1
|
Tian Y, Gao H, Li H, Li C, Li B. Evolutionary origin and distribution of leucine-rich repeat-containing G protein-coupled receptors in insects. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101318. [PMID: 39216279 DOI: 10.1016/j.cbd.2024.101318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/16/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Leucine-rich repeat-containing G protein-coupled receptors (LGRs) are crucial for animal growth and development. They were categorized into four types (A, B, C1, and C2) based on their sequence and domain structures. Despite the widespread distribution of LGRs across bilaterians, a thorough investigation of their distribution and evolutionary history remains elusive. Recent studies insect LGRs, especially the emergence of type C2 LGRs in various hemimetabolous insects, had prompted our study to address these problems. Initially, we traced the origins of LGRs by exploiting data from 99 species spanning 11 metazoan phyla, and discovered that type A and B LGRs originated from sponges, while type C LGRs originated from cnidarians. Subsequently, through comprehensive genomic and transcriptomic analyses across 565 species across 25 orders of insects, we found that both type A and C1 LGRs divided into two gene clusters. These clusters can be traced back to basal Insecta and an early ancestor of the Arthropoda, respectively. Furthermore, the absence of type B LGRs in wingless insects suggests a role in wing development, while the absence of type C2 LGRs in holometabolous insects hints at novel functions unrelated to insect metamorphosis. According to the origin of LGRs and the investigation of LGRs in insects, we speculated that type A and B LGRs appeared first among four types of LGRs, type A evolved into type C LGRs later, and type A and C1 LGRs independently duplicated during the evolutionary process. This study provides a more comprehensive view of the evolution of LGR genes than previously available, and sheds light on the evolutionary history and significance of LGRs in insect biology.
Collapse
Affiliation(s)
- Ying Tian
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Han Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Hong Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Chengjun Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
2
|
Hassan HA, Mazen I, Elaidy A, Kamel AK, Eissa NR, Essawi ML. Expanding the phenotypic spectrum of LHCGR signal peptide insertion variant: novel clinical and allelic findings causing Leydig cell hypoplasia type II. Hormones (Athens) 2024; 23:305-312. [PMID: 38526829 PMCID: PMC11219444 DOI: 10.1007/s42000-024-00546-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 03/11/2024] [Indexed: 03/27/2024]
Abstract
PURPOSE Leydig cell hypoplasia (LCH) type II is a rare disease with only a few cases reported. Patients presented with hypospadias, micropenis, undescended testes, or infertility. In this study, we report a new patient with compound heterozygous variants in the LHCGR gene and LCH type II phenotype. METHODS Whole exome sequencing (WES) was performed followed by Sanger sequencing to confirm the detected variants in the patient and his parents. RESULTS A novel missense variant (p.Phe444Cys) was identified in a highly conserved site and is verified to be in trans with the signal peptide's 33-bases insertion variant. CONCLUSION Our research provides a more comprehensive clinical and genetic spectrum of Leydig cell hypoplasia type II. It highlighted the importance of WES in the diagnosis of this uncommon genetic disorder as well as the expansion of the genotype of LCH type II.
Collapse
Affiliation(s)
- Heba Amin Hassan
- Department of Medical Molecular Genetics, Human Genetics & Genome Research Institute, National Research Centre, 33 El-Bohouth street, Cairo, 12311, Egypt.
| | - Inas Mazen
- Department of Clinical Genetics, Human Genetics & Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Aya Elaidy
- Department of Clinical Genetics, Human Genetics & Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Alaa K Kamel
- Department of Human Cytogenetics, Human Genetics & Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Noura R Eissa
- Department of Medical Molecular Genetics, Human Genetics & Genome Research Institute, National Research Centre, 33 El-Bohouth street, Cairo, 12311, Egypt
| | - Mona L Essawi
- Department of Medical Molecular Genetics, Human Genetics & Genome Research Institute, National Research Centre, 33 El-Bohouth street, Cairo, 12311, Egypt
| |
Collapse
|
3
|
Xia K, Wang F, Tan Z, Zhang S, Lai X, Ou W, Yang C, Chen H, Peng H, Luo P, Hu A, Tu X, Wang T, Ke Q, Deng C, Xiang AP. Precise Correction of Lhcgr Mutation in Stem Leydig Cells by Prime Editing Rescues Hereditary Primary Hypogonadism in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300993. [PMID: 37697644 PMCID: PMC10582410 DOI: 10.1002/advs.202300993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/20/2023] [Indexed: 09/13/2023]
Abstract
Hereditary primary hypogonadism (HPH), caused by gene mutation related to testosterone synthesis in Leydig cells, usually impairs male sexual development and spermatogenesis. Genetically corrected stem Leydig cells (SLCs) transplantation may provide a new approach for treating HPH. Here, a novel nonsense-point-mutation mouse model (LhcgrW495X ) is first generated based on a gene mutation relative to HPH patients. To verify the efficacy and feasibility of SLCs transplantation in treating HPH, wild-type SLCs are transplanted into LhcgrW495X mice, in which SLCs obviously rescue HPH phenotypes. Through comparing several editing strategies, optimized PE2 protein (PEmax) system is identified as an efficient and precise approach to correct the pathogenic point mutation in Lhcgr. Furthermore, delivering intein-split PEmax system via lentivirus successfully corrects the mutation in SLCs from LhcgrW495X mice ex vivo. Gene-corrected SLCs from LhcgrW495X mice exert ability to differentiate into functional Leydig cells in vitro. Notably, the transplantation of gene-corrected SLCs effectively regenerates Leydig cells, recovers testosterone production, restarts sexual development, rescues spermatogenesis, and produces fertile offspring in LhcgrW495X mice. Altogether, these results suggest that PE-based gene editing in SLCs ex vivo is a promising strategy for HPH therapy and is potentially leveraged to address more hereditary diseases in reproductive system.
Collapse
Affiliation(s)
- Kai Xia
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Fulin Wang
- Department of Urology and AndrologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Zhipeng Tan
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Suyuan Zhang
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xingqiang Lai
- Cardiovascular DepartmentThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdong518033China
| | - Wangsheng Ou
- State Key Laboratory of Ophthalmology Zhong Shan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouGuangdong510000China
| | - Cuifeng Yang
- Department of Urology and AndrologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Hong Chen
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Hao Peng
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Peng Luo
- Department of Urology and AndrologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Anqi Hu
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xiang'an Tu
- Department of Urology and AndrologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Qiong Ke
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Chunhua Deng
- Department of Urology and AndrologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue EngineeringKey Laboratory for Stem Cells and Tissue EngineeringMinistry of Education National‐Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| |
Collapse
|
4
|
Su J, Song Y, Yang Y, Li Z, Zhao F, Mao F, Wang D, Cao G. Study on the changes of LHR, FSHR and AR with the development of testis cells in Hu sheep. Anim Reprod Sci 2023; 256:107306. [PMID: 37541020 DOI: 10.1016/j.anireprosci.2023.107306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/06/2023]
Abstract
The process of testis development in mammals is accompanied by the proliferation and maturation of Sertoli, Leydig and germ cells. Spermatogenesis depends on hormone regulation, which must bind to a receptor to exert its biological effects. The changes in Hu sheep testis cell composition and FSHR, LHR and AR expression during different developmental stages are unclear (newborn, puberty and adulthood). To address this, using single-cell RNA sequencing, we analyzed testis cell composition and hormone receptor expression changes during three important developmental stages of Hu sheep. We observed significant changes in the composition of somatic and germ cells in different Hu sheep testis developmental stages. Furthermore, we analyzed the FSHR, LHR and AR distribution and expression changes at three important periods and verified them by qRT-PCR and immunofluorescence. Our results suggest that after birth, the proportion of germ cells increased gradually, peaking in adulthood; the proportion of Sertoli cells decreased gradually, reaching the lowest in adulthood; and the proportion of Leydig cells increased and then decreased, reaching the lowest in adulthood. In addition, FSHR, LHR and AR are mainly located in Sertoli, Leydig and germ cells. LHR and FSHR expression decreased with increasing age, while AR expression increased and then decreased with increasing age.
Collapse
Affiliation(s)
- Jie Su
- Department of Medical Neurobiology, Inner Mongolia Medical University, Huhhot 010030, China; Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Yongli Song
- Research Center for Animal Genetic Resources of Mongolia Plateau, Inner Mongolia University, Huhhot 010021, China
| | - Yanyan Yang
- Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Huhhot 010000, China
| | - Zhijun Li
- Department of Medical Neurobiology, Inner Mongolia Medical University, Huhhot 010030, China
| | - Feifei Zhao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Fei Mao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China
| | - Daqing Wang
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China; Inner Mongolia Academy of Agricultural & Animal Husbandry Sciences, Huhhot 010000, China
| | - Guifang Cao
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Inner Mongolia Agriculture University, Hohhot 010018, China.
| |
Collapse
|
5
|
Lund M, Pearson AC, Sage MAG, Duffy DM. Luteinizing hormone receptor promotes angiogenesis in ovarian endothelial cells of Macaca fascicularis and Homo sapiens†. Biol Reprod 2023; 108:258-268. [PMID: 36214501 PMCID: PMC9930396 DOI: 10.1093/biolre/ioac189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/26/2022] [Accepted: 10/04/2022] [Indexed: 11/14/2022] Open
Abstract
Angiogenesis within the ovarian follicle is an important component of ovulation. New capillary growth is initiated by the ovulatory surge of luteinizing hormone (LH), and angiogenesis is well underway at the time of follicle rupture. LH-stimulated follicular production of vascular growth factors has been shown to promote new capillary formation in the ovulatory follicle. The possibility that LH acts directly on ovarian endothelial cells to promote ovulatory angiogenesis has not been addressed. For these studies, ovaries containing ovulatory follicles were obtained from cynomolgus macaques and used for histological examination of ovarian vascular endothelial cells, and monkey ovarian microvascular endothelial cells (mOMECs) were enriched from ovulatory follicles for in vitro studies. mOMECs expressed LHCGR mRNA and protein, and immunostaining confirmed LHCGR protein in endothelial cells of ovulatory follicles in vivo. Human chorionic gonadotropin (hCG), a ligand for LHCGR, increased mOMEC proliferation, migration and capillary-like sprout formation in vitro. Treatment of mOMECs with hCG increased cAMP, a common intracellular signal generated by LHCGR activation. The cAMP analog dibutyryl cAMP increased mOMEC proliferation in the absence of hCG. Both the protein kinase A (PKA) inhibitor H89 and the phospholipase C (PLC) inhibitor U73122 blocked hCG-stimulated mOMEC proliferation, suggesting that multiple G-proteins may mediate LHCGR action. Human ovarian microvascular endothelial cells (hOMECs) enriched from ovarian aspirates obtained from healthy oocyte donors also expressed LHCGR. hOMECs also migrated and proliferated in response to hCG. Overall, these findings indicate that the LH surge may directly activate ovarian endothelial cells to stimulate angiogenesis of the ovulatory follicle.
Collapse
Affiliation(s)
- Merete Lund
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Andrew C Pearson
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Megan A G Sage
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia, USA
| |
Collapse
|
6
|
Umetsu A, Sato T, Watanabe M, Ida Y, Furuhashi M, Tsugeno Y, Ohguro H. Unexpected Crosslinking Effects of a Human Thyroid Stimulating Monoclonal Autoantibody, M22, with IGF1 on Adipogenesis in 3T3L-1 Cells. Int J Mol Sci 2023; 24:ijms24021110. [PMID: 36674625 PMCID: PMC9863235 DOI: 10.3390/ijms24021110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
To study the effects of the crosslinking of IGF1 and/or the human thyroid-stimulating monoclonal autoantibody (TSmAb), M22 on mouse adipocytes, two- and three-dimensional (2D or 3D) cultures of 3T3-L1 cells were prepared. Each sample was then subjected to the following analyses: (1) lipid staining, (2) a real-time cellular metabolic analysis, (3) analysis of the mRNA expression of adipogenesis-related genes and extracellular matrix (ECM) molecules including collagen (Col) 1, 4 and 6, and fibronectin (Fn), and (4) measurement of the size and physical properties of the 3D spheroids with a micro-squeezer. Upon adipogenic differentiation (DIF+), lipid staining and the mRNA expression of adipogenesis-related genes in the 2D- or 3D-cultured 3T3-L1 cells substantially increased. On adding IGF1 but not M22 to DIF+ cells, a significant enhancement in lipid staining and gene expressions of adipogenesis-related genes was detected in the 2D-cultured 3T3-L1 cells, although some simultaneous suppression or enhancement effects by IGF1 and M22 against lipid staining or Fabp4 expression, respectively, were detected in the 3D 3T3-L1 spheroids. Real-time metabolic analyses indicated that monotherapy with IGF1 or M22 shifted cellular metabolism toward energetic states in the 2D 3T3-L1 cells upon DIF+, although no significant metabolic changes were induced by DIF+ alone in 2D cultures. In addition, some synergistical effects on cellular metabolism by IGF1 and M22 were also observed in the 2D 3T3-L1 cells as well as in cultured non-Graves' orbitopathy-related human orbital fibroblasts (n-HOFs), but not in Graves' orbitopathy-related HOFs (GHOFs). In terms of the physical properties of the 3D 3T3-L1 spheroids, (1) their sizes significantly increased upon DIF+, and this increase was significantly enhanced by the presence of both IGF1 and M22 despite downsizing by monotreatment, and (2) their stiffness increased substantially, and no significant effects by IGF-1 and/or M22 were observed. Regarding the expression of ECM molecules, (1) upon DIF+, significant downregulation or upregulation of Col1 and Fn (3D), or Col4 and 6 (2D and 3D) were observed, and (2) in the presence of IGF-1 and/or M22, the mRNA expression of Col4 was significantly downregulated by M22 (2D and 3D), but the expression of Col1 was modulated in different manners by monotreatment (upregulation) or the combined treatment (downregulation) (3D). These collective data suggest that the human-specific TSmAb M22 induced some unexpected simultaneous crosslinking effects with IGF-1 with respect to the adipogenesis of 2D-cultured 3T3-L1 cells and the physical properties of 3D 3T3-L1 spheroids.
Collapse
Affiliation(s)
- Araya Umetsu
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yosuke Ida
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yuri Tsugeno
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
- Correspondence: ; Tel.: +81-611-2111
| |
Collapse
|
7
|
Single-Step Protocol for Isolating the Recombinant Extracellular Domain of the Luteinizing Hormone Receptor from the Ovis aries Testis. Curr Issues Mol Biol 2022; 44:5718-5727. [DOI: 10.3390/cimb44110387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
The luteinizing hormone receptor (LHR) is a glycoprotein member of the G protein-coupled receptors superfamily. It participates in corpus luteum formation and ovulation in females and acts in testosterone synthesis and spermatogenesis in males. In this study, we extracted RNA from sheep testicles and synthetized the cDNA to amplify the gene lhr-bed. This gene consists of 762 bp and encodes 273 amino acids of the extracellular domain of LHR. The lhr-bed was cloned into pJET1.2/blunt, then subcloned into pCOLD II, and finally, transformed in E. coli BL21 (DE3) cells. Because the induced rLHR-Bed protein was found in the insoluble fraction, we followed a modified purification protocol involving induction at 25 °C, subjection to denaturing conditions, and on-column refolding to increase solubility. We confirmed rLHR-Bed expression by means of Western blot and mass spectrometry analysis. It is currently known that the structure stem-loop 5′UTR on pCOLD II vector is stable at 15 °C. We predicted and obtained RNAfold stability at 25 °C. We successfully obtained the recombinant LHR extracellular domain, with protein yields of 0.2 mg/L, and purity levels of approximately 90%, by means of a single chromatographic purification step. The method described here may be used to obtain large quantities of rLHR-Bed in the future.
Collapse
|
8
|
Koganti PP, Zhao AH, Selvaraj V. Exogenous cholesterol acquisition signaling in LH-responsive MA-10 Leydig cells and in adult mice. J Endocrinol 2022; 254:187-199. [PMID: 35900012 PMCID: PMC9840751 DOI: 10.1530/joe-22-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 01/17/2023]
Abstract
MA-10 cells, established 4 decades ago from a murine Leydig cell tumor, has served as a key model system for studying steroidogenesis. Despite a precipitous loss in their innate ability to respond to luteinizing hormone (LH), the use of a cell-permeable cAMP analog for induction ensured their continued use. In parallel, a paradigm that serum-free conditions are essential for trophic steroidogenic stimulation was rationalized. Through the selection of LH-responsive single-cell MA-10Slip clones, we uncovered that Leydig cells remain responsive in the presence of serum in vitro and that exogenous cholesterol delivery by lipoproteins provided a significantly elevated steroid biosynthetic response (>2-fold). In scrutinizing the underlying regulation, systems biology of the MA-10 cell proteome identified multiple Rho-GTPase signaling pathways as highly enriched. Testing Rho function in steroidogenesis revealed that its modulation can negate the specific elevation in steroid biosynthesis observed in the presence of lipoproteins/serum. This signaling modality primarily linked to the regulation of endocytic traffic is evident only in the presence of exogenous cholesterol. Inhibiting Rho function in vivo also decreased hCG-induced testosterone production in mice. Collectively, our findings dispel a long-held view that the use of serum could confound or interfere with trophic stimulation and underscore the need for exogenous lipoproteins when dissecting physiological signaling and cholesterol trafficking for steroid biosynthesis in vitro. The LH-responsive MA-10Slip clones derived in this study present a reformed platform enabling biomimicry to study the cellular and molecular basis of mammalian steroidogenesis.
Collapse
Affiliation(s)
- Prasanthi P. Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Amy H. Zhao
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
- Correspondence should be addressed to: Vimal Selvaraj, Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853; ; Tel. 607-255-6138; Fax. 607-255-9829
| |
Collapse
|
9
|
Bello SF, Adeola AC, Nie Q. The study of candidate genes in the improvement of egg production in ducks – a review. Poult Sci 2022; 101:101850. [PMID: 35544958 PMCID: PMC9108513 DOI: 10.1016/j.psj.2022.101850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 11/01/2022] Open
Abstract
Duck is the second-largest poultry species aside from chicken. The rate of egg production is a major determinant of the economic income of poultry farmers. Among the reproductive organs, the ovary is a major part of the female reproductive system which is highly important for egg production. Based on the importance of this organ, several studies have been carried out to identify candidate genes at the transcriptome level, and also the expression level of these genes at different tissues or egg-laying conditions, and single nucleotide polymorphism (SNPs) of genes associated with egg production in duck. In this review, expression profile and association study analyses at SNPs level of different candidate genes with egg production traits of duck were highlighted. Furthermore, different studies on transcriptome analysis, Quantitative Trait Loci (QTL) mapping, and Genome Wide Association Study (GWAS) approach used to identify potential candidate genes for egg production in ducks were reported. This review would widen our knowledge on molecular markers that are associated or have a positive correlation to improving egg production in ducks, for the increasing world populace.
Collapse
|
10
|
The Roles of Luteinizing Hormone, Follicle-Stimulating Hormone and Testosterone in Spermatogenesis and Folliculogenesis Revisited. Int J Mol Sci 2021; 22:ijms222312735. [PMID: 34884539 PMCID: PMC8658012 DOI: 10.3390/ijms222312735] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
Spermatogenesis and folliculogenesis involve cell–cell interactions and gene expression orchestrated by luteinizing hormone (LH) and follicle-stimulating hormone (FSH). FSH regulates the proliferation and maturation of germ cells independently and in combination with LH. In humans, the requirement for high intratesticular testosterone (T) concentration in spermatogenesis remains both a dogma and an enigma, as it greatly exceeds the requirement for androgen receptor (AR) activation. Several data have challenged this dogma. Here we report our findings on a man with mutant LH beta subunit (LHβ) that markedly reduced T production to 1–2% of normal., but despite this minimal LH stimulation, T production by scarce mature Leydig cells was sufficient to initiate and maintain complete spermatogenesis. Also, in the LH receptor (LHR) knockout (LuRKO) mice, low-dose T supplementation was able to maintain spermatogenesis. In addition, in antiandrogen-treated LuRKO mice, devoid of T action, the transgenic expression of a constitutively activating follicle stimulating hormone receptor (FSHR) mutant was able to rescue spermatogenesis and fertility. Based on rodent models, it is believed that gonadotropin-dependent follicular growth begins at the antral stage, but models of FSHR inactivation in women contradict this claim. The complete loss of FSHR function results in the complete early blockage of folliculogenesis at the primary stage, with a high density of follicles of the prepubertal type. These results should prompt the reassessment of the role of gonadotropins in spermatogenesis, folliculogenesis and therapeutic applications in human hypogonadism and infertility.
Collapse
|
11
|
Schöneberg T, Liebscher I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol Rev 2020; 73:89-119. [PMID: 33219147 DOI: 10.1124/pharmrev.120.000011] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There are approximately 800 annotated G protein-coupled receptor (GPCR) genes, making these membrane receptors members of the most abundant gene family in the human genome. Besides being involved in manifold physiologic functions and serving as important pharmacotherapeutic targets, mutations in 55 GPCR genes cause about 66 inherited monogenic diseases in humans. Alterations of nine GPCR genes are causatively involved in inherited digenic diseases. In addition to classic gain- and loss-of-function variants, other aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, pseudogenes, gene fusion, and gene dosage, contribute to the repertoire of GPCR dysfunctions. However, the spectrum of alterations and GPCR involvement is probably much larger because an additional 91 GPCR genes contain homozygous or hemizygous loss-of-function mutations in human individuals with currently unidentified phenotypes. This review highlights the complexity of genomic alteration of GPCR genes as well as their functional consequences and discusses derived therapeutic approaches. SIGNIFICANCE STATEMENT: With the advent of new transgenic and sequencing technologies, the number of monogenic diseases related to G protein-coupled receptor (GPCR) mutants has significantly increased, and our understanding of the functional impact of certain kinds of mutations has substantially improved. Besides the classical gain- and loss-of-function alterations, additional aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, uniparental disomy, pseudogenes, gene fusion, and gene dosage, need to be elaborated in light of GPCR dysfunctions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| |
Collapse
|
12
|
Liu G, Zeng M, Li X, Rong Y, Hu Z, Zhang H, Liu X. Expression and analysis of ESR1, IGF-1, FSH, VLDLR, LRP, LH, PRLR genes in Pekin duck and Black Muscovy duck. Gene 2020; 769:145183. [PMID: 33007371 DOI: 10.1016/j.gene.2020.145183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/12/2020] [Accepted: 09/23/2020] [Indexed: 12/27/2022]
Abstract
In order to explore the influence of egg-laying regulatory genes on egg production in ducks at different laying stages, Pekin duck and Black Muscovy duck were used in this study, including early laying stage (20-30 weeks old), peak laying period (31-48 weeks old) and late laying stage (49-66 weeks old). Relative quantitative RT-PCR was used to detect the mRNA transcription level of selected egg-laying regulatory genes in the ovary tissues of ducks at different laying stages. Study shows: during the laying period of Pekin duck, ESR1, LRP1, IGF-1 and LHR were involved in the regulation of egg-laying, and the high expression of LRP1 in the late stage could inhibit egg production. Still, the expression products of the other three genes showed promoting effect. During the laying period of Black Muscovy duck, FSH, VLDLR, IGF-1, PRLR, LHR and LRP1 participated in the regulation of egg-laying, in which the expression products of the first five genes could promote egg production, while LRP1 showed inhibitory effect. Through our experiments, these data will provide strong theoretical support for the breeding of Pekin duck and Black Muscovy duck.
Collapse
Affiliation(s)
- Guangyu Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Mingfei Zeng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xingxing Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yu Rong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Zhigang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Huilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Xiaolin Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.
| |
Collapse
|
13
|
Luteinizing Hormone Action in Human Oocyte Maturation and Quality: Signaling Pathways, Regulation, and Clinical Impact. Reprod Sci 2020; 27:1223-1252. [PMID: 32046451 PMCID: PMC7190682 DOI: 10.1007/s43032-019-00137-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022]
Abstract
The ovarian follicle luteinizing hormone (LH) signaling molecules that regulate oocyte meiotic maturation have recently been identified. The LH signal reduces preovulatory follicle cyclic nucleotide levels which releases oocytes from the first meiotic arrest. In the ovarian follicle, the LH signal reduces cyclic nucleotide levels via the CNP/NPR2 system, the EGF/EGF receptor network, and follicle/oocyte gap junctions. In the oocyte, reduced cyclic nucleotide levels activate the maturation promoting factor (MPF). The activated MPF induces chromosome segregation and completion of the first and second meiotic divisions. The purpose of this paper is to present an overview of the current understanding of human LH signaling regulation of oocyte meiotic maturation by identifying and integrating the human studies on this topic. We found 89 human studies in the literature that identified 24 LH follicle/oocyte signaling proteins. These studies show that human oocyte meiotic maturation is regulated by the same proteins that regulate animal oocyte meiotic maturation. We also found that these LH signaling pathway molecules regulate human oocyte quality and subsequent embryo quality. Remarkably, in vitro maturation (IVM) prematuration culture (PMC) protocols that manipulate the LH signaling pathway improve human oocyte quality of cultured human oocytes. This knowledge has improved clinical human IVM efficiency which may become a routine alternative ART for some infertile patients.
Collapse
|
14
|
Xiong S, Mhawech-Fauceglia P, Tsao-Wei D, Roman L, Gaur RK, Epstein AL, Pinski J. Expression of the luteinizing hormone receptor (LHR) in ovarian cancer. BMC Cancer 2019; 19:1114. [PMID: 31729966 PMCID: PMC6857310 DOI: 10.1186/s12885-019-6153-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 09/11/2019] [Indexed: 11/10/2022] Open
Abstract
We investigated the association of LHR expression in epithelial ovarian cancer (OC) with clinical and pathologic characteristics of patients. LHR expression was examined immunohistochemically using tissue microarrays (TMAs) of specimens from 232 OC patients. Each sample was scored quantitatively evaluating LHR staining intensity (LHR-I) and percentage of LHR (LHR-P) staining cells in tumor cells examined. LHR-I was assessed as no staining (negative), weak (+ 1), moderate (+ 2), and strong positive (+ 3). LHR-P was measured as 1 to 5, 6 to 50% and > 50% of the tumor cells examined. Positive LHR staining was found in 202 (87%) patients' tumor specimens and 66% patients had strong intensity LHR expression. In 197 (85%) of patients, LHR-P was measured in > 50% of tumor cells. LHR-I was significantly associated with pathologic stage (p = 0.007). We found that 72% of stage III or IV patients expressed strong LHR-I in tumor cells. There were 87% of Silberberg's grade 2 or 3 patients compared to 70% of grade 1 patients with LHR expression observed in > 50% of tumor cells, p = 0.037. Tumor stage was significantly associated with overall survival and recurrence free survival, p < 0.001 for both analyses, even after adjustment for age, tumor grade and whether patient had persistent disease after therapy or not. Our study demonstrates that LHR is highly expressed in the majority of OC patients. Both LHR-I and LHR-P are significantly associated with either the pathologic stage or tumor grade.
Collapse
Affiliation(s)
- Shigang Xiong
- Department of Medicine/Medical Oncology Division, University of Southern California, 1441 Eastlake Ave, Los Angeles, CA, 90033, USA
| | - Paulette Mhawech-Fauceglia
- Aurora Diagnostics, Department of Pathology, Gynecologic Pathology Consultant, San Antonio, TX, 78209, USA
| | - Denice Tsao-Wei
- University of Southern California, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA
| | - Lynda Roman
- Department of Obstetrics & Gynecology, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Rajesh K Gaur
- Department of Medicine/Medical Oncology Division, University of Southern California, 1441 Eastlake Ave, Los Angeles, CA, 90033, USA
| | - Alan L Epstein
- Department of Pathology, University of Southern California, HMR 2011 Zonal Ave, Los Angeles, CA, 90033, USA
| | - Jacek Pinski
- Department of Medicine/Medical Oncology Division, University of Southern California, 1441 Eastlake Ave, Los Angeles, CA, 90033, USA. .,University of Southern California, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Los Angeles, CA, 90033, USA.
| |
Collapse
|
15
|
Li YX, Guo X, Gulappa T, Menon B, Menon KMJ. SREBP Plays a Regulatory Role in LH/hCG Receptor mRNA Expression in Human Granulosa-Lutein Cells. J Clin Endocrinol Metab 2019; 104:4783-4792. [PMID: 31150065 PMCID: PMC6736214 DOI: 10.1210/jc.2019-00913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/24/2019] [Indexed: 01/29/2023]
Abstract
CONTEXT LH receptor (LHR) expression has been shown to be regulated posttranscriptionally by LHR mRNA binding protein (LRBP) in rodent and human ovaries. LRBP was characterized as mevalonate kinase. The gene that encodes mevalonate kinase is a member of a family of genes that encode enzymes involved in lipid synthesis and are regulated by the transcription factor sterol regulatory element binding proteins (SREBPs). OBJECTIVE The current study examined the regulation of LHR mRNA expression in human granulosa-lutein cells in response to alterations in cholesterol metabolism. DESIGN Using atorvastatin, an inhibitor of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase to inhibit cholesterol biosynthesis, we examined its effect on LHR mRNA expression. The effect of atorvastatin on SREBP and mRNA expression as well as LHR mRNA binding protein expression was examined. Finally, the effect of atorvastatin on human chorionic gonadotropin (hCG)-stimulated progesterone production and the expression of key steroidogenic enzymes was also examined. RESULTS Statin treatment reduced LHR mRNA expression by increasing the levels of SREBP1a and SREBP2, leading to an increase in LRBP. RNA gel shift assay showed that increased binding of LHR mRNA to LRBP occurred in response to atorvastatin, leading to LHR mRNA degradation. The granulosa-lutein cells pretreated with atorvastatin also showed decreased responsiveness to hCG by decreasing the mRNA and protein expression of steroidogenic enzymes. Atorvastatin also attenuated LH/hCG-induced progesterone production. CONCLUSION These results imply that LHR mRNA expression by the human granulosa-lutein cells is regulated by cholesterol, through a mechanism involving SREBP and SREBP cleavage activating protein serving as the cholesterol sensor.
Collapse
Affiliation(s)
- Yin-Xia Li
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Xingzi Guo
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Thippeswamy Gulappa
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bindu Menon
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
| | - K M J Menon
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan
- Correspondence and Reprint Requests: K. M. J. Menon, PhD, Departments of Obstetrics/Gynecology and Biological Chemistry, University of Michigan Medical School, 6428 Medical Sciences Building I, 1301 Catherine Street, Ann Arbor, Michigan 48109. E-mail:
| |
Collapse
|
16
|
Hausken K, Levavi-Sivan B. Synteny and phylogenetic analysis of paralogous thyrostimulin beta subunits (GpB5) in vertebrates. PLoS One 2019; 14:e0222808. [PMID: 31536580 PMCID: PMC6752823 DOI: 10.1371/journal.pone.0222808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/06/2019] [Indexed: 01/02/2023] Open
Abstract
At some point early in the vertebrate lineage, two whole genome duplication events (1R, 2R) took place that allowed for the diversification and sub-/neo-functionalization of the glycoprotein hormones (GpHs). All jawed vertebrates possess the GpHs luteinizing hormone (LH), follicle stimulating hormone (FSH), and thyroid stimulating hormone (TSH), each of which are heterodimers with a common alpha subunit and unique beta subunits. In 2002, a novel glycoprotein hormone named thyrostimulin was described to have unique GpA2 and GpB5 subunits that were homologous to the vertebrate alpha and beta subunits. The presence of GpA2 and GpB5 in representative protostomes and deuterostomes indicates their ancestry in the GpH family. There are several reports of GpH subunit evolution, but none have included GpA2 and GpB5 for species in each major vertebrate class. Thus, we addressed the ancestry of two paralogous GpB5 subunits (GpB5a and GpB5b) that were previously only recognized in two teleost species. Our search for orthologous GpB5a and GpB5b sequences in representative vertebrates and phylogenetic analysis, in addition to the currently published evolutionary scenarios of the GpH family, supports that GpB5a and GpB5b are paralogs that arose from the first vertebrate whole genome duplication event (1R). Syntenic analysis supports lineage specific losses of GpB5a in chondrichthyes, basal actinopterygians, and tetrapods, and retention in coelacanth and teleosts. Additionally, we were unable to identify GpA2 transcripts from tilapia mRNA, suggesting that this species does not produce heterodimeric thyrostimulin. While the conserved or even species-specific functional role of thyrostimulin or its individual subunits are still unknown in vertebrates, the analyses presented here provide context for future studies on the functional divergence of the GpH family.
Collapse
Affiliation(s)
- Krist Hausken
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Berta Levavi-Sivan
- Department of Animal Sciences, Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
17
|
Babahajian A, Sarveazad A, Golab F, Vahabzadeh G, Alizadeh A, Rasoolijazi H, Amini N, Entezari M, Soleimani M, Katebi M, Haramshahi SMA. Neuroprotective Effects of Trolox, Human Chorionic Gonadotropin, and Carnosic Acid on Hippocampal Neurodegeneration After Ischemiareperfusion Injury. Curr Stem Cell Res Ther 2019; 14:177-183. [PMID: 30227824 DOI: 10.2174/1574888x13666180918093822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/20/2018] [Accepted: 08/21/2018] [Indexed: 11/22/2022]
Abstract
INTRODUCTION One of the serious complications of stroke is memory impairment, which is considered as one of the complications of reperfusion of tissue. The present study was designed to compare the effect of administration of Trolox, carnosic acid and Human Chorionic Gonadotropin (HCG) immediately after reperfusion of the stroke tissue on the memory and hippocampal histology. METHOD Ischemia-Reperfusion Model (IRI) was created by bilateral occlusion of the common carotid artery for 15 minutes and the first dose was administered immediately after reperfusion. 10 days after ischemia, passive avoidance memory test and apoptotic protein levels were evaluated. RESULTS Cerebral Ischemia perfusion reduced the time of latency in entering the dark box in the ischemic group. Administration of Trolox and HCG increased this latency time, while treatment with carnosic acid had no effect. Also, IRI significantly reduced the number of healthy cells in the hippocampus. Administration of Trolox, carnosic acid and HCG increased the number of healthy cells and decreased the expression of Caspase-3 and Bax, but significantly increased the expression of Bcl-2 compared to the ischemic group. CONCLUSION Findings indicate the beneficial effects of HCG and Trolox on the improvement of memory and the number of healthy cells in the hippocampal region. It is worth noting that the amount of apoptosis in the hippocampus was significantly reduced by Trolox, HCG and Carnosic acid.
Collapse
Affiliation(s)
- Asrin Babahajian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Liver and Digestive Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Gelareh Vahabzadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Homa Rasoolijazi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maedeh Entezari
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Katebi
- Department of Anatomy, Faculty of Medicine, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | - Seyed Mohammad Amin Haramshahi
- Department of Tissue Engineering & Regenerative Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Schug S, Baunacke A, Goeckenjan M, Horn LC, Pretzsch G, Zimmermann G, Alexander H. Endometrial human chorionic gonadotropin (hCG) expression is a marker for adequate secretory transformation of the endometrium. Arch Gynecol Obstet 2019; 299:1727-1736. [PMID: 30955059 DOI: 10.1007/s00404-019-05130-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 03/25/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Successful embryo implantation into the endometrium depends on embryonic characteristics and proper endometrial development. Reproductive medicine often focuses on embryo quality, whereas reliable diagnostic tests for endometrial receptivity are still needed. We previously found that human chorionic gonadotropin (hCG), one of the earliest proteins secreted by the embryo, was also expressed by the luteal phase endometrium around the implantation window. Here, we tested our hypothesis of endometrial hCG as an implantation marker. METHODS Endometrial biopsies and serum samples were taken from patients undergoing routine infertility diagnostics. Correlations of immunohistochemically detected endometrial hCG expression with adequate endometrial secretory transformation, the infiltration of CD45-positive leukocytes, clinical diagnostic parameters, and endometrial thickness were analyzed. RESULTS A highly significant correlation between the endometrial score, as a measurement for regular secretory transformation, and the intensity of hCG staining was found. The invasion of CD45-positive leukocytes increased with progressing endometrial secretory transformation and rising endometrial hCG expression. In addition, serum progesterone concentrations correlated with hCG expression by the endometrial glands. CONCLUSIONS Our results suggest endometrial hCG as a possible diagnostic parameter characterizing the endometrial secretory transformation and, thus, possibly also its implantation capability.
Collapse
Affiliation(s)
- Sindy Schug
- Research Laboratory of the Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, Medical School, University of Leipzig, Semmelweisstr. 14, 04103, Leipzig, Germany.
| | - Anja Baunacke
- Research Laboratory of the Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, Medical School, University of Leipzig, Semmelweisstr. 14, 04103, Leipzig, Germany
| | - Maren Goeckenjan
- Department of Gynecology and Obstetrics, University of Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Lars-Christian Horn
- Institute of Pathology, Department of Breast, Gynecological and Perinatal Pathology, University Hospital of Leipzig, Liebigstr. 24, 04103, Leipzig, Germany
| | - Gabriele Pretzsch
- Women's Hospital, University Hospital of Leipzig, Liebigstr. 20a, 04103, Leipzig, Germany
| | - Gerolf Zimmermann
- Research Laboratory of the Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, Medical School, University of Leipzig, Semmelweisstr. 14, 04103, Leipzig, Germany
| | - Henry Alexander
- Research Laboratory of the Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, Medical School, University of Leipzig, Semmelweisstr. 14, 04103, Leipzig, Germany.
| |
Collapse
|
19
|
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: 1.0] [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
|
20
|
Tapia-Pizarro A, Archiles S, Argandoña F, Valencia C, Zavaleta K, Cecilia Johnson M, González-Ramos R, Devoto L. hCG activates Epac-Erk1/2 signaling regulating Progesterone Receptor expression and function in human endometrial stromal cells. Mol Hum Reprod 2018; 23:393-405. [PMID: 28333280 DOI: 10.1093/molehr/gax015] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/09/2017] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION How does hCG signal in human endometrial stromal cells (ESCs) and what is its role in regulating ESC function? SUMMARY ANSWER hCG signaling in ESCs activates the extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) pathway through exchange protein activated by cyclic AMP (cAMP) (Epac) and transiently increases progesterone receptor (PR) transcript and protein expression and its transcriptional function. WHAT IS KNOWN ALREADY hCG is one of the earliest embryo-derived secreted signals in the endometrium, which abundantly expresses LH/hCG receptors. hCG signals through cAMP/protein kinase A (PKA) in gonadal cells, but in endometrial epithelial cells, hCG induces Erk1/2 activation independent of the cAMP/PKA pathway. Few data exist concerning the signal transduction pathways triggered by hCG in ESCs and their role in regulation of ESC function. STUDY DESIGN, SIZE, DURATION This is an in vitro study comprising patients undergoing benign gynecological surgery (n = 46). PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial samples were collected from normal cycling women during the mid-secretory phase for ESCs isolation. The study conducted in an academic research laboratory within a tertiary-care hospital. The activation of the Erk1/2 signal transduction pathway elicited by hCG was evaluated in ESC. Signaling pathway inhibitors were used to examine the roles of PKA, PI3K, PKC, adenylyl cyclase and Epac on the hCG-stimulated up-regulation of phospho-Erk1/2 (pErk1/2). Erk1/2 phosphorylation was determined by immunoblot. siRNA targeting Epac was used to investigate the molecular mechanisms. To assess the role of Erk1/2 signaling induced by hCG on ESC function, gene expression regulation was examined by immunofluorescence and real-time quantitative PCR. The role of PR on the regulation of transcript levels was studied using progesterone and the PR antagonist RU486. All experiments were conducted using at least three different cell culture preparations in triplicate. MAIN RESULTS AND THE ROLE OF CHANCE Addition of hCG to ESCs in vitro induced the phosphorylation of Erk1/2 through cAMP accumulation. Such induction could not be blocked by inhibitors for PKA, PKC and PI3K. Epac inhibition and knockdown with siRNA prevented pErk1/2 induction by hCG. ESCs stimulated with hCG for up to 72 h showed a significant increase in PR mRNA and immunofluorescent label at 48 h only; an effect that was abrogated with the mitogen-activated protein kinase kinase inhibitor UO126. In addition, the hCG-activated Erk1/2 pathway significantly decreased the mRNA levels for secreted frizzled-related protein 4 (SFRP4) at 24 h, whereas it increased those for homeobox A10 (HOXA10) at 48 h (P = 0.041 and P = 0.022 versus control, respectively). Prolactin mRNA levels were not significantly modified. HOXA10 mRNA up-regulation by hCG was not enhanced by co-stimulation with progesterone; however, it was completely abolished in the presence of RU486 (P = 0.036 hCG versus hCG + RU486). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This is an in vitro study utilizing stromal cell cultures from human endometrial tissues. Furthermore, results obtained should also be confirmed in vivo in the context of the whole human endometrial tissue and hormonal milieu. The in vitro experiments using hCG have been conducted without other hormones/factors that may also modulate the ESCs response to hCG. WIDER IMPLICATIONS OF THE FINDINGS We have determined that hCG induces the PR through the Erk1/2 pathway in ESCs which may render them more sensitive to progesterone, increasing our understanding about the effects of hCG at the embryo-maternal interface. The activation of such a pathway in the context of the hormonal milieu during the window of implantation might contribute to a successful dialog between the embryo and the uterus, leading to appropriate endometrial function. Defective hCG signaling in the endometrial stromal tissue may lead to an incomplete uterine response, compromising embryo implantation and early pregnancy. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Fund for Scientific and Technological Development, Government of Chile (FONDECYT) grants 11100443 and 1140614 (A.T.-P.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Alejandro Tapia-Pizarro
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Sebastián Archiles
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Felipe Argandoña
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Cecilia Valencia
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Keyla Zavaleta
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - M Cecilia Johnson
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Reinaldo González-Ramos
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| | - Luigi Devoto
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Av. Sta. Rosa 1234, 2do piso, Santiago 8360160, Chile
| |
Collapse
|
21
|
Bhatta S, Blair JA, Casadesus G. Luteinizing Hormone Involvement in Aging Female Cognition: Not All Is Estrogen Loss. Front Endocrinol (Lausanne) 2018; 9:544. [PMID: 30319538 PMCID: PMC6165885 DOI: 10.3389/fendo.2018.00544] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/28/2018] [Indexed: 01/29/2023] Open
Abstract
Pervasive age-related dysfunction in hypothalamic-pituitary-gonadal (HPG) axis is associated with cognitive impairments in aging as well as pathogenesis of age-related neurodegenerative diseases such as the Alzheimer's disease (AD). As a major regulator of the HPG axis, the steroid hormone estrogen has been widely studied for its role in regulation of memory. Although estrogen modulates both cognition as well as cognition associated morphological components in a healthy state, the benefits of estrogen replacement therapy on cognition and disease seem to diminish with advancing age. Emerging data suggests an important role for luteinizing hormone (LH) in CNS function, which is another component of the HPG axis that becomes dysregulated during aging, particularly in menopause. The goal of this review is to highlight the current existing literature on LH and provide new insights on possible mechanisms of its action.
Collapse
Affiliation(s)
- Sabina Bhatta
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jeffrey A. Blair
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
- Department of Biological Sciences, Kent State University, Kent, OH, United States
- *Correspondence: Gemma Casadesus
| |
Collapse
|
22
|
The effect of estradiol, testosterone, and human chorionic gonadotropin on the proliferation of Schwann cells with NF1
+/− or NF1
−/− genotype derived from human cutaneous neurofibromas. Mol Cell Biochem 2017; 444:27-33. [DOI: 10.1007/s11010-017-3227-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 11/24/2017] [Indexed: 10/18/2022]
|
23
|
Ventura-López C, Galindo-Torres PE, Arcos FG, Galindo-Sánchez C, Racotta IS, Escobedo-Fregoso C, Llera-Herrera R, Ibarra AM. Transcriptomic information from Pacific white shrimp (Litopenaeus vannamei) ovary and eyestalk, and expression patterns for genes putatively involved in the reproductive process. Gen Comp Endocrinol 2017; 246:164-182. [PMID: 27964922 DOI: 10.1016/j.ygcen.2016.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 11/27/2022]
Abstract
The increased use of massive sequencing technologies has enabled the identification of several genes known to be involved in different mechanisms associated with reproduction that so far have only been studied in vertebrates and other model invertebrate species. In order to further investigate the genes involved in Litopenaeus vannamei reproduction, cDNA and SSH libraries derived from female eyestalk and gonad were produced, allowing the identification of expressed sequences tags (ESTs) that potentially have a role in the regulation of gonadal maturation. In the present study, different transcripts involved in reproduction were identified and a number of them were characterized as full-length. These transcripts were evaluated in males and females in order to establish their tissue expression profiles during developmental stages (juvenile, subadult and adult), and in the case of females, their possible association with gonad maturation was assessed through expression analysis of vitellogenin. The results indicated that the expression of vitellogenin receptor (vtgr) and minichromosome maintenance (mcm) family members in the female gonad suggest an important role during previtellogenesis. Additionally, the expression profiles of genes such as famet, igfbp and gpcr in brain tissues suggest an interaction between the insulin/insulin-like growth factor signaling pathway (IIS) and methyl farnesoate (MF) biosynthesis for control of reproduction. Furthermore, the specific expression pattern of farnesoic acid O-methyltransferase suggests that final synthesis of MF is carried out in different target tissues, where it is regulated by esterase enzymes under a tissue-specific hormonal control. Finally, the presence of a vertebrate type steroid receptor in hepatopancreas and intestine besides being highly expressed in female gonads, suggest a role of that receptor during sexual maturation.
Collapse
Affiliation(s)
- Claudia Ventura-López
- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional No.195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur 23096, Mexico.
| | - Pavel E Galindo-Torres
- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional No.195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur 23096, Mexico.
| | - Fabiola G Arcos
- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional No.195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur 23096, Mexico.
| | - Clara Galindo-Sánchez
- Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada, Baja California CP 22860, Mexico.
| | - Ilie S Racotta
- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional No.195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur 23096, Mexico.
| | - Cristina Escobedo-Fregoso
- Consejo Nacional de Ciencia y Tecnología (CONACYT) - Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional 195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur C.P. 23096, Mexico.
| | - Raúl Llera-Herrera
- Consejo Nacional de Ciencia y Tecnología (CONACYT) - Centro de Investigación en Alimentación y Desarrollo A.C. (CIAD) Unidad Mazatlán, Av. Sábalo-Cerritos s/n. Estero del Yugo, Mazatlán, Sinaloa 82000, Mexico.
| | - Ana M Ibarra
- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Av. Instituto Politécnico Nacional No.195, Col. Playa Palo de Santa Rita, La Paz, Baja California Sur 23096, Mexico.
| |
Collapse
|
24
|
Long L, Wu SG, Yuan F, Zhang HJ, Wang J, Qi GH. Effects of dietary octacosanol supplementation on laying performance, egg quality, serum hormone levels, and expression of genes related to the reproductive axis in laying hens. Poult Sci 2017; 96:894-903. [PMID: 27665009 DOI: 10.3382/ps/pew316] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/29/2016] [Indexed: 11/20/2022] Open
Abstract
This experiment was conducted to evaluate the effects of dietary octacosanol supplementation on laying performance, egg quality, serum hormone levels, and gene expression related to reproductive axis in laying hens to confirm the reproduction-promoting function of octacosanol. In total, 360 Hy-Line Brown (67-wk-old) laying hens were randomly assigned to one of three treatments with 0, 5, and 10 mg octacosanol (extracted from rice bran, purity >92%)/kg feed. The feeding trial lasted for 10 weeks. The results showed that the dietary addition of 5 and 10 mg/kg octacosanol improved feed efficiency by 4.9% and 3.4% (P < 0.01), increased the albumen height by 20.5% and 13.3% (P < 0.01), the Haugh unit score by 12.9% and 8.7% (P < 0.01), and the eggshell strength by 39.5% and 24.5% (P < 0.01), respectively, compared with the control diet. Dietary octacosanol addition significantly affected serum triiodothyronine, estradiol, follicle-stimulating hormone levels (P < 0.05), and progesterone and luteinizing hormone level (P < 0.01). Compared with the control, dietary addition of octacosanol at 5 mg/kg promoted the follicle-stimulating hormone receptor (FSHR) mRNA expression in different-sized follicles, and significantly increased the FSHR mRNA expression of granulosa cells from the F2 and F3 follicles (P < 0.05). Dietary supplementation with both 5 and 10 mg/kg octacosanol promoted the mRNA expression of luteinizing hormone receptor and prolactin receptor in different-sized follicles, and significantly up-regulated the expression levels in F1 granulosa cells (P < 0.05). The ovarian weight was significantly increased with the dietary addition of 5 mg/kg octacosanol (P < 0.05). The numbers of small yellow follicles and large white follicles were increased with the addition of dietary 5 and 10 mg/kg octacosanol (P < 0.01). This study provides evidence that octacosanol has the capacity to improve reproductive performance, indicating that it is a potentially effective feed additive in egg production.
Collapse
Affiliation(s)
- L Long
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Tianjin Naer Biotechnology Co., Ltd., Tianjin 300457, China
| | - S G Wu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - F Yuan
- Tianjin Naer Biotechnology Co., Ltd., Tianjin 300457, China
| | - H J Zhang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - J Wang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - G H Qi
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
25
|
|
26
|
Wisden W. A Tribute to Peter H Seeburg (1944-2016): A Founding Father of Molecular Neurobiology. Front Mol Neurosci 2016; 9:133. [PMID: 27965536 PMCID: PMC5126100 DOI: 10.3389/fnmol.2016.00133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 11/15/2016] [Indexed: 11/25/2022] Open
Affiliation(s)
- William Wisden
- Department of Life Sciences and Centre for Neurotechnology, Imperial College London London, UK
| |
Collapse
|
27
|
Rao CV. Protective Effects of Human Chorionic Gonadotropin Against Breast Cancer: How Can We Use This Information to Prevent/Treat the Disease? Reprod Sci 2016; 24:1102-1110. [DOI: 10.1177/1933719116676396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- C. V. Rao
- Department of Cellular Biology and Pharmacology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Molecular and Human Genetics, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
28
|
Affiliation(s)
- Elizabeth A. Stewart
- Department of Obstetrics, Gynecology and Reproductive Biology, Bngham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Center for Uterine Fibroids, Brigham and Women's Hospital, 75 Francis Street, Boston, Massachusetts 02115
| |
Collapse
|
29
|
Abstract
Alzheimer disease (AD) is a slow progressive neurodegenerative disease that affects more elderly women than elderly men. It impairs memory, typically progresses into multidomain cognitive decline that destroys the quality of life, and ultimately leads to death. About 5.3 million older Americans are now living with this disease, and this number is projected to rise to 14 million by 2050. Annual health-care costs in the United States alone are projected to increase to about US$1.1 trillion by 2050. The initial theory that decreasing estrogen levels leads to AD development in postmenopausal women has been proven inconclusive. For example, Women's Health Research Initiative Memory Study and the population-based nested case-control study have failed to demonstrate that estrogen/progesterone (hormone replacement therapy [HRT]) or estrogen replacement therapy could prevent the cognitive decline or reduce the risk of AD. This led to the realization that AD development could be due to a progressive increase in luteinizing hormone (LH) levels in postmenopausal women. Accordingly, a large number of studies have demonstrated that an increase in LH levels is positively correlated with neuropathological, behavioral, and cognitive changes in AD. In addition, LH has been shown to promote amyloidogenic pathway of precursor protein metabolism and deposition of amyloid β plaques in the hippocampus, a region involved in AD. Cognate receptors that mediate LH effects are abundantly expressed in the hippocampus. Reducing the LH levels by treatment with gonadotropin-releasing hormone agonists could provide therapeutic benefits. Despite these advances, many questions remain and require further research.
Collapse
Affiliation(s)
- C V Rao
- 1 Department of Cellular Biology and Pharmacology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,2 Department of Molecular and Human Genetics, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.,3 Department of Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
30
|
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
|
31
|
STORAGE OF ACTIVITY OF THE HUMAN CHORIONIC GONADOTROPIN HORMONE IN SOLUTION AT ADDITION OF ORGANIC COMPOUNDS TO THE PHARMACOLOGICAL COMPOSITION. EUREKA: LIFE SCIENCES 2016. [DOI: 10.21303/2504-5695.2016.00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The more stable among the tested samples were samples with saccharose in the concentration of 50–75 mg per cm3. While adding of L- lysine to samples the most stable activity was discovered in the experimental series of samples with the content of lysine of 10 mg per cm3 – activity increased by 54 % as compared to theoretical initial activity of HCG during 8 weeks. While storing gonadotropin with L-glycine fluctuations of hormone activity in all series of samples were observed. Adding of 0.2 mg per cm3 of L-glycine had a more expressed stabilizing effect. Adding of 0.2 mg per cm3 of L-methionine produced relatively high and stable activity of gonadotropin during the 6 weeks storage. Adding of 0.25 mg / cm3 of L- glycine and 75.50 mg / cm3 of saccharose to experimental samples during 2 weeks at 40 °C provided 69.8 % and 60.7 % saving activity of hCG respectively. Activity of gonadotropin in a series of samples with the addition of L- glycine and mannitol was significantly lower and at the end of the study was at an appropriate rate with the control series models. The highest activity of gonadotropin was detected while adding fillers – 10 mg / cm3 L-lysine and 75 mg / cm3 saccharose and mannitol – to recipes as a stabilizer.
Collapse
|
32
|
Gharwan H, Bunch KP, Annunziata CM. The role of reproductive hormones in epithelial ovarian carcinogenesis. Endocr Relat Cancer 2015; 22:R339-63. [PMID: 26373571 DOI: 10.1530/erc-14-0550] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/15/2015] [Indexed: 12/12/2022]
Abstract
Epithelial ovarian cancer comprises ∼85% of all ovarian cancer cases. Despite acceptance regarding the influence of reproductive hormones on ovarian cancer risk and considerable advances in the understanding of epithelial ovarian carcinogenesis on a molecular level, complete understanding of the biologic processes underlying malignant transformation of ovarian surface epithelium is lacking. Various hypotheses have been proposed over the past several decades to explain the etiology of the disease. The role of reproductive hormones in epithelial ovarian carcinogenesis remains a key topic of research. Primary questions in the field of ovarian cancer biology center on its developmental cell of origin, the positive and negative effects of each class of hormones on ovarian cancer initiation and progression, and the role of the immune system in the ovarian cancer microenvironment. The development of the female reproductive tract is dictated by the hormonal milieu during embryogenesis. Intensive research efforts have revealed that ovarian cancer is a heterogenous disease that may develop from multiple extra-ovarian tissues, including both Müllerian (fallopian tubes, endometrium) and non-Müllerian structures (gastrointestinal tissue), contributing to its heterogeneity and distinct histologic subtypes. The mechanism underlying ovarian localization, however, remains unclear. Here, we discuss the role of reproductive hormones in influencing the immune system and tipping the balance against or in favor of developing ovarian cancer. We comment on animal models that are critical for experimentally validating existing hypotheses in key areas of endocrine research and useful for preclinical drug development. Finally, we address emerging therapeutic trends directed against ovarian cancer.
Collapse
Affiliation(s)
- Helen Gharwan
- National Cancer InstituteNational Institutes of Health, 10 Center Drive, Building 10, 12N226, Bethesda, Maryland 20892-1906, USAWomen's Malignancies BranchNational Cancer Institute, National Institutes of Health, Center for Cancer Research, Bethesda, Maryland, USADepartment of Gynecologic OncologyWalter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Kristen P Bunch
- National Cancer InstituteNational Institutes of Health, 10 Center Drive, Building 10, 12N226, Bethesda, Maryland 20892-1906, USAWomen's Malignancies BranchNational Cancer Institute, National Institutes of Health, Center for Cancer Research, Bethesda, Maryland, USADepartment of Gynecologic OncologyWalter Reed National Military Medical Center, Bethesda, Maryland, USA National Cancer InstituteNational Institutes of Health, 10 Center Drive, Building 10, 12N226, Bethesda, Maryland 20892-1906, USAWomen's Malignancies BranchNational Cancer Institute, National Institutes of Health, Center for Cancer Research, Bethesda, Maryland, USADepartment of Gynecologic OncologyWalter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Christina M Annunziata
- National Cancer InstituteNational Institutes of Health, 10 Center Drive, Building 10, 12N226, Bethesda, Maryland 20892-1906, USAWomen's Malignancies BranchNational Cancer Institute, National Institutes of Health, Center for Cancer Research, Bethesda, Maryland, USADepartment of Gynecologic OncologyWalter Reed National Military Medical Center, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Brüser A, Schulz A, Rothemund S, Ricken A, Calebiro D, Kleinau G, Schöneberg T. The Activation Mechanism of Glycoprotein Hormone Receptors with Implications in the Cause and Therapy of Endocrine Diseases. J Biol Chem 2015; 291:508-20. [PMID: 26582202 DOI: 10.1074/jbc.m115.701102] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Indexed: 11/06/2022] Open
Abstract
Glycoprotein hormones (GPHs) are the main regulators of the pituitary-thyroid and pituitary-gonadal axes. Selective interaction between GPHs and their cognate G protein-coupled receptors ensure specificity in GPH signaling. The mechanisms of how these hormones activate glycoprotein hormone receptors (GPHRs) or how mutations and autoantibodies can alter receptor function were unclear. Based on the hypothesis that GPHRs contain an internal agonist, we systematically screened peptide libraries derived from the ectodomain for agonistic activity on the receptors. We show that a peptide (p10) derived from a conserved sequence in the C-terminal part of the extracellular N terminus can activate all GPHRs in vitro and in GPHR-expressing tissues. Inactivating mutations in this conserved region or in p10 can inhibit activation of the thyroid-stimulating hormone receptor by autoantibodies. Our data suggest an activation mechanism where, upon extracellular ligand binding, this intramolecular agonist isomerizes and induces structural changes in the 7-transmembrane helix domain, triggering G protein activation. This mechanism can explain the pathophysiology of activating autoantibodies and several mutations causing endocrine dysfunctions such as Graves disease and hypo- and hyperthyroidism. Our findings highlight an evolutionarily conserved activation mechanism of GPHRs and will further promote the development of specific ligands useful to treat Graves disease and other dysfunctions of GPHRs.
Collapse
Affiliation(s)
| | | | | | - Albert Ricken
- Institute of Anatomy, Medical Faculty, University of Leipzig, 04103 Leipzig
| | - Davide Calebiro
- the Institute of Pharmacology and Toxicology & Bio-Imaging Center/Rudolf Virchow Center, University of Würzburg, 97078 Würzburg, and
| | - Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | | |
Collapse
|
34
|
Charmandari E, Guan R, Zhang M, Silveira LG, Fan QR, Chrousos GP, Sertedaki AC, Latronico AC, Segaloff DL. Misfolding Ectodomain Mutations of the Lutropin Receptor Increase Efficacy of Hormone Stimulation. Mol Endocrinol 2015; 30:62-76. [PMID: 26554443 DOI: 10.1210/me.2015-1205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We demonstrate 2 novel mutations of the LHCGR, each homozygous, in a 46,XY patient with severe Leydig cell hypoplasia. One is a mutation in the signal peptide (p.Gln18_Leu19ins9; referred to here as SP) that results in an alteration of the coding sequence of the N terminus of the mature mutant receptor. The other mutation (p.G71R) is also within the ectodomain. Similar to many other inactivating mutations, the cell surface expression of recombinant human LHR(SP,G71R) is greatly reduced due to intracellular retention. However, we made the unusual discovery that the intrinsic efficacy for agonist-stimulated cAMP in the reduced numbers of receptors on the cell surface was greatly increased relative to the same low number of cell surface wild-type receptor. Remarkably, this appears to be a general attribute of misfolding mutations in the ectodomains, but not serpentine domains, of the gonadotropin receptors. These findings suggest that there must be a common, shared mechanism by which disparate mutations in the ectodomain that cause misfolding and therefore reduced cell surface expression concomitantly confer increased agonist efficacy to those receptor mutants on the cell surface. Our data further suggest that, due to their increased agonist efficacy, extremely small changes in cell surface expression of misfolded ectodomain mutants cause larger than expected alterations in the cellular response to agonist. Therefore, for inactivating LHCGR mutations causing ectodomain misfolding, the numbers of cell surface mutant receptors on fetal Leydig cells of 46,XY individuals exert a more exquisite effect on the relative severity of the clinical phenotypes than already appreciated.
Collapse
Affiliation(s)
- E Charmandari
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - R Guan
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - M Zhang
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - L G Silveira
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Q R Fan
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - G P Chrousos
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - A C Sertedaki
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - A C Latronico
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - D L Segaloff
- Division of Endocrinology, Metabolism and Diabetes (E.C., G.P.C., A.C.S.), First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, and Division of Endocrinology and Metabolism (E.C., G.P.C., A.C.S.), Clinical Research Center, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece; Department of Pharmacology (Q.R.F.), Columbia University Medical Center, New York, New York 10032; Unidade de Endocrinologia do Desenvolvimento (L.G.S., A.C.L.), Laboratório de Hormônios e Genética Molecular LIM/42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; and Department of Molecular Physiology and Biophysics (M.Z., R.G., D.L.S.), The University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| |
Collapse
|
35
|
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
|
36
|
Blair JA, Bhatta S, McGee H, Casadesus G. Luteinizing hormone: Evidence for direct action in the CNS. Horm Behav 2015; 76:57-62. [PMID: 26172857 PMCID: PMC4741372 DOI: 10.1016/j.yhbeh.2015.06.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 01/09/2023]
Abstract
This article is part of a Special Issue "SBN 2014". Hormonal dysfunction due to aging, especially during menopause, plays a substantial role in cognitive decline as well as the progression and development of neurodegenerative diseases. The hypothalamic-pituitary-gonadal (HPG) axis has long been implicated in changes in behavior and neuronal morphology. Most notably, estrogens have proven beneficial in the healthy brain through a host of different mechanisms. Recently, luteinizing hormone (LH) has emerged as a candidate for further investigation for its role in the CNS. The basis of this is that both LH and the LH receptor are expressed in the brain, and serum levels of LH correlate with cognitive deficits and Alzheimer's disease (AD) incidence. The study of LH in cognition and AD primarily focuses on evaluating the effects of downregulation of this peptide. This literature has shown that decreasing peripheral LH, through a variety of pharmacological interventions, reduces cognitive deficits in ovariectomy and AD models. However, few studies have researched the direct actions of LH on neurons and glial cells. Here we summarize the role of luteinizing hormone in modulating cognition, and we propose a mechanism that underlies a role for brain LH in this process.
Collapse
Affiliation(s)
- Jeffrey A Blair
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Sabina Bhatta
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Henry McGee
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
37
|
Menon B, Gulappa T, Menon KMJ. miR-122 Regulates LH Receptor Expression by Activating Sterol Response Element Binding Protein in Rat Ovaries. Endocrinology 2015; 156:3370-80. [PMID: 26125464 PMCID: PMC4541618 DOI: 10.1210/en.2015-1121] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
LH/human chorionic gonadotropin receptor (LHR) undergoes down-regulation during preovulatory LH surge or in response to exposure to a supraphysiological concentration of its ligands through a posttranscriptional mechanism involving an RNA binding protein designated as LHR mRNA binding protein (LRBP). miR-122, a short noncoding RNA, has been shown to mediate the up-regulation of LRBP. In the present study, we show that inhibition of miR-122 using a locked nucleic acid (LNA)-conjugated antagomir suppressed human chorionic gonadotropin (hCG)-induced up-regulation of LRBP as well as its association with LHR mRNA, as analyzed by RNA EMSA. Most importantly, inhibition of miR-122 resulted in the abolishment of hCG-mediated LHR mRNA down-regulation. We also show that the transcription factor, sterol regulatory element binding protein (SREBP) (SREBP-1a and SREBP-2 isoforms), is an intermediate in miR-122-mediated LHR mRNA regulation. HCG-stimulated increase in the activation of both SREBP-1a and SREBP-2 was inhibited by pretreatment with the miR-122 antagomir. The inhibition of cAMP/protein kinase A (PKA) and ERK pathways, upstream activators of miR-122, abolished SREBP activation after hCG treatment. SREBP-mediated regulation of LRBP expression is mediated by recruitment of LRBP promoter element to SREBP-1a, because chromatin immunoprecipitation assay revealed that association of LRBP promoter to SREBP was increased by hCG treatment. Pretreatment with miR-122 antagomir suppressed this response. Inhibition of SREBP activation by pretreating the rats with a chemical compound, fatostatin abrogated hCG-induced up-regulation of LRBP mRNA and protein. Fatostatin also inhibited LHR-LRBP mRNA-protein complex formation and LHR down-regulation. These results conclusively show that miR-122 plays a regulatory role in LH/hCG-induced LHR mRNA down-regulation by increasing LRBP expression through the activation of SREBP pathway.
Collapse
Affiliation(s)
- Bindu Menon
- Departments of Obstetrics/Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0617
| | - Thippeswamy Gulappa
- Departments of Obstetrics/Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0617
| | - K M J Menon
- Departments of Obstetrics/Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109-0617
| |
Collapse
|
38
|
Maugars G, Dufour S. Demonstration of the Coexistence of Duplicated LH Receptors in Teleosts, and Their Origin in Ancestral Actinopterygians. PLoS One 2015; 10:e0135184. [PMID: 26271038 PMCID: PMC4536197 DOI: 10.1371/journal.pone.0135184] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/18/2015] [Indexed: 11/18/2022] Open
Abstract
Pituitary gonadotropins, FSH and LH, control gonad activity in vertebrates, via binding to their respective receptors, FSHR and LHR, members of GPCR superfamily. Until recently, it was accepted that gnathostomes possess a single FSHR and a single LHR, encoded by fshr and lhcgr genes. We reinvestigated this question, focusing on vertebrate species of key-phylogenetical positions. Genome analyses supported the presence of a single fshr and a single lhcgr in chondrichthyans, and in sarcopterygians including mammals, birds, amphibians and coelacanth. In contrast, we identified a single fshr but two lhgcr in basal teleosts, the eels. We further showed the coexistence of duplicated lhgcr in other actinopterygians, including a non-teleost, the gar, and other teleosts, e.g. Mexican tetra, platyfish, or tilapia. Phylogeny and synteny analyses supported the existence in actinopterygians of two lhgcr paralogs (lhgcr1/ lhgcr2), which do not result from the teleost-specific whole-genome duplication (3R), but likely from a local gene duplication that occurred early in the actinopterygian lineage. Due to gene losses, there was no impact of 3R on the number of gonadotropin receptors in extant teleosts. Additional gene losses during teleost radiation, led to a single lhgcr (lhgcr1 or lhgcr2) in some species, e.g. medaka and zebrafish. Sequence comparison highlighted divergences in the extracellular and intracellular domains of the duplicated lhgcr, suggesting differential properties such as ligand binding and activation mechanisms. Comparison of tissue distribution in the European eel, revealed that fshr and both lhgcr transcripts are expressed in the ovary and testis, but are differentially expressed in non-gonadal tissues such as brain or eye. Differences in structure-activity relationships and tissue expression may have contributed as selective drives in the conservation of the duplicated lhgcr. This study revises the evolutionary scenario and nomenclature of gonadotropin receptors, and opens new research avenues on the roles of duplicated LHR in actinopterygians.
Collapse
Affiliation(s)
- Gersende Maugars
- Muséum National d'Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208-IRD 207-UPMC-UCBN, Paris, France
| | - Sylvie Dufour
- Muséum National d'Histoire Naturelle, Sorbonne Universités, Research Unit BOREA, Biology of Aquatic Organisms and Ecosystems, CNRS 7208-IRD 207-UPMC-UCBN, Paris, France
| |
Collapse
|
39
|
Ma Q, Fan J, Wang J, Yang S, Cong Q, Wang R, Lv Q, Liu R, Ning G. High levels of chorionic gonadotrophin attenuate insulin sensitivity and promote inflammation in adipocytes. J Mol Endocrinol 2015; 54:161-70. [PMID: 25691497 DOI: 10.1530/jme-14-0284] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gestational diabetes mellitus (GDM) presents with moderate inflammation, insulin resistance and impaired glucose uptake, which may result from increased maternal fat mass and increased circulation of placental hormones and adipokines. In this study, we set out to test whether the surge in chorionic gonadotrophin (CG) secretion is a cause of inflammation and impaired insulin sensitivity in GDM. We first found that LH/chorionic gonadotrophin receptors (CG/LHR) were expressed at low levels in insulin-sensitive murine 3T3-L1 adipocytes and murine C2C12 myocytes. CG treatment not only directly reduced insulin-responsive gene expression, including that of glucose transporter 4 (GLUT4), but also impaired insulin-stimulated glucose uptake in 3T3-L1 cells. Moreover, CG treatment increased the expression of the proinflammatory cytokine monocyte chemotactic protein 1 (MCP1) and upregulated nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activity in 3T3-L1 cells. Clinically, pregnant women who had higher CG levels and elevated MCP1 developed GDM. Above all, apart from prepregnancy BMI and MCP1 level, CG level was associated with abnormal glucose tolerance. In summary, our findings confirmed that higher CG levels in pregnancy possibly played a role in GDM development partly by impairing the functions of insulin, such those involved in as glucose uptake, while promoting inflammation in adipocyte.
Collapse
Affiliation(s)
- Qinyun Ma
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Jianxia Fan
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Jiqiu Wang
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Shuai Yang
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Qing Cong
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Rui Wang
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Qianqian Lv
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Ruixin Liu
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| | - Guang Ning
- Shanghai Key Laboratory for Endocrine Tumors and E-Institute of Shanghai UniversitiesDepartment of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrinology and Metabolism, Shanghai Clinical Center for Endocrine and Metabolic Diseases, China National Clinical Research for Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, ChinaDepartment of Obstetrics and GynecologyInternational Peace Maternity and Child Health Hospital affiliated Shanghai Jiaotong University School of Medicine, 910 Hengshan Road, Shanghai 200030, ChinaDepartment of Obstetrics and GynecologyObstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai 200011, China
| |
Collapse
|
40
|
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: 0.8] [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
|
41
|
Cahoreau C, Klett D, Combarnous Y. Structure-function relationships of glycoprotein hormones and their subunits' ancestors. Front Endocrinol (Lausanne) 2015; 6:26. [PMID: 25767463 PMCID: PMC4341566 DOI: 10.3389/fendo.2015.00026] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/13/2015] [Indexed: 12/22/2022] Open
Abstract
Glycoprotein hormones (GPHs) are the most complex molecules with hormonal activity. They exist only in vertebrates but the genes encoding their subunits' ancestors are found in most vertebrate and invertebrate species although their roles are still unknown. In the present report, we review the available structural and functional data concerning GPHs and their subunits' ancestors.
Collapse
Affiliation(s)
- Claire Cahoreau
- Physiologie de la Reproduction et des Comportements (PRC), Centre National de la Recherche Scientifique, INRA, Nouzilly, France
| | - Danièle Klett
- Physiologie de la Reproduction et des Comportements (PRC), Centre National de la Recherche Scientifique, INRA, Nouzilly, France
| | - Yves Combarnous
- Physiologie de la Reproduction et des Comportements (PRC), Centre National de la Recherche Scientifique, INRA, Nouzilly, France
- *Correspondence: Yves Combarnous, Physiologie de la Reproduction et des Comportements (PRC), Centre National de la Recherche Scientifique, INRA, Nouzilly 37380, France e-mail:
| |
Collapse
|
42
|
Blair JA, McGee H, Bhatta S, Palm R, Casadesus G. Hypothalamic-pituitary-gonadal axis involvement in learning and memory and Alzheimer's disease: more than "just" estrogen. Front Endocrinol (Lausanne) 2015; 6:45. [PMID: 25859241 PMCID: PMC4373369 DOI: 10.3389/fendo.2015.00045] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/12/2015] [Indexed: 11/25/2022] Open
Abstract
Accumulating studies affirm the effects of age-related endocrine dysfunction on cognitive decline and increasing risk of neurodegenerative diseases. It is well known that estrogens can be protective for cognitive function, and more recently androgens and luteinizing hormone have also been shown to modulate learning and memory. Understanding the mechanisms underlying hypothalamic-pituitary-gonadal axis-associated cognitive dysfunction is crucial for therapeutic advancement. Here, we emphasize that reproductive hormones are influential in maintaining neuronal health and enhancing signaling cascades that lead to cognitive impairment. We summarize and critically evaluate age-related changes in the endocrine system, their implications in the development of Alzheimer's disease, and the therapeutic potential of endocrine modulation in the prevention of age-related cognitive decline.
Collapse
Affiliation(s)
- Jeffrey A. Blair
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Henry McGee
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Sabina Bhatta
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Russell Palm
- University of Toledo School of Medicine, Toledo, OH, USA
| | - Gemma Casadesus
- Department of Biological Sciences, Kent State University, Kent, OH, USA
- *Correspondence: Gemma Casadesus, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH 44242, USA e-mail:
| |
Collapse
|
43
|
Sisinni L, Landriscina M. The Role of Human Chorionic Gonadotropin as Tumor Marker: Biochemical and Clinical Aspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 867:159-76. [PMID: 26530366 DOI: 10.1007/978-94-017-7215-0_11] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Tumor markers are biological substances that are produced/released mainly by malignant tumor cells, enter the circulation in detectable amounts and are potential indicators of the presence of a tumor. The most useful biochemical markers are the tumor-specific molecules, i.e., receptors, enzymes, hormones, growth factors or biological response modifiers that are specifically produced by tumor cells and not, or minimally, by the normal counterpart (Richard et al. Principles and practice of gynecologic oncology. Wolters Kluwer Health, Philadelphia, 2009). Based on their specificity and sensitivity in each malignancy, biomarkers are used for screening, diagnosis, disease monitoring and therapeutic response assessment in clinical management of cancer patients.This chapter is focused on human chorionic gonadotropin (hCG), a hormone with a variety of functions and widely used as a tumor biomarker in selected tumors. Indeed, hCG is expressed by both trophoblastic and non-trophoblastic human malignancies and plays a role in cell transformation, angiogenesis, metastatization, and immune escape, all process central to cancer progression. Of note, hCG testing is crucial for the clinical management of placental trophoblastic malignancies and germ cell tumors of the testis and the ovary. Furthermore, the production of hCG by tumor cells is accompanied by varying degrees of release of the free subunits into the circulation, and this is relevant for the management of cancer patients (Triozzi PL, Stevens VC, Oncol Rep 6(1):7-17, 1999).The name chorionic gonadotropin was conceived: chorion derives from the latin chordate meaning afterbirth, gonadotropin indicates that the hormone is a gonadotropic molecule, acting on the ovaries and promoting steroid production (Cole LA, Int J Endocrinol Metab 9(2):335-352, 2011). The function, the mechanism of action and the interaction between hCG and its receptor continue to be the subject of intensive investigation, even though many issues about hCG have been well documented (Tegoni M et al., J Mol Biol 289(5):1375-1385, 1999).
Collapse
Affiliation(s)
- Lorenza Sisinni
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, Rionero in Vulture, PZ, Italy
| | - Matteo Landriscina
- Clinical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto, 1, 71100, Foggia, Italy.
| |
Collapse
|
44
|
Jiang X, Dias JA, He X. Structural biology of glycoprotein hormones and their receptors: insights to signaling. Mol Cell Endocrinol 2014; 382:424-451. [PMID: 24001578 DOI: 10.1016/j.mce.2013.08.021] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/20/2013] [Accepted: 08/24/2013] [Indexed: 01/18/2023]
Abstract
This article reviews the progress made in the field of glycoprotein hormones (GPH) and their receptors (GPHR) by several groups of structural biologists including ourselves aiming to gain insight into GPH signaling mechanisms. The GPH family consists of four members, with follicle-stimulating hormone (FSH) being the prototypic member. GPH members belong to the cystine-knot growth factor superfamily, and their receptors (GPHR), possessing unusually large N-terminal ectodomains, belong to the G-protein coupled receptor Family A. GPHR ectodomains can be divided into two subdomains: a high-affinity hormone binding subdomain primarily centered on the N-terminus, and a second subdomain that is located on the C-terminal region of the ectodomain that is involved in signal specificity. The two subdomains unexpectedly form an integral structure comprised of leucine-rich repeats (LRRs). Following the structure determination of hCG in 1994, the field of FSH structural biology has progressively advanced. Initially, the FSH structure was determined in partially glycosylated free form in 2001, followed by a structure of FSH bound to a truncated FSHR ectodomain in 2005, and the structure of FSH bound to the entire ectodomain in 2012. Comparisons of the structures in three forms led a proposal of a two-step monomeric receptor activation mechanism. First, binding of FSH to the FSHR high-affinity hormone-binding subdomain induces a conformational change in the hormone to form a binding pocket that is specific for a sulfated-tyrosine found as sTyr 335 in FSHR. Subsequently, the sTyr is drawn into the newly formed binding pocket, producing a lever effect on a helical pivot whereby the docking sTyr provides as the 'pull & lift' force. The pivot helix is flanked by rigid LRRs and locked by two disulfide bonds on both sides: the hormone-binding subdomain on one side and the last short loop before the first transmembrane helix on the other side. The lift of the sTyr loop frees the tethered extracellular loops of the 7TM domain, thereby releasing a putative inhibitory influence of the ectodomain, ultimately leading to the activating conformation of the 7TM domain. Moreover, the data lead us to propose that FSHR exists as a trimer and to present an FSHR activation mechanism consistent with the observed trimeric crystal form. A trimeric receptor provides resolution of the enigmatic, but important, biological roles played by GPH residues that are removed from the primary FSH-binding site, as well as several important GPCR phenomena, including negative cooperativity and asymmetric activation. Further reflection pursuant to this review process revealed additional novel structural characteristics such as the identification of a 'seat' sequence in GPH. Together with the 'seatbelt', the 'seat' enables a common heteodimeric mode of association of the common α subunit non-covalently and non-specifically with each of the three different β subunits. Moreover, it was possible to establish a dimensional order that can be used to estimate LRR curvatures. A potential binding pocket for small molecular allosteric modulators in the FSHR 7TM domain has also been identified.
Collapse
Affiliation(s)
- Xuliang Jiang
- EMD Serono Research & Development Institute, Billerica, MA 01821, United States.
| | - James A Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany-SUNY, Albany, NY 12222, United States
| | - Xiaolin He
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| |
Collapse
|
45
|
Law NC, Weck J, Kyriss B, Nilson JH, Hunzicker-Dunn M. Lhcgr Expression in Granulosa Cells: Roles for PKA-Phosphorylated β-Catenin, TCF3, and FOXO1. Mol Endocrinol 2013; 27:1295-310. [PMID: 23754802 PMCID: PMC3725343 DOI: 10.1210/me.2013-1025] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ovarian follicles lacking FSH or FSH receptors fail to progress to a preovulatory stage, resulting in infertility. One hallmark of the preovulatory follicle is the presence of luteinizing hormone/choriogonadotropin receptors (LHCGR) on granulosa cells (GCs). However, the mechanisms by which FSH induces Lhcgr gene expression are poorly understood. Our results show that protein kinase A (PKA) and phosphoinositide 3-kinase (PI3K)/AKT pathways are required for FSH to activate both the murine Lhcgr-luciferase reporter and expression of Lhcgr mRNA in rat GCs. Based on results showing that an adenovirus (Ad) expressing a steroidogenic factor 1 (SF1) mutant that cannot bind β-catenin abolished FSH-induced Lhcgr mRNA, we evaluated the role of β-catenin in the regulation of Lhcgr gene expression. FSH promoted the PKA-dependent, PI3K-independent phosphorylation of β-catenin on Ser552 and Ser665. FSH activated the β-catenin/T-cell factor (TCF) artificial promoter-reporter TOPFlash via a PKA-dependent, PI3K-independent pathway, and dominant-negative (DN) TCF abolished FSH-activated Lhcgr-luciferase reporter and induction of Lhcgr mRNA. Microarray analysis of GCs treated with Ad-DN-TCF and FSH identified the Lhcgr as the most down-regulated gene. Chromatin immunoprecipitation results placed β-catenin phosphorylated on Ser552 and Ser675 and SF1 on the Lhcgr promoter in FSH-treated GCs; TCF3 was constitutively associated with the Lhcgr promoter. Transduction with an Ad-phospho-β-catenin mutant (Ser552/665/Asp) enhanced Lhcgr mRNA expression in FSH-treated cells greater than 3-fold. Finally, we identified a recognized PI3K/AKT target, forkhead box O1, as a negative regulator of Lhcgr mRNA expression. These results provide new understanding of the complex regulation of Lhcgr gene expression in GCs.
Collapse
Affiliation(s)
- Nathan C Law
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-7520, USA
| | | | | | | | | |
Collapse
|
46
|
de Jong J, Nap M, Merz WE, Leunissen JE, Jäger W. Direct Gold-Silver Method for HCG Binding to Specific Tissue Structures. J Histotechnol 2013. [DOI: 10.1179/his.1992.15.4.275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
47
|
Kogure K, Nakamura K, Ikeda S, Kitahara Y, Nishimura T, Iwamune M, Minegishi T. Glucose-Regulated Protein, 78-Kilodalton Is a Modulator of Luteinizing Hormone Receptor Expression in Luteinizing Granulosa Cells in Rats1. Biol Reprod 2013; 88:8. [DOI: 10.1095/biolreprod.112.101873] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
48
|
Dukic-Stefanovic S, Walther J, Wosch S, Zimmermann G, Wiedemann P, Alexander H, Claudepierre T. Chorionic gonadotropin and its receptor are both expressed in human retina, possible implications in normal and pathological conditions. PLoS One 2012; 7:e52567. [PMID: 23285091 PMCID: PMC3526580 DOI: 10.1371/journal.pone.0052567] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 11/20/2012] [Indexed: 11/18/2022] Open
Abstract
Extra-gonadal role of gonadotropins has been re-evaluated over the last 20 years. In addition to pituitary secretion of luteinizing hormone (LH) and follicle stimulating hormone (FSH), the CNS has been clearly identified as a source of hCG acting locally to influence behaviour. Here we demonstrated that human retina is producing this gonadotropin that acts as a neuroactive molecule. Müller glial and retinal pigmented epithelial (RPE) cells are producing hCG that may affects neighbour cells expressing its receptor, namely cone photoreceptors. It was previously described that amacrine and retinal ganglion (RGC) cells are targets of the gonadotropin releasing hormone that control the secretion of all gonadotropins. Therefore our findings suggest that a complex neuroendocrine circuit exists in the retina, involving hCG secreting cells (glial and RPE), hCG targets (photoreceptors) and hCG-release controlling cells (amacrine and RGC). The exact physiological functions of this circuit have still to be identified, but the proliferation of photoreceptor-derived tumor induced by hCG demonstrated the need to control this neuroendocrine loop.
Collapse
MESH Headings
- Blotting, Western
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Chorionic Gonadotropin, beta Subunit, Human/metabolism
- Chorionic Gonadotropin, beta Subunit, Human/pharmacology
- Gene Expression Regulation
- Humans
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, LH/metabolism
- Retina/metabolism
- Retina/pathology
- Retinoblastoma/metabolism
- Retinoblastoma/pathology
Collapse
Affiliation(s)
| | - Jan Walther
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Sebastian Wosch
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Gerolf Zimmermann
- Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, University of Leipzig, Leipzig, Germany
| | - Peter Wiedemann
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Henry Alexander
- Department of Obstetrics and Gynecology, Division of Human Reproduction and Endocrinology, University of Leipzig, Leipzig, Germany
| | - Thomas Claudepierre
- Department of Ophthalmology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
49
|
Yu Y, Pang Y, Zhao H, Xu X, Wu Z, An L, Tian J. Association of a missense mutation in the luteinizing hormone/choriogonadotropin receptor gene (LHCGR) with superovulation traits in Chinese Holstein heifers. J Anim Sci Biotechnol 2012; 3:35. [PMID: 23140330 PMCID: PMC3502498 DOI: 10.1186/2049-1891-3-35] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 08/07/2012] [Indexed: 11/22/2022] Open
Abstract
Background Upon binding luteinizing hormone in the ovary, the luteinizing hormone/choriogonadotropin receptor (LHCGR) is necessary for follicular maturation and ovulation, as well as luteal function. We detected mutations in the LHCGR gene and evaluated their association with superovulation. Methods Using polymerase chain reaction-single strand conformation polymorphism (PCR-SSCP) and DNA sequencing, we examined polymorphisms in LHCGR and the genotypes associated with superovulation traits in 127 Chinese Holstein heifers. Results A G/T polymorphism (ss52050737) in exon 11 was significantly associated with the total number of ova and the number of transferable embryos. Conclusions LHCGR may be a new predictor for superovulation in Chinese Holstein heifers.
Collapse
Affiliation(s)
- Yong Yu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Yunwei Pang
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Haichao Zhao
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Xiaoling Xu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Zhonghong Wu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Lei An
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| | - Jianhui Tian
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, PR China
| |
Collapse
|
50
|
Yamaguchi M, Ohba T, Tashiro H, Yamada G, Katabuchi H. Human chorionic gonadotropin induces human macrophages to form intracytoplasmic vacuoles mimicking Hofbauer cells in human chorionic villi. Cells Tissues Organs 2012; 197:127-35. [PMID: 23128164 DOI: 10.1159/000342806] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2012] [Indexed: 11/19/2022] Open
Abstract
The most characteristic morphological feature of macrophages in the stroma of placental villi, known as Hofbauer cells, is their highly vacuolated appearance. They also show positive immunostaining for human chorionic gonadotropin (hCG) and express messenger ribonucleic acid of the luteinizing hormone/chorionic gonadotropin receptor with a deletion of exon 9 (LH/CG-R Δ9). Maternal hCG enters fetal plasma through the mesenchyme of the placental villi and promotes male sexual differentiation in early pregnancy; therefore, excess hCG may induce aberrant genital differentiation and hCG must be adjusted at the fetomaternal interface. We hypothesized that hCG is regulated by Hofbauer cells and that their peculiar vacuoles are involved in a cell-specific function. To assess the morphological modification and expression of LH/CG-R Δ9 in human macrophages after hCG exposure, the present study examined phorbol 12-myristate 13-acetate (PMA)-treated THP-1 cells, a human monocyte-macrophage cell line. hCG induced transient vacuole formation in PMA-treated THP-1 cells, morphologically mimicking Hofbauer cells. Immunocytochemistry showed that PMA-treated THP-1 cells incorporated hCG but not luteinizing hormone or follicle-stimulating hormone. Western blotting analyses demonstrated that PMA-treated THP-1 cells expressed an immunoreactive 60-kDa protein, designated as endogenous LH/CG-R Δ9. hCG induced a transient reduction in the LH/CG-R Δ9, which was synchronous with the appearance of cytoplasmic vacuoles. In conclusion, human macrophages regulating hCG via cytoplasmic LH/CG-R Δ9 mimic the morphological characteristics of Hofbauer cells. Their vacuoles may be associated with their cell-specific function to protect the fetus from exposure to excess maternal hCG during pregnancy.
Collapse
Affiliation(s)
- Munekage Yamaguchi
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | | | | | | | | |
Collapse
|