHR) have contributed to these new emerging models. We now understand that receptor signaling is highly organized at a spatial level, whereby signaling not only occurs from the plasma membrane but distinct intracellular compartments. Recent studies in the role of membrane trafficking and spatial organization of GPCR signaling in regulating gonadotropin hormone receptor activity has identified novel intracellular compartments, which are tightly linked with receptor signaling and reciprocally regulated by the cellular trafficking machinery. Understanding the impact of these cell biological mechanisms to physiology and pathophysiology is emerging for certain GPCRs. However, for FSHR, the potential impact in both health and disease and the therapeutic possibilities of these newly identified systems is currently unknown, but offers the potential to reassess prior strategies, or unveil novel opportunities, in targeting this receptor.
Collapse
|
38
|
Ulloa-Aguirre A, Zariñán T, Jardón-Valadez E, Gutiérrez-Sagal R, Dias JA. Structure-Function Relationships of the Follicle-Stimulating Hormone Receptor. Front Endocrinol (Lausanne) 2018; 9:707. [PMID: 30555414 PMCID: PMC6281744 DOI: 10.3389/fendo.2018.00707] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
The follicle-stimulating hormone receptor (FSHR) plays a crucial role in reproduction. This structurally complex receptor is a member of the G-protein coupled receptor (GPCR) superfamily of membrane receptors. As with the other structurally similar glycoprotein hormone receptors (the thyroid-stimulating hormone and luteinizing hormone-chorionic gonadotropin hormone receptors), the FSHR is characterized by an extensive extracellular domain, where binding to FSH occurs, linked to the signal specificity subdomain or hinge region. This region is involved in ligand-stimulated receptor activation whereas the seven transmembrane domain is associated with receptor activation and transmission of the activation process to the intracellular loops comprised of amino acid sequences, which predicate coupling to effectors, interaction with adapter proteins, and triggering of downstream intracellular signaling. In this review, we describe the most important structural features of the FSHR intimately involved in regulation of FSHR function, including trafficking, dimerization, and oligomerization, ligand binding, agonist-stimulated activation, and signal transduction.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Alfredo Ulloa-Aguirre
| | - Teresa Zariñán
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Eduardo Jardón-Valadez
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana Unidad Lerma, Lerma, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México and Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - James A. Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, United States
| |
Collapse
|
39
|
Jamilian M, Farhat P, Foroozanfard F, Afshar Ebrahimi F, Aghadavod E, Bahmani F, Badehnoosh B, Jamilian H, Asemi Z. Comparison of myo-inositol and metformin on clinical, metabolic and genetic parameters in polycystic ovary syndrome: A randomized controlled clinical trial. Clin Endocrinol (Oxf) 2017; 87:194-200. [PMID: 28485095 DOI: 10.1111/cen.13366] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/09/2017] [Accepted: 04/27/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To our knowledge, data on comparison of myo-inositol and metformin on clinical, metabolic and genetic parameters in subjects with polycystic ovary syndrome (PCOS) are limited. This study was carried out to compare myo-inositol and metformin on clinical, metabolic and genetic parameters in subjects with PCOS. DESIGN, PATIENTS AND MEASUREMENTS This randomized controlled trial was conducted among 60 subjects with PCOS aged 18-40 years. Subjects were randomly allocated into two groups to receive either myo-inositol (N=30) or metformin (N=30) for 12 weeks. Gene expression of inflammatory cytokines was assessed in peripheral blood mononuclear cells (PBMCs) of PCOS women by RT-PCR. RESULTS After the 12-week intervention, compared with metformin, myo-inositol intake significantly decreased serum total testosterone (-1.4±4.2 vs +0.7±1.4 nmol/L, P=.03), modified Ferriman-Gallwey (mF-G) scores (-1.1±0.7 vs -0.5±0.8, P=.01) and serum high-sensitivity C-reactive protein (hs-CRP) levels (-2.6±3.9 vs +0.2±1.5 mg/L, P<.001). RT-PCR demonstrated that compared with metformin, myo-inositol downregulated gene expression of interleukin-1 (IL-1) (P=.02) in PBMCs of subjects with PCOS. We did not observe any significant effect of myo-inositol intake compared with metformin on other hormonal profiles, plasma nitric oxide (NO) or gene expression of IL-8 and tumour necrosis factor alpha (TNF-α). CONCLUSIONS Overall, taking myo-inositol, compared with metformin, for 12 weeks in patients with PCOS with hyperinsulinism and normoinsulinism had beneficial effects on total testosterone, mFG scores, serum hs-CRP levels and gene expression of IL-1, but did not affect other hormonal profiles, NO levels or gene expression of IL-8 and TNF-α.
Collapse
Affiliation(s)
- Mehri Jamilian
- Endocrinology and Metabolism Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Pegah Farhat
- Endocrinology and Metabolism Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Fatemeh Foroozanfard
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Faraneh Afshar Ebrahimi
- Department of Gynecology and Obstetrics, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Fereshteh Bahmani
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Bita Badehnoosh
- Department of Gynecology and Obstetrics, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamidreza Jamilian
- Department of Psychiatry, Arak University of Medical Sciences, Arak, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
40
|
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: 8.0] [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
|
41
|
|
42
|
Genazzani AD. Inositol as putative integrative treatment for PCOS. Reprod Biomed Online 2016; 33:770-780. [DOI: 10.1016/j.rbmo.2016.08.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 01/28/2023]
|
43
|
Stelmaszewska J, Chrusciel M, Doroszko M, Akerfelt M, Ponikwicka-Tyszko D, Nees M, Frentsch M, Li X, Kero J, Huhtaniemi I, Wolczynski S, Rahman NA. Revisiting the expression and function of follicle-stimulation hormone receptor in human umbilical vein endothelial cells. Sci Rep 2016; 6:37095. [PMID: 27848975 PMCID: PMC5111068 DOI: 10.1038/srep37095] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/21/2016] [Indexed: 12/29/2022] Open
Abstract
Expression of follicle-stimulation hormone receptor (FSHR) is confined to gonads and at low levels to some extragonadal tissues like human umbilical vein endothelial cells (HUVEC). FSH-FSHR signaling was shown to promote HUVEC angiogenesis and thereafter suggested to have an influential role in pregnancy. We revisited hereby the expression and functionality of FSHR in HUVECs angiogenesis, and were unable to reproduce the FSHR expression in human umbilical cord, HUVECs or immortalized HUVECs (HUV-ST). Positive controls as granulosa cells and HEK293 cells stably transfected with human FSHR cDNA expressed FSHR signal. In contrast to positive control VEGF, FSH treatment showed no effects on tube formation, nitric oxide production, wound healing or cell proliferation in HUVEC/HUV-ST. Thus, it remains open whether the FSH-FSHR activation has a direct regulatory role in the angiogenesis of HUVECs.
Collapse
Affiliation(s)
- Joanna Stelmaszewska
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, 15276 Bialystok, Poland
| | - Marcin Chrusciel
- Institute of Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland.,Department of Biology and Pathology of Human Reproduction, Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10714 Olsztyn, Poland
| | - Milena Doroszko
- Institute of Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland
| | - Malin Akerfelt
- Institute of Biomedicine, Department of Cell Biology and Anatomy, University of Turku, 20520 Turku, Finland
| | - Donata Ponikwicka-Tyszko
- Department of Biology and Pathology of Human Reproduction, Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10714 Olsztyn, Poland
| | - Matthias Nees
- Institute of Biomedicine, Department of Cell Biology and Anatomy, University of Turku, 20520 Turku, Finland
| | - Marco Frentsch
- Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies, CVK Charité University Medicine, 13353 Berlin, Germany
| | - Xiangdong Li
- State Key Lab for Agrobiotechnology, China Agriculture University, 100193 Beijing, China
| | - Jukka Kero
- Institute of Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland
| | - Ilpo Huhtaniemi
- Institute of Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland.,Institute of Reproductive and Developmental Biology, Imperial College London, W12 ONN London, UK
| | - Slawomir Wolczynski
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, 15276 Bialystok, Poland.,Department of Biology and Pathology of Human Reproduction, Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10714 Olsztyn, Poland
| | - Nafis A Rahman
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, 15276 Bialystok, Poland.,Institute of Biomedicine, Department of Physiology, University of Turku, 20520 Turku, Finland
| |
Collapse
|
44
|
Bevilacqua A, Bizzarri M. Physiological role and clinical utility of inositols in polycystic ovary syndrome. Best Pract Res Clin Obstet Gynaecol 2016; 37:129-139. [DOI: 10.1016/j.bpobgyn.2016.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/09/2016] [Indexed: 11/16/2022]
|
45
|
|
46
|
Sacchi S, Marinaro F, Tondelli D, Lui J, Xella S, Marsella T, Tagliasacchi D, Argento C, Tirelli A, Giulini S, La Marca A. Modulation of gonadotrophin induced steroidogenic enzymes in granulosa cells by d-chiroinositol. Reprod Biol Endocrinol 2016; 14:52. [PMID: 27582109 PMCID: PMC5006365 DOI: 10.1186/s12958-016-0189-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/23/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND d-chiroinositol (DCI) is a inositolphosphoglycan (IPG) involved in several cellular functions that control the glucose metabolism. DCI functions as second messenger in the insulin signaling pathway and it is considered an insulin sensitizer since deficiency in tissue availability of DCI were shown to cause insulin resistance (IR). Polycystic ovary syndrome (PCOS) is a pathological condition that is often accompanied with insulin resistance. DCI can positively affects several aspect of PCOS etiology decreasing the total and free testosterone, lowering blood pressure, improving the glucose metabolism and increasing the ovulation frequency. The purpose of this study was to evaluate the effects of DCI and insulin combined with gonadotrophins namely follicle-stimulating hormone (FSH) and luteinizing hormone (LH) on key steroidogenic enzymes genes regulation, cytochrome P450 family 19 subfamily A member 1 (CYP19A1) and cytochrome P450 side-chain cleavage (P450scc) in primary cultures of human granulosa cells (hGCs). We also investigated whether DCI, being an insulin-sensitizer would be able to counteract the expected stimulator activity of insulin on human granulosa cells (hGCs). METHODS The study was conducted on primary cultures of hGCs. Gene expression was evaluated by RT-qPCR method. Statistical analysis was performed applying student t-test, as appropriate (P < 0.05) set for statistical significance. RESULTS DCI is able to reduce the gene expression of CYP19A1, P450scc and insulin-like growth factor 1 receptor (IGF-1R) in dose-response manner. The presence of DCI impaired the increased expression of steroidogenic enzyme genes generated by the insulin treatment in gonadotrophin-stimulated hGCs. CONCLUSIONS Insulin acts as co-gonadotrophin increasing the expression of steroidogenic enzymes genes in gonadotrophin-stimulated granulosa cells. DCI is an insulin-sensitizer that counteracts this action by reducing the expression of the genes CYP19A1, P450scc and IGF-1R. The ability of DCI to modulate in vitro ovarian activity of insulin could in part explain its beneficial effect when used as treatment for conditions associated to insulin resistance.
Collapse
Affiliation(s)
- Sandro Sacchi
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Federica Marinaro
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Debora Tondelli
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Jessica Lui
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Susanna Xella
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Tiziana Marsella
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Daniela Tagliasacchi
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Cindy Argento
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Alessandra Tirelli
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Simone Giulini
- Mother-Infant Department, University of Modena and Reggio Emilia, Via del pozzo 41, 41100 Modena, Italy
| | - Antonio La Marca
- University of Modena and Reggio Emilia and Clinica Eugin Modena, Modena, Italy
| |
Collapse
|
47
|
Ulloa-Aguirre A, Zariñán T. The Follitropin Receptor: Matching Structure and Function. Mol Pharmacol 2016; 90:596-608. [DOI: 10.1124/mol.116.104398] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/28/2016] [Indexed: 12/19/2022] Open
|
48
|
Stilley JAW, Guan R, Santillan DA, Mitchell BF, Lamping KG, Segaloff DL. Differential Regulation of Human and Mouse Myometrial Contractile Activity by FSH as a Function of FSH Receptor Density. Biol Reprod 2016; 95:36. [PMID: 27335068 PMCID: PMC5029472 DOI: 10.1095/biolreprod.116.141648] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/15/2016] [Indexed: 01/05/2023] Open
Abstract
Previous studies from our laboratory revealed that the follicle-stimulating hormone receptor (FSHR) is expressed at low levels in nonpregnant human myometrium and that it is up-regulated in pregnant term nonlaboring myometrium; however, the physiological relevance of these findings was unknown. Herein, we examined signaling pathways stimulated by FSH in immortalized uterine myocytes expressing recombinant FSHR at different densities and showed that cAMP accumulation is stimulated in all cases but that inositol phosphate accumulation is stimulated only at high FSHR densities. Because an increase in cAMP quiets myometrial contractile activity but an increase in 1,4,5-triphosphoinositol stimulates contractile activity, we hypothesized that FSHR density dictates whether FSH quiets or stimulates myometrial contractility. Indeed, in human and mouse nonpregnant myometrium, which express low levels of FSHR, application of FSH resulted in a quieting of contractile activity. In contrast, in pregnant term nonlaboring myometrium, which expresses higher levels of FSHR, application of FSH resulted in increased contractile activity. Examination of pregnant mouse myometrium from different stages of gestation revealed that FSHR levels remained low throughout most of pregnancy. Accordingly, through mid-gestation, the application of FSH resulted in a quieting of contractile activity. At Pregnancy Day (PD) 16.5, FSHR was up-regulated, although not yet sufficiently to mediate stimulation of contractility in response to FSH. This outcome was not observed until PD 19.5, when FSHR was further up-regulated. Our studies describe a novel FSHR signaling pathway that regulates myometrial contractility, and suggest that myometrial FSHR levels dictate the quieting vs. stimulation of uterine contractility in response to FSH.
Collapse
Affiliation(s)
- Julie A W Stilley
- Department of Molecular Physiology and Biophysics, the University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Rongbin Guan
- Department of Molecular Physiology and Biophysics, the University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, the University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Bryan F Mitchell
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Kathryn G Lamping
- Departments of Internal Medicine and Pharmacology, the University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Deborah L Segaloff
- Department of Molecular Physiology and Biophysics, the University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
49
|
Zhai JS, Song JG, Zhu CH, Wu K, Yao Y, Li N. Expression of APPL1 is correlated with clinicopathologic characteristics and poor prognosis in patients with gastric cancer. ACTA ACUST UNITED AC 2016; 23:e95-e101. [PMID: 27122990 DOI: 10.3747/co.23.2775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although appl1 is overexpressed in many cancers, its status in gastric cancer (gc) is not known. In the present study, we used relevant pathologic and clinical data to investigate appl1 expression in patients with gc. METHODS In 47 gc and 27 non-gc surgical specimens, immunohistochemistry was used to detect the expression of appl1, and reverse-transcriptase polymerase chain reaction (rt-pcr) was used to detect messenger rna (mrna). A scatterplot visualized the relationship between survival time and mrna expression in gc patients. The log-rank test and other survival statistics were used to determine the association of appl1 expression with the pathologic features of the cancer and clinical outcomes. RESULTS In gc, appl1 was expressed in 28 of 47 specimens (59.6%), and in non-gc, it was expressed in 7 of 23 specimens (30.4%, p < 0.05). The expression of mrna in gc was 0.82 [95% confidence interval (ci): 0.78 to 0.86], and in non-gc, it was 0.73 (95% ci: 0.69 to 0.77; p < 0.05). Immunohistochemistry demonstrated that, in gc, appl1 expression was correlated with depth of infiltration (p = 0.005), lymph node metastasis (p = 0.017), and TNM stage (p = 0.022), but not with pathologic type (p = 0.41). Testing by rt-pcr demonstrated that, in gc, appl1 mrna expression was correlated with depth of infiltration (p = 0.042), lymph node metastasis (p = 0.031), and TNM stage (p = 0.04), but again, not with pathologic type (p = 0.98). The correlation coefficient between survival time and mrna expression was -0.83 (p < 0.01). Overexpression of appl1 protein (hazard ratio: 3.88; 95% ci: 1.07 to 14.09) and mrna (hazard ratio: 4.23; 95% ci: 3.09 to 15.11) was a risk factor for death in patients with gc. CONCLUSIONS Expression of appl1 is increased in gc. Overexpression is prognostic for a lethal outcome.
Collapse
Affiliation(s)
- J S Zhai
- Postgraduate Team, Chinese pla General Hospital, Medical School of Chinese pla, Beijing, P.R.C.;; Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| | - J G Song
- Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| | - C H Zhu
- Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| | - K Wu
- Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| | - Y Yao
- Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| | - N Li
- Postgraduate Team, Chinese pla General Hospital, Medical School of Chinese pla, Beijing, P.R.C.;; Department of Gastroenterology, Chinese pla 309 Hospital, Beijing, P.R.C
| |
Collapse
|
50
|
Lindgren I, Bååth M, Uvebrant K, Dejmek A, Kjaer L, Henic E, Bungum M, Bungum L, Cilio C, Leijonhufvud I, Skouby S, Andersen CY, Giwercman YL. Combined assessment of polymorphisms in the LHCGR and FSHR genes predict chance of pregnancy after in vitro fertilization. Hum Reprod 2016; 31:672-83. [PMID: 26769719 DOI: 10.1093/humrep/dev342] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/17/2015] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Can gonadotrophin receptor variants separately or in combination, be used for the prediction of pregnancy chances in in vitro fertilization (IVF) trials? SUMMARY ANSWER The luteinizing hormone/human chorionic gonadotrophin receptor (LHCGR) variant N312S and the follicle-stimulating hormone receptor (FSHR) variant N680S can be utilized for the prediction of pregnancy chances in women undergoing IVF. WHAT IS KNOWN ALREADY The FSHR N680S polymorphism has been shown to affect the ovarian response in response to gonadotrophin treatment, while no information is currently available regarding variants of the LHCGR in this context. STUDY DESIGN, SIZE, DURATION Cross-sectional study, duration from September 2010 to February 2015. Women undergoing IVF were consecutively enrolled and genetic variants compared between those who became pregnant and those who did not. The study was subsequently replicated in an independent sample. Granulosa cells from a subset of women were investigated regarding functionality of the genetic variants. PARTICIPANTS/MATERIALS, SETTING, METHODS Women undergoing IVF (n = 384) were enrolled in the study and genotyped. Clinical variables were retrieved from medical records. For replication, an additional group of n = 233 women was utilized. Granulosa cells from n = 135 women were isolated by flow cytometry, stimulated with Follitropin alpha or Menotropin, and the downstream targets 3',5'-cyclic adenosine monophosphate (cAMP) and inositol 1,4,5-trisphosphate (IP3) measured with enzyme-linked immunosorbent assay. MAIN RESULTS AND THE ROLE OF CHANCE Women homozygous for serine (S) in both polymorphisms displayed higher pregnancy rates than women homozygous asparagine (N) (OR = 14.4, 95% CI: [1.65, 126], P = 0.016). Higher pregnancy rates were also evident for women carrying LHCGR S312, regardless of FSHR variant (OR = 1.61, 95% CI: [1.13, 2.29], P = 0.008). These women required higher doses of FSH for follicle recruitment than women homozygous N (161 versus 148 IU, P = 0.030). When combining the study cohort with the replication cohort (n = 606), even stronger associations with pregnancy rates were noted for the combined genotypes (OR = 11.5, 95% CI: [1.86, 71.0], P = 0.009) and for women carrying LHCGR S312 (OR = 1.49, 95% CI: [1.14, 1.96], P = 0.004). A linear significant trend with pregnancy rate and increasing number of G alleles was also evident in the merged study population (OR = 1.34, 95% CI: [1.10, 1.64], P = 0.004). A lower cAMP response in granulosa cells was noted following Follitropin alpha stimulation for women homozygous N in both polymorphisms, compared with women with other genotypes (0.901 pmol cAMP/mg total protein versus 2.19 pmol cAMP/mg total protein, P = 0.035). LIMITATIONS, REASONS FOR CAUTION Due to racial differences in LHCGR genotype distribution, these results may not be applicable for all populations. WIDER IMPLICATIONS OF THE FINDINGS Despite that >250 000 cycles of gonadotrophin stimulations are performed annually worldwide prior to IVF, it has not been possible to predict neither the pregnancy outcome, nor the response to the hormone with accuracy. If LHCGR and FSHR variants are recognized as biomarkers for chance of pregnancy, more individualized and thereby more efficient treatment modalities can be developed. STUDY FUNDING, COMPETING INTERESTS This work was supported by Interreg IV A, EU (grant 167158) and ALF governments grant (F2014/354). Merck-Serono (Darmstadt, Germany) supported the enrollment of the subjects. The authors declare no conflict of interest.
Collapse
Affiliation(s)
- I Lindgren
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - M Bååth
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - K Uvebrant
- Department of Clinical Sciences, Cellular Autoimmunity Unit, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - A Dejmek
- Department of Clinical Pathology, University and Regional Laboratories, Skåne University Hospital, Jan Waldenströms gata 59, SE 20502 Malmö, Sweden
| | - L Kjaer
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - E Henic
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - M Bungum
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - L Bungum
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - C Cilio
- Department of Clinical Sciences, Cellular Autoimmunity Unit, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| | - I Leijonhufvud
- Reproductive Medicine Centre, Skåne University Hospital, Jan Waldenströms gata 47, Plan 3, SE 20502 Malmö, Sweden
| | - S Skouby
- Department of Obstetrics and Gynecology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - C Yding Andersen
- Laboratory of Reproductive Biology, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Y Lundberg Giwercman
- Department of Translational Medicine, Molecular Genetic Reproductive Medicine, Clinical Research Centre, Lund University, Jan Waldenströms gata 35, Building 91, Plan 10, SE 20502 Malmö, Sweden
| |
Collapse
|