1
|
Zaleska M, Waclawik A, Bodek G, Zezula-Szpyra A, Li X, Janowski T, Hansel WH, Rahman NA, Ziecik AJ. Growth Repression in Diethylstilbestrol/Dimethylbenz[a]anthracene–Induced Rat Mammary Gland Tumor Using Hecate-CGβ Conjugate. Exp Biol Med (Maywood) 2016; 229:335-44. [PMID: 15044717 DOI: 10.1177/153537020422900408] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Recently, we have shown that Hecate-CGβ conjugate, which is a fusion of the lytic peptide Hecate and a 15–amino acid fragment of the β-chain of chorionic gonadotropin (CGβ), selectively destroys mammary gland carcinoma cells that possess luteinizing hormone receptors (LHR) in vitro. We induced mammary gland tumors using combined prenatal exposure to synthetic diethylstilbestrol (DES) and additional postnatal exposure to dimethylbenz[a]anthracene (DMBA). Rats with tumors were equally randomized (10/group) and treated with either sham (control) or 12 mg/kg body wt of either Hecate or Hecate-CGβ once a week for 3 weeks by tail vein injections. One week after the last injection, rats were kilted. Reverse-transcription–nested polymerase chain reaction/Southern blotting revealed alternatively spliced mRNA for LHR in tumor tissues of 5 of 30 females, which was further confirmed by Western blot analysis. The percentage of tumor volume increase was lowest in the group treated with Hecate-CGβ (45.3 ± 27.6), compared with Hecate- and shamtreated, control group (324.8 ± 78.1 and 309.9 ± 51.2, respectively; P < 0.001). Hecate-CGβ induced a significant decrease in tumor burden compared with controls (9.5 ± 2.1 mg/g body wt vs. 21.6 ± 2.9; P < 0.01). A smaller reduction in tumor burden was also observed in Hecate-treated females (17.6 ± 1.6 mg/g body wt vs. 21.6 ± 2.9; P < 0.05). Our results prove the principle that Hecate-CGβ conjugate is able to repress mammary gland tumor growth, even in tumor tissues that lack or have very low levels of LHR. The mechanism of Hecate-CGβ conjugate action in repression of DES/DMBA-induced tumor growth needs to be further analyzed to clarify the molecular mechanisms of Hecate-CGβ conjugate action in vivo.
Collapse
Affiliation(s)
- Monika Zaleska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Barros CM, Satrapa RA, Castilho ACS, Fontes PK, Razza EM, Ereno RL, Nogueira MFG. Effect of superstimulatory treatments on the expression of genes related to ovulatory capacity, oocyte competence and embryo development in cattle. Reprod Fertil Dev 2013; 25:17-25. [DOI: 10.1071/rd12271] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple ovulation (superovulation) and embryo transfer has been used extensively in cattle. In the past decade, superstimulatory treatment protocols that synchronise follicle growth and ovulation, allowing for improved donor management and fixed-time AI (FTAI), have been developed for zebu (Bos indicus) and European (Bos taurus) breeds of cattle. There is evidence that additional stimulus with LH (through the administration of exogenous LH or equine chorionic gonadotrophin (eCG)) on the last day of the superstimulatory treatment protocol, called the ‘P-36 protocol’ for FTAI, can increase embryo yield compared with conventional protocols that are based on the detection of oestrus. However, inconsistent results with the use of hormones that stimulate LH receptors (LHR) have prompted further studies on the roles of LH and its receptors in ovulatory capacity (acquisition of LHR in granulosa cells), oocyte competence and embryo quality in superstimulated cattle. Recent experiments have shown that superstimulation with FSH increases mRNA expression of LHR and angiotensin AT2 receptors in granulosa cells of follicles >8 mm in diameter. In addition, FSH decreases mRNA expression of growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) in oocytes, but increases the expression of both in cumulus cells, without diminishing the capacity of cumulus–oocyte complexes to generate blastocysts. Although these results indicate that superstimulation with FSH is not detrimental to oocyte competence, supplementary studies are warranted to investigate the effects of superstimulation on embryo quality and viability. In addition, experiments comparing the cellular and/or molecular effects of adding eCG to the P-36 treatment protocol are being conducted to elucidate the effects of superstimulatory protocols on the yield of viable embryos.
Collapse
|
3
|
George JW, Dille EA, Heckert LL. Current concepts of follicle-stimulating hormone receptor gene regulation. Biol Reprod 2011; 84:7-17. [PMID: 20739665 PMCID: PMC4480823 DOI: 10.1095/biolreprod.110.085043] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 05/04/2010] [Accepted: 08/16/2010] [Indexed: 12/25/2022] Open
Abstract
Follicle-stimulating hormone (FSH), a pituitary glycoprotein hormone, is an integral component of the endocrine axis that regulates gonadal function and fertility. To transmit its signal, FSH must bind to its receptor (FSHR) located on Sertoli cells of the testis and granulosa cells of the ovary. Thus, both the magnitude and the target of hormone response are controlled by mechanisms that determine FSHR levels and cell-specific expression, which are supported by transcription of its gene. The present review examines the status of FSHR/Fshr gene regulation, emphasizing the importance of distal sequences in FSHR/Fshr transcription, new insights gained from the influx of genomics data and bioinformatics, and emerging trends that offer direction in deciphering the FSHR/Fshr regulatory landscape.
Collapse
Affiliation(s)
- Jitu W. George
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Elizabeth A. Dille
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Leslie L. Heckert
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
|
5
|
Mittelholzer C, Andersson E, Taranger GL, Consten D, Hirai T, Senthilkumaran B, Nagahama Y, Norberg B. Molecular characterization and quantification of the gonadotropin receptors FSH-R and LH-R from Atlantic cod (Gadus morhua). Gen Comp Endocrinol 2009; 160:47-58. [PMID: 18992749 DOI: 10.1016/j.ygcen.2008.10.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 09/29/2008] [Accepted: 10/15/2008] [Indexed: 10/21/2022]
Abstract
In order to elucidate regulatory mechanisms during puberty final oocyte maturation and spawning, full-length sequences coding for the receptors for follicle-stimulating hormone (FSH-R) and luteinizing hormone (LH-R) were isolated from female Atlantic cod (Gadus morhua) by a RACE-PCR based strategy. The nucleotide and amino acid sequences showed high homologies with the corresponding sequences of other fish species but contained some distinct differences. Conserved features important for functionality, such as a long N-terminal extracellular domain (ECD), seven transmembrane domains and a short C-terminal intracellular domain, were identified in both predicted proteins. Partial genomic sequences for these genes were also determined, allowing the design of mRNA-specific quantitative PCR assays. Due to suspected alternative splicing during expression of these genes, additional real-time PCR assays detecting variants containing the membrane-anchoring domain were established. Besides the expected expression of FSH-R and LH-R mRNA in the gonads similarly strong signals for LH-R were also obtained in male gill, and in female and male brain. When relative expression was analysed at different stages of sexual maturation, levels for FSH-R increased moderately during gonadal growth whereas those of LH-R showed a high peak at spawning.
Collapse
Affiliation(s)
- C Mittelholzer
- Institute of Marine Research Austevoll, Storebø, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Ikeda S, Nakamura K, Kogure K, Omori Y, Yamashita S, Kubota K, Mizutani T, Miyamoto K, Minegishi T. Effect of estrogen on the expression of luteinizing hormone-human chorionic gonadotropin receptor messenger ribonucleic acid in cultured rat granulosa cells. Endocrinology 2008; 149:1524-33. [PMID: 18174289 DOI: 10.1210/en.2007-1163] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Estrogen has been considered to enhance FSH actions in the ovary, including the induction of the LH receptor (LHR). In this study, we elucidated the mechanism underlying the effect of estrogen on the induction of LHR by FSH in rat granulosa cells. Estradiol clearly enhanced the FSH-induced LHR mRNA increase in a time- and dose-dependent manner, with a maximum increase of approximately 3.5-fold at 72 h, compared with the level of LHR mRNA solely induced by FSH. We then investigated whether the effect of estrogen on LHR mRNA was due to increased transcription and/or altered mRNA stability. A luciferase assay with the plasmid containing the LHR 5'-flanking region did not show that estradiol increased the promoter activity induced by FSH. In contrast, the decay curves for LHR mRNA showed a significant increase in half-life with FSH and estradiol, suggesting that the increased stability of LHR mRNA is at least responsible for the regulation of LHR mRNA by estrogen. Recently mevalonate kinase (Mvk) was identified as a trans-factor that binds to LHR mRNA and alters LHR mRNA stability in the ovary. We found that estradiol, with FSH, decreased Mvk mRNA levels in rat granulosa cell culture, resulting in up-regulation of LHR mRNA that was inversely correlated to Mvk mRNA expression. Furthermore, the augmentation of FSH-induced LHR expression in the presence of estrogen was erased with the overexpression of Mvk by transient transfection. Taken together, these data indicate that LHR mRNA is up-regulated due to increased stability when estrogen negatively controls Mvk.
Collapse
Affiliation(s)
- Sadatomo Ikeda
- Department of Gynecology and Reproductive Medicine, Gunma University Graduate School of Medicine, Gunma, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ziecik AJ, Kaczmarek MM, Blitek A, Kowalczyk AE, Li X, Rahman NA. Novel biological and possible applicable roles of LH/hCG receptor. Mol Cell Endocrinol 2007; 269:51-60. [PMID: 17367919 DOI: 10.1016/j.mce.2006.08.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 08/01/2006] [Accepted: 08/01/2006] [Indexed: 11/17/2022]
Abstract
Luteinizing hormone/human chorionic gonadotropin (LH/hCG) receptors are widely expressed in gonadal cells, however, the presence of these receptors has also been demonstrated in several other non-gonadal female and male tissues. The expression level of non-gonadal LH/hCG receptors is much lower than in gonads, although their expression is regulated by similar mechanisms and they also exert biological effects using similar signaling pathways. Hormonally regulated LH/hCG receptor expression in the oviduct suggests that LH could be involved in the regulation of its contraction, gametes/embryos transport and synchronization of the fertilization. One of the major roles of the myometrial LH/hCG receptors may also be the stimulation of growth and maintenance of the uterine relaxation during pregnancy. In pigs, LH seems to be one of the pleiotropic factors which influence the endometrial prostaglandin F(2alpha) synthesis and initiation of the luteolysis. The LH/hCG receptor expression in several cancer cells provides new possibilities for developing new strategies for targeted cancer therapy based on lytic LH/hCG conjugates.
Collapse
Affiliation(s)
- Adam J Ziecik
- Division of Reproductive Endocrinology and Pathophysiology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-747 Olsztyn, Poland.
| | | | | | | | | | | |
Collapse
|
8
|
Menon KMJ, Nair AK, Wang L. A novel post-transcriptional mechanism of regulation of luteinizing hormone receptor expression by an RNA binding protein from the ovary. Mol Cell Endocrinol 2006; 246:135-41. [PMID: 16406262 DOI: 10.1016/j.mce.2005.11.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Luteinizing hormone/human chorionic gonadotropin (LH/hCG) receptor, a member of the rhodopsin/beta(2) adrenergic receptor subfamily of G-protein coupled receptors, is expressed primarily in the gonads and essential for the regulation of reproductive function. In the ovary, the expression of the receptor is post-transcriptionally regulated under physiological conditions. Studies from our laboratory showed that the ligand-induced down-regulation of the receptor occurs by accelerated degradation of the mRNA rather than by decreased transcription. We have identified a cytoplasmic LHR mRNA binding protein (LRBP) as a trans-acting factor in regulating LHR mRNA levels. LRBP binds to the coding region of LHR mRNA and causes accelerated degradation of mRNA. The RNA binding activity of LRBP was found to be inversely correlated to LH/hCG receptor mRNA levels. LRBP was purified to homogeneity and its identity was established as mevalonate kinase by N-terminal microsequencing and MALDI analysis. Mevalonate kinase, an enzyme involved in de novo synthesis of cholesterol, belongs to the GHMP family of kinases having a potential RNA binding fold. The expression of MVK mRNA and MVK protein levels were induced in response to hCG treatment prior to the down-regulation of LH/hCG receptor mRNA expression. A model for the post-transcriptional regulation of LH/hCG receptor in the ovary by mevalonate kinase is proposed.
Collapse
Affiliation(s)
- K M J Menon
- Department of Obstetrics/Gynecology, University of Michigan Medical Center, Ann Arbor, MI 48109 0617, USA.
| | | | | |
Collapse
|
9
|
Saxena D, Safi R, Little-Ihrig L, Zeleznik AJ. Liver receptor homolog-1 stimulates the progesterone biosynthetic pathway during follicle-stimulating hormone-induced granulosa cell differentiation. Endocrinology 2004; 145:3821-9. [PMID: 15117876 DOI: 10.1210/en.2004-0423] [Citation(s) in RCA: 44] [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: 11/19/2022]
Abstract
FSH-stimulated granulosa cell differentiation is associated with the induction of the LH receptor (LHr) as well as induction of the estrogen and progesterone biosynthetic pathways. Although activation of the cAMP-protein kinase A pathway is sufficient to stimulate progesterone production, additional pathways are required for the induction of the LHr and p450 aromatase. The orphan nuclear receptor, liver receptor homolog-1 (LRH-1), is expressed in granulosa cells and has been shown to synergize with the cAMP signaling system to regulate the gonadal type II aromatase promoter in transient transfection assays. To determine whether LRH-1 can interact with the cAMP pathway in the induction of aromatase and the LHr, we examined the effects of an adenoviral vector that directs the expression of human LRH-1 (Ad-LRH-1) on FSH-stimulated granulosa cell differentiation. Infection of undifferentiated granulosa cells with LRH-1 alone had no effect on estrogen production, progesterone production, or the expression of the LHr. However, combination of FSH stimulation and Ad-LRH-1 infection led to significantly greater progesterone production and increases in mRNA for p450 side-chain cleavage and 3beta-hydroxysteroid dehydrogenase than granulosa cells stimulated by FSH alone. However, infection with Ad-LRH-1 did not stimulate estradiol production or increases in mRNA for p450 aromatase or the LHr above that seen with FSH treatment alone. Moreover, infection with Ad-LRH-1 was able to overcome H-89 inhibition of FSH-stimulated progesterone but not estrogen production. Collectively, these observations support a direct role for LRH-1 in the induction of the progesterone but not the estrogen biosynthetic pathway during granulosa cell differentiation.
Collapse
Affiliation(s)
- Deeksha Saxena
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
10
|
Menon KMJ, Munshi UM, Clouser CL, Nair AK. Regulation of luteinizing hormone/human chorionic gonadotropin receptor expression: a perspective. Biol Reprod 2004; 70:861-6. [PMID: 14668203 DOI: 10.1095/biolreprod.103.024471] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The LH/hCG receptor, a member of the G protein coupled receptor family mediates the cellular actions of LH in the ovary. A considerable amount of information regarding its structure, mechanism of activation, and regulation of expression has emerged in recent years. Here we provide a brief overview of the current information on the structural organization of the receptor and the mechanism of receptor mediated signaling as well as an in-depth discussion on recent developments pertaining to the regulation of receptor expression. Specifically, we describe studies from our laboratory showing that the posttranscriptional regulation of the receptor involves an LH/hCG receptor mRNA-binding protein. We also propose a model to explain the loss of steady-state LH/hCG receptor mRNA levels during receptor down-regulation.
Collapse
Affiliation(s)
- K M J Menon
- Departments of Obstetrics and Gynecology and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.
| | | | | | | |
Collapse
|
11
|
Zhang Y, Dufau ML. Dual mechanisms of regulation of transcription of luteinizing hormone receptor gene by nuclear orphan receptors and histone deacetylase complexes. J Steroid Biochem Mol Biol 2003; 85:401-14. [PMID: 12943729 DOI: 10.1016/s0960-0760(03)00230-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The luteinizing hormone receptor (LHR), a member of the G protein-coupled, seven transmembrane receptor family, is essential for normal sexual development and reproductive function. LHR are expressed primarily in the gonads, but also are found in non-gonadal and cancer tissues. LH acts through LH receptors in Leydig cells to maintain general metabolic processes and steroidogenic enzymes, and in the ovary enhances follicular development and steroidogenesis in granulosa and luteal cells. The major transcriptional start sites of the LHR gene are located within the 176bp promoter domain. In the rat, the LHR gene is constitutively inhibited by upstream sequences (-176/-2056bp) in several cell systems, while in the human only a minor inhibitory effect was observed in JAR and HeLa cells (>20%). The TATA-less human promoter is driven by Sp1 and Sp3 transactivators that bind to two Sp1 domains at -79bp [Sp1(I)] and -119bp [Sp1(II)] (from ATG) with additive effects. An imperfect estrogen receptor half-site response element direct-repeat within the LHR promoter is an inhibitory locus. Endogenous orphan receptors, EAR2 and EAR3/COUP-TFI, bind this motif and repress promoter activity by 70%. TR4 also binds this motif and stimulates promoter activity (up to 2.5-fold). This is reversed by coexpression of EAR2 or EAR3/COUP-TFI through competitive binding to this site. Comparative studies of hDR and rDR orphan receptors binding and function revealed sequence-specific requirements. The A/C mismatch between hDR and rDR is responsible for the lack of TR4 binding and function in the rat. The G 3'-adjacent to the hDR core is important for EAR2/EAR3-COUP-TFI high-affinity binding. The Sp1-1 site is critical for EAR3/COUP-TFI repression, with minor participation for EAR2, and is not involved in the TR4 effect. Interaction of EAR3/COUP-TFI with Sp1 perturbs association of TFIIB with Sp1, independently of HDACs, and caused impairment of LHR transcription. Other aspect of control is through HDAC/mSin3A mechanism. Inhibition of HDACs by TSA increases LHR promoter activity in JAR cells (40-fold), association of acetylated H3/H4 with the LHR promoter, recruitment of Pol II to the promoter, and LHR mRNA levels. A multiprotein complex is recruited to the hLHR promoter via interaction with Sp1/Sp3: HDACs dock directly to Sp1-1 bound DNA and indirectly to Sp3-1 bound DNA through RbAp48, while mSin3A interacts HDACs and potentiates HDAC1-mediated repression. Our studies have demonstrated that orphan receptor-ERE complexes, and the HDAC1-HDAC2-mSin3A complex have important roles in the regulation of LHR gene transcription by interaction with Sp1/Sp3, and by region-specific changes in histone acetylation and Pol II recruitment within the LHR promoter.
Collapse
Affiliation(s)
- Ying Zhang
- Molecular Endocrinology Section, Endocrinology and Reproduction Research Branch, National Institute of Child Health & Human Development/NIH, 49 Convent Drive, Building 49, Room 6A-36, Bethesda, MD 20892-4510, USA
| | | |
Collapse
|
12
|
Hämäläinen T, Kero J, Poutanen M, Huhtaniemi I. Transgenic mice harboring murine luteinizing hormone receptor promoter/beta-galactosidase fusion genes: different structural and hormonal requirements of expression in the testis, ovary, and adrenal gland. Endocrinology 2002; 143:4096-103. [PMID: 12239121 DOI: 10.1210/en.2002-220162] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In vivo regulation of the LH receptor (LHR) promoter was studied using transgenic (TG) mice harboring fusion genes containing three different lengths of the LHR promoter (7.4 kb, 2.1 kb, and 173 bp), fused with coding sequence of the Escherichia coli beta-galactosidase (beta-GAL) reporter gene. The length of the LHR promoter significantly affected the pattern of beta-GAL expression. In the testis the shortest promoter directed expression primarily of the full-length beta-GAL mRNA, but mainly truncated messages were transcribed from the longer LHR promoter/beta-GAL constructs. The case was reversed in the ovary and adrenal gland. Furthermore, we have recently detected strong LHR expression in the adrenal gland of female mice with chronically elevated serum LH. Therefore, the regulation of the adrenal LHR expression was addressed in the present study using the LHR/beta-GAL TG mice. Elevated LH levels were achieved in the LHR/beta-GAL mice either by gonadectomy or cross-breeding them with TG mice overexpressing a chimeric protein of bovine LH beta-subunit and the C-terminal fragment of human chorionic gonadotropin-beta. In both models, beta-GAL mRNA was found in the adrenal cortex when the 7.4-kb LHR promoter was applied but not in mice carrying the 173-bp LHR promoter. The 7.4-kb construct was activated also in the ovaries in the double TG LHR(beta-GAL)/bovine LH beta-subunit/C-terminal fragment of human chorionic gonadotropin-betamice in some theca-interstitial cells surrounding the follicles. Hence, the LHR promoter elements essential for directing beta-GAL expression to the adrenal gland and ovary (7.4 kb) are different from those recently shown to be essential for the testicular expression (173 bp). In conclusion, elevated serum LH concentrations were found seminal for the LHR promoter activation in the ovaries and adrenals, and different lengths of the promoter are responsible for reporter gene expression in the testis, ovary, and adrenal gland.
Collapse
|
13
|
Yoshino M, Mizutani T, Yamada K, Tsuchiya M, Minegishi T, Yazawa T, Kawata H, Sekiguchi T, Kajitani T, Miyamoto K. Early growth response gene-1 regulates the expression of the rat luteinizing hormone receptor gene. Biol Reprod 2002; 66:1813-9. [PMID: 12021067 DOI: 10.1095/biolreprod66.6.1813] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
LH receptor gene expression is primarily regulated via specific interactions of trans-acting proteins and cis-acting DNA sequences in the upstream region of the gene. In this study, we report, using luciferase assays, that the region between -171 and -137 base pairs (bp) is essential for basal expression of the rat LH receptor gene. To identify factors that interact with the region between -171 and -137 bp and regulate expression of the gene, a rat granulosa cell cDNA library was screened using a yeast one-hybrid system. A positive clone, isolated by the screening, encodes a transcription factor early growth response gene-1 (Egr-1). To determine the sequence to which Egr-1 protein binds, electrophoretic mobility shift assay (EMSA) was employed. The Egr-1 protein was produced by an in vitro transcription/translation system using a full-length rat Egr-1 cDNA. The upstream region between -171 and -137 bp contains 2 overlapping Egr-1 consensus sequences. The EMSA revealed that Egr-1 binds independently to both sites. The overexpression of Egr-1 in MA-10 cells caused an approximately 2-fold increase in reporter luciferase activity. However, no induction of the luciferase activity was observed when luciferase constructs that lacked or had mutations in either or both of the Egr-1 sites were used, indicating that Egr-1 positively regulates LH receptor gene expression. In differentiated granulosa cells that had been pretreated with FSH for 48 h, the levels of both mRNA and Egr-1 protein were induced by hCG or cAMP, reaching maximal levels approximately 1.5 h after treatment and then returning to basal levels 8 h thereafter. No Egr-1 mRNA or protein was detected in undifferentiated granulosa cells, even after stimulation with 8-bromoadenosine-cAMP. These results suggest that Egr-1 functions only in luteinized granulosa cells after stimulation with hCG or cAMP. In conclusion, the findings demonstrate that Egr-1 actually binds to the regulatory upstream region of the LH receptor gene and positively regulates receptor gene expression. In addition, Egr-1 expression was observed only in luteinized granulosa cells after stimulation with hCG or cAMP. The present study provides further support to the hypothesis that Egr-1 plays important roles in the pituitary-gonadal axis.
Collapse
Affiliation(s)
- Miki Yoshino
- Department of Biochemistry, Fukui Medical University, Shimoaizuki, Matsuoka, Fukui 910-, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
Reproduction cannot take place without the proper functioning of the lutropin/choriogonadotropin receptor (LHR). When the LHR does not work properly, ovulation does not occur in females and Leydig cells do not develop normally in the male. Also, because the LHR is essential for sustaining the elevated levels of progesterone needed to maintain pregnancy during the first trimester, disruptions in the functions of the LHR during pregnancy have catastrophic consequences. As such, a full understanding of the biology of the LHR is essential to the survival of our species. In this review we summarize our current knowledge of the structure, functions, and regulation of this important receptor.
Collapse
Affiliation(s)
- Mario Ascoli
- Department of Pharmacology, The University of Iowa, Iowa City, 52242-1109, USA.
| | | | | |
Collapse
|
15
|
Yoshida M, Yoshida S, Sugawara I, Takeda K. Maternal Exposure to Diesel Exhaust Decreases Expression of Steroidogenic Factor-1 and Muellerian Inhibiting Substance in the Murine Fetus. ACTA ACUST UNITED AC 2002. [DOI: 10.1248/jhs.48.317] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Miki Yoshida
- Department of Hygiene Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Seiichi Yoshida
- Department of Hygiene Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science
- CREST, JST (Japan Science and Technology Corporation)
| | - Isamu Sugawara
- Department of Molecular Pathology, The Research Institute of Tuberculosis
- CREST, JST (Japan Science and Technology Corporation)
| | - Ken Takeda
- Department of Hygiene Chemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science
- CREST, JST (Japan Science and Technology Corporation)
| |
Collapse
|
16
|
Otsuka F, Moore RK, Shimasaki S. Biological function and cellular mechanism of bone morphogenetic protein-6 in the ovary. J Biol Chem 2001; 276:32889-95. [PMID: 11447221 DOI: 10.1074/jbc.m103212200] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The process of ovarian folliculogenesis is composed of proliferation and differentiation of the constitutive cells in developing follicles. Growth factors emitted by oocytes integrate and promote this process. Growth differentiation factor-9 (GDF-9), bone morphogenetic protein (BMP)-15, and BMP-6 are oocyte-derived members of the transforming growth factor-beta superfamily. In contrast to the recent studies on GDF-9 and BMP-15, nothing is known about the biological function of BMP-6 in the ovary. Here we show that, unlike BMP-15 and GDF-9, BMP-6 lacks mitogenic activity on rat granulosa cells (GCs) and produces a marked decrease in follicle-stimulating hormone (FSH)-induced progesterone (P(4)) but not estradiol (E(2)) production, demonstrating not only the first identification of GCs as BMP-6 targets in the ovary but also its selective modulation of FSH action in steroidogenesis. This BMP-6 activity resembles BMP-15 but differs from GDF-9 activities. BMP-6 also exhibited similar action to BMP-15 by attenuating the steady state mRNA levels of FSH-induced steroidogenic acute regulatory protein (StAR) and P450 side-chain cleavage enzyme (P450scc), without affecting P450 aromatase mRNA level, supporting its differential function on FSH-regulated P(4) and E(2) production. However, unlike BMP-15, BMP-6 inhibited forskolin- but not 8-bromo-cAMP-induced P(4) production and StAR and P450scc mRNA expression. BMP-6 also decreased FSH- and forskolin-stimulated cAMP production, suggesting that the underlying mechanism by which BMP-6 inhibits FSH action most likely involves the down-regulation of adenylate cyclase activity. This is clearly distinct from the mechanism of BMP-15 action, which causes the suppression of basal FSH receptor (FSH-R) expression, without affecting adenylate cyclase activity. As assumed, BMP-6 did not alter basal FSH-R mRNA levels, whereas it inhibited FSH- and forskolin- but not 8-bromo-cAMP-induced FSH-R mRNA accumulation. These studies provide the first insight into the biological function of BMP-6 in the ovary and demonstrate its unique mechanism of regulating FSH action.
Collapse
Affiliation(s)
- F Otsuka
- Department of Reproductive Medicine, University of California San Diego, School of Medicine, La Jolla, California 92093-0633, USA
| | | | | |
Collapse
|
17
|
Otsuka F, Yamamoto S, Erickson GF, Shimasaki S. Bone morphogenetic protein-15 inhibits follicle-stimulating hormone (FSH) action by suppressing FSH receptor expression. J Biol Chem 2001; 276:11387-92. [PMID: 11154695 DOI: 10.1074/jbc.m010043200] [Citation(s) in RCA: 211] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have recently reported that oocyte-derived bone morphogenetic protein-15 (BMP-15) can directly modulate follicle-stimulating hormone (FSH) action in rat granulosa cells. Here, we investigate underlying mechanisms of this BMP-15 effect. Treatment with BMP-15 alone exerted no significant effect on the basal expression of mRNAs encoding steroidogenic acute regulatory protein, P450 side chain cleavage enzyme, P450 aromatase, 3beta-hydroxysteroid dehydrogenase, luteinization hormone receptor, and inhibin/activin subunits. However, BMP-15 markedly inhibited the FSH-induced increases in these messages. In striking contrast, BMP-15 did not change the forskolin-induced levels of these transcripts. Thus, the inhibitory effect of BMP-15 on FSH action must be upstream of cAMP signaling. We next examined changes in FSH receptor mRNA expression. Interestingly, BMP-15 severely reduced the levels of FSH receptor mRNA in both basal and FSH-stimulated cells. To determine whether this effect was at the level of FSH function, we investigated the effect of BMP-15 on FSH bioactivity. Consistent with the mRNA data, BMP-15 inhibited the biological response of FSH, but not that of forskolin. Based on these results, we propose that BMP-15 is an important determinant of FSH action through its ability to inhibit FSH receptor expression. Because FSH plays an essential role in follicle growth and development, our findings could have new implications for understanding how oocyte growth factors contribute to folliculogenesis.
Collapse
Affiliation(s)
- F Otsuka
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California 92093-0633, USA
| | | | | | | |
Collapse
|
18
|
Zeng H, Phang T, Song YS, Ji I, Ji TH. The role of the hinge region of the luteinizing hormone receptor in hormone interaction and signal generation. J Biol Chem 2001; 276:3451-8. [PMID: 10964934 DOI: 10.1074/jbc.m007488200] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Luteinizing hormone receptor, a G protein-coupled receptor, consists of two halves, the N-terminal extracellular hormone binding domain (exodomain) and the C-terminal membrane-associated, signal-generating domain (endodomain). The exodomain has seven to nine Leu-rich repeats, which are generally thought to form a 1/3 donut-like structure and interact with human choriogonadotropin (hCG). The resulting hCG-exodomain complex adjusts the structure and its association with the endodomain, which results in signal generation in the endodomain. It is unclear whether the rigid 1/3 donut structure could provide the agility and versatility of this dynamic action. In addition, there is no clue as to where the endodomain contact point (the signal modulator) in the exodomain is. To address these issues, the exodomain was examined by Ala scan and multiple substitutions, while receptor peptides were used for photoaffinity labeling and affinity cross-linking. Our results show that the C-flanking sequence (hinge region), Thr(250)-Gln(268), of the Leu-rich repeats (LRRs) specifically interacts with hCG, preferentially hCGalpha. This interaction is inhibited by exoloop 2 of the endodomain but not by exoloops 1 and 3, suggesting an intimate relationship between Thr(250)-Gln(268), exoloop 2, and hCG. Taken together, our observations in this article suggest a new paradigm that the LRRs contact the front of hCG, while both flanking regions of the LRRs interact with the sides of hCG. This would trap hCG in the 1/3 donut structure of the LRRs and enhance the binding affinity. In addition, mutations of conserved Ser(255) in the sequence can constitutively activate the receptor. This provides a clue for the signal modulator in the exodomain. In contrast, a phenyl or phenolic group is necessary at conserved Tyr(253) for targeting the receptor to the surface.
Collapse
Affiliation(s)
- H Zeng
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, USA
| | | | | | | | | |
Collapse
|
19
|
Bao B, Kumar N, Karp RM, Garverick HA, Sundaram K. Estrogen receptor-beta expression in relation to the expression of luteinizing hormone receptor and cytochrome P450 enzymes in rat ovarian follicles. Biol Reprod 2000; 63:1747-55. [PMID: 11090445 DOI: 10.1095/biolreprod63.6.1747] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Changes in mRNA expression for estrogen receptor (ER beta) in relation to mRNAs for LH receptor (LHr) and cytochrome P450 enzymes were examined in granulosa and theca cells from proestrous rat ovarian follicles. Of the 30 ovaries harvested from 15 adult rats, 24 were processed for in situ hybridization, and the remaining were used for reverse transcription-polymerase chain reaction. Messenger RNAs for ER beta, LHr, cytochrome P450 side-chain cleavage enzyme (P450(scc)), 17 alpha-hydroxylase (P450(c17)), aromatase (P450(arom)), and steroidogenic acute regulatory protein (StAR) were localized in cross sections of ovaries by in situ hybridization and quantified in granulosa and theca cell layers by a computer-image analyzing system. Ovarian follicles were classified as healthy or atretic. Healthy follicles were divided into four size groups: very small (40-100 microm), small (101-275 microm), medium (276-450 microm), and large (451-850 microm). Atretic follicles were divided into medium (276-450 microm) or large follicles (451-850 microm). A low level of ER beta mRNA expression was first detected in granulosa cells of very small healthy follicles, and the expression increased progressively up to medium-sized follicles. The expression of ER beta mRNA was highest (P < 0.01) in medium-sized follicles that was followed by a decrease (P < 0.01) in large follicles. Messenger RNAs for LHr, P450(scc), and P450(arom) were first detected in granulosa cells of medium-sized healthy follicles, while mRNAs for LHr, P450(scc), P450(c17), and StAR were first detected in theca cells associated with very small follicles. The highest expression of LHr, P450(scc), P450(c17), P450(arom), and StAR was seen in granulosa and/or theca cells of large healthy follicles. In atretic follicles, level of gene expression was relatively low in both granulosa and theca cells. In conclusion, stage-specific expression of ER beta mRNA was observed in granulosa cells during follicular development. The increased expression of ER beta and a concomitant initiation of LHr, P450(scc), and P450(arom) expression in granulosa cells of medium follicles may signify a role for estrogen in follicular development. Also, a strong correlation between ER beta mRNA expression in granulosa cells, and the expression of mRNAs for LHr, P450(scc), P450(c17), and StAR in theca cells associated with growing follicles suggests a possible role for estrogen in steroidogenesis.
Collapse
Affiliation(s)
- B Bao
- Center for Biomedical Research, The Population Council, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|
20
|
Eriksen KK, Hauser F, Schiøtt M, Pedersen KM, Søndergaard L, Grimmelikhuijzen CJ. Molecular cloning, genomic organization, developmental regulation, and a knock-out mutant of a novel leu-rich repeats-containing G protein-coupled receptor (DLGR-2) from Drosophila melanogaster. Genome Res 2000; 10:924-38. [PMID: 10899142 PMCID: PMC310913 DOI: 10.1101/gr.10.7.924] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
After screening the Berkeley Drosophila Genome Project database with sequences from a recently characterized Leu-rich repeats-containing G protein-coupled receptor (LGR) from Drosophila (DLGR-1), we identified a second gene for a different LGR (DLGR-2) and cloned its cDNA. DLGR-2 is 1360 amino acid residues long and shows a striking structural homology with members of the glycoprotein hormone [thyroid-stimulating hormone (TSH); follicle-stimulating hormone (FSH); luteinizing hormone/choriogonadotropin (LH/CG)] receptor family from mammals and with two additional, recently identified mammalian orphan LGRs (LGR-4 and LGR-5). This homology includes the seven transmembrane region (e.g., 49% amino acid identity with the human TSH receptor) and the very large extracellular amino terminus. This amino terminus contains 18 Leu-rich repeats-in contrast with the 3 mammalian glycoprotein hormone receptors and DLGR-1 that contain 9 Leu-rich repeats, but resembling the mammalian LGR-4 and LGR-5 that each have 17 Leu-rich repeats in their amino termini. The DLGR-2 gene is >18.6 kb pairs long and contains 15 exons and 14 introns. Four intron positions coincide with the intron positions of the three mammalian glycoprotein hormone receptors and have the same intron phasing, showing that DLGR-2 is evolutionarily related to these mammalian receptors. The DLGR-2 gene is located at position 34E-F on the left arm of the second chromosome and is expressed in embryos and pupae but not in larvae and adult flies. Homozygous knock-out mutants, where the DLGR-2 gene is interrupted by a P element insertion, die around the time of hatching. This finding, together with the expression data, strongly suggests that DLGR-2 is exclusively involved in development.
Collapse
Affiliation(s)
- K K Eriksen
- Department of Cell Biology, Zoological Institute, Institute of Molecular Biology, University of Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
21
|
You S, Kim H, Hsu CC, El Halawani ME, Foster DN. Three different turkey luteinizing hormone receptor (tLH-R) isoforms I: characterization of alternatively spliced tLH-R isoforms and their regulated expression in diverse tissues. Biol Reprod 2000; 62:108-16. [PMID: 10611074 DOI: 10.1095/biolreprod62.1.108] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Using combinations of reverse transcription-polymerase chain reaction (RT-PCR) and 5'- and 3'-rapid amplification of cDNA ends, three different, alternatively spliced, partial turkey LH receptor (tLH-R) cDNA isoforms were characterized from ovarian mRNA. The first cDNA (tLH-R(intact)) showed 98% and 72-75% similarity with chicken and mammalian LH-R sequences, respectively. The second cloned cDNA isoform (tLH-R(insert)) contained an in-frame TGA stop codon within an 86-base pair insertion that was located in the extracellular domain of the seven-transmembrane region. The tLH-R(insert) isoform could encode a truncated soluble protein isoform that lacked the transmembrane region. The third cDNA isoform truncated the transmembrane region (tLH-R(trunc)) and was derived by the deletion of the last exon by incomplete splicing. Generation of multiple transcripts by alternative splicing was elucidated by partial characterization of tLH-R genomic sequences. The differentially regulated expression of the tLH-R mRNA isoforms in nongonadal tissues and ovarian stromal tissues during various reproductive stages was quantified and analyzed by Northern blot and/or RT-PCR. Alternatively spliced tLH-R isoforms were differentially expressed in a tissue-specific manner in most of the tissues examined. The steady-state levels of tLH-R mRNA isoforms were relatively high in the hypothalamus and optic nerve and relatively low in the cortex, pituitary, and cerebellum when compared to levels in ovarian follicles. In nongonadal reproductive tissues, the steady-state levels of tLH-R mRNA isoforms were relatively high in the uterus and infundibulum and relatively low in the isthmus, oviduct, and magnum. In addition, in the nongonadal peripheral tissues, the steady-state levels of tLH-R isoforms were relatively high in the thyroid gland and relatively low in the spleen, adrenal gland, kidney, skin, bursa, and muscle. The present study suggests that the alternative splicing of LH-R transcripts occurs in a tissue-specific manner and has been evolutionarily conserved (similar results were obtained in chicken and swine). These results raise fundamental questions as to the function of LH-R isoforms in nongonadal tissues.
Collapse
Affiliation(s)
- S You
- Division of Animal Physiology, Department of Animal Science, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | | | | | |
Collapse
|
22
|
Hämäläinen T, Poutanen M, Huhtaniemi I. Age- and sex-specific promoter function of a 2-kilobase 5'-flanking sequence of the murine luteinizing hormone receptor gene in transgenic mice. Endocrinology 1999; 140:5322-9. [PMID: 10537163 DOI: 10.1210/endo.140.11.7115] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A transgenic (TG) mouse model carrying a 2-kb murine LH receptor (LHR) promoter/beta-galactosidase (beta-GAL) fusion gene was created to study the regulatory function of the 5'-flanking region of the murine LHR gene. Of the five TG mouse lines produced, three displayed high beta-GAL expression in the testis, but none showed any expression in the ovary. In addition, all mouse lines of both sexes expressed beta-GAL consistently in the brain, most prominently in hippocampus, hypothalamus, midbrain, and cortex. Weak staining was found in a few pituitary samples. All other tissues examined were negative for transgene expression. In support of sex-specific gonadal expression of the transgene, transient transfection of the LHR/beta-GAL gene construct into immortalized mouse granulosa (KK-1) and Leydig (mLTC-1) tumor cells revealed a more than 5-fold higher expression level in the Leydig cells. Histological examination of the TG testes demonstrated strong beta-GAL expression in Leydig cells, but, unexpectedly, also in elongating spermatids of adult age and in some spermatogonia of the neonatal testis. The functional significance of the latter findings remains open. The transgene was only expressed in adult Leydig cells; no expression was found in the fetal population of these cells. Hence, these findings indicate that the immediate 2-kb fragment of the murine LHR 5'-flanking sequence is transcriptionally active only in adult Leydig cells and certain brain areas, but other promoter sequences are apparently needed for ovarian and fetal testicular expression of the LHR gene.
Collapse
Affiliation(s)
- T Hämäläinen
- Department of Physiology, University of Turku, Finland
| | | | | |
Collapse
|
23
|
Geng Y, Tsai-Morris CH, Zhang Y, Dufau ML. The human luteinizing hormone receptor gene promoter: activation by Sp1 and Sp3 and inhibitory regulation. Biochem Biophys Res Commun 1999; 263:366-71. [PMID: 10491299 DOI: 10.1006/bbrc.1999.1374] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To understand the transcriptional mechanism(s) of human LH receptor (LHR) gene expression, we have identified the dominant functional cis-elements that regulate the activity of the promoter domain (-1 to -176 bp from ATG). Mutagenesis demonstrated that the promoter activity was dependent on two Sp1 domains (-79 bp, -120 bp) in a transformed normal placental cell (PLC) and the choriocarcinoma JAR cell. Both elements interacted with endogenous Sp1 and Sp3 factors but not with Sp2 or Sp4. In Drosophila SL2 cells, the promoter was activated by either Sp1 or Sp3. An ERE half-site (EREhs) at -174 bp was inhibitory (by 100%), but was unresponsive to estradiol and did not bind the estrogen receptor or orphan receptors ERR1 and SF-1. The 5' upstream sequence (-177 to -2056 bp) inhibited promoter activity in PLC by 60%, but only minimally in JAR cells. Activation of the human LHR promoter through Sp1/3 factors is negatively regulated through EREhs and upstream sequences to exert control of gene expression.
Collapse
Affiliation(s)
- Y Geng
- Endocrinology and Reproduction Research Branch, National Institutes of Child Health and Human Development, Bethesda, Maryland, 20892, USA
| | | | | | | |
Collapse
|
24
|
Ryu K, Gilchrist R, Koo Y, Ji I, Ji T. Gene, interaction, signal generation, signal divergence and signal transduction of the LH/CG receptor. Int J Gynaecol Obstet 1999; 60 Suppl 1:S9-S20. [DOI: 10.1016/s0020-7292(98)80001-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
25
|
Hondo E, Kobayashi T, Ishiguro N, Kurohmaru M, Kitamura N, Yamada J, Nagahama Y. Prolactin Induces Protamine 2 mRNA Expression in Rat Testis. J Reprod Dev 1999. [DOI: 10.1262/jrd.45.205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Eiichi Hondo
- Department of Veterinary Anatomy, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Tohru Kobayashi
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki 444-8585, Japan
| | - Naotaka Ishiguro
- Department of Veterinary Public Health, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Masamichi Kurohmaru
- Department of Veterinary Anatomy, Graduate School of Agricultural and Life Sciences, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Nobuo Kitamura
- Department of Veterinary Anatomy, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Junzo Yamada
- Department of Veterinary Anatomy, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Yoshitaka Nagahama
- Laboratory of Reproductive Biology, National Institute for Basic Biology, Okazaki 444-8585, Japan
| |
Collapse
|
26
|
Hsu SY, Liang SG, Hsueh AJ. Characterization of two LGR genes homologous to gonadotropin and thyrotropin receptors with extracellular leucine-rich repeats and a G protein-coupled, seven-transmembrane region. Mol Endocrinol 1998; 12:1830-45. [PMID: 9849958 DOI: 10.1210/mend.12.12.0211] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The receptors for LH, FSH, and TSH belong to the large G protein-coupled, seven-transmembrane (TM) protein family and are unique in having a large N-terminal extracellular (ecto-) domain containing leucine-rich repeats important for interaction with the glycoprotein ligands. We have identified two new leucine-rich repeat-containing, G protein-coupled receptors and named them as LGR4 and LGR5, respectively. The ectodomains of both receptors contain 17 leucine-rich repeats together with N- and C-terminal flanking cysteine-rich sequences, compared with 9 repeats found in known glycoprotein hormone receptors. The leucine-rich repeats in LGR4 and LGR5 are arrays of 24 amino acids showing similarity to repeats found in the acid labile subunit of the insulin-like growth factor (IGF)/IGF binding protein complexes as well as slit, decorin, and Toll proteins. The TM region and the junction between ectodomain and TM 1 are highly conserved in LGR4, LGR5, and seven other LGRs from sea anemone, fly, nematode, mollusk, and mammal, suggesting their common evolutionary origin. In contrast to the restricted tissue expression of gonadotropin and TSH receptors in gonads and thyroid, respectively, LGR4 is expressed in diverse tissues including ovary, testis, adrenal, placenta, thymus, spinal cord, and thyroid, whereas LGR5 is found in muscle, placenta, spinal cord, and brain. Hybridization analysis of genomic DNA indicated that LGR4 and LGR5 genes are conserved in mammals. Comparison of overall amino acid sequences indicated that LGR4 and LGR5 are closely related to each other but diverge, during evolution, from the homologous receptor found in snail and the mammalian glycoprotein hormone receptors. The identification and characterization of new members of the LGR subfamily of receptor genes not only allow future isolation of their ligands and understanding of their physiological roles but also reveal the evolutionary relationship of G protein-coupled receptors with leucine-rich repeats.
Collapse
Affiliation(s)
- S Y Hsu
- Department of Gynecology and Obstetrics, Stanford University Medical School, California 94305-5317, USA
| | | | | |
Collapse
|
27
|
Vibede N, Hauser F, Williamson M, Grimmelikhuijzen CJ. Genomic organization of a receptor from sea anemones, structurally and evolutionarily related to glycoprotein hormone receptors from mammals. Biochem Biophys Res Commun 1998; 252:497-501. [PMID: 9826559 DOI: 10.1006/bbrc.1998.9661] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cnidarians (e.g., sea anemones and corals) are the lowest animal group having a nervous system. Previously, we cloned a receptor from sea anemones that showed a strong structural similarity to the glycoprotein hormone (TSH, FSH, LH/CG) receptors from mammals. Here, we determine the genomic organization of this sea anemone receptor. The receptor gene contains eight introns that are all localized within a region coding for the large extracellular N terminus. These introns occur at the same positions and have the same intron phasing as eight introns in the genes coding for the mammalian glycoprotein hormone receptors, indicating that the cnidarian and mammalian receptor genes are evolutionarily related. As with the mammalian receptor genes, the sea anemone receptor gene does not contain introns in the region coding for the transmembrane and intracellular domains. Southern blot analyses show that the cnidarian receptor is coded for by a single gene.
Collapse
Affiliation(s)
- N Vibede
- Department of Cell Biology and Anatomy, Zoological Institute, University of Copenhagen, Universitetsparken 15, Copenhagen O, DK-2100, Denmark
| | | | | | | |
Collapse
|
28
|
Zhang FP, El-Hefnawy T, Huhtaniemi I, El-Hafnawy T. Regulation of luteinizing hormone receptor gene expression by insulin-like growth factor-I in an immortalized murine Leydig tumor cell line (BLT-1). Biol Reprod 1998; 59:1116-23. [PMID: 9780317 DOI: 10.1093/biolreprod/59.5.1116] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It is postulated that insulin-like growth factor-I (IGF-I), a 70-amino acid mitogenic polypeptide, regulates Leydig cell steroidogenesis. In the present study, we assessed the effect of IGF-I on LH receptor (LHR) gene expression in an immortalized murine Leydig tumor cell line (BLT-1). Culture of BLT-1 cells in the presence of IGF-I (0.1-100 ng/ml) for 24 or 48 h increased their [125I]iodo-hCG binding in a dose-dependent manner up to 275% of the control level. Northern hybridization analysis revealed four major transcripts of LHR mRNA in BLT-1 cells (6.9, 2.6, 1.7, and 1.2 kilobases), and treatment at 10-100 ng/ml of IGF-I increased steady-state levels of LHR mRNAs in coordinate fashion up to 2. 2-fold. IGF-I (30 ng/ml) induced a time-dependent increase in [125I]hCG binding after a lag period of 2-6 h when studied up to 48 h, with a subsequent decrease. A similar response with steady increase up to 72 h was observed in total LHR mRNA. To elucidate the molecular mechanism of IGF-I action on LHR mRNA expression, we measured the transcription rate of the LHR gene by nuclear run-off assay and assessed transcript stability by the actinomycin D blocking method. The results showed that IGF-I treatment had no effect on the transcription rate of the LHR gene, whereas the half-life (t1/2) of LHR mRNA was significantly prolonged (IGF-I-treated cells, 30 +/- 3.8 h; controls, 17 +/- 2.5 h). Furthermore, IGF-I at 30 ng/ml and 100 ng/ml increased the expression of LHR promoter-driven luciferase and cytomegalovirus-promoter driven ss-galactosidase activities in BLT-1 cells; however, the former increased only marginally more than the latter. This suggests that the increase of LHR mRNA by IGF-I in Leydig cells is mainly due to increased mRNA stability.
Collapse
Affiliation(s)
- F P Zhang
- Department of Physiology, University of Turku, FIN-20520 Turku, Finland
| | | | | | | |
Collapse
|
29
|
Thompson DA, Othman MI, Lei Z, Li X, Huang ZH, Eadie DM, Rao CV. Localization of receptors for luteinizing hormone/chorionic gonadotropin in neural retina. Life Sci 1998; 63:1057-64. [PMID: 9749828 DOI: 10.1016/s0024-3205(98)00367-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although the expression of the luteinizing hormone (LH)/human chorionic gonadotropin (CG) receptor gene has been traditionally thought to be restricted to gonadal tissue, recent studies have shown that LH/CG receptors are present in many regions of the central nervous system (CNS), as well as in peripheral tissues. We now report the characterization of LH/CG receptor gene expression in the neural retina, a component of the CNS. Transcript levels in the retina are approximately equal to levels present in the cerebral cortex, but are at least 100 fold lower than the levels in testis. The density of LH/CG receptor transcripts, receptor protein and 125I-CG binding is the highest in the photoreceptor cells and then decreased throughout the inner retina. Our study is the first to demonstrate the presence of LH/CG receptors in the neural retina. This finding raises the possibility that photoreceptor cells have the potential to mount cellular responses to LH/CG that may impact on visual processing, and poses an intriguing connection to the proposed role of gonadotropins in the progression of proliferative retinopathy.
Collapse
Affiliation(s)
- D A Thompson
- Department of Ophthalmology, University of Michigan Medical School, Ann Arbor 48105, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhang FP, Kero J, Huhtaniemi I. The unique exon 10 of the human luteinizing hormone receptor is necessary for expression of the receptor protein at the plasma membrane in the human luteinizing hormone receptor, but deleterious when inserted into the human follicle-stimulating hormone receptor. Mol Cell Endocrinol 1998; 142:165-74. [PMID: 9783912 DOI: 10.1016/s0303-7207(98)00108-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The LH receptor (LHR) is a member of the family of G protein-coupled seven-times plasma membrane transversing receptors. Its gene consists of 11 exons, the last one encoding the transmembrane and intracellular domains of the receptor. The FSHR, and its gene, resemble structurally those of the LHR, with the exception that the sequences corresponding to exon 10 in LHR are missing in FSHR, which is thus encoded by a total of ten exons. Our recent studies on the marmoset monkey testis LHR cDNA indicated that an 81 bp nucleotide sequence, encoding the complete exon 10 of the LHR gene in other mammalian species, is absent in this species without affecting the LHR function. To study further the role of the exon 10 encoded sequences of the LHR in the gonadotropin receptor function, a deletion of exon 10 from the human LHR (hLHdeltaexon10R), and a chimeric hFSHR with exon 10 from hLHR inserted (hFSHLHexon10R), were constructed in expression vectors. The results presented here demonstrate that 293 cells transfected with the hLHdeltaexon10R display a decrease in the proportion of the receptor binding at the cell surface, compared with cells transfected with wild-type hLHR. However, the cells expressing hLHdeltaexon10R showed similar high affinity binding of [125I]iodo-hCG as those transfected with wild-type hLHR, in either intact cells or their detergent extracts. In addition, cells expressing the hLHdeltaexon10R and wild-type hLHR displayed similar dose-response of cAMP production to hCG stimulation. Cells transfected with chimeric hFSHLHexon10R showed barely detectable [125I]iodo-FSH binding in intact cells compared with those transfected with wild-type hFSHR. The FSH binding detected in cellular detergent extracts displayed 10-fold lower binding activity than wild-type receptors, in spite of similar level of immunoreactive FSHR protein expression in the transfected cells. The hFSHLHexon10R had a modest 5-fold lower binding affinity for FSH as compared with wild-type hFSHR. In conclusion, the present study indicates that the sequences encoding exon 10 of the hLHR are essential for the LHR expression at the plasma membrane, but deleterious for function if inserted into the hFSHR.
Collapse
Affiliation(s)
- F P Zhang
- Department of Physiology, University of Turku, Finland
| | | | | |
Collapse
|
31
|
Hong S, Phang T, Ji I, Ji TH. The amino-terminal region of the luteinizing hormone/choriogonadotropin receptor contacts both subunits of human choriogonadotropin. I. Mutational analysis. J Biol Chem 1998; 273:13835-40. [PMID: 9593728 DOI: 10.1074/jbc.273.22.13835] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The luteinizing hormone/choriogonadotropin receptor is a seven-transmembrane receptor. Unlike most seven-transmembrane receptors, it is composed of two halves of equal size, the N-terminal extracellular exodomain and the C-terminal membrane-associated endodomain. The exodomain is exclusively responsible for high affinity hormone binding, whereas receptor activation occurs only in the endodomain. This mutually exclusive physical separation of the two functional domains sets the lutropin receptor and its subfamily of receptors apart from all other seven-transmembrane receptors. The mechanisms of hormone binding and receptor activation also appear to be different from those of other receptors in that binding occurs in at least two steps. However, the precise hormone contact sites in the exodomain are unknown. To determine the hormone/receptor contact sites, we have examined the receptor using progressive truncation from the C terminus, Ala scanning, immunofluorescence microscopy, and antibody binding. Progressive truncation from the C terminus of the receptor indicates several discrete regions that impact hormone binding. These regions are around the boundaries of exons 1-2, 4-5, 6-7, and 9-10. Ala scanning of the Asp17-Arg26 region near the exon 1-2 junction uncovered three alternating residues (Leu20, Cys22, and Gly24) crucial for hormone binding. Ala substitution for any one of these residues abolished hormone binding, although the resulting mutant receptors were successfully expressed on the cell surface. In contrast, Ala substitution for their flanking and intervening residues did not impair hormone binding. These results and the data in the accompanying article (Phang, T., Kundu, G., Hong, S., Ji, I., and Ji, T. (1998) J. Biol. Chem. 273, 13841-13847) indicate that this region directly contacts the hormone and suggest a novel mode of embracing the hormone.
Collapse
Affiliation(s)
- S Hong
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071-3944, USA
| | | | | | | |
Collapse
|
32
|
Abstract
The luteinizing hormone receptor (LHR) is a member of the subfamily of glycoprotein hormone receptors within the superfamily of G protein-coupled receptor (GPCR)/seven-transmembrane domain receptors. Over the past eight years, major advances have been made in determining the structure and function of the LHR and its gene. The hormone-binding domain has been localized to exons 1-7 in the extracellular (EC) domain/region of the receptor, which contains several leucine-rich repeats. High-affinity binding of LH and human chorionic gonadotrophin (hCG) causes secondary hormone or receptor contacts to be established with regions of the EC loop/transmembrane module that initiate signal transduction. Models of hormone-receptor interaction have been derived from the crystal structures of hCG and of the ribonuclease inhibitor, which also contains leucine-rich repeats. Such models provide a framework for the interpretation of mutational studies and for further experiments. The extracellular domain of the receptor has been overexpressed in vitro, which will facilitate crystallographic resolution of the structure of the receptor-binding site. The transmembrane domain/loop/cytoplasmic module transduces the signal for coupling to G proteins. Several constitutive, activating mutations that cause human disease have been found in helix VI and adjacent structures. These mutations have provided valuable information about mechanisms of signal transfer and G protein coupling. The structure of the LHR gene has been elucidated, and the regulation of its transcription is beginning to be understood. Valuable insights into receptor evolution have been derived from analysis of sequence homologies, the gene structure of glycoprotein hormone receptors and other members of the GPCR family, and the glycoprotein hormone receptor-like precursors identified in several invertebrate species.
Collapse
Affiliation(s)
- M L Dufau
- Molecular Endocrinology Section, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
33
|
El-Hefnawy T, Huhtaniemi I. Progesterone can participate in down-regulation of the luteinizing hormone receptor gene expression and function in cultured murine Leydig cells. Mol Cell Endocrinol 1998; 137:127-38. [PMID: 9605514 DOI: 10.1016/s0303-7207(98)00002-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The intratesticular concentration of progesterone (P) rises up to the micromolar range during high-dose luteinizing hormone (LH)/hCG stimulation. The aim of this study was to examine whether P is involved in the concomitant down-regulation of the LH receptor (R) function. The effects were tested in a mouse Leydig tumor cell line (mLTC-1) and in Percoll-purified adult mouse Leydig cells. Pre-incubation of the mLTC-1 cells for 48 h with P (1-10 micromol/l) decreased in dose-dependent fashion their specific binding of [125I]iodo-hCG as well as the hCG-induced cAMP production (down to 65 and 40% respectively, of controls, P < 0.01). Similar effect of P on hCG-induced cAMP production was observed in adult mouse Leydig cells following a 24 h incubation in the presence of P (0.3-10 micromol/l). In addition, P treatment significantly inhibited the expression of a transiently transfected murine LHR promoter (715 or 950 bp of the 5' untranslated region)-luciferase fusion constructs in mLTC-1 cells (down to 50% of control, P < 0.01). In accordance, a 6-12 h culture in the presence of 5-10 micromol/l of P showed significant down-regulatory effects on the steady state levels of LHR-mRNA in mLTC-1 cells. These inhibitory effects of P on the LHR expression and function were mimicked by similar concentrations of cortisol, but not by testosterone or estradiol. Blocking the steroid synthesis of mLTC-1 cells with 86 micromol/l of aminoglutethimide (AMG) partially reversed the down-regulating effect of hCG on the LHR-mRNA. Moreover, a 24 h culture in the presence of AMG showed an up-regulating effect on expression of the LHR promoter-luciferase constructs, and including hCG (50 microg/l) in the culture medium enhanced this effect. Hence, in the absence of steroidogenesis, hCG up-regulates the LHR promoter expression. In conclusion, we present here a novel short-loop regulatory mechanism in murine Leydig cells where P exerts a negative effect on LHR expression and function. Since Leydig cell P production is dramatically increased during high-dose stimulation with LH/hCG, due to blockade of C21 steroid side chain cleavage, the present findings offer a function for this steroid in the LHR down-regulation.
Collapse
Affiliation(s)
- T El-Hefnawy
- Department of Physiology, University of Turku, Finland
| | | |
Collapse
|
34
|
Racine C, Rey R, Forest MG, Louis F, Ferré A, Huhtaniemi I, Josso N, di Clemente N. Receptors for anti-müllerian hormone on Leydig cells are responsible for its effects on steroidogenesis and cell differentiation. Proc Natl Acad Sci U S A 1998; 95:594-9. [PMID: 9435237 PMCID: PMC18465 DOI: 10.1073/pnas.95.2.594] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Strong overexpression of anti-Müllerian hormone (AMH) in transgenic mice leads to incomplete fetal virilization and decreased serum testosterone in the adult. Conversely, AMH-deficient mice exhibit Leydig cell hyperplasia. To probe the mechanism of action of AMH on Leydig cell steroidogenesis, we have studied the expression of mRNA for steroidogenic proteins in vivo and in vitro and performed a morphometric analysis of testicular tissue in mice overexpressing the hormone. We show that overexpression of AMH in male transgenic mice blocks the differentiation of Leydig cell precursors. Expression of steroidogenic protein mRNAs, mainly cytochrome P450 17 alpha-hydroxylase/C17-20 lyase (P450c17), is decreased in transgenic mice overexpressing AMH and in AMH-treated purified Leydig cells. In contrast, transgenic mice in whom the AMH locus has been disrupted show increase expression of P450c17. In vitro, but not in vivo, AMH also decreases the expression of the luteinizing hormone receptor. The effect of AMH is explained by the presence of its receptor on Leydig cells. Our results provide insight into the action of AMH as a negative modulator of Leydig cell differentiation and function.
Collapse
Affiliation(s)
- C Racine
- Institut National de la Santé et de la Recherche Médicale, Ecole Normale Supérieure, Département de Biologie, Montrouge, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Simoni M, Gromoll J, Nieschlag E. The follicle-stimulating hormone receptor: biochemistry, molecular biology, physiology, and pathophysiology. Endocr Rev 1997; 18:739-73. [PMID: 9408742 DOI: 10.1210/edrv.18.6.0320] [Citation(s) in RCA: 187] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- M Simoni
- Institute of Reproductive Medicine of the University, Münster, Germany
| | | | | |
Collapse
|
36
|
Sairam MR, Subbarayan VS. Characterization of the 5' flanking region and potential control elements of the ovine follitropin receptor gene. Mol Reprod Dev 1997; 48:480-7. [PMID: 9364442 DOI: 10.1002/(sici)1098-2795(199712)48:4<480::aid-mrd8>3.0.co;2-m] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the preceding two reports, we presented evidence for the structure and functional characteristics of two different, yet related variants of the sheep testicular follicle-stimulating hormone receptor (oFSH receptor) cDNA. To shed further light on the structural basis of the formation of such receptor forms with different motifs and the eventual understanding of gene regulation, we initiated studies to clone the gene. An 8 kb EcoR I fragment containing the exon-1 and 5' flanking sequence was cloned and characterized from among the 14 clones that were isolated from the genomic library. Although not all other clones were fully characterized, we believe that the entire gene of 85-100 kb has been secured as we adopted a successive screening strategy to accommodate currently known alternatively spliced variants of the receptor in this species. This has led us to propose a revised model that includes an 11th exon for the oFSH receptor gene. The 11th exon that lies beyond the currently postulated 10th exon contributes important DNA sequence that results in two different structural/functional motifs. One creates a dominant negative receptor and the other leads to the formation of a growth factor type I receptor for the hormone. In the 2.1 kb 5'-upstream region, there are a number of potentially interesting regulatory elements that resemble sites for estrogen response element (ERE-like), CRE, and orphan receptor (SF-1/ NGF I-A) transcription factors among others. Other interesting features include the presence of potential germ cell specific and methylation sites. By performing primer extension with testicular RNA, we could identify a single major transcription start site at -163 relative to +1ATG. The availability of the structure of FSH-receptor gene in this domestically important seasonal breeder could spur investigations into the control of receptor gene expression.
Collapse
Affiliation(s)
- M R Sairam
- Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), Illkirch, France.
| | | |
Collapse
|
37
|
Kakinuma A, Chazenbalk GD, Tanaka K, Nagayama Y, McLachlan SM, Rapoport B. An N-linked glycosylation motif from the noncleaving luteinizing hormone receptor substituted for the homologous region (Gly367 to Glu369) of the thyrotropin receptor prevents cleavage at its second, downstream site. J Biol Chem 1997; 272:28296-300. [PMID: 9353284 DOI: 10.1074/jbc.272.45.28296] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The thyrotropin receptor (TSHR) exists in two forms (single polypeptide and two subunits), whereas the lutropin/chorionic gonadotropin receptor (LH/CGR) is a single chain. Recent data suggest that the TSHR cleaves at two sites. We mutagenized selected chimeric TSH-LH/CGR to localize the cleavage sites in the TSHR. All 23 receptors mutated in the estimated vicinity of the upstream site cleaved into two subunits as determined by 125I-TSH cross-linking to intact cells. In contrast, in a series of mutations homologous to the noncleaving LH/CGR, the downstream TSHR cleavage site localized to three amino acids (GQE367-369). Remarkably, group substitution of these residues, but not substitution of individual residues, abolished cleavage. Moreover, the mutation that prevented cleavage (GQE367-369NET) transposed a motif (NET291-293) that is glycosylated in the LH/CGR. TSHR cleavage or noncleavage after substitution of GQE367-369 with other triplets (AAA, NQE, and NQT) was consistent with a role for N-linked glycosylation at this site. In summary, our data (i) support the concept that the TSHR cleaves at two sites, (ii) relate TSHR residues GQE367-369 to cleavage at the second, downstream site, and (iii) suggest that cleavage or noncleavage at site two is related to N-linked glycosylation. These findings provide new insight into the evolutionary divergence of two closely related receptors.
Collapse
Affiliation(s)
- A Kakinuma
- Thyroid Molecular Biology Unit, Veterans Administration Medical Center and the University of California, San Francisco, California 94121, USA
| | | | | | | | | | | |
Collapse
|
38
|
Mandai M, Konishi I, Kuroda H, Fukumoto M, Komatsu T, Yamamoto S, Nanbu K, Rao CV, Mori T. Messenger ribonucleic acid expression of LH/hCG receptor gene in human ovarian carcinomas. Eur J Cancer 1997; 33:1501-7. [PMID: 9337696 DOI: 10.1016/s0959-8049(97)00166-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The mRNA expression of luteinizing hormone (LH)/human chorionic gonadotropin (hCG) receptors was analysed by the RT-nested PCR method in five normal ovarian tissues, 62 ovarian tumours (5 benign, 7 borderline and 43 malignant epithelial tumours, 3 sex cord-stromal tumours and 4 germ cell tumours) and in 2 ovarian cancer cell lines. In normal ovaries, two cDNA fragments of different sizes were detected using primers designed to amplify a region including exon 9. Sequencing revealed that the larger fragment was derived from a full-length receptor, while the smaller fragment was a splice variant lacking exon 9. In ovarian tumours, the larger fragment of LH/hCG receptors was detected in 40% of the epithelial ovarian carcinomas, none of the germ cell tumours, all of the sex cord-stromal tumours and one of the 2 ovarian cancer cell lines. Immunohistochemistry confirmed the localisation of LH/hCG receptor protein in the tumour cells which correlated with mRNA expression. Patients with full-length LH/hCG receptors in carcinomas showed a better prognosis compared with those without the receptors.
Collapse
Affiliation(s)
- M Mandai
- Department of Gynecology and Obstetrics, Kyoto University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Reinhart J, Xiao S, Arora KK, Catt KJ. Structural organization and characterization of the promoter region of the rat gonadotropin-releasing hormone receptor gene. Mol Cell Endocrinol 1997; 130:1-12. [PMID: 9220016 DOI: 10.1016/s0303-7207(97)00064-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The gene encoding the rat gonadotropin-releasing hormone (GnRH) receptor was isolated, and its structural organization and promoter region were characterized. The gene was found to consist of three exons that encode the receptor protein, and spanned about 20 kb. Of two genomic clones analyzed, one contained the 5'-untranslated region and the first exon, and the other contained the second and third exons. The sizes of the first, second, and third exons are 625, 217, and 1476 nt, respectively. The first intron is at least 12 kb in length and is located between nucleotides 522 and 523 of the cDNA reading frame, in the middle of the fourth transmembrane domain. The second intron is about 2.5 kb and is also located in the reading frame between nucleotides 739 and 740, separating the fifth and sixth transmembrane domains. Genomic blots in combination with cloning and sequencing suggested that a single GnRH receptor gene is present in the rat genome. Primer extension indicated that the transcription start site is located 103 nt upstream of the translational start codon. A putative TATA box is positioned 23 nt in front of the transcription initiation site. The 1.8 kb 5' flanking sequence contains an SF-1 site, an AP-1 site, CCAAT sequences, a Pit-1 binding site, and a potential CRE-like sequence. To evaluate promoter activity, the 1.8 kb and two 5' deleted fragments of 1.2 and 0.6 kb were fused to the luciferase reporter gene and transiently expressed in immortalized pituitary gonadotrophs (alphaT3-1 cells) and hypothalamic neurons (GT1-7 cells), and in nonpituitary (COS-7) cells. Luciferase gene expression was significantly increased by all three fragments in pituitary and hypothalamic cells, but not in COS-7 cells. The promoter activity of the 1.2 kb fragment was higher than that of the other fragments. Forskolin and cAMP analogs increased luciferase gene expression in both alphaT3-1 and GT1-7 cells, but activation of protein kinase C by phorbol myristate acetate had no effect. These studies indicate that positive and negative regulatory elements are present within the 1.8 kb 5' flanking sequence of the GnRH receptor. Knowledge of the genomic organization and analysis of the promoter region of the rat GnRH receptor gene will facilitate the elucidation of its transcriptional control in pituitary gonadotrophs and hypothalamic neurons.
Collapse
Affiliation(s)
- J Reinhart
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
40
|
Zhang FP, Rannikko AS, Manna PR, Fraser HM, Huhtaniemi IT. Cloning and functional expression of the luteinizing hormone receptor complementary deoxyribonucleic acid from the marmoset monkey testis: absence of sequences encoding exon 10 in other species. Endocrinology 1997; 138:2481-90. [PMID: 9165039 DOI: 10.1210/endo.138.6.5196] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Based on sequence homologies among the human, porcine, rat, and mouse genes for the LH receptor (LHR), overlapping partial fragments of LHR complementary DNAs (cDNAs) were multiplied from marmoset monkey testicular RNA using reverse transcription-PCR. Ligations of the individual cDNA fragments generated a full-length monkey LHR cDNA (2031 bp) containing the complete amino acid-coding sequence (676 amino acids). Northern hybridization analysis of monkey testicular RNA, using a complementary RNA probe corresponding to the full-length cDNA, demonstrated major transcripts of 5.5 and 1.4 kilobases and minor ones of 4.0, 2.7, and 1.9 kilobases. Sequence analysis of the monkey LHR cDNA revealed a striking feature, i.e. the absence of an 81-bp nucleotide sequence corresponding to exon 10, present in the LHR cDNAs of all other species studied to date. The monkey LHR cDNA displayed 83-94% overall sequence homology with the other mammalian LHR cDNAs. Reverse transcription-PCR with human exon 10-specific primers demonstrated the total absence of this sequence from the monkey LHR messenger RNA. Southern hybridization of monkey genomic DNA using a human exon 10 probe demonstrated its presence in the monkey gene and that it is totally spliced out from the primary transcript. COS cells transfected with the monkey LHR cDNA showed similar high affinity (Kd = 0.25 nmol/liter) of [125I]iodo-hCG binding as those transfected with human LHR cDNA (Kd = 0.20 nmol/liter). The cells expressing the recombinant monkey and human LHR displayed similar responses of extracellular cAMP and inositol trisphosphate to hCG. In conclusion, marmoset monkey LHR seems to lack the sequence corresponding to exon 10 of the LHR gene in other mammalian species. The truncation does not alter LHR function, as the monkey receptor protein bound hCG and evoked cAMP and inositol trisphosphate responses comparable to those of the human LHR containing the exon 10-encoded structure. As the sequence homologous to exon 10 is missing in the other two glycoprotein receptors, i.e. those of FSH and TSH, this extra exon is apparently inserted into the LHR messenger RNA of some species during evolution from intronic sequences by a change in alternative splicing.
Collapse
Affiliation(s)
- F P Zhang
- Department of Physiology, University of Turku, Finland
| | | | | | | | | |
Collapse
|
41
|
Hauser F, Nothacker HP, Grimmelikhuijzen CJ. Molecular cloning, genomic organization, and developmental regulation of a novel receptor from Drosophila melanogaster structurally related to members of the thyroid-stimulating hormone, follicle-stimulating hormone, luteinizing hormone/choriogonadotropin receptor family from mammals. J Biol Chem 1997; 272:1002-10. [PMID: 8995395 DOI: 10.1074/jbc.272.2.1002] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Using oligonucleotide probes derived from consensus sequences for glycoprotein hormone receptors, we have cloned an 831-amino acid residue-long receptor from Drosophila melanogaster that shows a striking structural homology with members of the glycoprotein hormone (thyroid-stimulating hormone (TSH); follicle-stimulating hormone (FSH); luteinizing hormone/choriogonadotropin (LH/CG)) receptor family from mammals. This homology includes a very large, extracellular N terminus (20% sequence identity with rat TSH, 19% with rat FSH, and 20% with the rat LH/CG receptor) and a seven-transmembrane region (53% sequence identity with rat TSH, 50% with rat FSH, and 52% with the rat LH/CG receptor). The Drosophila receptor gene is >7.5 kilobase pairs long and contains 17 exons and 16 introns. Seven intron positions coincide with introns in the mammalian glycoprotein hormone receptor genes and have the same intron phasing. This indicates that the Drosophila receptor is evolutionarily related to the mammalian receptors. The Drosophila receptor gene is located at position 90C on the right arm of the third chromosome. The receptor is strongly expressed starting 8-16 h after oviposition, and the expression stays high until after pupation. Adult male flies express high levels of receptor mRNA, but female flies express about 6 times less. The expression pattern in embryos and larvae suggests that the receptor is involved in insect development. This is the first report on the molecular cloning of a glycoprotein hormone receptor family member from insects.
Collapse
Affiliation(s)
- F Hauser
- Department of Cell Biology and Anatomy, Zoological Institute, University of Copenhagen, Denmark
| | | | | |
Collapse
|
42
|
Le F, Groshan K, Zeng XP, Richelson E. Characterization of the genomic structure, promoter region, and a tetranucleotide repeat polymorphism of the human neurotensin receptor gene. J Biol Chem 1997; 272:1315-22. [PMID: 8995438 DOI: 10.1074/jbc.272.2.1315] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In the present study, we have cloned the human neurotensin receptor (NTR) gene, determined its structure, demonstrated that its promoter is functional in transfection experiments, and identified the start site of transcription and a tetranucleotide repeat polymorphism that locates at less than 3 kilobase pairs from the gene. The gene contains three introns, all in the coding regions. Several differences in genomic clones and previously characterized cDNA sequences are reconciled. The 5' regulatory region, which is rich in presumptive transcription factors, can drive luciferase expression in transfected CHO-K1 cells. Stepwise 5' deletions identify a positive modulator between -782 and -1309 and a negative modulator between -1309 and -1563. Southern blot analyses demonstrate a single copy gene for the NTR. The tetranucleotide repeat polymorphism is highly informative with at least 23 alleles and might serve as a very useful marker for genetic study of the relationship between the NTR and neuropsychiatric disorders.
Collapse
Affiliation(s)
- F Le
- Laboratory of Neuropsychopharmacology, Mayo Clinic, Jacksonville, Florida 32224, USA
| | | | | | | |
Collapse
|
43
|
O'Shaughnessy PJ, Dudley K, Rajapaksha WR. Expression of follicle stimulating hormone-receptor mRNA during gonadal development. Mol Cell Endocrinol 1996; 125:169-75. [PMID: 9027355 DOI: 10.1016/s0303-7207(96)03957-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Receptors for follicle-stimulating hormone (FSH) are found only in the gonads and have been localised to the Sertoli cells of the testis and the granulosa cells of the ovary. During gonadal development, functional signal transduction systems are present before gonadotrophin receptors appear indicating the expression of the receptors is the crucial step in development of gonadal responsiveness to gonadotrophins. The FSH receptor gene contains a single large exon which encodes the transmembrane and intracellular domains and nine smaller exons which encode most of the extracellular domain. In all species studied so far the FSH-receptor primary transcript has been shown to undergo alternate splicing. The function of these alternate transcripts is unclear but changes in alternate splicing appear to be associated with development of receptor mRNA expression. In the rat transcripts encoding only the extracellular domain of the receptor are detectable 2 days before transcripts encoding the full length receptor. In the mouse ovary FSH-receptor mRNA levels and alternate splicing has been measured during development. Results show that FSH-receptor mRNA is detectable in day 1 ovaries which contain only primordial follicles. At this stage mRNA levels are low but a significant increase in FSH-receptor mRNA is seen around day 5 when primary follicles first appear. This correlates with in situ hybridisation studies which first detect FSH-receptor transcripts in primary follicles. At all stages of development the level of transcripts encoding the extracellular domain was significantly greater than that encoding for the transmembrane and intracellular regions suggesting that significant levels of shortened transcripts are produced. In the hypogonadal (hpg) mouse which lacks circulating gonadotrophins levels of FSH-receptor mRNA appeared normal up to 15 days. This shows that gonadotrophins ar not require for development of FSH-receptor mRNA levels. Studies on FSH-receptor mRNA levels during granulosa cell luteinization show that there is complete loss of full-length transcripts soon after luteinization. Transcripts encoding the extracellular domain remain present, however, up to at least mid-cycle. Thus, changes in receptor transcript splicing during loss of FSH-receptors appear to mimic, in reverse, changes occurring during development. It may be that the FSH-receptor gene is constitutively expressed in follicular (pre-granulosa) cells, granulosa cells and granulosa-luteal cells but that control of RNA splicing regulates levels of full-length FSH-receptor transcript.
Collapse
Affiliation(s)
- P J O'Shaughnessy
- Department of Veterinary Physiology, University of Glasgow Veterinary School, UK
| | | | | |
Collapse
|
44
|
Remy JJ, Couture L, Pantel J, Haertlé T, Rabesona H, Bozon V, Pajot-Augy E, Robert P, Troalen F, Salesse R, Bidart JM. Mapping of HCG-receptor complexes. Mol Cell Endocrinol 1996; 125:79-91. [PMID: 9027346 DOI: 10.1016/s0303-7207(96)03955-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Molecular forms of the porcine LH/CG receptor (pLHR) and complexes between hCG and either the full-length pLHR or its extracellular domain (ectodomain) have been produced in various recombinant systems. In COS cells and in the baculovirus insect cells system, the co-expression of the ecto- and endo-domains reconstituted a functional receptor where the association of the two domains seems to depend upon the presence of disulfide bridges. According to previous observations [39], synthetic peptides mimicking three regions of the ectodomain (21-38, 100-115, 250-272) were found to inhibit hormone binding and stimulation of cAMP production. Antisera raised against these peptides contained anti-peptide antibodies (Ab) able to interfere with hormone signalling. Moreover, the results of peptide mapping indicated that some peptides stretches may be more involved in signalling rather than in binding. Immunochemical mapping based on monoclonal antibodies (mAbs) was used to probe the hCG-ectodomain complex. It appeared that mAbs directed to epitopes present on the 'beta-tip' of hCG (assembled from the beta subunit loops 3 and 1, and previously designated site IIIb) and on the 'alpha-tip' (alpha subunit loops 1 and 3, site IIIa) bound to hCG-receptor complexes, whereas a conformational epitope (defined by the alpha-beta interface between beta seat belt C-terminus and alpha loop 2, site II) was masked. Interestingly, we and others previously reported that, in the hCG-full length receptor complex, site IIIa was shielded to mAb binding. A peptide mimicking the second extracellular loop (EL2) of the receptor endodomain was found to prevent the binding of a mAb directed to site IIIa, suggesting that this region of the endodomain may be interacting with the 'alpha-tip'. In the full-length, membrane anchored pLHR, the EL2 peptide inhibited hCG-induced cAMP production, but not binding. The possibility of inhibiting stimulation without inhibition of binding gives support to the 'negative specificity' hypothesis [6]. Thus, the ectodomain of the glycoprotein hormone receptors might be considered as a screening device preventing access of any glycoprotein hormone to the signalling peptide keys of the endodomain, which otherwise would be sensitive to any alpha subunit stimulation. Finally, antibody binding to site IIIa on the hCG-ectodomain complex was also hindered by an anti-peptide mAb directed against a peptide encoded by the eighth exon (pE x 8) of the LHR. This suggests that pEx8 is vicinal to the alpha-tip of hCG and to EL2 in the hCG-full length receptor complex. Altogether, these observations help to build up a topological model of the hCG-receptor complex.
Collapse
Affiliation(s)
- J J Remy
- Unité Récepteurs et Communication Cellulaire, Biologie Cellulaire et Moléculaire, INRA-Biotechnologies, Jouy-en-Josas, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gilchrist RL, Ryu KS, Ji I, Ji TH. The luteinizing hormone/chorionic gonadotropin receptor has distinct transmembrane conductors for cAMP and inositol phosphate signals. J Biol Chem 1996; 271:19283-7. [PMID: 8702611 DOI: 10.1074/jbc.271.32.19283] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The luteinizing hormone/chorionic gonadotropin receptor is a member of the seven-transmembrane receptor family. It is coupled, presumably via Gs and Gq, to two signal pathways involving adenylyl cyclase/cAMP and phospholipase C/inositol phosphate (IP). Little is known about the events prior to G-protein coupling: for example, whether these signals are generated from a single or multiple independent origins and mechanisms, when and where they diverge, and how they are transduced. We report novel observations that the cAMP signal and the IP signal originate and diverge upstream of G-protein coupling. The generation of these two signals independently involves Lys583 in exoloop 3 of the rat receptor. For this study, Lys583 of the receptor was substituted with a panel of amino acids, and mutant receptors were assayed for hormone binding and induction of cAMP, inositol monophosphate, inositol bisphosphate, and inositol trisphosphate. No substitutions for Lys583 were permissible for cAMP induction, despite successful surface expression and hormone binding. In contrast, several substitutions were permissible for IP induction. Our results suggest two distinct transmembrane signal conductors for cAMP and inositol phosphate signals and imply particular models of receptor activation not previously suggested.
Collapse
Affiliation(s)
- R L Gilchrist
- Department of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071-3944, USA
| | | | | | | |
Collapse
|
46
|
Tsai-Morris CH, Buczko E, Geng Y, Gamboa-Pinto A, Dufau ML. The genomic structure of the rat corticotropin releasing factor receptor. A member of the class II G protein-coupled receptors. J Biol Chem 1996; 271:14519-25. [PMID: 8662941 DOI: 10.1074/jbc.271.24.14519] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Isolation and structural characterization of the rat corticotropin releasing factor receptor (CRFR) gene was performed to determine the exon/intron organization of the coding region and the potential for splice variants. The CRFR gene contains 13 exons and 12 introns, and the positions of the exon/intron junctions are similar to those of other Class II G protein-coupled receptor genes including the parathyroid hormone and glucagon receptors. The promoter resides within 593 base pairs of the initiation codon and the major transcriptional start site at nucleotide -238. This domain does not possess a TATA box but contains multiple Sp1 and AP-2 sites upstream and downstream of the major transcriptional start site. Intron junctions were identified in the extracellular, transmembrane (TM), and cytoplasmic (C) domains of the CRFR, giving the potential for differential signal transduction by splice variants. CRFR cDNAs derived from rat Leydig cell mRNA included the pituitary Form A, which spans exons 1-13, and two splice variants with deletion of exon 3 or exons 7, 11, and 12. An evolutionary link between the intronless TM/C module of the glycoprotein hormone receptors and the intron-containing TM/C module of the CRFR is suggested by the common position of the luteinizing hormone receptor Form D alternate acceptor splice site and the CRFR intron 12.
Collapse
Affiliation(s)
- C H Tsai-Morris
- Section on Molecular Endocrinology, Endocrinology and Reproduction Research Branch, NICHD, National Institutes of Health, Bethesda, Maryland 20892-4510, USA
| | | | | | | | | |
Collapse
|
47
|
El-Hefnawy T, Krawczyk Z, Nikula H, Viherä I, Huhtaniemi I. Regulation of function of the murine luteinizing hormone receptor promoter by cis- and trans-acting elements in mouse Leydig tumor cells. Mol Cell Endocrinol 1996; 119:207-17. [PMID: 8807640 DOI: 10.1016/0303-7207(96)03815-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The transcriptional activity of various lengths of the 5'-untranslated region (UTR) of the murine LH receptor (R) gene were studied using the luciferase reporter gene in transiently transfected mouse Leydig tumor cells (mLTC-1). Chinese hamster ovarian (CHO) and HeLa cells were used as controls. The basal transcriptional promoter activity in mLTC-1 cells resided within the first 173 base pairs (bp) of the 5'-UTR. Placing an LHR promoter fragment (bases -715/ -56) in front of the Herpes simplex virus thymidine kinase (TK) minimal promoter resulted in a 7-fold increase in luciferase activity. Deletion of bases -56 to -173 of the above construct totally abolished the increased luciferase activity, brought about by the LHR promoter sequences. Basically similar results on LHR promoter function were observed using CHO cells. In contrast, no LHR promoter activity was detected in HeLa cells, indicating a cell specific nature of its function. The first 173 bp promoter domain is GC-rich, with several SP-1 binding domains, and it bound specifically nuclear proteins isolated from mLTC-1 and HeLa cells. RNAse protection assays reconfirmed the presence of several transcription initiation sites within the first 310 bp of the 5'-UTR, also in the absence of the cognate LHR coding sequences. The most distal site at bp -310 did not function in the absence of the first 173 bp of the 5'-UTR. Other transcription initiation sites were identified closer to the translation initiation site. hCG (50 micrograms/l), 8-bromo (Br)-cAMP (100 mumol/l) and cholera toxin (100 microgram/l) displayed qualitatively similar negative effects on the LHR promoter activity in the transfected mLTC-1 cells when the constructs containing at least the first 565 bp of the LHR 5'-UTR were used, but the inhibitory effects were greatly decreased in constructs containing < or = 304 bp of the promoter region. Hence, the hCG/cAMP associated inhibitory effects interact with region(s) located mainly between bp -565 and -305 of the LHR promoter. The inhibitory role of cAMP on LHR gene expression was also confirmed at the level of hCO-binding and hCG stimulated cAMP production of mLTC-1 cells. In conclusion, the current results elucidate the cis- and trans-acting elements in the regulation of expression of the murine LHR gene in cultured mouse Leydig cells. The minimal basal promoter activity is within the first 173 nucleotides of the 5'-UTR and the structural elements of the negative LHR regulation by the cognate hormone and elevated cAMP levels are mainly located within nucleotides -305 to -565 of the 5'-UTR. The function of the murine LHR promoter is similar to, though not identical with that of the rat, but at variance with that of the human LHR gene.
Collapse
Affiliation(s)
- T El-Hefnawy
- Department of Physiology, University of Turku, Finland
| | | | | | | | | |
Collapse
|
48
|
Zhang R, Buczko E, Dufau ML. Requirement of cysteine residues in exons 1-6 of the extracellular domain of the luteinizing hormone receptor for gonadotropin binding. J Biol Chem 1996; 271:5755-60. [PMID: 8621442 DOI: 10.1074/jbc.271.10.5755] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The functional importance of cysteine residues in the extracellular domain and the extracellular loops (EL1 and EL2) to hormone binding of the rat luteinizing hormone receptor (LHR) was investigated. For this purpose, cysteines in the seven-transmembrane holoreceptor (Form A) and its hormone-binding splice variant (Form B) were replaced by serine residues, and mutant receptors were expressed in COS1 and/or insect cells. Within the extracellular domain, individual replacement of all four cysteines from Exon 1 abolished hormone binding activity, and replacement of Cys-109 and Cys-134 from exons 5 and 6 caused a 75% decrease in both cell surface and total cellular solubilized LHR hormone binding activity. Mutations of Cys-257 and -258 (Exon 9), Cys-321 and -331, and Cys-417 and -492 of EL1 and EL2, respectively (Exon 11), showed no surface hormone binding activity on intact cells, but exhibited wild type levels of total hormone binding activity when recovered from detergent-solubilized cellular extracts. This finding indicated that expression of high affinity LHR binding activity at the cell surface is independent of the acquisition of the high affinity binding conformation. Other cysteine residues, including Cys-282 (exon 10), and Cys-314 (exon 11) were not essential for hormone binding activity or plasma membrane insertion. This study demonstrates that the functional hormone binding domain utilizes all cysteines N-terminal to exon 7 and localizes the binding site to this N-terminal region of the extracellular domain.
Collapse
Affiliation(s)
- R Zhang
- Section on Molecular Endocrinology, Endocrinology and Reproduction Research Branch, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
49
|
Jiang X, Dreano M, Buckler DR, Cheng S, Ythier A, Wu H, Hendrickson WA, el Tayar N. Structural predictions for the ligand-binding region of glycoprotein hormone receptors and the nature of hormone-receptor interactions. Structure 1995; 3:1341-53. [PMID: 8747461 DOI: 10.1016/s0969-2126(01)00272-6] [Citation(s) in RCA: 149] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Glycoprotein hormones influence the development and function of the ovary, testis and thyroid by binding to specific high-affinity receptors. The extracellular domains of these receptors are members of the leucine-rich repeat (LRR) protein superfamily and are responsible for the high-affinity binding. The crystal structure of a glycoprotein hormone, namely human choriogonadotropin (hCG), is known, but neither the receptor structure, mode of hormone binding, nor mechanism for activation, have been established. RESULTS Despite very low sequence similarity between exon-demarcated LRRs in the receptors and the LRRs of porcine ribonuclease inhibitor (RI), the secondary structures for the two repeat sets are found to be alike Constraints on curvature and beta-barrel geometry from the sequence pattern for repeated beta alpha units suggest that the receptors contain three-dimensional structures similar to that of RI. With the RI crystal structure as a template, models were constructed for exons 2-8 of the receptors. The model for this portion of the choriogonadotropin receptor is complementary in shape and electrostatic characteristics to the surface of hCG at an identified focus of hormone-receptor interaction. CONCLUSIONS The predicted models for the structures and mode of hormone binding of the glycoprotein hormone receptors are to a large extent consistent with currently available biochemical and mutational data. Repeated sequences in beta-barrel proteins are shown to have general implications for constraints on structure. Averaging techniques used here to recognize the structural motif in these receptors should also apply to other proteins with repeated sequences.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- Chemical Phenomena
- Chemistry, Physical
- Chorionic Gonadotropin/metabolism
- Cystine/chemistry
- Follicle Stimulating Hormone/metabolism
- GTP-Binding Proteins/metabolism
- Glycosylation
- Hormones/metabolism
- Humans
- Luteinizing Hormone/metabolism
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis
- Protein Binding
- Protein Structure, Secondary
- Rats
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/metabolism
- Receptors, FSH/chemistry
- Receptors, FSH/genetics
- Receptors, FSH/metabolism
- Receptors, LH/chemistry
- Receptors, LH/genetics
- Receptors, LH/metabolism
- Receptors, Thyrotropin/chemistry
- Receptors, Thyrotropin/genetics
- Receptors, Thyrotropin/metabolism
- Repetitive Sequences, Nucleic Acid
- Sequence Alignment
- Sequence Homology, Amino Acid
- Structure-Activity Relationship
- Swine
- Thyrotropin/metabolism
Collapse
Affiliation(s)
- X Jiang
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Huang J, Puett D. Identification of two amino acid residues on the extracellular domain of the lutropin/choriogonadotropin receptor important in signaling. J Biol Chem 1995; 270:30023-8. [PMID: 8530405 DOI: 10.1074/jbc.270.50.30023] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The lutropin/choriogonadotropin receptor (LH/CG-R) is a G protein-coupled receptor with a relatively large extracellular domain. The cDNAs of LH/CG-R wild type and 15 point and double mutants, which encoded residues of opposite charge to that of wild type, were transiently transfected into COS-7 cells. Human choriogonadotropin (hCG) binding was determined, as was hCG-mediated cAMP production. Most of the replacements resulted in no substantive effect on the binding affinity of hCG to LH/CG-R or on hCG-stimulated cAMP production, although the mutants expressed at a lower level than LH/CG-R wild type. The most interesting observation was noted with two point mutants of LH/CG-R, Glu332-->Lys and Asp333-->Lys, which bound hCG but failed to give increased cAMP production. Several of the mutant forms of LH/CG-R that expressed at low levels were further analyzed by soluble binding assays and Western blots. There was no evidence of any significant degree of intracellular trapping of hCG-binding mutant receptors. The expected major (93 kDa) and minor (78 kDa) forms were found for LH/CG-R wild type and several of the mutants. The Lys235-->Asp and Asp333-->Lys mutants exhibited primarily the lower M(r) form, indicating that receptor processing was impaired or that the mutant higher M(r) form was more rapidly degraded than LH/CG-R wild type. These results demonstrate that Glu332 and Asp333, which are located near the first transmembrane helix, are important in receptor activation, while other conserved ionizable residues of LH/CG-R appear important in cell surface expression or stability but not in binding or signaling.
Collapse
Affiliation(s)
- J Huang
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens 30602, USA
| | | |
Collapse
|