1
|
Wu PS, Wong TH, Hou CW, Chu TP, Lee JW, Lou BS, Lin MH. Cold Atmospheric Plasma Jet Promotes Wound Healing Through CK2-Coordinated PI3K/AKT and MAPK Signaling Pathways. Mol Cell Proteomics 2025; 24:100962. [PMID: 40187493 DOI: 10.1016/j.mcpro.2025.100962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 03/03/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
The promising role of cold atmospheric plasma jet (CAPJ) treatment in promoting wound healing has been widely documented in therapeutic implications. However, the fact that not all subjects respond equally to CAPJ necessitates the investigation of the underlying cellular mechanisms, which have been rarely understood so far. Given that wound healing is a complex and prolonged process, post plasma-activated medium (PAM) treated keratinocytes were collected at two time points, 2 h (receiving) and 24 h (recovery), for (phospho)proteomic analysis to systematically dissect the molecular basis of CAPJ-promoted wound healing. The receiving (phospho)proteomics datasets, referred to the time point of 2 h, revealed an apparent increase in the phosphorylation of CK2 and its-mediated PI3K/AKT and MAPK signaling pathways, accompanied by a prompted downstream physiological response of cell migration. Additionally, incorporating the network analysis of predicted kinases and their direct interactors, we reiterated that CAPJ influenced cell growth and migration, thereby paving the way for its role in subsequent wound healing processes. Further determining the proteome profiles at recovery phase, which is the time point of 24 h, displayed a totally different view from the receiving proteome which had almost no change. The upregulation of ROBOs/SLITs expression and vesicle trafficking and fusion-related proteins, along with the abundant presence of 14-3-3 family proteins, indicated that the persistent effect of PAM on the wound healing process could potentially promote keratinocyte-fibroblast cross talk and stimulate extracellular matrix synthesis upon epithelialization. Consistent with proteome patterns, CAPJ-treated wound tissues indeed showed a denser and well-organized extracellular matrix architecture, implying hastened epithelialization during wound healing. Collectively, we delineated the molecular basis of CAPJ-accelerated wound healing at early and late responses, providing valuable insights for treatment selection and the development of therapeutic strategies to achieve better outcomes.
Collapse
Affiliation(s)
- Pei-Shan Wu
- Chemistry Division, Center for General Education, Chang Gung University, Taoyuan, Taiwan; Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tzu-Hsuan Wong
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chun-Wei Hou
- Chemistry Division, Center for General Education, Chang Gung University, Taoyuan, Taiwan
| | - Teng-Ping Chu
- Center for Plasma and Thin Film Technologies, Ming Chi University of Technology, New Taipei, Taiwan; International PhD. Program in Plasma and Thin Film Technology, Ming Chi University of Technology, New Taipei, Taiwan
| | - Jyh-Wei Lee
- Center for Plasma and Thin Film Technologies, Ming Chi University of Technology, New Taipei, Taiwan; International PhD. Program in Plasma and Thin Film Technology, Ming Chi University of Technology, New Taipei, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, New Taipei, Taiwan; High Entropy Materials Center, National Tsing Hua University, Hsinchu, Taiwan; College of Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Bih-Show Lou
- Chemistry Division, Center for General Education, Chang Gung University, Taoyuan, Taiwan; Department of Orthopaedic Surgery, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Miao-Hsia Lin
- Department of Microbiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
2
|
Cannarella R, Crafa A, Curto R, Condorelli RA, La Vignera S, Calogero AE. Obesity and male fertility disorders. Mol Aspects Med 2024; 97:101273. [PMID: 38593513 DOI: 10.1016/j.mam.2024.101273] [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: 02/22/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Often associated with obesity, male infertility represents a widespread condition that challenges the wellbeing of the couple. In this article, we provide a comprehensive and critical analysis of studies exploring the association between obesity and male reproductive function, to evaluate the frequency of this association, and establish the effects of increased body weight on conventional and biofunctional sperm parameters and infertility. In an attempt to find possible molecular markers of infertility in obese male patients, the numerous mechanisms responsible for infertility in overweight/obese patients are reviewed in depth. These include obesity-related functional hypogonadism, insulin resistance, hyperinsulinemia, chronic inflammation, adipokines, irisin, gut hormones, gut microbiome, and sperm transcriptome. According to meta-analytic evidence, excessive body weight negatively influences male reproductive health. This can occurr through a broad array of molecular mechanisms. Some of these are not yet fully understood and need to be further elucidated in the future. A better understanding of the effects of metabolic disorders on spermatogenesis and sperm fertilizing capacity is very useful for identifying new diagnostic markers and designing therapeutic strategies for better clinical management of male infertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
3
|
Glebov-McCloud AGP, Saide WS, Gaine ME, Strack S. Protein Kinase A in neurological disorders. J Neurodev Disord 2024; 16:9. [PMID: 38481146 PMCID: PMC10936040 DOI: 10.1186/s11689-024-09525-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/29/2024] [Indexed: 03/17/2024] Open
Abstract
Cyclic adenosine 3', 5' monophosphate (cAMP)-dependent Protein Kinase A (PKA) is a multi-functional serine/threonine kinase that regulates a wide variety of physiological processes including gene transcription, metabolism, and synaptic plasticity. Genomic sequencing studies have identified both germline and somatic variants of the catalytic and regulatory subunits of PKA in patients with metabolic and neurodevelopmental disorders. In this review we discuss the classical cAMP/PKA signaling pathway and the disease phenotypes that result from PKA variants. This review highlights distinct isoform-specific cognitive deficits that occur in both PKA catalytic and regulatory subunits, and how tissue-specific distribution of these isoforms may contribute to neurodevelopmental disorders in comparison to more generalized endocrine dysfunction.
Collapse
Affiliation(s)
- Alexander G P Glebov-McCloud
- Department of Neuroscience and Pharmacology, Bowen Science Building, University of Iowa, Carver College of Medicine, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Walter S Saide
- Department of Neuroscience and Pharmacology, Bowen Science Building, University of Iowa, Carver College of Medicine, 51 Newton Road, Iowa City, IA, 52242, USA
| | - Marie E Gaine
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy Building, College of Pharmacy, University of Iowa, 180 S. Grand Ave, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, Intellectual and Developmental Disabilities Research Center, Iowa City, IA, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, Bowen Science Building, University of Iowa, Carver College of Medicine, 51 Newton Road, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, Intellectual and Developmental Disabilities Research Center, Iowa City, IA, USA.
| |
Collapse
|
4
|
Guner JZ, Monsivais D, Yu H, Stossi F, Johnson HL, Gibbons WE, Matzuk MM, Palmer S. Oral follicle-stimulating hormone receptor agonist affects granulosa cells differently than recombinant human FSH. Fertil Steril 2023; 120:1061-1070. [PMID: 37532169 PMCID: PMC10659100 DOI: 10.1016/j.fertnstert.2023.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE To determine whether TOP5300, a novel oral follicle-stimulating hormone (FSH) receptor (FSHR) allosteric agonist, elicits a different cellular response than recombinant human FSH (rh-FSH) in human granulosa cells from patients undergoing in vitro fertilization. DESIGN Basic science research with a preclinical allosteric FSHR agonist. SETTING University hospital. PATIENT(S) Patients with infertility at a single academic fertility clinic were recruited under an Institutional Review Board-approved protocol. Primary granulosa cell cultures were established for 41 patients, of whom 8 had normal ovarian reserve (NOR), 17 were of advanced reproductive age (ARA), 12 had a diagnosis of polycystic ovary syndrome (PCOS), and 4 had a combination of diagnoses, such as ARA and PCOS. INTERVENTION(S) Primary granulosa-lutein (GL) cell cultures were treated with rh-FSH, TOP5300, or vehicle. MAIN OUTCOME MEASURE(S) Estradiol (E2) production using enzyme-linked immunosorbent assay, steroid pathway gene expression of StAR and aromatase using quantitative polymerase chain reaction, and FSHR membrane localization using immunofluorescence were measured in human GL cells. RESULT(S) TOP5300 consistently stimulated E2 production among patients with NOR, ARA, and PCOS. Recombinant FSH was the more potent ligand in GL cells from patients with NOR but was ineffective in cells from patients with ARA or PCOS. The lowest level of FSHR plasma membrane localization was seen in patients with ARA, although FSHR localization was more abundant in cells from patients with PCOS; the highest levels were present in cells from patients with NOR. The localization of FSHR was not affected by TOP5300 relative to rh-FSH in any patient group. TOP5300 stimulated greater expression of StAR and CYP19A1 across cells from all patients with NOR, ARA, and PCOS combined, although rh-FSH was unable to stimulate StAR and aromatase (CYP19A1) expression in cells from patients with PCOS. TOP5300-induced expression of StAR and CYP19A1 mRNA among patients with ARA and NOR was consistently lower than that observed in cells from patients with PCOS. CONCLUSION(S) TOP5300 appears to stimulate E2 production and steroidogenic gene expression from GL cells more than rh-FSH in PCOS, relative to patients with ARA and NOR. It does not appear that localization of FSHR at cell membranes is a limiting step for TOP5300 or rh-FSH stimulation of steroidogenic gene expression and E2 production.
Collapse
Affiliation(s)
- Joie Z Guner
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, California
| | - Diana Monsivais
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Henry Yu
- CanWell Pharma, Wellesley, Massachusetts
| | - Fabio Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; Integrated Microscopy Core and GCC Center for Advanced Microscopy and Image Informatics, Baylor College of Medicine, Houston, Texas
| | - Hannah L Johnson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas; Integrated Microscopy Core and GCC Center for Advanced Microscopy and Image Informatics, Baylor College of Medicine, Houston, Texas
| | - William E Gibbons
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Baylor College of Medicine, and Texas Children's Hospital Family Fertility Center, Houston, Texas
| | - Martin M Matzuk
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Stephen Palmer
- Center for Drug Discovery, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas.
| |
Collapse
|
5
|
Madsen JF, Amoushahi M, Choi CP, Bundgaard S, Heuck A, Lykke-Hartmann K. Inhibition of phosphodiesterase PDE8B reduces activation of primordial follicles in mouse ovaries. Mol Reprod Dev 2023; 90:378-388. [PMID: 37499226 DOI: 10.1002/mrd.23699] [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: 11/20/2022] [Revised: 06/12/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
In the ovaries, cyclic adenosine 3',5'-monophosphate (cAMP) is a second messenger supporting the generation of steroids. Phosphodiesterases (PDEs) are regulators of intracellular cAMP, and therefore, potential regulators of ovarian function. Interestingly, the family of PDE genes are differentially expressed in human oocytes and granulosa cells from primordial and primary follicles, suggesting diverse roles. In this study, we addressed the functions of PDE3B and PDE8B in primordial follicle regulation using inhibitors of PDE3B and PDE8B in murine ovary primary in vitro cultures. Inhibition of PDE8B in ovarian cultures prevented primordial follicle activation, while inhibition of PDE3B had no effect on follicle distribution in the ovary, under the tested conditions. As cAMP levels may increase steroid levels, we assessed the protein levels of the steroidogenic acute regulatory protein (StAR) and aromatase enzymes, and found that inhibition of PDE3B reduced StAR protein levels, whereas inhibition of PDE8 did not alter StAR expression in our murine ovary culture system conditions. Our results showed that ketotifen-induced inhibition of PDE8B can decrease primordial follicle activation, whereas we observed no effect of follicle distribution, when PDE3B was inhibited. Expression of the StaR enzyme was not altered when PDE8B was inhibited, which might reflect not sufficient inhibition by ketotifen to induce StAR alterations, or redundant mechanisms.
Collapse
Affiliation(s)
| | | | | | - Stine Bundgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Heuck
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
6
|
Mizutani T, Orisaka M, Kawabe S, Morichika R, Uesaka M, Yoshida Y. YAP/TAZ-TEAD is a novel transcriptional regulator of genes encoding steroidogenic enzymes in rat granulosa cells and KGN cells. Mol Cell Endocrinol 2023; 559:111808. [PMID: 36309205 DOI: 10.1016/j.mce.2022.111808] [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] [Received: 06/04/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Steroidogenesis in ovarian granulosa cells is regulated by the follicle-stimulating hormone (FSH) via transcriptional regulation of its related genes. We herein showed the involvement of the Hippo pathway in this regulation. In KGN granulosa cell, repression of YAP/TAZ activity induced the expression of CYP11A1, HSD3B2, and CYP19A1 in a TEAD-dependent manner without cAMP stimulation. A selective inhibitor of p38 MAP kinase, suppressed YAP/TAZ knockdown-indued the expression of these genes, suggesting this signal could be involved. The expression of these genes was induced by 8Br-cAMP, whereas that of CYR61 and ADATS1, typical YAP/TAZ-TEAD target genes, was suppressed, suggesting that the cellular signaling of cAMP reduced YAP/TAZ-TEAD activity. The constitutively active mutant YAP canceled the FSH- and 8Br-cAMP-mediated induction of these genes in primary rat granulosa and KGN cells, respectively. Moreover, regulation of steroidogenesis-related genes by YAP/TAZ-TEAD was independent of steroidogenic factor 1, a master gene regulator of steroidogenesis. These results suggest that YAP/TAZ-TEAD is a negative regulator of steroidogenesis and that suppression of YAP/TAZ-TEAD activity by FSH is involved in ovarian steroidogenesis.
Collapse
Affiliation(s)
- Tetsuya Mizutani
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Japan.
| | - Makoto Orisaka
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Shinya Kawabe
- Department of Food Science and Technology, National Fisheries University, Japan
| | - Ririko Morichika
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Japan
| | - Miki Uesaka
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| |
Collapse
|
7
|
Allethrin Promotes Apoptosis and Autophagy Associated with the Oxidative Stress-Related PI3K/AKT/mTOR Signaling Pathway in Developing Rat Ovaries. Int J Mol Sci 2022; 23:ijms23126397. [PMID: 35742842 PMCID: PMC9224321 DOI: 10.3390/ijms23126397] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
The increased concern regarding the reduction in female fertility and the impressive numbers of women undergoing fertility treatment support the existence of environmental factors beyond inappropriate programming of developing ovaries. Among these factors are pyrethroids, which are currently some of the most commonly used pesticides worldwide. The present study was performed to investigate the developmental effects of the pyrethroid-based insecticide allethrin on ovarian function in rat offspring in adulthood. We mainly focused on the roles of oxidative stress, apoptosis, autophagy and the related pathways in ovarian injury. Thirty-day-old Wistar albino female rats were intragastrically administered 0 (control), 34.2 or 68.5 mg/kg body weight allethrin after breeding from Day 6 of pregnancy until delivery. We found that allethrin-induced ovarian histopathological damage was accompanied by elevations in oxidative stress and apoptosis. Interestingly, the number of autophagosomes in allethrin-treated ovaries was higher, and this increase was correlated with the upregulated expression of genes and proteins related to the autophagic marker LC-3. Furthermore, allethrin downregulated the expression of PI3K, AKT and mTOR in allethrin-treated ovaries compared with control ovaries. Taken together, the findings of this study suggest that exposure to the pyrethroid-based insecticide allethrin adversely affects both the follicle structure and function in rat offspring during adulthood. Specifically, allethrin can induce excessive oxidative stress and defective autophagy-related apoptosis, probably through inactivation of the PI3K/AKT/mTOR signaling pathway, and these effects may contribute to ovarian dysfunction and impaired fertility in female offspring.
Collapse
|
8
|
Zhan CH, Ding DS, Zhang W, Wang HL, Mao ZY, Liu GJ. The cancer-testis antigen a-kinase anchor protein 3 facilitates breast cancer progression via activation of the PTEN/PI3K/AKT/mTOR signaling. Bioengineered 2022; 13:8478-8489. [PMID: 35322748 PMCID: PMC9161980 DOI: 10.1080/21655979.2022.2051687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The cancer-testis antigen A-kinase anchor protein 3 (AKAP3) has been shown to have a strong association with breast cancer (BC). However, its role in BC progression received scant attention. We aimed to explore the prognostic implication of aberrant AKAP3 expression for a better knowledge of BC progression and improved treatment. AKAP3 expression was quantitated using tissue microarrays and immunohistochemistry (IHC). Cell viability, invasion, migration, apoptosis, and expressions of PTEN/PI3K/AKT/mTOR signaling components were assessed in AKAP3-overexpressed or si-AKAP3-transfected BC cells. Finally, elevated AKAP3 expression was observed in BC versus paracancerous tissues. BC patients with high AKAP3 expression showed a worse prognosis than low expression patients (P < 0.0001). AKAP3 overexpressions fueled cell growth, proliferation, migration, and invasion in HCC1937 and MDA-MB-468 BC cell lines, alongside increased expressions of PI3K/AKT/mTOR signaling components and PTEN suppression. These effects were pronouncedly reversed, together with elevated apoptosis, in cells transfected with si-AKAP3. Therefore, AKAP3 is upregulated in BC and promotes BC cell growth, invasion, and migration via PTEN/PI3K/AKT/mTOR signaling activation. It may serve as a prognosis indicator for BC survival.
Collapse
Affiliation(s)
- Chuan-Hua Zhan
- Department of Clinical Laboratory, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China
| | - Dong-Shen Ding
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China.,Department of Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China
| | - Wei Zhang
- Department of Clinical Laboratory, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China
| | - Hong-Liang Wang
- Department of Clinical Laboratory, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China.,Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China
| | - Zhe-Yu Mao
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China.,Department of Breast Cancer Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China
| | - Guo-Jun Liu
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, P.R. China.,Department of Breast Cancer Surgery, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, P.R. China
| |
Collapse
|
9
|
Morris G, Walder K, Berk M, Carvalho AF, Marx W, Bortolasci CC, Yung AR, Puri BK, Maes M. Intertwined associations between oxidative and nitrosative stress and endocannabinoid system pathways: Relevance for neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2022; 114:110481. [PMID: 34826557 DOI: 10.1016/j.pnpbp.2021.110481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/19/2021] [Accepted: 11/21/2021] [Indexed: 12/12/2022]
Abstract
The endocannabinoid system (ECS) appears to regulate metabolic, cardiovascular, immune, gastrointestinal, lung, and reproductive system functions, as well as the central nervous system. There is also evidence that neuropsychiatric disorders are associated with ECS abnormalities as well as oxidative and nitrosative stress pathways. The goal of this mechanistic review is to investigate the mechanisms underlying the ECS's regulation of redox signalling, as well as the mechanisms by which activated oxidative and nitrosative stress pathways may impair ECS-mediated signalling. Cannabinoid receptor (CB)1 activation and upregulation of brain CB2 receptors reduce oxidative stress in the brain, resulting in less tissue damage and less neuroinflammation. Chronically high levels of oxidative stress may impair CB1 and CB2 receptor activity. CB1 activation in peripheral cells increases nitrosative stress and inducible nitric oxide (iNOS) activity, reducing mitochondrial activity. Upregulation of CB2 in the peripheral and central nervous systems may reduce iNOS, nitrosative stress, and neuroinflammation. Nitrosative stress may have an impact on CB1 and CB2-mediated signalling. Peripheral immune activation, which frequently occurs in response to nitro-oxidative stress, may result in increased expression of CB2 receptors on T and B lymphocytes, dendritic cells, and macrophages, reducing the production of inflammatory products and limiting the duration and intensity of the immune and oxidative stress response. In conclusion, high levels of oxidative and nitrosative stress may compromise or even abolish ECS-mediated redox pathway regulation. Future research in neuropsychiatric disorders like mood disorders and deficit schizophrenia should explore abnormalities in these intertwined signalling pathways.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Wolf Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| | - Alison R Yung
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia; School of Health Science, University of Manchester, UK.
| | - Basant K Puri
- University of Winchester, UK, and C.A.R., Cambridge, UK.
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
10
|
Cannarella R, Mancuso F, Arato I, Lilli C, Bellucci C, Gargaro M, Curto R, Aglietti MC, La Vignera S, Condorelli RA, Luca G, Calogero AE. Sperm-carried IGF2 downregulated the expression of mitogens produced by Sertoli cells: A paracrine mechanism for regulating spermatogenesis? Front Endocrinol (Lausanne) 2022; 13:1010796. [PMID: 36523595 PMCID: PMC9744929 DOI: 10.3389/fendo.2022.1010796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 2 (IGF2) mRNA has been found in human and mouse spermatozoa. It is currently unknown whether the IGF2 protein is expressed in human spermatozoa and, if so, its possible role in the cross-talk between germ and Sertoli cells (SCs) during spermatogenesis. METHODS To accomplish this, we analyzed sperm samples from four consecutive Caucasian men. Furthermore, to understand its role during the spermatogenetic process, porcine SCs were incubated with increasing concentrations (0.33, 3.33, and 10 ng/mL) of recombinant human IGF2 (rhIGF2) for 48 hours. Subsequently, the experiments were repeated by pre-incubating SCs with the non-competitive insulin-like growth factor 1 receptor (IGF1R) inhibitor NVP-AEW541. The following outcomes were evaluated: 1) Gene expression of the glial cell-line derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and stem cell factor (SCF) mitogens; 2) gene and protein expression of follicle-stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), and inhibin B; 3) SC proliferation. RESULTS We found that the IGF2 protein was present in each of the sperm samples. IGF2 appeared as a cytoplasmic protein localized in the equatorial and post-acrosomal segment and with a varying degree of expression in each cell. In SCs, IGF2 significantly downregulated GDNF gene expression in a concentration-dependent manner. FGF2 and SCF were downregulated only by the highest concentration of IGF2. Similarly, IGF2 downregulated the FSHR gene and FSHR, AMH, and inhibin B protein expression. Finally, IGF2 significantly suppressed the SC proliferation rate. All these findings were reversed by pre-incubation with NVP-AEW541, suggesting an effect mediated by the interaction of IGF2 with the IGFR. CONCLUSION In conclusion, sperm IGF2 seems to downregulate the expression of mitogens, which are known to be physiologically released by the SCs to promote gonocyte proliferation and spermatogonial fate adoption. These findings suggest the presence of paracrine regulatory mechanisms acting on the seminiferous epithelium during spermatogenesis, by which germ cells can influence the amount of mitogens released by the SCs, their sensitivity to FSH, and their rate of proliferation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- *Correspondence: Rossella Cannarella,
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria C. Aglietti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovani Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
11
|
Utomo B, Rimayanti R, Triana IN, Fadholly A. Melanocortin-4 receptor and leptin as genes for the selection of superior Madrasin cattle. Vet World 2021; 14:3224-3228. [PMID: 35153416 PMCID: PMC8829405 DOI: 10.14202/vetworld.2021.3224-3228] [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: 08/15/2021] [Accepted: 11/24/2021] [Indexed: 11/27/2022] Open
Abstract
Background and Aim: The genetic improvement of cattle through livestock section is based on quantitative, qualitative, and molecular characteristics. This study examined polymorphisms of the melanocortin-4 receptor (MC4R) and leptin genes as a reference for the selection of superior breeds in Madrasin cattle. Materials and Methods: The leptin and MC4R genes of Madrasin cattle were amplified using polymerase chain reaction (PCR); then, restriction fragment length polymorphism of the leptin gene was performed using the restriction enzyme BsaA1, at site 2793 with ACGT point position. Results: The leptin gene was divided into three bands, namely, AA with one fragment (522 bp), CG with two fragments (441 bp and 81 bp), and AG with three fragments (522 bp, 441 bp, and 81 bp). The MCR-4 gene was divided into three bands, namely, 493 bp, 318 bp, and 175 bp. Conclusion: The MC4R and leptin genes can act as molecular markers for growth traits in Madrasin cattle and can be used to genetically optimize and improve growth. The GG allele of the MC4R gene and the AA allele of the leptin gene can be used in Madrasin cattle.
Collapse
Affiliation(s)
- Budi Utomo
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Rimayanti Rimayanti
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Indah Norma Triana
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, 60115, Indonesia
| | - Amaq Fadholly
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, 60115, Indonesia
| |
Collapse
|
12
|
Long H, Yu W, Yu S, Yin M, Wu L, Chen Q, Cai R, Suo L, Wang L, Lyu Q, Kuang Y. Progesterone affects clinic oocyte yields by coordinating with follicle stimulating hormone via PI3K/AKT and MAPK pathways. J Adv Res 2021; 33:189-199. [PMID: 34603789 PMCID: PMC8463924 DOI: 10.1016/j.jare.2021.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 11/28/2022] Open
Abstract
Progesterone reduced oocyte yields in clinic. Yields were rescued by the higher dose of hMG. Progesterone downregulated follicle growth and consequently reduced oocyte yields. Progesterone inhibited granular cell proliferation via MAPK and PI3K/AKT pathways. Progesterone and FSH coordinated follicle growth via signalling crosstalk in granular cells.
Introduction As an effective inhibitor of premature ovulation, progestin was introduced to a novel ovarian stimulation regimen for infertility treatment. However, the local action of progestin on the ovary and its effect on clinical outcomes have not been described. Objectives The influence of progesterone administration on clinical oocyte outcomes and the mechanisms involved in the coordination of progesterone and follicle stimulating hormone (FSH) on follicle growth and oocyte yields were investigated. Methods Clinical outcomes of patients undergoing ovarian stimulation for in vitro fertilization were analyzed. The murine ovarian stimulation model and follicle culture system were used to evaluate the effects of progesterone on oocyte yield, follicle development, granular cell proliferation, and hormone secretion. Phospho-specific protein microarrays were used to explore involved signaling pathways. Results Progesterone decreased clinical oocyte yields, and yields were rescued with an increased dose of human menopausal gonadotropin. Administration of progesterone inhibited murine granular cell proliferation and reduced the growth rate of follicles; both of which were rescued by FSH. The phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) and mitogen-activated protein kinase (MAPK) were identified as pivotal signaling pathways to integrate progesterone into the FSH signaling network in granular cells. Conclusion Progesterone inhibited granular cell proliferation and antral follicle growth during ovarian stimulation, and subsequently influenced oocyte outcomes in the clinical setting. Progesterone coordinated with FSH to regulate follicle growth through PI3K/AKT and MAPK signaling pathways. These findings advance our knowledge regarding the ovarian response to gonadotropins during progestin-primed ovarian stimulation and create an opportunity to manipulate individual oocyte yields.
Collapse
Affiliation(s)
- Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Weina Yu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Sha Yu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Mingru Yin
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Qiuju Chen
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Renfei Cai
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Lun Suo
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Li Wang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Zhizaoju Road No. 639, Huangpu District, Shanghai 200011, People's Republic of China
| |
Collapse
|
13
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
14
|
Pseudo-Starvation Driven Energy Expenditure Negatively Affects Ovarian Follicle Development. Int J Mol Sci 2021; 22:ijms22073557. [PMID: 33808081 PMCID: PMC8036485 DOI: 10.3390/ijms22073557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 11/30/2022] Open
Abstract
In the present investigation, we examined whether a change in whole body energy fluxes could affect ovarian follicular development, employing mice ectopically expressing uncoupling protein 1 in skeletal muscle (UCP1-TG). Female UCP1-TG and wild-type (WT) mice were dissected at the age of 12 weeks. Energy intake and expenditure, activity, body weight and length, and body composition were measured. Plasma insulin, glucose, leptin, plasma fibroblast growth factor 21 (FGF21) and plasma insulin-like growth factor 1 (IGF1) levels were analyzed and ovarian follicle and corpus luteum numbers were counted. IGF1 signaling was analyzed by immunohistochemical staining for the activation of insulin receptor substrate 1/2 (IRS1/2) and AKT. UCP1-TG female mice had increased energy expenditure, reduced body size, maintained adiposity, and decreased IGF1 concentrations compared to their WT littermates, while preantral and antral follicle numbers were reduced by 40% and 60%, respectively. Corpora lutea were absent in 40% of the ovaries of UCP1-TG mice. Phospho-IRS1, phospho-AKT -Ser473 and -Thr308 immunostaining was present in the granulosa cells of antral follicles in WT ovaries, but faint to absent in the antral follicles of UCP1-TG mice. In conclusion, the reduction in circulating IGF1 levels due to the ectopic expression of UCP1 is associated with reduced immunostaining of the IRS1-PI3/AKT pathway, which may negatively affect ovarian follicle development and ovulation.
Collapse
|
15
|
Chahal N, Geethadevi A, Kaur S, Lakra R, Nagendra A, Shrivastav TG, De Pascali F, Reiter E, Crépieux P, Devi MG, Malhotra N, Muralidhar K, Singh R. Direct impact of gonadotropins on glucose uptake and storage in preovulatory granulosa cells: Implications in the pathogenesis of polycystic ovary syndrome. Metabolism 2021; 115:154458. [PMID: 33278413 DOI: 10.1016/j.metabol.2020.154458] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is often associated with higher levels of LH, and arrested ovarian follicular growth. The direct impact of high LH on FSH mediated metabolic responses in PCOS patients is not clearly understood. METHOD In order to investigate the impact of FSH and LH on glucose metabolism in preovulatory granulosa cells (GCs), we used [U14C]-2 deoxyglucose, D-[U14C]-glucose or 2-NBD glucose to analyse glucose uptake and its incorporation into glycogen. To reproduce the high androgenic potential in PCOS patients, we administered hCG both in vitro and in vivo. The role of IRS-2/PI3K/Akt2 pathway was studied after knockdown with specific siRNA. Immunoprecipitation and specific assays were used for the assessment of IRS-2, glycogen synthase and protein phosphatase 1. Furthermore, we examined the in vivo effects of hCG on FSH mediated glycogen increase in normal and PCOS rat model. HEK293 cells co-expressing FSHR and LHR were used to demonstrate glucose uptake and BRET change by FSH and hCG. RESULTS In normal human and rat granulosa cells, FSH is more potent than hCG in stimulating glucose uptake, however glycogen synthesis was significantly upregulated only by FSH through increase in activity of glycogen synthase via IRS-2/PI3K/Akt2 pathway. On the contrary, an impaired FSH-stimulated glucose uptake and glycogen synthesis in granulosa cells of PCOS-patients indicated a selective defect in FSHR activation. Further, in normal human granulosa cells, and in immature rat model, the impact of hCG on FSH responses was such that it inhibited the FSH-mediated glucose uptake as well as glycogen synthesis through inhibition of FSH-stimulated IRS-2 expression. These findings were further validated in HEK293 cells overexpressing Flag-LHR and HA-FSHR, where high hCG inhibited the FSH-stimulated glucose uptake. Notably, an increased BRET change was observed in HEK293 cells expressing FSHR-Rluc8 and LHR-Venus possibly suggesting increased heteromerization of LHR and FSHR in the presence of both hCG and FSH in comparison to FSH or hCG alone. CONCLUSION Our findings confirm a selective attenuation of metabolic responses to FSH such as glucose uptake and glycogen synthesis by high activation level of LHR leading to the inhibition of IRS-2 pathway, resulting in depleted glycogen stores and follicular growth arrest in PCOS patients.
Collapse
Affiliation(s)
- Nidhi Chahal
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali Geethadevi
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India; Medical College of Wisconsin, Milwaukee 53226, USA
| | - Surleen Kaur
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India; Ferticity Fertility Clinics, Delhi, India
| | - Ruchi Lakra
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali Nagendra
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India
| | - T G Shrivastav
- National Institute of Health and Family Welfare, Delhi, India
| | - Francesco De Pascali
- Physiologie de la Reproduction et des Comportements, INRAE UMR-0085, CNRS UMR-7247, Université de Tours, IFCE, F-37380 Nouzilly, France
| | - Eric Reiter
- Physiologie de la Reproduction et des Comportements, INRAE UMR-0085, CNRS UMR-7247, Université de Tours, IFCE, F-37380 Nouzilly, France
| | - Pascale Crépieux
- Physiologie de la Reproduction et des Comportements, INRAE UMR-0085, CNRS UMR-7247, Université de Tours, IFCE, F-37380 Nouzilly, France
| | | | - Neena Malhotra
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Delhi, India
| | - K Muralidhar
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India
| | - Rita Singh
- Division of Molecular Endocrinology and Reproduction, Department of Zoology, University of Delhi, Delhi, India.
| |
Collapse
|
16
|
Nataraja S, Yu H, Guner J, Palmer S. Discovery and Preclinical Development of Orally Active Small Molecules that Exhibit Highly Selective Follicle Stimulating Hormone Receptor Agonism. Front Pharmacol 2021; 11:602593. [PMID: 33519465 PMCID: PMC7845544 DOI: 10.3389/fphar.2020.602593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/17/2020] [Indexed: 12/17/2022] Open
Abstract
An orally active follicle stimulating hormone receptor allosteric agonist would provide a preferred treatment for over 16 million infertile women of reproductive age in low complexity methods (ovulation induction-intrauterine insemination) or in high complexity methods (controlled ovarian stimulation-in vitro fertilization). We present two oral follicle stimulating hormone receptor allosteric agonist compounds that have the desired pharmacology, drug metabolism, pharmacokinetics, and safety profile for clinical use. These molecules provide a single agent suitable for ovulation induction-intrauterine insemination or controlled ovarian stimulation-in vitro fertilization that is more convenient for patients and achieves similar preclinical efficacy as rec-hFSH. TOP5668, TOP5300 were evaluated in vitro in Chinese hamster ovary cells transfected with individual glycoprotein receptors measuring cAMP (FSHR, LH/CGR, thyroid stimulating hormone receptor). TOP5668 was found to have solely follicle stimulating hormone receptor allosteric agonist activity while TOP5300 was found to have mixed follicle stimulating hormone receptor allosteric agonist and LHR-AA activity. Both compounds stimulated concentration-dependent increases in estradiol production from cultured rat granulosa cells in the presence or absence of low dose rec-hFSH, while only TOP5300 stimulated testosterone production from rat primary Leydig cells. In pooled human granulosa cells obtained from patients undergoing controlled ovarian stimulation-in vitro fertilization, TOP5300 stimulated 7-fold greater maximal estradiol response than rec-hFSH and TOP5668 was 10-fold more potent than TOP5300. Both TOP5300 and TOP5668 stimulated follicular development in immature rat to the same efficacy as recombinant follicle stimulating hormone. In mice treated with TOP5300, in the presence of low dose of follicle stimulating hormone, there were no differences in oocyte number, fertilization rate, and hatched blastocyst rate in mice with TOP5300 and low dose follicle stimulating hormone vs. reference proteins pregnant mare serum gonadotropin or high dose rec-hFSH. ADME/PK and safety profiles were favorable. In addition, there was no appreciable activity on thyroid hormones by TOP5300 in 14-days toxicological study in rat or dog. The selected lead compound, TOP5300 stimulated a more robust increase in estradiol production from granulosa-lutein cells from women with polycystic ovarian syndrome patient compared to rec-hFSH. Conclusions: Two novel oral FSHR allosteric agonist, TOP5668 and TOP5300, were found to mimic the biological activity of rec hFSH in preclinical studies. Both compounds led to folliculogenesis and superovulation in rat and mice. Specifically, TOP5300 led to a similar number of ovulated oocytes that fertilized and developed into hatched blastocysts in mice when compared to rec-hFSH. The safety profile demonstrated lack of toxicity.
Collapse
Affiliation(s)
| | | | - Joie Guner
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Stephen Palmer
- TocopheRx, Inc., Groton, MA, United States
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
17
|
Tokmakov AA, Stefanov VE, Sato KI. Dissection of the Ovulatory Process Using ex vivo Approaches. Front Cell Dev Biol 2020; 8:605379. [PMID: 33363163 PMCID: PMC7755606 DOI: 10.3389/fcell.2020.605379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Ovulation is a unique physiological phenomenon that is essential for sexual reproduction. It refers to the entire process of ovarian follicle responses to hormonal stimulation resulting in the release of mature fertilization-competent oocytes from the follicles and ovaries. Remarkably, ovulation in different species can be reproduced out-of-body with high fidelity. Moreover, most of the molecular mechanisms and signaling pathways engaged in this process have been delineated using in vitro ovulation models. Here, we provide an overview of the major molecular and cytological events of ovulation observed in frogs, primarily in the African clawed frog Xenopus laevis, using mainly ex vivo approaches, with the focus on meiotic oocyte maturation and follicle rupture. For the purpose of comparison and generalization, we also refer extensively to ovulation in other biological species, most notoriously, in mammals.
Collapse
Affiliation(s)
| | - Vasily E Stefanov
- Department of Biochemistry, Saint Petersburg State University, Saint Petersburg, Russia
| | - Ken-Ichi Sato
- Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| |
Collapse
|
18
|
Tremblay PG, Sirard MA. Gene analysis of major signaling pathways regulated by gonadotropins in human ovarian granulosa tumor cells (KGN)†. Biol Reprod 2020; 103:583-598. [PMID: 32427331 DOI: 10.1093/biolre/ioaa079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/17/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
The female reproductive function largely depends on timing and coordination between follicle-stimulating hormone (FSH) and luteinizing hormone. Even though it was suggested that these hormones act on granulosa cells via shared signaling pathways, mainly protein kinases A, B, and C (PKA, PKB, and PKC), there is still very little information available on how these signaling pathways are regulated by each hormone to provide such differences in gene expression throughout folliculogenesis. To obtain a global picture of the principal upstream factors involved in PKA, PKB, and PKC signaling in granulosa cells, human granulosa-like tumor cells (KGN) were treated with FSH or specific activators (forskolin, SC79, and phorbol 12-myristate 13-acetate) for each pathway to analyze gene expression with RNA-seq technology. Normalization and cutoffs (FC 1.5, P ≤ 0.05) revealed 3864 differentially expressed genes between treatments. Analysis of major upstream regulators showed that PKA is a master kinase of early cell differentiation as its activation resulted in the gene expression profile that accompanies granulosa cell differentiation. Our data also revealed that the activation of PKC in granulosa cells is also a strong differentiation signal that could control "advanced" differentiation in granulosa cells and the inflammatory cascade that occurs in the dominant follicle. According to our results, PKB activation provides support for PKA-stimulated gene expression and is also involved in granulosa cell survival throughout follicular development. Taken together, our results provide new information on PKA, PKB, and PKC signaling pathways and their roles in stimulating a follicle at the crossroad between maturation/ovulation and atresia.
Collapse
Affiliation(s)
- Patricia G Tremblay
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marc-André Sirard
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
19
|
Utomo B, Putranto ED, Fadholly A. Profile of follicle-stimulating hormone and polymorphism of follicle-stimulating hormone receptor in Madrasin cattle with ovarian hypofunction. Vet World 2020; 13:879-883. [PMID: 32636582 PMCID: PMC7311873 DOI: 10.14202/vetworld.2020.879-883] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 04/07/2020] [Indexed: 11/16/2022] Open
Abstract
Background and Aim The follicle-stimulating hormone (FSH) gene is an essential regulator of fertility in livestock. This study aims to provide information on the genetic makeup of Madrasin cattle experiencing hypofunction by the FSH profile and FSH receptors (FSHR) polymorphism. Materials and Methods Blood samples were collected from the Bangkalan regency in Indonesia. DNA was isolated and purified following the extraction protocol of polymerase chain reaction (PCR) and PCR-restriction fragment length polymorphism. Results Our results showed that the FSH gene had a band length of 310 bp and produce two alleles (A and B) with restriction enzymes at 250 bp, 230 bp, and 145 bp. Furthermore, the FSHR gene had a band length of 303 bp and produced two homozygous genotypes: GG at bp 239 and CC at bp 188. Conclusion Based on these differences, there was no change in allele frequency and genotype between Madura and Madrasin cattle due to crossbreeding with Limousin cattle. Thus, further detailed investigations of Madrasin cattle are required to elucidate the profile of the LH and LHR genes.
Collapse
Affiliation(s)
- Budi Utomo
- Department of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Emmanuel Djoko Putranto
- Department of Veterinary Clinical, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Amaq Fadholly
- Department of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
20
|
Coss D. Commentary on the Recent FSH Collection: Known Knowns and Known Unknowns. Endocrinology 2020; 161:5683667. [PMID: 31865385 PMCID: PMC6986550 DOI: 10.1210/endocr/bqz035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Follicle-stimulating hormone (FSH) is a dimeric glycoprotein secreted by the anterior pituitary gonadotrope that is necessary for reproductive function in mammals. FSH primarily regulates granulosa cells and follicular growth in females, and Sertoli cell function in males. Since its identification in the 1930s and sequencing in the 1970s, significant progress has been made in elucidating its regulation and downstream function. Recent advances provide deeper insight into FSH synthesis, and effects in the gonads suggest potential roles in extragonadal tissues and examine pharmacological approaches and clinical applications in infertility treatment that now affect 18% of couples. These advances were discussed in detail in a number of reviews published in the last 2 years in Endocrinology. In this brief commentary, we summarize these reviews and point to the outstanding questions that should be answered in the near future to bridge a gap in our understanding of this hormone.
Collapse
Affiliation(s)
- Djurdjica Coss
- Division of Biomedical Sciences; School of Medicine, University of California, Riverside; Riverside, California
- Correspondence: Djurdjica Coss, Division of Biomedical Sciences, School of Medicine, University of California, Riverside; Riverside, California. E-mail:
| |
Collapse
|
21
|
Cannarella R, Arato I, Condorelli RA, Mongioì LM, Lilli C, Bellucci C, La Vignera S, Luca G, Mancuso F, Calogero AE. Effects of Insulin on Porcine Neonatal Sertoli Cell Responsiveness to FSH In Vitro. J Clin Med 2019; 8:jcm8060809. [PMID: 31174276 PMCID: PMC6617126 DOI: 10.3390/jcm8060809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
There is ongoing debate as to whether the decline of sperm production in recent times may be related to a parallel increase in the rate of obesity and diabetes. Lower anti-Müllerian hormone (AMH) and inhibin B secretion have been observed in young hyperinsulinemic patients compared to healthy controls, suggesting a Sertoli cell (SC) dysfunction. The pathophysiological mechanisms underlying SC dysfunction in these patients are poorly understood. To the best of our knowledge, no evidence is available on the effects of insulin on SC function. Therefore, this study was undertaken to assess the effects of insulin on basal and follicle-stimulating hormone (FSH)-stimulated SC function in vitro. To accomplish this, we evaluated the expression of AMH, inhibin B and FSHR genes, the secretion of AMH and inhibin B and the phosphorylation of AKT473 and SC proliferation on neonatal porcine SC after incubation with FSH and/or insulin. We found that similar to FSH, the expression and secretion of AMH is suppressed by insulin. Co-incubation with FSH and insulin decreased AMH secretion significantly more than with FSH alone. Insulin had no effect on the expression and secretion of the inhibin B gene, but co-incubation with FSH and insulin had a lower effect on inhibin B secretion than that found with FSH alone. FSH and/or insulin increased AKT473 phosphorylation and SC proliferation. In conclusion, the results of this study showed that insulin modulates SC function. We hypothesize that hyperinsulinemia may therefore influence testicular function even before puberty begins. Therefore, particular care should be taken to avoid the onset of hyperinsulinemia in children to prevent a future deleterious effect on fertility.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
22
|
Cannarella R, Arato I, Condorelli RA, Luca G, Barbagallo F, Alamo A, Bellucci C, Lilli C, La Vignera S, Calafiore R, Mancuso F, Calogero AE. The IGF1 Receptor Is Involved in Follicle-Stimulating Hormone Signaling in Porcine Neonatal Sertoli Cells. J Clin Med 2019; 8:jcm8050577. [PMID: 31035547 PMCID: PMC6571966 DOI: 10.3390/jcm8050577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Experimental evidence has shown that the IGF1 receptor (IGF1R) is involved in testicular development during embryogenesis. More recently, data gathered from mice granulosa cells and zebrafish spermatogonia suggest that IGF1R has a role in Follicle-stimulating hormone (FSH) signaling. No evidence has been reported on this matter in Sertoli cells (SCs) so far. The aim of the study was to evaluate the role, if any, of the IGF1R in FSH signaling in SCs. The effects of FSH exposure on myosin-phosphatase 1 (MYPT1), ERK 1/2, AKT308, AKT473, c-Jun N-terminal kinase (JNK) phosphorylation and on anti-Müllerian hormone (AMH), inhibin B and FSH receptor (FSHR) mRNA levels were assessed with and without the IGF1R inhibitor NVP-AEW541 in purified and functional porcine neonatal SCs. Pre-treatment with NVP-AEW541 inhibited the FSH-induced MYPT1 and ERK 1/2 phosphorylation, decreased the FSH-dependent Protein kinase B (AKT)308 phosphorylation, but did not affect the FSH-induced AKT473 and JNK phosphorylation rate. It also interfered with the FSH-induced AMH and FSHR down-regulation. No influence was observed on the FSH-stimulated Inhibin B gene expression. Conclusion. These findings support the role of theIGF1R in FSH signaling in porcine SCs. The possible influence of IGF1 stimulation on the FSH-mediated effects on SCs should be further explored.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Federica Barbagallo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Angela Alamo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | - Catia Bellucci
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Cinzia Lilli
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| | | | - Francesca Mancuso
- Department of Experimental Medicine, University of Perugia, 06123 Perugia, Italy.
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy.
| |
Collapse
|
23
|
Lai KP, Wang SY, Li JW, Tong Y, Chan TF, Jin N, Tse A, Zhang JW, Wan MT, Tam N, Au DWT, Lee BY, Lee JS, Wong AST, Kong RYC, Wu RSS. Hypoxia Causes Transgenerational Impairment of Ovarian Development and Hatching Success in Fish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:3917-3928. [PMID: 30844260 DOI: 10.1021/acs.est.8b07250] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Hypoxia is a pressing environmental problem in both marine and freshwater ecosystems globally, and this problem will be further exacerbated by global warming in the coming decades. Recently, we reported that hypoxia can cause transgenerational impairment of sperm quality and quantity in fish (in F0, F1, and F2 generations) through DNA methylome modifications. Here, we provide evidence that female fish ( Oryzias melastigma) exposed to hypoxia exhibit reproductive impairments (follicle atresia and retarded oocyte development), leading to a drastic reduction in hatching success in the F2 generation of the transgenerational group, although they have never been exposed to hypoxia. Further analyses show that the observed transgenerational impairments in ovarian functions are related to changes in the DNA methylation and expression pattern of two gene clusters that are closely associated with stress-induced cell cycle arrest and cell apoptosis. The observed epigenetic and transgenerational alterations suggest that hypoxia may pose a significant threat to the sustainability of natural fish populations.
Collapse
Affiliation(s)
- Keng Po Lai
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
- State Key Laboratory of Marine Pollution , The City University of Hong Kong , Hong Kong SAR , China
| | - Simon Yuan Wang
- Division of Newborn Medicine , Children's Hospital Boston , 300 Longwood Avenue , Boston , Massachusetts 02115 , United States
- Department of Pediatrics , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jing Woei Li
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
| | - Yin Tong
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Ting Fung Chan
- School of Life Sciences , The Chinese University of Hong Kong , Hong Kong SAR , China
- Partner State Key Laboratory of Agrobiotechnology , The Chinese University of Hong Kong , Hong Kong SAR , China
| | - Nana Jin
- School of Life Sciences , The Chinese University of Hong Kong , Hong Kong SAR , China
| | - Anna Tse
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Jiang Wen Zhang
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Miles Teng Wan
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
| | - Nathan Tam
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
| | - Doris Wai Ting Au
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
- State Key Laboratory of Marine Pollution , The City University of Hong Kong , Hong Kong SAR , China
| | - Bo-Young Lee
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon , South Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science , Sungkyunkwan University , Suwon , South Korea
| | - Alice Sze Tsai Wong
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Richard Yuen Chong Kong
- Department of Chemistry , The City University of Hong Kong , Hong Kong SAR , China
- State Key Laboratory of Marine Pollution , The City University of Hong Kong , Hong Kong SAR , China
| | - Rudolf Shiu Sun Wu
- State Key Laboratory of Marine Pollution , The City University of Hong Kong , Hong Kong SAR , China
- Department of Science and Environmental Studies , The Education University of Hong Kong , Hong Kong SAR , China
| |
Collapse
|
24
|
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
|
25
|
Abedel-Majed MA, Romereim SM, Davis JS, Cupp AS. Perturbations in Lineage Specification of Granulosa and Theca Cells May Alter Corpus Luteum Formation and Function. Front Endocrinol (Lausanne) 2019; 10:832. [PMID: 31849844 PMCID: PMC6895843 DOI: 10.3389/fendo.2019.00832] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/14/2019] [Indexed: 12/31/2022] Open
Abstract
Anovulation is a major cause of infertility, and it is the major leading reproductive disorder in mammalian females. Without ovulation, an oocyte is not released from the ovarian follicle to be fertilized and a corpus luteum is not formed. The corpus luteum formed from the luteinized somatic follicular cells following ovulation, vasculature cells, and immune cells is critical for progesterone production and maintenance of pregnancy. Follicular theca cells differentiate into small luteal cells (SLCs) that produce progesterone in response to luteinizing hormone (LH), and granulosa cells luteinize to become large luteal cells (LLCs) that have a high rate of basal production of progesterone. The formation and function of the corpus luteum rely on the appropriate proliferation and differentiation of both granulosa and theca cells. If any aspect of granulosa or theca cell luteinization is perturbed, then the resulting luteal cell populations (SLC, LLC, vascular, and immune cells) may be reduced and compromise progesterone production. Thus, many factors that affect the differentiation/lineage of the somatic cells and their gene expression profiles can alter the ability of a corpus luteum to produce the progesterone critical for pregnancy. Our laboratory has identified genes that are enriched in somatic follicular cells and luteal cells through gene expression microarray. This work was the first to compare the gene expression profiles of the four somatic cell types involved in the follicle-to-luteal transition and to support previous immunofluorescence data indicating theca cells differentiate into SLCs while granulosa cells become LLCs. Using these data and incorporating knowledge about the ways in which luteinization can go awry, we can extrapolate the impact that alterations in the theca and granulosa cell gene expression profiles and lineages could have on the formation and function of the corpus luteum. While interactions with other cell types such as vascular and immune cells are critical for appropriate corpus luteum function, we are restricting this review to focus on granulosa, theca, and luteal cells and how perturbations such as androgen excess and inflammation may affect their function and fertility.
Collapse
Affiliation(s)
| | - Sarah M. Romereim
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - John S. Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, United States
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Andrea S. Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
- *Correspondence: Andrea S. Cupp
| |
Collapse
|
26
|
Ivell R, Anand-Ivell R. Insulin-like peptide 3 (INSL3) is a major regulator of female reproductive physiology. Hum Reprod Update 2018; 24:639-651. [DOI: 10.1093/humupd/dmy029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/31/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Richard Ivell
- School of Biosciences, University of Nottingham, Sutton Bonington, UK
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| | | |
Collapse
|
27
|
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: 70] [Impact Index Per Article: 10.0] [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
|
28
|
Ogundele OM, Pardo J, Francis J, Goya RG, Lee CC. A Putative Mechanism of Age-Related Synaptic Dysfunction Based on the Impact of IGF-1 Receptor Signaling on Synaptic CaMKIIα Phosphorylation. Front Neuroanat 2018; 12:35. [PMID: 29867375 PMCID: PMC5960681 DOI: 10.3389/fnana.2018.00035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 04/18/2018] [Indexed: 01/13/2023] Open
Abstract
Insulin-like growth factor 1 receptor (IGF-1R) signaling regulates the activity and phosphorylation of downstream kinases linked to inflammation, neurodevelopment, aging and synaptic function. In addition to the control of Ca2+ currents, IGF-1R signaling modulates the activity of calcium-calmodulin-dependent kinase 2 alpha (CaMKIIα) and mitogen activated protein kinase (MAPK/ErK) through multiple signaling pathways. These proteins (CaMKIIα and MAPK) regulate Ca2+ movement and long-term potentiation (LTP). Since IGF-1R controls the synaptic activity of Ca2+, CaMKIIα and MAPK signaling, the possible mechanism through which an age-dependent change in IGF-1R can alter the synaptic expression and phosphorylation of these proteins in aging needs to be investigated. In this study, we evaluated the relationship between an age-dependent change in brain IGF-1R and phosphorylation of CaMKIIα/MAPK. Furthermore, we elucidated possible mechanisms through which dysregulated CaMKIIα/MAPK interaction may be linked to a change in neurotransmitter processing and synaptic function. Male C57BL/6 VGAT-Venus mice at postnatal days 80 (P80), 365 and 730 were used to study age-related neural changes in two brain regions associated with cognitive function: hippocampus and prefrontal cortex (PFC). By means of high throughput confocal imaging and quantitative immunoblotting, we evaluated the distribution and expression of IGF-1, IGF-1R, CaMKIIα, p-CaMKIIα, MAPK and p-MAPK in whole brain lysate, hippocampus and cortex. Furthermore, we compared protein expression patterns and regional changes at P80, P365 and P730. Ultimately, we determined the relative phosphorylation pattern of CaMKIIα and MAPK through quantification of neural p-CaMKIIα and p-MAPK/ErK, and IGF-1R expression for P80, P365 and P730 brain samples. In addition to a change in synaptic function, our results show a decrease in neural IGF-1/IGF-1R expression in whole brain, hippocampus and cortex of aged mice. This was associated with a significant upregulation of phosphorylated neural MAPK (p-MAPK) and decrease in total brain CaMKIIα (i.e., CaMKIIα and p-CaMKIIα) in the aged brain. Taken together, we showed that brain aging is associated with a change in neural IGF-1/IGF-1R expression and may be linked to a change in phosphorylation of synaptic kinases (CaMKIIα and MAPK) that are involved in the modulation of LTP.
Collapse
Affiliation(s)
- Olalekan M. Ogundele
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Joaquin Pardo
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Rodolfo G. Goya
- Institute for Biochemical Research of La Plata, School of Medicine, National University of La Plata, La Plata, Argentina
| | - Charles C. Lee
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
29
|
Depression promotes prostate cancer invasion and metastasis via a sympathetic-cAMP-FAK signaling pathway. Oncogene 2018. [PMID: 29515233 DOI: 10.1038/s41388-018-0177-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Depression drives cancer progression and induces poor clinical outcome. However, the mechanisms underlying depression and cancer outcomes are unclear. In this work, we investigated 98 prostate cancer patients and found that patients with high score of psychological depression were correlated with tumor invasion and metastasis. We found focal adhesion kinase (FAK) was increased in cancer patients with metastatic features and high score of depression. FAK knockdown completely blocked depression-promoted tumor invasion in orthotopic transplantation tumors. In Hi-myc mice and a murine model of depression, sympathetic activation was detected in the prostate tissue. Further we showed that FAK activation was dependent on a cAMP-PKA signaling pathway. Our results demonstrated that the activation of a sympathetic-FAK signaling pathway in prostate cancer patients with high degrees of depression facilitates tumor invasion. We suggest that blocking β2AR with propranolol or inhibiting FAK activation with PF562 271 may be novel strategies for depressed patients with invasive prostate cancer.
Collapse
|
30
|
Cannarella R, Condorelli RA, La Vignera S, Calogero AE. Effects of the insulin-like growth factor system on testicular differentiation and function: a review of the literature. Andrology 2017; 6:3-9. [PMID: 29195026 DOI: 10.1111/andr.12444] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
We recently described the occurrence of cryptorchidism, oligoasthenoteratozoospermia, and genital abnormalities in patients with distal 15q chromosome structural abnormalities. This observation brought us to hypothesize that insulin-like growth factor (IGF) receptor (IGF1R), mapping on the 15q 26.3 chromosomal band, may be involved in testicular function. To further evaluate this topic, we reviewed in vitro and in vivo studies exploring the role of the IGF system [IGF1, IGF2, IGF1R, insulin receptor substrates (IRS)] at the testicular level both in animals and in humans. In animals, IGF1/IGF1R has been found to be involved in testicular development during embryogenesis, in Sertoli cell (SC) proliferation, and in germ cell (GS) proliferation and differentiation. Interestingly, IGF1R seems to mediate follicle-stimulating hormone (FSH) effects through the PI3K/AKT pathway. In humans, IGF1 directly increases testicular volume. The molecular pathways responsible for testicular differentiation and IGF1/IGF1R signaling are highly conserved among species; therefore, the IGF system may be involved in FSH signaling also in humans. We suggest a possible molecular pathway occurring in human SCs, which involves both IGF1 and FSH through the PI3K/AKT pathway. The acknowledgment of an IGF1 mediation of the FSH-induced effects may open new ways for a targeted therapy in idiopathic non-FSH-responder oligoasthenoteratozoospermia.
Collapse
Affiliation(s)
- R Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - R A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - S La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - A E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|