1
|
Huhtaniemi IT. Luteinizing hormone receptor knockout mouse: What has it taught us? Andrology 2025. [PMID: 39840520 DOI: 10.1111/andr.70000] [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: 11/06/2024] [Revised: 12/19/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
Luteinizing hormone (LH), along with its agonist choriongonadotropin (hCG) in humans, is the key hormone responsible for the tropic regulation of the gonadal function. LH and hCG act through their cognate receptor, the luteinizing hormone/choriongonadotropin receptor (LHCGR; more appropriately LHR in rodents lacking CG), located in the testis in Leydig cells and in the ovary in theca, luteal, and luteinizing granulosa cells. Low levels in LHCGR are also expressed in numerous extragonadal sites. Hypogonadism is observed in humans expressing inactivating mutations in the LHβ-subunit (LHB)and LHCGR genes, confirming the crucial role of LH and LHCGR in gonadal development and function. Unraveling of the LHR structure and the advent of gene manipulation techniques enabled the production of mouse models with inactivated LHR function, that is, the LHR knockout (LuRKO) mouse, some 20 years ago. This mouse model has thereafter been instrumental in various experimental settings, alone or combined with other genetically modified mouse models, in providing novel, and in some cases unexpected, details about the LH/LHR function. We will review here the salient findings of these studies.
Collapse
Affiliation(s)
- Ilpo T Huhtaniemi
- Department of Digestion, Metabolism and Reproduction, Institute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, UK
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu, Turku, Finland
| |
Collapse
|
2
|
Biomediators in Polycystic Ovary Syndrome and Cardiovascular Risk. Biomolecules 2021; 11:biom11091350. [PMID: 34572562 PMCID: PMC8467803 DOI: 10.3390/biom11091350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is extremely heterogeneous in terms of clinical manifestations. The variability of the syndrome's phenotype is derived from the genetic and molecular heterogeneity, with a great deal of environmental factors that may have long-term health consequences, such as metabolic and cardiovascular (CV) diseases. There is no doubt that women with PCOS suffer from metabolic complications more than their age-matched counterparts in the general population and at an earlier age. Obesity, low steroid hormone-binding globulin (SHBG), hyperandrogenemia, insulin resistance, and compensatory hyperinsulinemia are biomediators and early predictors of metabolic complications in PCOS. Doubts remain about the real risk of CV diseases in PCOS and the molecular mechanisms at the basis of CV complications. Based on that assumption, this review will present the available evidence on the potential implications of some biomediators, in particular, hyperandrogenism, estrogen-progesterone imbalance, insulin resistance, and low SHBG, in the processes leading to CV disease in PCOS, with the final aim to propose a more accurate CV risk assessment.
Collapse
|
3
|
Juel Mortensen L, Lorenzen M, Jørgensen A, Albrethsen J, Jørgensen N, Møller S, Andersson AM, Juul A, Blomberg Jensen M. Possible Relevance of Soluble Luteinizing Hormone Receptor during Development and Adulthood in Boys and Men. Cancers (Basel) 2021; 13:cancers13061329. [PMID: 33809538 PMCID: PMC7999540 DOI: 10.3390/cancers13061329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary The reproductive hormones luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are both agonists for the luteinizing hormone receptor (LHCGR) and essential for male reproduction during development and adulthood. LHCGR is expressed and stimulates testosterone production from the testicular Leydig cells. In this study, we demonstrate the presence of soluble LHCGR in blood, urine, and seminal fluid in both healthy boys and men, and patients with aberrations in sex-chromosomes. We show how circulating levels of sLHCGR are associated with pubertal development, testicular function, and semen quality and demonstrate that LHCGR is released from fetal human non-gonadal tissue. sLHCGR is released into serum by testis and other organs, which suggests possible extra-gonadal effects of LH or hCG in boys and men. Abstract Luteinizing hormone (LH) and human chorionic gonadotropin (hCG) are agonists for the luteinizing hormone receptor (LHCGR) which regulates male reproductive function. LHCGR may be released into body fluids. We wish to determine whether soluble LHCGR is a marker for gonadal function. Cross-sectional, longitudinal, and intervention studies on 195 healthy boys and men and 396 men with infertility, anorchia, or Klinefelter Syndrome (KS) were used to correlate LHCGR measured in serum, seminal fluid, urine, and hepatic/renal artery and vein with gonadal function. LHCGR was determined in fluids from in vitro and in vivo models of human testicular tissue and cell lines, xenograft mouse models, and human fetal kidney and adrenal glands. Western blot showed LHCGR fragments in serum and gonadal tissue of similar size using three different antibodies. The LHCGR-ELISA had no species cross-reactivity or unspecific reaction in mouse serum even after human xenografting. Instead, sLHCGR was released into the media after the culture of a human fetal kidney and adrenal glands. Serum sLHCGR decreased markedly during puberty in healthy boys (p = 0.0001). In healthy men, serum sLHCGR was inversely associated with the Inhibin B/FSH ratio (β −0.004, p = 0.027). In infertile men, seminal fluid sLHCGR was inversely associated with serum FSH (β 0.006, p = 0.009), sperm concentration (β −3.5, p = 0.003) and total sperm count (β −3.2, p = 0.007). The injection of hCG lowered sLHCGR in serum and urine of healthy men (p < 0.01). In conclusion, sLHCGR is released into body-fluids and linked with pubertal development and gonadal function. Circulating sLHCGR in anorchid men suggests that sLHCGR in serum may originate from and possibly exert actions in non-gonadal tissues. (ClinicalTrials: NTC01411527, NCT01304927, NCT03418896).
Collapse
Affiliation(s)
- Li Juel Mortensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
| | - Mette Lorenzen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
| | - Anne Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Jakob Albrethsen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Niels Jørgensen
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Søren Møller
- Center for Functional and Diagnostic Imaging and Research, Department of Clinical Physiology and Nuclear Medicine 260, Hvidovre Hospital, 2650 Copenhagen, Denmark;
- Department of Clinical Medicine, Faculty of Health Sciences, Copenhagen University, 2200 Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
| | - Anders Juul
- Department of Growth and Reproduction and International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; (A.J.); (J.A.); (N.J.); (A.-M.A.); (A.J.)
- Department of Clinical Medicine, Faculty of Health Sciences, Copenhagen University, 2200 Copenhagen, Denmark
| | - Martin Blomberg Jensen
- Group of Skeletal, Mineral and Gonadal Endocrinology, University Department of Growth and Reproduction, Rigshospitalet, 2100 Copenhagen, Denmark; (L.J.M.); (M.L.)
- Division of Bone and Mineral Research, Harvard School of Dental Medicine/Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +45-3545-5064
| |
Collapse
|
4
|
Lobo J, Leão R, Jerónimo C, Henrique R. Liquid Biopsies in the Clinical Management of Germ Cell Tumor Patients: State-of-the-Art and Future Directions. Int J Mol Sci 2021; 22:ijms22052654. [PMID: 33800799 PMCID: PMC7961393 DOI: 10.3390/ijms22052654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Liquid biopsies constitute a minimally invasive means of managing cancer patients, entailing early diagnosis, follow-up and prediction of response to therapy. Their use in the germ cell tumor field is invaluable since diagnostic tissue biopsies (which are invasive) are often not performed, and therefore only a presumptive diagnosis can be made, confirmed upon examination of the surgical specimen. Herein, we provide an overall review of the current liquid biopsy-based biomarkers of this disease, including the classical, routinely used serum tumor markers—the promising microRNAs rapidly approaching the introduction into clinical practice—but also cell-free DNA markers (including DNA methylation) and circulating tumor cells. Finally, and importantly, we also explore novel strategies and challenges for liquid biopsy markers and methodologies, providing a critical view of the future directions for liquid biopsy tests in this field, highlighting gaps and unanswered questions.
Collapse
Affiliation(s)
- João Lobo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Ricardo Leão
- Faculty of Medicine, University of Coimbra, Rua Larga, 3000-370 Coimbra, Portugal;
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
- Correspondence: (C.J.); (R.H.); Tel.: +351-22-225084000 (C.J. & R.H.); Fax: +351-22-5084199 (C.J. & R.H.)
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
- Correspondence: (C.J.); (R.H.); Tel.: +351-22-225084000 (C.J. & R.H.); Fax: +351-22-5084199 (C.J. & R.H.)
| |
Collapse
|
5
|
Mondaca JM, Uzair ID, Castro Guijarro AC, Flamini MI, Sanchez AM. Molecular Basis of LH Action on Breast Cancer Cell Migration and Invasion via Kinase and Scaffold Proteins. Front Cell Dev Biol 2021; 8:630147. [PMID: 33614634 PMCID: PMC7893099 DOI: 10.3389/fcell.2020.630147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/24/2020] [Indexed: 11/13/2022] Open
Abstract
Breast cancer (BC) is a major public health problem affecting women worldwide. Approximately 80% of diagnosed cases are hormone-dependent breast cancers. These hormones are known to stimulate tumor development and progression. In this setting, tentative evidence suggests that luteinizing hormone (LH) may also play a role in tumors. In BC cells that express functional LH receptors (LHR), this hormone regulates cell migration and invasion by controlling several kinases that activate actin cytoskeletal proteins. In this article, we show that LH induces phosphorylation of paxillin and its translocation toward the plasmatic membrane, where focal adhesion complexes are assembled. This process is triggered via a rapid extra-gonadal LHR signaling to Src/FAK/paxillin, which results in the phosphorylation/activation of the nucleation promoter factors cortactin and N-WASP. As a consequence, Arp2/3 complexes induce actin polymerization, essential to promote cell adhesion, migration, and invasion, thus enhancing metastatic spread of tumoral cells. Our findings provide relevant information about how gonadotrophins exert their action in BC. This information helps us understand the extragonadal effects of LH on BC metastasis. It may provide new perspectives for therapeutic treatment, especially for women with high serum levels of gonadotrophins.
Collapse
Affiliation(s)
- Joselina Magali Mondaca
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ivonne Denise Uzair
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ana Carla Castro Guijarro
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marina Inés Flamini
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Angel Matias Sanchez
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
6
|
Albrecht ED, Pepe GJ. Regulation of Uterine Spiral Artery Remodeling: a Review. Reprod Sci 2020; 27:1932-1942. [PMID: 32548805 PMCID: PMC7452941 DOI: 10.1007/s43032-020-00212-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/06/2020] [Indexed: 12/31/2022]
Abstract
Extravillous trophoblast remodeling of the uterine spiral arteries is essential for promoting blood flow to the placenta and fetal development, but little is known about the regulation of this process. A defect in spiral artery remodeling underpins adverse conditions of human pregnancy, notably early-onset preeclampsia and fetal growth restriction, which result in maternal and fetal morbidity and mortality. Many in vitro studies have been conducted to determine the ability of growth and other factors to stimulate trophoblast cells to migrate across a synthetic membrane. Clinical studies have investigated whether the maternal levels of various factors are altered during abnormal human pregnancy. Animal models have been established to assess the ability of various factors to recapitulate the pathophysiological symptoms of preeclampsia. This review analyzes the results of the in vitro, clinical, and animal studies and describes a nonhuman primate experimental paradigm of defective uterine artery remodeling to study the regulation of vessel remodeling.
Collapse
Affiliation(s)
- Eugene D Albrecht
- Bressler Research Laboratories, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, 655 West Baltimore St., Baltimore, MD, USA.
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
7
|
Fujiwara H, Ono M, Sato Y, Imakawa K, Iizuka T, Kagami K, Fujiwara T, Horie A, Tani H, Hattori A, Daikoku T, Araki Y. Promoting Roles of Embryonic Signals in Embryo Implantation and Placentation in Cooperation with Endocrine and Immune Systems. Int J Mol Sci 2020; 21:ijms21051885. [PMID: 32164226 PMCID: PMC7084435 DOI: 10.3390/ijms21051885] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Embryo implantation in the uterus is an essential process for successful pregnancy in mammals. In general, the endocrine system induces sufficient embryo receptivity in the endometrium, where adhesion-promoting molecules increase and adhesion-inhibitory molecules decrease. Although the precise mechanisms remain unknown, it is widely accepted that maternal–embryo communications, including embryonic signals, improve the receptive ability of the sex steroid hormone-primed endometrium. The embryo may utilize repulsive forces produced by an Eph–ephrin system for its timely attachment to and subsequent invasion through the endometrial epithelial layer. Importantly, the embryonic signals are considered to act on maternal immune cells to induce immune tolerance. They also elicit local inflammation that promotes endometrial differentiation and maternal tissue remodeling during embryo implantation and placentation. Additional clarification of the immune control mechanisms by embryonic signals, such as human chorionic gonadotropin, pre-implantation factor, zona pellucida degradation products, and laeverin, will aid in the further development of immunotherapy to minimize implantation failure in the future.
Collapse
Affiliation(s)
- Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8641, Japan; (M.O.); (T.I.); (K.K.)
- Correspondence: or ; Tel.: +81-(0)76-265-2425; Fax: +81-(0)76-234-4266
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8641, Japan; (M.O.); (T.I.); (K.K.)
| | - Yukiyasu Sato
- Department of Obstetrics and Gynecology, Takamatsu Red Cross Hospital, Takamatsu 760-0017, Japan;
| | - Kazuhiko Imakawa
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan;
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8641, Japan; (M.O.); (T.I.); (K.K.)
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8641, Japan; (M.O.); (T.I.); (K.K.)
| | - Tomoko Fujiwara
- Department of Home Science and Welfare, Kyoto Notre Dame University, Kyoto 606-0847, Japan;
| | - Akihito Horie
- Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (A.H.); (H.T.)
| | - Hirohiko Tani
- Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (A.H.); (H.T.)
| | - Akira Hattori
- Department of System Chemotherapy and Molecular Sciences, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto 606-8501, Japan;
| | - Takiko Daikoku
- Division of Transgenic Animal Science, Advanced Science Research Center, Kanazawa University, Kanazawa 920-8640, Japan;
| | - Yoshihiko Araki
- Institute for Environmental and Gender-specific Medicine, Juntendo University Graduate School of Medicine, Urayasu 279-0021, Japan;
- Department of Obstetrics and Gynecology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
8
|
Ibeto L, Antonopoulos A, Grassi P, Pang PC, Panico M, Bobdiwala S, Al-Memar M, Davis P, Davis M, Norman Taylor J, Almeida P, Johnson MR, Harvey R, Bourne T, Seckl M, Clark G, Haslam SM, Dell A. Insights into the hyperglycosylation of human chorionic gonadotropin revealed by glycomics analysis. PLoS One 2020; 15:e0228507. [PMID: 32045434 PMCID: PMC7012436 DOI: 10.1371/journal.pone.0228507] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/16/2020] [Indexed: 12/13/2022] Open
Abstract
Human chorionic gonadotropin (hCG) is a glycoprotein hormone that is essential for the maintenance of pregnancy. Glycosylation of hCG is known to be essential for its biological activity. "Hyperglycosylated" variants secreted during early pregnancy have been proposed to be involved in initial implantation of the embryo and as a potential diagnostic marker for gestational diseases. However, what constitutes "hyperglycosylation" is not yet fully understood. In this study, we perform comparative N-glycomic analysis of hCG expressed in the same individuals during early and late pregnancy to help provide new insights into hCG function, reveal new targets for diagnostics and clarify the identity of hyperglycosylated hCG. hCG was isolated in urine collected from women at 7 weeks and 20 weeks' gestation. hCG was also isolated in urine from women diagnosed with gestational trophoblastic disease (GTD). We used glycomics methodologies including matrix assisted laser desorption/ionisation-time of flight (MALDI-TOF) mass spectrometry (MS) and MS/MS methods to characterise the N-glycans associated with hCG purified from the individual samples. The structures identified on the early pregnancy (EP-hCG) and late pregnancy (LP-hCG) samples corresponded to mono-, bi-, tri-, and tetra-antennary N-glycans. A novel finding was the presence of substantial amounts of bisected type N-glycans in pregnancy hCG samples, which were present at much lower levels in GTD samples. A second novel observation was the presence of abundant LewisX antigens on the bisected N-glycans. GTD-hCG had fewer glycoforms which constituted a subset of those found in normal pregnancy. When compared to EP-hCG, GTD-hCG samples had decreased signals for tri- and tetra-antennary N-glycans. In terms of terminal epitopes, GTD-hCG had increased signals for sialylated structures, while LewisX antigens were of very minor abundance. hCG carries the same N-glycans throughout pregnancy but in different proportions. The N-glycan repertoire is more diverse than previously reported. Bisected and LewisX structures are potential targets for diagnostics. hCG isolated from pregnancy urine inhibits NK cell cytotoxicity in vitro at nanomolar levels and bisected type glycans have previously been implicated in the suppression of NK cell cytotoxicity, suggesting that hCG-related bisected type N-glycans may directly suppress NK cell cytotoxicity.
Collapse
Affiliation(s)
- Linda Ibeto
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | | | - Paola Grassi
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maria Panico
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Shabnam Bobdiwala
- Tommys' National Centre for Miscarriage Research, Queen Charlottes' & Chelsea Hospital, Imperial College, London, United Kingdom
| | - Maya Al-Memar
- Tommys' National Centre for Miscarriage Research, Queen Charlottes' & Chelsea Hospital, Imperial College, London, United Kingdom
| | - Paul Davis
- Mologic LTD, Bedford Technology Park, Bedfordshire, United Kingdom
| | - Mark Davis
- Mologic LTD, Bedford Technology Park, Bedfordshire, United Kingdom
| | - Julian Norman Taylor
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Paula Almeida
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Mark R. Johnson
- Academic Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital, London, United Kingdom
| | - Richard Harvey
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Tom Bourne
- Tommys' National Centre for Miscarriage Research, Queen Charlottes' & Chelsea Hospital, Imperial College, London, United Kingdom
| | - Michael Seckl
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Gary Clark
- Department of Obstetrics, Gynaecology and Women's Health, University of Missouri, Columbia, Missouri, United States of America
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Schumacher A, Zenclussen AC. Human Chorionic Gonadotropin-Mediated Immune Responses That Facilitate Embryo Implantation and Placentation. Front Immunol 2019; 10:2896. [PMID: 31921157 PMCID: PMC6914810 DOI: 10.3389/fimmu.2019.02896] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022] Open
Abstract
Human chorionic gonadotropin (hCG) serves as one of the first signals provided by the embryo to the mother. Exactly at the time when the first step of the implantation process is initiated and the blastocyst adheres to the maternal endometrium, the embryonic tissue starts to actively secrete hCG. Shortly thereafter, the hormone can be detected in the maternal circulation where its concentration steadily increases throughout early pregnancy as it is continuously released by the forming placenta. Accumulating evidence underlines the critical function of hCG for embryo implantation and placentation. hCG not only regulates biological aspects of these early pregnancy events but also supports maternal immune cells in their function as helpers in the establishment of an adequate embryo-endometrial relationship. In view of its early presence in the maternal circulation, hCG has the potential to influence both local uterine immune cell populations as well as peripheral ones. The current review aims to summarize recent literature on the participation of innate and adaptive immune cells in embryo implantation and placentation with a specific focus on their regulation by hCG.
Collapse
Affiliation(s)
- Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
10
|
Meng X, Li X, Xu X, Li P, Chen Y, Fu X, Xu X. Elevated luteinizing hormone contributes to atherosclerosis formation by inhibiting nitric oxide synthesis via PI3K/Akt pathway. Vascul Pharmacol 2019; 121:106582. [PMID: 31437529 DOI: 10.1016/j.vph.2019.106582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/17/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The contentious effects of estrogen therapy on the risk of postmenopausal cardiovascular disease (CVD) indicate that this type of atherosclerosis is not solely induced by estrogen deficiency. Other sex hormones such as elevated luteinizing hormone (LH) may also affect CVD risk in this population. We therefore explored the relationship between LH and atherosclerosis in ovariectomized (OVX) female mice. METHODS Aortic atherosclerotic lesions were assessed in OVX ApoE knock out (ApoE-/-) female mice administered with LH. Human umbilical vascular endothelial cells (HUVECs) were cultured as cell model. The influence of LH on NO release, phosphorylated endothelial nitric oxide synthase (eNOS) and Akt levels were evaluated. Immunoprecipitation and lentiviral particle transfection were applied to assess the role of Gαq on PI3K activity. RESULTS LH increased the atherosclerotic lesion area and carotid artery intima-media thickness (IMT) in OVX ApoE-/- female mice. High levels of LH attenuated vasodilation induced by Ach and inhibited NO release from HUVECs. These effects were related to the findings that LH enhanced interaction between Gαq and p110α, which subsequently inhibited PI3K activity and suppressed the phosphorylation of Akt and eNOS. CONCLUSIONS Elevated LH promotes atherosclerosis formation in OVX ApoE-/- female mice. This effect may be mediated by inhibiting endothelial NO synthesis via PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Xuan Meng
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaosa Li
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xingyan Xu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Ping Li
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yiwen Chen
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| | - Xiaoyang Xu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
11
|
Xu Z, Chen W, Chen C, Xiao Y, Chen X. Effect of intrauterine injection of human chorionic gonadotropin before frozen-thawed embryo transfer on pregnancy outcomes in women with endometriosis. J Int Med Res 2019; 47:2873-2880. [PMID: 31119991 PMCID: PMC6683888 DOI: 10.1177/0300060519848928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the effect of human chorionic gonadotropin (hCG) intrauterine injection before frozen–thawed embryo transfer (FET) in women with endometriosis. Methods This retrospective cohort study included 45 women with endometriosis who underwent hCG intrauterine injection before FET; each woman was matched with three patients with endometriosis who did not receive hCG intrauterine injection (controls). Data on pregnancy and prenatal outcomes were extracted from medical records and compared. Results Patients in the hCG intrauterine injection group had significantly higher rates of pregnancy and clinical pregnancy (64.4% and 57.8%, respectively) than controls (47.4% and 39.3%, respectively). Neonatal birth weight for both singletons and twins was significantly higher in the hCG group (3486 ± 458 g and 2710 ± 437 g, respectively) than in the control group (3195 ± 401 g and 2419 ± 370 g, respectively). Conclusion Pregnancy rate, clinical pregnancy rate, and birth weight were improved in women with endometriosis who underwent intrauterine hCG injection compared with those who did not receive hCG before FET.
Collapse
Affiliation(s)
- Zhihui Xu
- 1 Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wenming Chen
- 2 Department of Obstetrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chune Chen
- 3 School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yikang Xiao
- 3 School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xia Chen
- 1 Reproductive Medicine Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
12
|
Anderson RC, Newton CL, Anderson RA, Millar RP. Gonadotropins and Their Analogs: Current and Potential Clinical Applications. Endocr Rev 2018; 39:911-937. [PMID: 29982442 DOI: 10.1210/er.2018-00052] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022]
Abstract
The gonadotropin receptors LH receptor and FSH receptor play a central role in governing reproductive competency/fertility. Gonadotropin hormone analogs have been used clinically for decades in assisted reproductive therapies and in the treatment of various infertility disorders. Though these treatments are effective, the clinical protocols demand multiple injections, and the hormone preparations can lack uniformity and stability. The past two decades have seen a drive to develop chimeric and modified peptide analogs with more desirable pharmacokinetic profiles, with some displaying clinical efficacy, such as corifollitropin alfa, which is now in clinical use. More recently, low-molecular-weight, orally active molecules with activity at gonadotropin receptors have been developed. Some have excellent characteristics in animals and in human studies but have not reached the market-largely as a result of acquisitions by large pharma. Nonetheless, such molecules have the potential to mitigate risks currently associated with gonadotropin-based fertility treatments, such as ovarian hyperstimulation syndrome and the demands of injection-based therapies. There is also scope for novel use beyond the current remit of gonadotropin analogs in fertility treatments, including application as novel contraceptives; in the treatment of polycystic ovary syndrome; in the restoration of function to inactivating mutations of gonadotropin receptors; in the treatment of ovarian and prostate cancers; and in the prevention of bone loss and weight gain in postmenopausal women. Here we review the properties and clinical application of current gonadotropin preparations and their analogs, as well as the development of novel orally active, small-molecule nonpeptide analogs.
Collapse
Affiliation(s)
- Ross C Anderson
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Claire L Newton
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert P Millar
- Centre for Neuroendocrinology, University of Pretoria, Pretoria, South Africa.,Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
13
|
Koch Y, Wimberger P, Grümmer R. Human chorionic gonadotropin induces decidualization of ectopic human endometrium more effectively than forskolin in an in-vivo endometriosis model. Exp Biol Med (Maywood) 2018; 243:953-962. [PMID: 29886768 PMCID: PMC6108049 DOI: 10.1177/1535370218782658] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/21/2018] [Indexed: 12/28/2022] Open
Abstract
Endometriosis, characterized by the presence of endometrial tissue at ectopic sites, is a leading cause of pelvic pain and subfertility in women. The stromal compartment of the endometrium is considered to play a pivotal role in the establishment and persistence of endometriotic lesions, thus impaired decidualization of these cells may result in enhanced invasion capacity at ectopic sites. Consequently, stimulation of decidualization may alleviate this disease. To analyze the effect of systemically applied compounds on decidualization of ectopic endometrial tissue, endometriosis was induced by suturing human eutopic endometrium to the peritoneum of 22 NOD/SCID mice. Each mouse received four tissue fragments from the same patient. Mice were randomly allocated either to one control and three experimental groups ( n = 4/group) which were treated with progesterone alone or in combination with forskolin or human chorionic gonadotropin for seven days or to one control and one experimental group ( n = 3/group) which was treated with progesterone and human chorionic gonadotropin for 10 days followed by 7 days without treatment. At the end of the experiments, lesions were measured and analyzed for markers of decidualization (FOXO-1, prolactin) and proliferation (Ki-67). Decidualization was induced in the ectopic lesions by systemic treatment in vivo. This induction was significantly stronger after treatment with progesterone in combination with human chorionic gonadotropin than with forskolin or with progesterone alone. Only the combination with human chorionic gonadotropin led to induction of FOXO1 protein expression and a significant physiologic transformation of the ectopic endometrial stromal cells after seven days of treatment. After termination of human chorionic gonadotropin treatment, the decidualization process continued, leading to a significant inhibition of proliferation. Thus, decidualization of human ectopic endometrial tissue can be induced in a humanized endometriosis mouse model in vivo. This model may help to decipher the signal pathways involved in this decidualization process and to develop novel therapeutical approaches to alleviate this painful disease. Impact statement Impaired decidualization of endometrial stromal cells may contribute to the development of endometriosis, and an increased decidualization reaction may prevent or alleviate this prevalent gynecological disease. Human chorionic gonadotropin (hCG) has been shown to promote decidualization in eutopic endometrium. Up to now in vitro studies mainly used cAMP for successful induction of decidualization of isolated endometrial stromal cells. Here, for the first time, decidualization of ectopic endometrial lesions is induced in an experimental endometriosis mouse model, comparing the effectiveness of hCG with that of the direct adenylyl cyclase activator Forskolin. In this 3D-organ structure in vivo, hCG proved to be more effective in the induction of decidualization than forskolin. Particularly in case of progesterone resistance, alternative pathways inducing decidualization could alleviate endometriosis, and the sophisticated hCG action could constitute a therapeutical tool to induce terminal differentiation in ectopic endometrial lesions.
Collapse
Affiliation(s)
- Yvonne Koch
- Institute of Anatomy, University Hospital, Universität Duisburg-Essen, Essen 45147, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Ruth Grümmer
- Institute of Anatomy, University Hospital, Universität Duisburg-Essen, Essen 45147, Germany
| |
Collapse
|
14
|
Shen X, Ma Y, Zeng Q, Huang J, Tao J, Wang L. Ultrasensitive Determination of Human Chorionic Gonadotropin using a Molecularly Imprinted Electrochemical Sensor. ChemistrySelect 2017. [DOI: 10.1002/slct.201700962] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Xiaolei Shen
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| | - Ya Ma
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| | - Qiang Zeng
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| | - Jianzhi Huang
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| | - Jia Tao
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| | - Lishi Wang
- Key Laboratory of Fuel Cell Technology of Guangdong Province School of Chemistry and Chemical Engineering; South China University of Technology, Guangdong Province; P.R. China
| |
Collapse
|
15
|
Steinmetz C, Kashyap A, Zhivkova N, Alizor H, Ernst I, Gottfried-Brand D, Janssen H, Teufel A, Schulze-Bergkamen H, Lotz J, Kuball J, Theobald M, Heise M, Lang H, Galle PR, Strand D, Strand S. Activation of silent mating type information regulation 2 homolog 1 by human chorionic gonadotropin exerts a therapeutic effect on hepatic injury and inflammation. Hepatology 2017; 65:2074-2089. [PMID: 28108987 DOI: 10.1002/hep.29072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 01/12/2017] [Accepted: 01/16/2017] [Indexed: 12/30/2022]
Abstract
UNLABELLED Incidence and prevalence of inflammatory liver diseases has increased over the last years, but therapeutic options are limited. Pregnancy induces a state of immune tolerance, which can result in spontaneous improvement of clinical symptoms of certain autoimmune diseases including autoimmune hepatitis (AIH). We investigated the immune-suppressive mechanisms of the human pregnancy hormone, chorionic gonadotropin (hCG), in the liver. hCG signaling activates silent mating type information regulation 2 homolog 1 (SIRT1), which deacetylates forkhead box o3 (FOXO3a), leading to repression of proapoptotic gene expression, because the immunosuppressive consequence attributed to the absence of caspase-3 activity of hepatocellular interleukin 16 (IL-16) is no longer processed and released. Thus, serum levels of IL-16, a key chemotactic factor for CD4+ lymphocytes, were reduced and migration to injured hepatocytes prevented. Furthermore, elevated IL-16 levels are found in the sera from patients with AIH, hepatitis B virus, hepatitis C virus, and nonalcoholic steatohepatitis. CONCLUSION Here, we report that hCG regulates the SIRT1/FOXO3a axis in hepatocytes, resulting in immune suppression by attenuating caspase-3-dependent IL-16 processing and release, which concomitantly prevents autoaggressive T-cell infiltration of the liver. Considering the low toxicity profile of hCG in humans, interrupting the inflammatory cycle by hCG opens new perspectives for therapeutic intervention of inflammatory liver diseases. (Hepatology 2017;65:2074-2089).
Collapse
Affiliation(s)
- Caroline Steinmetz
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Anubha Kashyap
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Nataliya Zhivkova
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Henry Alizor
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Isabell Ernst
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | | | - Henning Janssen
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Andreas Teufel
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | | | - Johannes Lotz
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Jürgen Kuball
- Department of Immunology, Department of Hematology and Van Creveld Clinic University Medical Center Utrecht, Utrecht, The Netherlands
| | - Matthias Theobald
- Department of Hematology and Oncology, Johannes Gutenberg University, Mainz, Germany
| | - Michael Heise
- General, Visceral and Transplant Surgery, Johannes Gutenberg-University, Mainz, Germany
| | - Hauke Lang
- General, Visceral and Transplant Surgery, Johannes Gutenberg-University, Mainz, Germany
| | - Peter R Galle
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Dennis Strand
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Susanne Strand
- I. Department of Internal Medicine, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
16
|
Hershko Klement A, Shulman A. hCG Triggering in ART: An Evolutionary Concept. Int J Mol Sci 2017; 18:E1075. [PMID: 28513550 PMCID: PMC5454984 DOI: 10.3390/ijms18051075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 12/02/2022] Open
Abstract
Human chorionic gonadotropin (hCG) is no longer a single, omnipotent ovulation triggering option. Gonadotropin releasing hormone (GnRH) agonist, initially presented as a substitute for hCG, has led to a new era of administering GnRH agonist followed by hCG triggering. According to this new concept, GnRH agonist enables successful ovum maturation, while hCG supports the luteal phase and pregnancy until placental shift.
Collapse
Affiliation(s)
- Anat Hershko Klement
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba 4428164, Israel.
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel.
| | - Adrian Shulman
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba 4428164, Israel.
- The Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel.
| |
Collapse
|
17
|
Sanchez AM, Flamini MI, Russo E, Casarosa E, Pacini S, Petrini M, Genazzani AR, Simoncini T. LH and FSH promote migration and invasion properties of a breast cancer cell line through regulatory actions on the actin cytoskeleton. Mol Cell Endocrinol 2016; 437:22-34. [PMID: 27502036 DOI: 10.1016/j.mce.2016.08.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 07/19/2016] [Accepted: 08/05/2016] [Indexed: 02/07/2023]
Abstract
Reproductive hormones influence breast cancer development and progression. While the actions of sex steroids in this setting are established, tentative evidence suggests that follicle-stimulating hormone (FSH) and luteinizing hormone (LH) may also play a role, yet this remains elusive. We here identify that T-47D breast cancer cells express functional receptors for FSH and LH, and that these hormones regulate breast cancer cell motility and invasion through the control of the actin cytoskeleton and the formation of cortical actin aggregates and focal adhesion complexes. Such actions are mediated by the cytoskeletal controllers Moesin and focal adhesion kinase (FAK). Moesin is recruited rapidly by FSH and LH through a signaling cascade requiring the G protein Gα13 and the Rho-associated kinase, ROCK-2. FSH and LH activate FAK via a Gαi/β and c-Src-dependent signaling cascade. Both cascades involve signaling to phosphatidylinositol-3 kinase and Akt. FSH and LH receptors and the related signaling intermediates are necessary for the actions of gonadotrophins on breast cancer cell cytoskeletal rearrangement, migration and invasion. These findings provide original information on the actions of gonadotrophins on breast cancer cells and may have clinical implications for the use of drugs that modulate gonadotrophins in breast cancer patients.
Collapse
Affiliation(s)
- Angel Matias Sanchez
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy; Institute of Medicine and Experimental Biology of Cuyo (IMBECU), CCT-CONICET Mendoza, National University of Cuyo, Parque General San Martin s/n, Mendoza, CP:5500, Argentina
| | - Marina Ines Flamini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy; Institute of Medicine and Experimental Biology of Cuyo (IMBECU), CCT-CONICET Mendoza, National University of Cuyo, Parque General San Martin s/n, Mendoza, CP:5500, Argentina
| | - Eleonora Russo
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy
| | - Elena Casarosa
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy
| | - Simone Pacini
- Division of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy
| | - Mario Petrini
- Division of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy
| | - Andrea Riccardo Genazzani
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy
| | - Tommaso Simoncini
- Molecular and Cellular Gynecological Endocrinology Laboratory (MCGEL), Department of Clinical and Experimental Medicine, University of Pisa, Pisa, 56100, Italy.
| |
Collapse
|
18
|
López-Doval S, Salgado R, Lafuente A. The expression of several reproductive hormone receptors can be modified by perfluorooctane sulfonate (PFOS) in adult male rats. CHEMOSPHERE 2016; 155:488-497. [PMID: 27151425 DOI: 10.1016/j.chemosphere.2016.04.081] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 06/05/2023]
Abstract
This study was undertaken to evaluate the possible role of several reproductive hormone receptors on the disruption of the hypothalamic-pituitary-testis (HPT) axis activity induced by perfluorooctane sulfonate (PFOS). The studied receptors are the gonadotropin-releasing hormone receptor (GnRHr), luteinizing hormone receptor (LHr), follicle-stimulating hormone receptor (FSHr), and the androgen receptor (Ar). Adult male rats were orally treated with 1.0; 3.0 and 6.0 mg of PFOS kg(-1) d(-1) for 28 days. In general terms, PFOS can modify the relative gene and protein expressions of these receptors in several tissues of the reproductive axis. At the testicular level, apart from the expected inhibition of both gene and protein expressions of FSHr and Ar, PFOS also stimulates the GnRHr protein and the LHr gene expression. The receptors of the main hormones involved in the HPT axis may have an important role in the disruption exerted by PFOS on this axis.
Collapse
MESH Headings
- Alkanesulfonic Acids/chemistry
- Alkanesulfonic Acids/pharmacology
- Animals
- Blotting, Western
- Fluorocarbons/chemistry
- Fluorocarbons/pharmacology
- Follicle Stimulating Hormone/metabolism
- Gene Expression Regulation/drug effects
- Gonadotropin-Releasing Hormone/metabolism
- Luteinizing Hormone/metabolism
- Male
- Polymerase Chain Reaction
- Rats
- Rats, Sprague-Dawley
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Receptors, LHRH/genetics
- Receptors, LHRH/metabolism
- Reproduction/drug effects
- Testis/drug effects
- Testis/metabolism
Collapse
Affiliation(s)
- S López-Doval
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas s/n, 32004 Ourense, Spain
| | - R Salgado
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas s/n, 32004 Ourense, Spain
| | - A Lafuente
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas s/n, 32004 Ourense, Spain.
| |
Collapse
|
19
|
Rao CV. Therapeutic Potential of Human Chorionic Gonadotropin Against Painful Bladder Syndrome/Interstitial Cystitis. Reprod Sci 2016; 23:1451-1458. [PMID: 27004802 DOI: 10.1177/1933719116639139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Painful bladder syndrome/interstitial cystitis is a debilitating chronic bladder disease that primarily affects women. The disease is due to a damage of urothelial cell lining. As a result, potassium particles and other toxic substances in urine can leak into bladder mucosa, causing the symptoms of lower abdominal/pelvic discomfort, pain, increased urination frequency, urgency, nocturia, and so on, all of which can substantially reduce the quality of daily life. There are multiple symptom reliving therapies. Among them, only pentosan polysulfate sodium, sold under the brand name of Elmiron, has been approved for oral use by US Food and Drug Administration. It provides the relief after several months of use. Based on the scientific leads presented in this article, we propose that human chorionic gonadotropin has a therapeutic potential that is worth investigating for the treatment of this disease.
Collapse
Affiliation(s)
- C V Rao
- Departments of Cellular Biology and Pharmacology, Molecular and Human Genetics and Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
20
|
Overexpression of PRL7D1 in Leydig Cells Causes Male Reproductive Dysfunction in Mice. Int J Mol Sci 2016; 17:ijms17010096. [PMID: 26771609 PMCID: PMC4730338 DOI: 10.3390/ijms17010096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/03/2016] [Accepted: 01/06/2016] [Indexed: 01/02/2023] Open
Abstract
Prolactin family 7, subfamily d, member 1 (PRL7D1) is found in mouse placenta. Our recent work showed that PRL7D1 is also present in mouse testis Leydig cells, and the expression of PRL7D1 in the testis exhibits an age-related increase. In the present study, we generated transgenic mice with Leydig cell-specific PRL7D1 overexpression to explore its function during male reproduction. Prl7d1 male mice exhibited subfertility as reflected by reduced sperm counts and litter sizes. The testes from Prl7d1 transgenic mice appeared histologically normal, but the frequency of apoptotic germ cells was increased. Prl7d1 transgenic mice also had lower testosterone concentrations than wild-type mice. Mechanistic studies revealed that Prl7d1 transgenic mice have defects in the testicular expression of steroidogenic acute regulatory protein (STAR) and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase cluster (HSD3B). Further studies revealed that PRL7D1 overexpression affected the expression of transferrin (TF) in Sertoli cells. These results suggest that PRL7D1 overexpression could lead to increased germ cell apoptosis and exert an inhibitory effect on testosterone production in Leydig cells by reducing the expression of certain steroidogenic-related genes. In addition, PRL7D1 appears to have important roles in the function of Sertoli cells, which, in turn, affects male fertility. We conclude that the expression level of PRL7D1 is associated with the reproductive function of male mice.
Collapse
|
21
|
Ohlsson B. Gonadotropin-Releasing Hormone and Its Physiological and Pathophysiological Roles in Relation to the Structure and Function of the Gastrointestinal Tract. Eur Surg Res 2016; 57:22-33. [DOI: 10.1159/000445717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
<b><i>Background:</i></b> Gonadotropin-releasing hormone (GnRH), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) are involved in the reproductive cycle and regulate the secretion of sex steroids from the gonads. In mammals, GnRH1 is secreted as a hormone from the hypothalamus, whereas both GnRH1 and GnRH2 are present as neuropeptides in a variety of tissues. This review describes the role of GnRH in the gastrointestinal tract. <b><i>Summary:</i></b> GnRH1, GnRH2, and LH receptors in humans and rats, and GnRH receptors in rats, have been described in the gastrointestinal tract, where they affect motility, gastric and hormone secretion, and cell proliferation. GnRH analogs are clinically used in the treatment of sex hormone-dependent diseases, i.e., endometriosis and malignancies, and as pretreatments for in vitro fertilization. Severe gastrointestinal dysmotility has been shown to develop in some women after such treatment, along with a reduction in the number of enteric neurons and autoantibodies against GnRH. Consequently, a rat model of enteric neurodegeneration has been developed based on the administration of the GnRH analog buserelin. Serum IgM antibodies against GnRH1, the GnRH2 precursor progonadoliberin-2, and the GnRH receptor have also been described in patients with irritable bowel syndrome and dysmotility, as well as in patients with gastrointestinal disorders associated with diabetes mellitus, posterior laryngitis, and primary Sjögren's syndrome, although no treatments using GnRH analogs have been administered. <b><i>Conclusion:</i></b> GnRH and receptors for GnRH and LH are present in the human and rat gastrointestinal tract. Treatment with GnRH analogs may induce severe dysmotility, and a rat model of enteric neurodegeneration has been developed based on stimulation by the GnRH analog buserelin. Autoantibodies against GnRH and its receptor are found in a subgroup of patients with functional bowel disorders and dysmotility, independent of treatment with GnRH analogs.
Collapse
|
22
|
Rao CV. Why are We Waiting to Start Large Scale Clinical Testing of Human Chorionic Gonadotropin for the Treatment of Preterm Births? Reprod Sci 2015; 23:830-7. [PMID: 26692543 DOI: 10.1177/1933719115620498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Preterm births are an expensive global health problem. Despite the basic science and clinical research advances to better understand and prevent preterm births, the rates are increasing. There are several therapeutic options. While some options such as progestins work for selected women, others such as magnesium sulfate can only be used for delaying births for 24 to 48 hours so that the patients can be treated with corticosteroids to promote fetal lung maturity. Based on the scientific and clinical evidence, we recommend testing human chorionic gonadotropin in a large multicenter, randomized, double-blind, and placebo-controlled clinical trials in women with active preterm labor and those with a previous history of preterm births. Human chorionic gonadotropin is not only inexpensive but also has not shown any side effects so far in the infants or in the mothers.
Collapse
Affiliation(s)
- C V Rao
- Departments of Cellular Biology and Pharmacology, Molecular and Human Genetics and Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
23
|
Abstract
Overactive bladder (OAB) is a common form of urinary incontinence, resulting from spontaneous and random contractions of the urinary bladder. The affected individuals have an uncontrollable urge to urinate and experience incontinence and nocturia, which can greatly reduce the quality of daily life. There are several drugs for the treatment, and all of them have serious side effects. The following findings suggested that human chorionic gonadotropin (hCG) has a therapeutic potential that is worth investigating for the treatment of OAB. The finding are (1) human detrusor muscle contains hCG receptors, (2) detrusor muscle becomes quiescent during pregnancy, (3) hCG can inhibit detrusor muscle contractions induced by cholinergic stimulation in rats, and (4) hCG can mimic the anticholinergic drug on detrusor muscle contractions.
Collapse
Affiliation(s)
- C V Rao
- Departments of Cellular Biology and Pharmacology, Molecular and Human Genetics, and Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
24
|
Balakrishnan SR, Hashim U, Gopinath SCB, Poopalan P, Ramayya HR, Veeradasan P, Haarindraprasad R, Ruslinda AR. Polysilicon nanogap lab-on-chip facilitates multiplex analyses with single analyte. Biosens Bioelectron 2015; 84:44-52. [PMID: 26560969 DOI: 10.1016/j.bios.2015.10.075] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/15/2015] [Accepted: 10/26/2015] [Indexed: 12/27/2022]
Abstract
Rationally designed biosensing system supports multiplex analyses is warranted for medical diagnosis to determine the level of analyte interaction. The chemically functionalized novel multi-electrode polysilicon nanogap (PSNG) lab-on-chip is designed in this study, facilitates multiplex analyses for a single analyte. On the fabricated 69nm PSNG, biocompatibility and structural characteristics were verified for the efficient binding of Human Chorionic Gonadotropin (hCG). With the assistance of microfluidics, hCG sample was delivered via single-injection to 3-Aminopropyl(triethoxy)silane (APTES) and Glycidoxypropyl(trimethoxy)silane (GPMS) modified PSNG electrodes and the transduced signal was used to investigate the dielectric mechanisms for multiplex analyses. The results from amperometric response and impedance measurement delivered the scale of interaction between anti-hCG antibody and hCG that exhibited 6.5 times higher sensitivity for the chemical linker, APTES than GPMS. Under optimized experimental conditions, APTES and GPMS modified immunosensor has a limit of detection as 0.56mIU/ml and 2.93mIU/ml (at S/N=3), with dissociation constants (Kd) of 5.65±2.5mIU/ml and 7.28±2.6mIU/ml, respectively. These results suggest that multiplex analysis of single target could enhance the accuracy of detection and reliable for real-time comparative analyses. The designed PSNG is simple, feasible, requires low sample consumption and could be applied for any given multiplex analyses.
Collapse
Affiliation(s)
- Sharma Rao Balakrishnan
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia
| | - U Hashim
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia; School of Microelectronic Engineering, University Malaysia Perlis (UniMAP), Kuala Perlis, Perlis, Malaysia.
| | - Subash C B Gopinath
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia
| | - P Poopalan
- School of Microelectronic Engineering, University Malaysia Perlis (UniMAP), Kuala Perlis, Perlis, Malaysia
| | - H R Ramayya
- Department of Obstetrics and Gynaecology, Hospital Tuanku Fauziah, Kangar, Perlis, Malaysia
| | - P Veeradasan
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia
| | - R Haarindraprasad
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia
| | - A R Ruslinda
- Biomedical Nano Diagnostics Research Group, Institute of Nano Electronic Engineering (INEE), Universiti Malaysia Perlis (UniMAP), Kangar, Perlis, Malaysia
| |
Collapse
|
25
|
Sand E, Voss U, Ohlsson B, Ekblad E. Luteinizing hormone receptors are expressed in rat myenteric neurons and mediate neuronal loss. Auton Neurosci 2015; 193:104-7. [PMID: 26480825 DOI: 10.1016/j.autneu.2015.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/09/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Clinical observations have suggested repeated gonadotropin-releasing hormone (GnRH) exposure to cause intestinal dysfunction and loss of enteric neurons. This has been further studied and confirmed in a rat in vivo model involving iterated GnRH treatments. Mechanisms behind are enigmatic since no GnRH receptors are found to be expressed in enteric neurons neither in man nor rat. Both species, however, harbor substantial subpopulations of luteinizing hormone (LH) receptor-immunoreactive myenteric neurons which suggests that intestinal GnRH-induced neuropathy may be mediated by LH release. AIMS To reveal if exposures of GnRH or LH to rat myenteric neurons in vitro cause neuronal loss. METHODS Primary cultured adult rat myenteric neurons were exposed to single or repeated treatments of the GnRH analog buserelin or the LH analog lutrotropin alpha, and neuronal survival was determined by cell counting. Possible presence of GnRH- or LH receptor -immunoreactive neurons was determined by immunocytochemistry. RESULTS Exposure to the LH, but not the GnRH, analog caused significantly reduced neuronal survival. LH, but not GnRH, receptors were found to be expressed on cultured myenteric neurons. CONCLUSION Myenteric neurons express LH receptors in vitro and LH exposure causes reduced neuronal survival. This suggests that GnRH-induced enteric neuropathy in vivo is mediated by way of LH release and activation of enteric neuronal LH receptors.
Collapse
Affiliation(s)
- Elin Sand
- Department of Experimental Medical Science, Unit Neurogastroenterology, BMC B11, Lund University, Sölvegatan 19, SE 22184 Lund, Sweden.
| | - Ulrikke Voss
- Department of Experimental Medical Science, Unit Neurogastroenterology, BMC B11, Lund University, Sölvegatan 19, SE 22184 Lund, Sweden.
| | - Bodil Ohlsson
- Department of Clinical Sciences, Division of Internal Medicine, Lund University, Sweden Lund University, Inga Marie Nilssons gata 32, SE 21428 Malmö, Sweden.
| | - Eva Ekblad
- Department of Experimental Medical Science, Unit Neurogastroenterology, BMC B11, Lund University, Sölvegatan 19, SE 22184 Lund, Sweden.
| |
Collapse
|
26
|
Rao CV. Potential Therapy for Rheumatoid Arthritis and Sjögren Syndrome With Human Chorionic Gonadotropin. Reprod Sci 2015; 23:566-71. [PMID: 26239386 DOI: 10.1177/1933719115597765] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autoimmune diseases such as rheumatoid arthritis (RA) and Sjögren syndrome (SS) ameliorate during pregnancy, through dampening (immunotolerance) of the maternal immune system which protects the fetus from rejection. A large number of studies have shown that human chorionic gonadotropin (hCG) contributes to this tolerance. Studies on animal models have reaffirmed that hCG treatment mimics the benefits of pregnancy. Based on the scientific evidence, randomized clinical trials comparing hCG with current therapies and/or placebo are recommended for RA, SS, and for other autoimmune diseases such as, type 1 diabetes and ankylosing spondylitis, which also get better during pregnancy and hCG treatment seems to help.
Collapse
Affiliation(s)
- C V Rao
- Department of Cellular Biology and Pharmacology, Molecular and Human Genetics and Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
27
|
Rao CV. Potential Therapy for Neisseria Gonorrhoeae Infections With Human Chorionic Gonadotropin. Reprod Sci 2015; 22:1484-7. [PMID: 25868582 DOI: 10.1177/1933719115580998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The scientific evidence suggests that Neisseria gonorrhoeae (NG) infects human fallopian tubes by molecular mimicry in which pathogens act like a ligand to bind to epithelial cell surface human chorionic gonadotrophin (hCG)/luteinizing hormone (LH) receptors. The hCG-like molecule has been identified as ribosomal protein L12 in NG coat surface. Human fallopian tube epithelial cells have been shown to contain functional hCG/LH receptors. As previously shown in human fallopian tube organ and cell culture studies, cellular invasion and infection can be prevented by exposing the cells to excess hCG, which would outnumber and outcompete NG for receptor binding. Based on these data, we suggest testing hCG in clinical trials on infected women.
Collapse
Affiliation(s)
- C V Rao
- Departments of Cellular Biology and Pharmacology, Molecular and Human Genetics and Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
28
|
Parkash J, Lei Z, Rao CV. The Presence of Human Chorionic Gonadotropin/Luteinizing Hormone Receptors in Pancreatic β-Cells. Reprod Sci 2015; 22:1000-7. [PMID: 25670721 DOI: 10.1177/1933719115570910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We investigated the possible presence of functional human chorionic gonadotropin (hCG)/luteinizing hormone (LH) receptors in β-cells of pancreas, using a combination of techniques on hCG/LH receptor knockout mice, immortalized rat insulinoma cells, and human pancreatic islets. The results showed the presence of receptors and their activation resulted in a dose-dependent increase in glucose-induced release of insulin. These findings place hCG and LH among the regulators of insulin release with potential implications for insulin-level changes during the periods of altered hCG and LH secretion.
Collapse
Affiliation(s)
- Jai Parkash
- Department of Environmental and Occupational Health, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, USA Keiser University, Fort Myers, FL, USA
| | - Zhenmin Lei
- Department of Obstetrics and Gynecology, University of Louisville Medical Center, Louisville, KY, USA
| | - C V Rao
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
29
|
Rao C, Ticconi C. Why does human chorionic gonadotropin have such a broad regulatory roles in the body and are they totally unexpected? ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.jrhm.2014.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
|
31
|
McGee SR, Narayan P. Precocious puberty and Leydig cell hyperplasia in male mice with a gain of function mutation in the LH receptor gene. Endocrinology 2013; 154:3900-13. [PMID: 23861372 PMCID: PMC3776872 DOI: 10.1210/en.2012-2179] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The LH receptor (LHR) is critical for steroidogenesis and gametogenesis. Its essential role is underscored by the developmental and reproductive abnormalities that occur due to genetic mutations identified in the human LHR. In males, activating mutations are associated with precocious puberty and Leydig cell hyperplasia. To generate a mouse model for the human disease, we have introduced an aspartic acid to glycine mutation in amino acid residue 582 (D582G) of the mouse LHR gene corresponding to the most common D578G mutation found in boys with familial male-limited precocious puberty (FMPP). In transfected cells, mouse D582G mLHR exhibited constitutive activity with a 23-fold increase in basal cAMP levels compared with the wild-type receptor. A temporal study of male mice from 7 days to 24 weeks indicated that the knock-in mice with the mutated receptor (KiLHR(D582G)) exhibited precocious puberty with elevated testosterone levels as early as 7 days of age and through adulthood. Leydig cell-specific genes encoding LHR and several steroidogenic enzymes were up-regulated in KiLHR(D582G) testis. Leydig cell hyperplasia was detected at all ages, whereas Sertoli and germ cell development appeared normal. A novel finding from our studies, not previously reported in the FMPP cases, is that extensive hyperplasia is commonly found around the periphery of the testis. We further demonstrate that the hyperplasia is due to premature proliferation and precocious differentiation of adult Leydig cells in the KiLHR(D582G) testis. The KiLHR(D582G) mice provide a mouse model for FMPP, and we suggest that it is a useful model for studying pathologies associated with altered LHR signaling.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- Cell Proliferation
- Crosses, Genetic
- Disease Models, Animal
- Gene Knock-In Techniques
- Humans
- Hyperplasia
- Leydig Cells/metabolism
- Leydig Cells/pathology
- Male
- Mice
- Mice, 129 Strain
- Mice, Mutant Strains
- Mutagenesis, Site-Directed
- Mutant Proteins/metabolism
- Puberty, Precocious/blood
- Puberty, Precocious/genetics
- Puberty, Precocious/metabolism
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Testicular Diseases/blood
- Testicular Diseases/metabolism
- Testicular Diseases/pathology
- Testosterone/blood
- Up-Regulation
Collapse
Affiliation(s)
- Stacey R McGee
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, Illinois 62901.
| | | |
Collapse
|
32
|
Transcriptome analysis reveals new insights into the modulation of endometrial stromal cell receptive phenotype by embryo-derived signals interleukin-1 and human chorionic gonadotropin: possible involvement in early embryo implantation. PLoS One 2013; 8:e64829. [PMID: 23717664 PMCID: PMC3661534 DOI: 10.1371/journal.pone.0064829] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 04/19/2013] [Indexed: 01/08/2023] Open
Abstract
The presence of the conceptus in uterine cavity necessitates an elaborate network of interactions between the implanting embryo and a receptive endometrial tissue. We believe that embryo-derived signals play an important role in the remodeling and the extension of endometrial receptivity period. Our previous studies provided original evidence that human Chorionic Gonadotropin (hCG) modulates and potentiates endometrial epithelial as well as stromal cell responsiveness to interleukin 1 (IL1), one of the earliest embryonic signals, which may represent a novel pathway by which the embryo favors its own implantation and growth within the maternal endometrial host. The present study was designed to gain a broader understanding of hCG impact on the modulation of endometrial cell receptivity, and in particular, cell responsiveness to IL1 and the acquisition of growth-promoting phenotype capable of receiving, sustaining, and promoting early and crucial steps of embryonic development. Our results showed significant changes in the expression of genes involved in cell proliferation, immune modulation, tissue remodeling, apoptotic and angiogenic processes. This points to a relevant impact of these embryonic signals on the receptivity of the maternal endometrium, its adaptation to the implanting embryo and the creation of an environment that is favorable for the implantation and the growth of this latter within a new and likely hostile host tissue. Interestingly our data further identified a complex interaction between IL1 and hCG, which, despite a synergistic action on several significant endometrial target genes, may encompass a tight control of endogenous IL1 and extends to other IL1 family members.
Collapse
|
33
|
Bourdiec A, Bédard D, Rao CV, Akoum A. Human Chorionic Gonadotropin Regulates Endothelial Cell Responsiveness to Interleukin 1 and Amplifies the Cytokine-Mediated Effect on Cell Proliferation, Migration and the Release of Angiogenic Factors. Am J Reprod Immunol 2013; 70:127-38. [DOI: 10.1111/aji.12080] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/01/2013] [Indexed: 01/07/2023] Open
Affiliation(s)
- Amélie Bourdiec
- Endocrinologie de la reproduction, Centre de recherche-HSFA, CHUQ, Faculté de médecine; Université Laval; Québec; QC; Canada
| | - David Bédard
- Endocrinologie de la reproduction, Centre de recherche-HSFA, CHUQ, Faculté de médecine; Université Laval; Québec; QC; Canada
| | - C. V. Rao
- Herbert Wertheim College of Medicine; Florida International University; Miami; FL; USA
| | - Ali Akoum
- Endocrinologie de la reproduction, Centre de recherche-HSFA, CHUQ, Faculté de médecine; Université Laval; Québec; QC; Canada
| |
Collapse
|
34
|
Wang LH, Zhang W, Gao QX, Wang F. Expression of the luteinizing hormone receptor (LHR) gene in ovine non-gonadal tissues during estrous cycle. GENETICS AND MOLECULAR RESEARCH 2012; 11:3766-80. [PMID: 23096696 DOI: 10.4238/2012.october.15.8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Luteinizing hormone (LH) is an important glycoprotein hormone that regulates gonadal function in mammals and in turn regulates physiological status changes during the estrous cycle. The function of LH is mediated by luteinizing hormone receptor (LHR). In order to examine the expression patterns of the LHR gene in non-gonadal tissues during the 4 phases of the ovine estrous cycle, tissues from healthy non-pregnant adult Hu sheep were examined according to the estrous cycle for normal ovaries using real-time fluorescence quantitative PCR and ELISA methods with GAPDH as the reference gene. LHR mRNA expression levels were significantly correlated with protein concentrations and the LHR gene was abundantly expressed in olfactory bulb, hypothalamus, rumen, small intestine, kidney, and uterine tissues. When comparing the expression levels of LHR during the 4 estrous phases in particular tissues, the results showed that LHR expression levels were significantly different and relatively lower at the estrous stage in a number of non-gonadal tissues. The trends of change in LHR expression levels were highly significantly correlated between hypothalamus and rectum, hypophysis and oviduct, ileum and uterus, and among jejunum, olfactory bulb, and kidney (P < 0.01), and there was also significant correlation between duodenum and oviduct, hypothalamus and medulla oblongata, jejunum and uterus, omasum and abomasum, and reticulum and colon (P < 0.05). These results indicate that the ovine LHR gene (or LH) might control important mechanisms in non-gonadal tissues and that the level of LH activity in some tissues may be influenced by hormonal status during the estrous cycle.
Collapse
Affiliation(s)
- L H Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | | | | | | |
Collapse
|
35
|
Menon KMJ, Menon B. Structure, function and regulation of gonadotropin receptors - a perspective. Mol Cell Endocrinol 2012; 356:88-97. [PMID: 22342845 PMCID: PMC3327826 DOI: 10.1016/j.mce.2012.01.021] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 01/13/2012] [Accepted: 01/24/2012] [Indexed: 11/19/2022]
Abstract
Luteinizing hormone receptor and follicle stimulating hormone receptor play a crucial role in female and male reproduction. Significant new information has emerged about the structure, mechanism of activation, and regulation of expression of these receptors. Here we provide an overview of the current information on those aspects with an in-depth discussion of the recent developments in the post-transcriptional mechanism of LH receptor expression mediated by a specific LH receptor mRNA binding protein, designated as LRBP. LRBP was identified by electrophoretic gel mobility shift assay using cytosolic fractions from ovaries in the down regulated state. LRBP was purified, its binding site on LH receptor mRNA was identified and characterized. During ligand-induced down regulation, LRBP expression is increased through the cAMP/PKA and ERK signaling pathway, is translocated to translating ribosomes, binds LH receptor mRNA and forms an untranslatable ribonucleoprotein complex. This complex is then routed to the mRNA degradation machinery resulting in diminished levels of both LHR mRNA and cell surface expression of LH receptor. The studies leading to these conclusions are presented.
Collapse
Affiliation(s)
- K M J Menon
- Departments of Obstetrics/Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109-0617, United States.
| | | |
Collapse
|
36
|
Brouillet S, Hoffmann P, Chauvet S, Salomon A, Chamboredon S, Sergent F, Benharouga M, Feige JJ, Alfaidy N. Revisiting the role of hCG: new regulation of the angiogenic factor EG-VEGF and its receptors. Cell Mol Life Sci 2012; 69:1537-50. [PMID: 22138749 PMCID: PMC11115148 DOI: 10.1007/s00018-011-0889-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 11/02/2011] [Accepted: 11/14/2011] [Indexed: 11/25/2022]
Abstract
Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor reported to be specific for endocrine tissues, including the placenta. Its biological activity is mediated via two G protein-coupled receptors, prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). We have recently shown that (i) EG-VEGF expression peaks between the 8th and 11th weeks of gestation, (ii) its mRNA and protein levels are up-regulated by hypoxia, (iii) EG-VEGF is a negative regulator of trophoblast invasion and (iv) its circulating levels are increased in preeclampsia (PE), the most threatening pathology of pregnancy. Here, we investigated the regulation of the expression of EG-VEGF and its receptors by hCG, a key pregnancy hormone that is also deregulated in PE. During the first trimester of pregnancy, hCG and EG-VEGF exhibit the same pattern of expression, suggesting that EG-VEGF is potentially regulated by hCG. Both placental explants (PEX) and primary cultures of trophoblasts from the first trimester of pregnancy were used to investigate this hypothesis. Our results show that (i) LHCGR, the hCG receptor, is expressed both in cyto- and syncytiotrophoblasts, (ii) hCG increases EG-VEGF, PROKR1 and PROKR2 mRNA and protein expression in a dose- and time-dependent manner, (iii) hCG increases the release of EG-VEGF from PEX conditioned media, (iv) hCG effects are transcriptional and post-transcriptional and (v) the hCG effects are mediated by cAMP via cAMP response elements present in the EG-VEGF promoter region. Altogether, these results demonstrate a new role for hCG in the regulation of EG-VEGF and its receptors, an emerging regulatory system in placental development.
Collapse
MESH Headings
- Base Sequence
- Cells, Cultured
- Chorionic Gonadotropin/metabolism
- Chorionic Gonadotropin/pharmacology
- DNA Primers/genetics
- Female
- Gene Expression/drug effects
- Humans
- In Vitro Techniques
- Models, Biological
- Molecular Sequence Data
- Placenta/drug effects
- Placenta/metabolism
- Placentation
- Pregnancy
- Pregnancy Trimester, First
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, LH/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Trophoblasts/drug effects
- Trophoblasts/metabolism
- Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
- Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
Collapse
Affiliation(s)
- S Brouillet
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Laboratoire Biologie du Cancer et de l'Infection, Grenoble, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
BACKGROUND hCG is a wonder. Firstly, because hCG is such an extreme molecule. hCG is the most acidic glycoprotein containing the highest proportion of sugars. Secondly, hCG exists in 5 common forms. Finally, it has so many functions ranging from control of human pregnancy to human cancer. This review examines these molecules in detail. CONTENT These 5 molecules, hCG, sulfated hCG, hyperglycosylated hCG, hCG free beta and hyperglycosylated free beta are produced by placental syncytiotrophoblast cells and pituitary gonadotrope cells (group 1), and by placental cytotrophoblast cells and human malignancies (group 2). Group 1 molecules are both hormones that act on the hCG/LH receptor. These molecules are central to human menstrual cycle and human pregnancy. Group 2 molecules are autocrines, that act by antagonizing a TGF beta receptor. These molecules are critical to all advanced malignancies. CONCLUSIONS The hCG groups are molecules critical to both the molecules of pregnancy or human life, and to the advancement of cancer, or human death.
Collapse
|
38
|
Abstract
The primary embryonic signal in primates is chorionic gonadotropin (CG, designated hCG in humans), that is classically associated with corpus luteum rescue and progesterone production. However, research over the past decade has revealed the presence of the hCG receptor in a variety of extragonadal tissues. Additionally, discoveries of the multiple variants of hCG, namely, native hCG, hyperglycosylated hCG (hyp-hCG) and the β- subunit of the hyperglycosylated hCG (hCG-free β) has established a role for extragonadal actions of hCG. For the initiation and maintenance of pregnancy, hCG mediates multiple placental, uterine and fetal functions. Some of these include development of syncytiotrophoblast cells, mitotic growth and differentiation of the endometrium, localized suppression of the maternal immune system, modulation of uterine morphology and gene expression and coordination of intricate signal transduction between the endometrium. Recurrent pregnancy loss, pre-eclampsia and endometriosis are associated with altered responses of hCG, all of which have a detrimental effect on pregnancy. A role for hyp-hCG in mediating the development of both trophoblastic and non-trophoblastic tumors has also been suggested. Other significant non-gonadal applications of hCG include predicting preeclampsia, determining the risk of Down's syndrome and gestational trophoblastic disease, along with relaxing myometrial contractility and preventing recurrent miscarriages. Presence of hCG free-β in serum of cancer patients enables its usage as a diagnostic tumor marker. Thus, the extragonadal functions of hCG encompasses a wide spectrum of applications and is an open area for continued investigation.
Collapse
Affiliation(s)
- Prajna Banerjee
- Department of Medicine/Oncology, Stanford University, Stanford, CA 94305, USA
| | - Asgerally T. Fazleabas
- Department of Obstetrics and Gynecology, and Reproductive Biology, College of Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| |
Collapse
|
39
|
Cole LA. hCG, five independent molecules. Clin Chim Acta 2011; 413:48-65. [PMID: 22027338 DOI: 10.1016/j.cca.2011.09.037] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 09/11/2011] [Accepted: 09/26/2011] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The hCG amino acid sequence supports 5 glycoproteins. All are called hCG forms. This review examines all 5 molecules, the hormone as produced by the placental syncytiotrophoblast cells, the sulfated hormone produced by the pituitary gonadotrope cells, the hyperglycosylated hCG autocrine made by placental cytotrophoblast cells, and the autocrine cancer promoters hyperglycosylated hCG, hCGß and hyperglycosylated hCGß as made by all malignancies. This review examines all the molecules and multiple proven functions, ranging from evolution to cancer promotion to hormone action. RESULTS AND DISCUSSION hCG forms are critical super-growth factors in humans, with an exceptional wide range of functions.
Collapse
Affiliation(s)
- Laurence A Cole
- USA hCG Reference Service Reference Service, Division of Women's Health Research, Albuquerque, NM 87104, United States.
| |
Collapse
|
40
|
Luteinizing hormone receptor deficiency increases the susceptibility to alkylating agent-induced lymphomagenesis in mice. Discov Oncol 2011; 1:256-64. [PMID: 21666843 DOI: 10.1007/s12672-010-0045-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Previous studies have revealed a close link between luteinizing hormone (LH)/human chorionic gonadotropin (hCG) signaling and oncogenesis in gonadal and nongonadal tissues. To investigate whether genetic ablation of LH receptor (Lhr) affects the animal's oncogenic susceptibility, adult female wild-type (wt), heterozygous, and homozygous Lhr knockout (LhrKO) mice were intraperitoneally injected with an alkylating agent, N-methyl-N-nitrosourea (MNU, 50 mg/kg of body weight). The mice were sacrificed when they were short of breath or 10 months after the injection. The results showed that MNU induced non-Hodgkin's thymic and lymphonodus lymphomas in 70.6% and 100% of heterozygous and homozygous animals, respectively, compared with 35.7% in wt siblings. The tumor development was rapid; they were more aggressive and metastasized to the spleen, liver, and kidney in Lhr-deficient mice compared to wt siblings. All tumors were immunostained-positive for a T-cell specific marker, CD3, but not for a B-cell marker, CD22, suggesting that all the lymphomas arose from T-cells, which are known to be LH/hCG receptor-positive. There was no rearrangement of the Lhr gene locus or differences in thymic cell proliferation among the genotypes. However, apoptosis was lower in the Lhr-deficient thymuses. The thymic Bcl-2 levels were elevated and caspase-3 activation was reduced in Lhr heterozygous and homozygous animals. In conclusion, MNU induced a higher incidence and an earlier onset of aggressive lymphomas in LhrKO animals, which may be associated with a reduction in apoptosis of thymocytes.
Collapse
|
41
|
Abstract
BACKGROUND hCG is a term referring to 4 independent molecules, each produced by separate cells and each having completely separate functions. These are hCG produced by villous syncytiotrophoblast cells, hyperglycosylated hCG produced by cytotrophoblast cells, free beta-subunit made by multiple primary non-trophoblastic malignancies, and pituitary hCG made by the gonadotrope cells of the anterior pituitary. RESULTS AND DISCUSSION hCG has numerous functions. hCG promotes progesterone production by corpus luteal cells; promotes angiogenesis in uterine vasculature; promoted the fusion of cytotrophoblast cell and differentiation to make syncytiotrophoblast cells; causes the blockage of any immune or macrophage action by mother on foreign invading placental cells; causes uterine growth parallel to fetal growth; suppresses any myometrial contractions during the course of pregnancy; causes growth and differentiation of the umbilical cord; signals the endometrium about forthcoming implantation; acts on receptor in mother's brain causing hyperemesis gravidarum, and seemingly promotes growth of fetal organs during pregnancy. Hyperglycosylated hCG functions to promote growth of cytotrophoblast cells and invasion by these cells, as occurs in implantation of pregnancy, and growth and invasion by choriocarcinoma cells. hCG free beta-subunit is produced by numerous non-trophoblastic malignancies of different primaries. The detection of free beta-subunit in these malignancies is generally considered a sign of poor prognosis. The free beta-subunit blocks apoptosis in cancer cells and promotes the growth and malignancy of the cancer. Pituitary hCG is a sulfated variant of hCG produced at low levels during the menstrual cycle. Pituitary hCG seems to mimic luteinizing hormone actions during the menstrual cycle.
Collapse
Affiliation(s)
- Laurence A Cole
- USA hCG Reference Service, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
42
|
The LH/hCG Axis in Endometrial Cancer: A New Target in the Treatment of Recurrent or Metastatic Disease. Obstet Gynecol Int 2010; 2010. [PMID: 20706654 PMCID: PMC2913851 DOI: 10.1155/2010/486164] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 05/26/2010] [Accepted: 06/16/2010] [Indexed: 11/17/2022] Open
Abstract
Endometrial cancer (EC) is a hormone-dependent cancer that currently represents the most frequent malignancy of the female reproductive tract. The involvement of steroid hormones in EC etiology and progression has been reported. More recently, gonadotropins, and, in particular LH/hCG, are emerging as novel regulators of tumor progression. In the present review, we discuss the role of the LH/hCG axis (i.e. LH/hCG and its receptors, LH/hCG-R) in both gonadal and nongonadal tissues, in physiological and neoplastic conditions. In cancer cells, LH/hCG mainly controls cell proliferation and apoptosis. In particular, in EC LH/hCG improves cell invasiveness, through a mechanism which involves the LH/hCG-R, which in turn activate protein kinase A and modulate integrin adhesion receptors. Indeed, the LH/hCG-R mRNA is expressed in primary ECs and this expression correlates with LH/hCG-induced cell invasiveness in vitro. These results lead to hypothesize that recurrent and metastatic ECs, which express LH/hCG-R, could benefit from therapies aimed at decreasing LH levels, through Gn-RH analogues. Hence, the LH/hCG axis could represent a prognostic factor and a new therapeutic target in EC.
Collapse
|
43
|
Rizk DEE, Osman NA, Shafiullah MM, Nagelkerke NJD, Fahim MA. Effect of human chorionic gonadotrophin on in vitro contractions of stimulated detrusor muscle strips of female rats. J Obstet Gynaecol Res 2010; 35:835-41. [PMID: 20149029 DOI: 10.1111/j.1447-0756.2009.01052.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS We studied the effect of human chorionic gonadotrophin (hCG) on the in vitro detrusor muscle contractions in female rats. METHODS Two adjacent detrusor muscle strips from the bladder dome of 18 female Wistar rats (230-250 gm) were mounted in an organ bath for the recording of isometric tension. Carbachol (10(-9)-10(-3) M), alpha,beta methylene adenosine 5'-triphosphate (ATP) (10(-9)-10(-3) M) and potassium chloride (KCl) (10(-4)-10(-3) M) were applied (n = 6 x 3 groups). Concentration-response curves, before and after the addition of hCG (100 iu/mL) or oxybutynin (10(-5) M) to either muscle strip, were compared. RESULTS All curves were displaced to the right by hCG in a concentration-dependent manner with significant inhibition of contractions induced by carbachol (P < 0.001) and KCl (P = 0.016) but not those induced by alpha,beta-methylene ATP (P = 0.4). Estimated order of potency of inhibition was carbachol>KCl>alpha,beta-methylene ATP. The overall inhibitory effect of hCG was significantly less than oxybutynin (P < 0.001). CONCLUSIONS hCG significantly inhibited in vitro detrusor contractions induced by depolarization (KCl) and cholinergic (carbachol) but not purinergic (alpha,beta-methylene ATP) stimulation in a dose-dependent manner in female rats.
Collapse
Affiliation(s)
- Diaa E E Rizk
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| | | | | | | | | |
Collapse
|
44
|
Rahman NA, Rao C. Recent progress in luteinizing hormone/human chorionic gonadotrophin hormone research. Mol Hum Reprod 2009; 15:703-11. [DOI: 10.1093/molehr/gap067] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
45
|
Kumar TR. FSHbeta knockout mouse model: a decade ago and into the future. Endocrine 2009; 36:1-5. [PMID: 19387872 PMCID: PMC4074305 DOI: 10.1007/s12020-009-9199-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 02/24/2009] [Accepted: 04/09/2009] [Indexed: 10/20/2022]
Abstract
In 1997, more than 10 years ago now, we first reported the phenotypes of follicle stimulating hormone (FSH) beta null mice. Since then, these mice have been useful for various physiological and genetic studies in reproductive biology. More recently, extra-gonadal functions of FSH have been discovered in bone. These studies opened up exciting avenues of new research on osteoporosis in postmenopausal women. Several genomics and proteomics tools and novel strategies to spatio-temporally restricting gene expression in vivo are available now. It is hoped that with the aid of these and other emerging technologies, an integrated network of FSH signaling pathways in various tissues would emerge in the near future. Undoubtedly, the coming 10 years should be more exciting to explore this "fertile" area of reproductive physiology research.
Collapse
Affiliation(s)
- T Rajendra Kumar
- Department of Molecular, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
46
|
Is the zona pellucida an intrinsic source of signals activating maternal recognition of the developing mammalian embryo? J Reprod Immunol 2009; 81:1-8. [DOI: 10.1016/j.jri.2009.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Revised: 03/12/2009] [Accepted: 04/03/2009] [Indexed: 11/30/2022]
|
47
|
Abstract
When considering human chorionic gonadotropin (hCG) and hCG tests, it is important to realize that it is not a single biological molecule. The regular form of hCG produced by differentiated syncytotrophoblast cells (regular hCG) is a hormone made with the primary function of maintaining the myometrial and decidual spiral arteries and the vascular supply of the placenta during the full course of pregnancy. Hyperglycosylated hCG (hCG with double-size O-linked oligosaccharides) is made by undifferentiated cytotrophoblast cells. This is an autocrine hormone with separate functions, it maintains invasion as in implantation of pregnancy and malignancy in gestational trophoblastic diseases. A hyperglycosylated free beta-subunit is produced by a high proportion of all malignancies. This functions as an autocrine hormone to promote the growth and invasion of the malignancy. It is important to realize when ordering an hCG test what you are measuring and whether the test ordered will detect appropriately these three variant of hCG as well as their degradation products. Most automated commercial laboratory tests, point-of-care test and over-the-counter tests are limited in what is detected, focusing only on regular hCG. This is in part due to the US FDA, who only consider hCG as a pregnancy test, and to whom only detection of regular hCG is critical. This may be a cause of test errors since primarily hyperglycosylated hCG is produced in early pregnancy, choriocarcinoma and germ cell testicular malignancies, and only free beta-subunit may be produced in other germ cell malignancies (all applications for hCG test). The exceptions are the older style hCGbeta radioimmunoassay and the Siemen's Immulite platform hCG test which detect all variant and their degradation product appropriately. Regardless of test specificity limitations, assays for hCG variants are widely used clinically in pregnancy detection, early pregnancy detection, prediction of spontaneously aborting and ectopic pregnancies and prediction of trisomy pregnancies. hCG tests are essential in managing gestational trophoblastic diseases, whether hydatidiform mole, invasive mole or choriocarcinoma, and are very useful in management of testicular malignancies and other germ cell malignancies.
Collapse
Affiliation(s)
- Laurence A Cole
- Department of Obstetrics and Gynecology, University of New Mexico, Albuquerque NM 87131, USA.
| |
Collapse
|
48
|
de Medeiros S, Norman R. Human choriogonadotrophin protein core and sugar branches heterogeneity: basic and clinical insights. Hum Reprod Update 2008; 15:69-95. [DOI: 10.1093/humupd/dmn036] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Prast J, Saleh L, Husslein H, Sonderegger S, Helmer H, Knöfler M. Human chorionic gonadotropin stimulates trophoblast invasion through extracellularly regulated kinase and AKT signaling. Endocrinology 2008; 149:979-87. [PMID: 18063683 PMCID: PMC2974217 DOI: 10.1210/en.2007-1282] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chorionic gonadotropin (CG) is indispensable for human pregnancy because it controls implantation, decidualization, and placental development. However, its particular role in the differentiation process of invasive trophoblasts has not been fully unraveled. Here we demonstrate that the hormone promotes trophoblast invasion and migration in different trophoblast model systems. RT-PCR and Western blot analyses revealed expression of the LH/CG receptor in trophoblast cell lines and different trophoblast primary cultures. In vitro, CG increased migration and invasion of trophoblastic SGHPL-5 cells through uncoated and Matrigel-coated transwells, respectively. The hormone also increased migration of first-trimester villous explant cultures on collagen I. Proliferation of the trophoblast cell line and villous explant cultures measured by cumulative cell numbers and in situ 5-bromo-2'-deoxyuridine labeling, respectively, was unaffected by CG. Addition of the hormone activated ERK-1/2 and AKT in SGHPL-5 cells and pure, extravillous trophoblasts. Inhibition of MAPK kinase/ERK and phosphatidylinositide 3-kinase/AKT blocked phosphorylation of the kinases and attenuated CG-dependent invasion of SGHPL-5 cells. Similarly, the inhibitors decreased hormone-stimulated migration in villous explant cultures. Western blot analyses and gelatin zymography suggested that CG increased matrix metalloproteinase (MMP)-2 protein levels and activity in both culture systems. Inhibition of ERK or AKT diminished CG-induced MMP-2 expression. In summary, the data demonstrate that CG promotes trophoblast invasion and migration through activation of ERK and AKT signaling involving their downstream effector MMP-2. Because the increase of CG during the first trimester of pregnancy correlates with rising trophoblast motility, the hormone could be a critical regulator of the early invasion process.
Collapse
Affiliation(s)
- Johanna Prast
- Department of Obstetrics and Fetal-Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
50
|
Number of children is associated with neuropathology of Alzheimer's disease in women. Neurobiol Aging 2008; 30:1184-91. [PMID: 18079025 DOI: 10.1016/j.neurobiolaging.2007.11.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Revised: 10/16/2007] [Accepted: 11/08/2007] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To examine the association between number of born children and neuropathology of Alzheimer's disease (AD). METHODS The brains of 86 subjects with data on the number of biological children born, were studied postmortem. Primary analyses included 73 subjects (average age at death=80; 42 women) devoid of cerebrovascular disease associated lesions (i.e., infarcts) or of non-AD related neuropathology. Women were significantly older at death than men (85.6 vs. 73.4; p<.0005) but did not differ significantly from men in number of children or dementia severity. Secondary analyses included 13 additional subjects who had concomitant cerebrovascular disease. Density of neuritic plaques (NPs) and neurofibrillary tangles (NFTs) in the hippocampus, entorhinal cortex, amygdala and multiple regions of the cerebral cortex, as well as composites of these indices reflecting overall neuropathology, were analyzed. For men and women separately, partial correlations, controlling for age at death and dementia severity, were used to assess the associations of number of children with these neuropathological variables. RESULTS Among women, all the partial correlations were positive, with statistical significance for overall neuropathology (r=.37; p=.02), overall NPs (r=.36; p=.02), and for NPs in the amygdala (r=.47; p=.002). Among men, none of the partial correlations were statistically significant. Results of the secondary analyses were similar. CONCLUSIONS Since the associations between number of children and neuropathology of AD were found for women only, they might reflect sex-specific mechanisms (such as variations in estrogen or luteinizing hormone levels) rather than social, economic, biological or other mechanisms common to both men and women.
Collapse
|