Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 107(5): 2319-24, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20080658

ABSTRACT

G protein-coupled receptors (GPCRs) are ubiquitous mediators of signaling of hormones, neurotransmitters, and sensing. The old dogma is that a one ligand/one receptor complex constitutes the functional unit of GPCR signaling. However, there is mounting evidence that some GPCRs form dimers or oligomers during their biosynthesis, activation, inactivation, and/or internalization. This evidence has been obtained exclusively from cell culture experiments, and proof for the physiological significance of GPCR di/oligomerization in vivo is still missing. Using the mouse luteinizing hormone receptor (LHR) as a model GPCR, we demonstrate that transgenic mice coexpressing binding-deficient and signaling-deficient forms of LHR can reestablish normal LH actions through intermolecular functional complementation of the mutant receptors in the absence of functional wild-type receptors. These results provide compelling in vivo evidence for the physiological relevance of intermolecular cooperation in GPCR signaling.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, LH/chemistry , Receptors, LH/metabolism , Animals , Cell Line , Chorionic Gonadotropin/metabolism , Genetic Complementation Test , Humans , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Models, Molecular , Mutation , Phenotype , Protein Binding , Protein Multimerization , Receptors, LH/deficiency , Receptors, LH/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Testis/metabolism , Testis/pathology
2.
J Alzheimers Dis ; 12(3): 271-81, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18057561

ABSTRACT

Recently, it has been reported that Kalirin gene transcripts are under-expressed in AD hippocampal specimens compared to the controls. The Kalirin gene generates a dozen Kalirin isoforms. Kalirin-7 is the predominant protein expressed in the adult brain and plays crucial roles in growth and maintenance of neurons. Yet its role in human diseases is unknown. We report that Kalirin-7 is significantly diminished both at the mRNA and protein levels in the hippocampus specimens from 19 AD patients compared to the specimens from 15 controls. Kalirin-7 associates with iNOS in the hippocampus, and therefore, Kalirin-7 is complexed with iNOS less in AD hippocampus extracts than in control hippocampus extracts. In cultured cells, Kalirin-7 associates with iNOS and down-regulates the enzyme activity. The down-regulation is attributed to the highly conserved 33 amino acid sequence, K(617) -H(649), of the 1,663 amino acids long Kalirin-7. Remarkably, the iNOS activity is considerably higher in the hippocampus specimens from AD patients than the specimens from 15 controls. These observations suggest that the under-expression of Kalirin-7 in AD hippocampus correlates to the elevated iNOS activity.


Subject(s)
Alzheimer Disease , Gene Expression/genetics , Guanine Nucleotide Exchange Factors/genetics , Hippocampus/metabolism , Hippocampus/pathology , Nitric Oxide Synthase Type II/genetics , Protein Serine-Threonine Kinases/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cells, Cultured , DNA, Complementary/analysis , Genome/genetics , Guanine Nucleotide Exchange Factors/immunology , Humans , Immunoblotting , Immunoprecipitation , Protein Serine-Threonine Kinases/immunology , RNA/analysis , Reverse Transcriptase Polymerase Chain Reaction
3.
J Alzheimers Dis ; 11(3): 385-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17851188

ABSTRACT

To identify genes aberrantly expressed in the brain of individuals with Alzheimer's Disease (AD), we analyzed RNA extracts from the hippocampus and cerebellum from 19 AD patients and 15 age- and sex-matched control subjects. Our analysis identified a number of genes that were over-expressed or under-expressed specifically in AD hippocampus. Among these genes, kalirin was the most consistently under-expressed in AD hippocampus, which was verified by semi-quantitative RT-PCR and real time PCR. Kalirin is predominantly expressed in the brain, particularly in the hippocampus, and plays crucial roles in neuronal stability and growth. Our observation is the first to relate kalirin to AD and a human disease. In addition to kalirin, the genes for voltage-gated Ca++ channel gamma subunit 3 and visinin-like protein 1 (a Ca++ sensor protein) were under-expressed, whereas inositol 1,4,5-triphosphate 3-kinase B was over-expressed in AD hippocampus. Collectively, these differential expressions could severely impair calcium homeostasis. Remarkably, these aberrant gene expressions in AD hippocampus were not observed in AD cerebellum. Furthermore, housekeeping genes such as ribosomal protein genes are not affected by AD. These results provide new insights into the biochemistry of AD.


Subject(s)
Alzheimer Disease/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Protein Serine-Threonine Kinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Apolipoprotein E4/metabolism , Cerebellum/metabolism , Cerebellum/pathology , Female , Gene Expression , Guanine Nucleotide Exchange Factors/genetics , Hippocampus/pathology , Humans , Male , Protein Serine-Threonine Kinases/genetics , RNA , Reverse Transcriptase Polymerase Chain Reaction
4.
Mol Cell Endocrinol ; 260-262: 137-43, 2007 Jan 02.
Article in English | MEDLINE | ID: mdl-17055146

ABSTRACT

It has been thought that when a hormone binds to a receptor, the liganded receptor activates itself and generates hormone signals, such as the cAMP signal and the inositol phosphate signal (cis-activation). We describe that a liganded LH receptor or FSH receptor molecule is capable of intermolecularly activating nonliganded receptors (trans-activation). Particularly, intriguing is the possibility that a pair of compound heterozygous mutants, one defective in binding and the other defective in signaling, may cooperate and rescue signaling. Furthermore, trans-activation of the binding deficient receptors examined in our studies generates either the cAMP signal or the IP signal, but not both. Trans-activation and selective signal generation have broad implications on signal generation mechanisms, and suggest new therapeutic approaches.


Subject(s)
Gonadotropins/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction , Transcriptional Activation , Amino Acid Sequence , Cyclic AMP/biosynthesis , Gonadotropins/pharmacology , Humans , Leucine-Rich Repeat Proteins , Models, Genetic , Molecular Sequence Data , Photoaffinity Labels , Protein Structure, Tertiary/drug effects , Proteins/chemistry , Signal Transduction/drug effects , Transcriptional Activation/drug effects
5.
Mol Endocrinol ; 19(5): 1318-28, 2005 May.
Article in English | MEDLINE | ID: mdl-15677713

ABSTRACT

Expression of the rat LH receptor (rLHR) is characterized by a dynamic response to a variety of hormonal stimuli. In addition to activation, the pattern of rLHR expression is also modulated by repression. In this report, an upstream initiator-like element (UInr-lE), CTCACTCTAA, of which the CTC direct repeat motif (CTCACTC) is conserved in the rat, mouse, and human, was identified as a suppressor element. Disruption of the element resulted in a 2-fold enhancement of promoter activity in the LHR-expressing murine Leydig tumor cells. The sequences of the two major initiators (Inr), Inr3 and Inr4, of the rLHR core promoter are similar to UInr-lE and competed efficiently with UInr-lE in the formation of specific protein complexes, suggesting that the same proteins interact with both UInr-lE and the Inrs in vivo. The Inrs are necessary for full promoter activity because a mutant promoter lacking Inrs showed a 70% reduction in activity. UInr-lE also further suppressed the activity of a mutant promoter lacking Inrs. UInr-lE interacted with transcription factor II-I (TFII-I) and an unidentified nuclear protein. However, dominant-negative inhibition experiments using p70 indicated that TFII-I positively regulates LHR promoter activity through UInr-lE and Inrs, suggesting that TFII-I can compromise the suppression of promoter activity mediated by UInr-lE. UInr-lE also showed binding properties distinct from that of the upstream initiator-like suppressor element (upstream regulatory element: CACTCTCC) of rat and human dynorphin promoters. Transfection assays using mutated promoters indicate that the suppression of rLHR promoter activity could be regulated via specific interactions between UInr-lE and trans-acting factors.


Subject(s)
5' Flanking Region , Gene Expression Regulation/physiology , Luteinizing Hormone/genetics , Transcription, Genetic/physiology , Animals , Base Sequence , DNA/metabolism , DNA-Binding Proteins/metabolism , Humans , Luteinizing Hormone/biosynthesis , Mice , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Rats , Transcription Factors, TFII/metabolism , Transcription Initiation Site
6.
Mol Endocrinol ; 18(4): 968-78, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14726491

ABSTRACT

Previously, we reported that a liganded LH receptor (LHR) is capable of activating itself (cis-activation) and other nonliganded LHRs to induce cAMP (trans-activation). Trans-activation of the LHR raises two crucial questions. Is trans-activation unique to LHR or common to other G protein-coupled receptors? Does trans-activation stimulate phospholipase Cbeta as it does adenylyl cyclase? To address these questions, two types of novel FSH receptors (FSHRs) were constructed, one defective in hormone binding and the other defective in signal generation. The FSHR, a G protein-coupled receptor, comprises two major domains, the N-terminal extracellular exodomain that binds the hormone and the membrane-associated endodomain that generates the hormone signals. For signal defective receptors, the exodomain was attached to glycosyl phosphatidylinositol (ExoGPI) or the transmembrane domain of CD8 immune receptor (ExoCD). ExoGPI and ExoCD can trans-activate another nonliganded FSH. Surprisingly, the trans-activation generates a signal to activate either adenylyl cyclase or phospholipase Cbeta, but not both. These results indicate that trans-activation in these mutant receptors is selective and limited in signal generation, thus providing new approaches to investigating the generation of different hormone signals and a novel means to selectively generate a particular hormone signal. Our data also suggest that the FSHR's exodomain could not trans-activate LHR.


Subject(s)
Adenylyl Cyclases/metabolism , Isoenzymes/metabolism , Receptors, FSH/metabolism , Type C Phospholipases/metabolism , Cyclic AMP/metabolism , Gene Transfer Techniques , Humans , Iodine Radioisotopes/metabolism , Mutation , Phospholipase C beta , Receptors, FSH/genetics
7.
Endocrine ; 25(2): 111-5, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15711023

ABSTRACT

Asp578Gly is the major mutation of luteinizing hormone (LH) receptors in humans. It is a dominant mutant, constitutively activates Galphas, and induces cAMP production in the absence of the cognate hormone, causing the familial male precocious puberty. The mechanism of the elevated basal cAMP level is unclear. Our data show strikingly different mechanisms between the elevated basal cAMP induced by the activating mutant and the cAMP induced by the wild-type receptor activated by human chorionic gonadotropin (hCG) binding. The study suggests an approach to attenuating the elevated basal cAMP of the activating mutant LH receptor, which could be useful for controlling the familial male precocious puberty. For the study, we used the C-terminal peptides of Galphas and Galphai2, which couple to the receptor.


Subject(s)
Chorionic Gonadotropin/metabolism , Cyclic AMP/biosynthesis , Receptors, LH/physiology , Cell Line , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Expression Regulation , Humans , Male , Point Mutation , Puberty, Precocious/genetics , Receptors, LH/genetics , Receptors, LH/metabolism , Transfection
8.
J Biol Chem ; 278(48): 47868-76, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-12963710

ABSTRACT

Follicle-stimulating hormone (FSH) comprises an alpha subunit and a beta subunit, whereas the FSH receptor consists of two halves with distinct functions: the N-terminal extracellular exodomain and C-terminal membrane-associated endodomain. FSH initially binds to exodomain, and the resulting FSH/exodomain complex modulates the endodomain and generates signal. However, it has been difficult to determine which subunit of FSH contacts the exodomain or endodomain and in what orientation FSH interacts with them. To address these crucial issues, the receptor was Ala-scanned and the hormone subunits were probed with photoaffinity labeling with receptor peptides corresponding to the N-terminal region of the exodomain and exoloop 3 of the endodomain. Our results show that both regions of the receptors are important for hormone binding and signal generation. In addition, the FSH beta subunit is specifically labeled with the N-terminal peptide, whereas the alpha subunit is labeled with the exoloop 3 peptide. These contrasting results show that the FSH beta subunit is close to the N-terminal region and that the alpha subunit is projected toward exoloop 3 in the endodomain. The results raise the fundamental question whether the alpha subunit, common among the glycoprotein hormones, plays a major role in generating the hormone signal common to all glycoprotein hormones.


Subject(s)
Follicle Stimulating Hormone/chemistry , Amino Acid Sequence , Cell Line , Crystallography, X-Ray , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/metabolism , Glycoproteins/chemistry , Glycosylation , Humans , Immunoblotting , Kinetics , Light , Models, Molecular , Molecular Sequence Data , Mutagenesis , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transfection , Ultraviolet Rays
9.
J Clin Endocrinol Metab ; 88(7): 3360-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12843188

ABSTRACT

Loss of function by 11 of 13 naturally occurring mutations in the human GnRH receptor (hGnRHR) was thought to result from impaired ligand binding or effector coupling, but actually results from receptor misrouting. Homo- or heterodimerization of mutant receptors with wild-type (WT) receptors occurs for other G protein-coupled receptors and may result in dominant-negative or -positive effects on the WT receptor. We tested the hypothesis that WT hGnRHR function was affected by misfolded hGnRHR mutants. hGnRHR mutants were found to inhibit the function of WT GnRHR (measured by activation of effector and ligand binding). Inhibition varied depending on the particular hGnRHR mutant coexpressed and the ratio of hGnRHR mutant to WT hGnRHR cDNA cotransfected. The hGnRHR mutants did not interfere with the function of genetically modified hGnRHRs bearing either a deletion of primate-specific Lys(191) or the carboxyl-terminal tail of the catfish GnRHR; these show intrinsically enhanced expression. Moreover, a peptidomimetic antagonist of GnRH enhanced the expression of WT hGnRHR, but not of genetically modified hGnRHR species. The dominant-negative effect of the naturally occurring receptor mutants occurred only for the WT hGnRHR, which has intrinsic low maturation efficiency. The data suggest that this dominant negative effect accompanies the diminished plasma membrane expression as a recent evolutionary event.


Subject(s)
Cell Membrane/metabolism , Evolution, Molecular , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Amino Acid Sequence , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Buserelin/metabolism , Buserelin/pharmacology , COS Cells , Fertility Agents, Female/metabolism , Fertility Agents, Female/pharmacology , Gene Expression , Humans , Indoles/pharmacology , Inositol Phosphates/biosynthesis , Iodine Radioisotopes , Molecular Sequence Data , Mutagenesis , Protein Binding/drug effects , Protein Structure, Tertiary , Pyridines/pharmacology , Receptors, LHRH/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
Endocrinology ; 144(6): 2360-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746296

ABSTRACT

FSH plays crucial roles in differentiation of granulosa cells and development of follicles. Considering the broad scope of FSH effects, a large number of genes are likely responsive to the hormone. However, only a limited number of genes have been identified as FSH-regulated genes, particularly during the preantral stage. In an attempt to better define genes involved in follicular development, we examined primary granulosa cell cultures, an undifferentiated rat ovarian granulosa cell line and rat ovaries, using differential display, quantitative RT-PCR, Northern blot analysis, and in situ hybridization. We report, for the first time, that nicotinamide adenine dinucleotide phosphate-dependent cytosolic T(3)-binding protein mRNA is expressed in the ovary, particularly in the granulosa cell layer of preantral and early antral follicles, but not in large preovulatory follicles. Its expression markedly declines in response to FSH, which is dependent on the period of the exposure. This FSH-responsive down-regulation is dependent on granulosa cell differentiation and follicular development. FSH down-regulates the mRNA via the adenylyl cyclase/cAMP pathway, and the down-regulation requires de novo synthesis of a regulatory protein(s). The cytosolic T(3)-binding protein may play a significant role in the regulation of steroidogenesis and follicular development in the mammalian ovary.


Subject(s)
Carrier Proteins/genetics , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/physiology , Membrane Proteins/genetics , Thyroid Hormones/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cytosol/physiology , Female , Gene Expression/drug effects , Gene Expression Profiling , Granulosa Cells/cytology , Granulosa Cells/drug effects , Molecular Sequence Data , Ovarian Follicle/cytology , Ovarian Follicle/physiology , RNA, Messenger/analysis , Rats , Thyroid Hormone-Binding Proteins
11.
Endocrinology ; 144(1): 29-39, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12488327

ABSTRACT

FSH regulates gene expression for granulosa cell differentiation and follicular development. Therefore, FSH-responsive genes are crucial, but only a few genes have been identified for the early stage of follicular development. In particular, little is known about cytoskeletal genes, which likely play essential roles in the morphological changes such as the antrum formation, a major landmark. FSH is also known to induce the differentiation of an immature, undifferentiated rat ovary granulosa (ROG) cell line. Our data show that FSH induced massive yet distinct reorganization of microtubules and the actin cytoskeletons as well as morphological changes. To identify those genes responding to FSH during the differentiation, differential display was performed on ROG cells. Of the 80 FSH-responsive genes identified, there were three cytoskeleton-related genes (class I beta-tubulin, tropomyosin 4, and kinesin heavy chain), which are crucial for intracellular morphogenesis, transport, and differentiation. Northern blots show that the level of these gene transcripts reached a peak at 6 h after FSH treatment and subsided at 24 h. FSH induced the similar temporal expression not only in granulosa cells isolated from immature rats, but also in vivo. For instance, in situ hybridization showed that beta-tubulin mRNA was transiently expressed in the granulosa cells of large preantral and early antral follicles. Despite the same temporal expression, the regulatory mechanisms of the three genes were strikingly different. As an example, cycloheximide blocked the beta-tubulin mRNA expression, whereas it increased tropomyosin-4 (TM4) mRNA. Yet, it did not impact kinesin heavy chain (Khc) mRNA. In conclusion, FSH induces the massive reorganization of the cytoskeletons and morphological changes by the selective regulation of the gene expression, protein synthesis, and rearrangement of the cytoskeletal proteins in the ROG cells and probably, specific follicles and granulosa cells.


Subject(s)
Cytoskeleton/ultrastructure , Follicle Stimulating Hormone/pharmacology , Gene Expression/drug effects , Granulosa Cells/ultrastructure , Kinesins/genetics , Tropomyosin/genetics , Tubulin/genetics , Animals , Blotting, Northern , Cycloheximide/pharmacology , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Female , Gene Expression Profiling , In Situ Hybridization , Kinetics , Microscopy, Fluorescence , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tropomyosin/analysis , Tubulin/analysis
12.
J Biol Chem ; 277(51): 50165-75, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12374801

ABSTRACT

The human follicle-stimulating hormone (FSH) receptor consists of two distinct domains of approximately 330 amino acids, the N-terminal extracellular exodomain and membrane-associated endodomain including three exoloops and seven transmembrane helices. The exodomain binds the hormone with high affinity, and the resulting hormone/exodomain complex modulates the endodomain where receptor activation occurs. It has been an enigma whether the hormone interacts with the endodomain. In a step to address the question, exoloop 3 of (580)KVPLITVSKAK(590) was examined by Ala scan, multiple substitution, assays for hormone binding, cAMP and inositol phosphate (IP) induction, and photoaffinity labeling. We present the evidence for the interaction of FSH and exoloop 3. A peptide mimic of exoloop 3 specifically and saturably photoaffinity-labels FSH alpha but not FSH beta. This is in contrast to photoaffinity labeling of FSH beta by the peptide mimic of the N-terminal region of the receptor. Leu(583) and Ile(584) are crucial for the interaction of FSH and exoloop 3. Substitutions of these two residues enhanced the hormone binding affinity. This is due to the loss of the original side chains but not the introduction of new side chains. The Leu(583) and Ile(584) side chains appear to project in opposite directions. Ile(584) appears to be so specific and to require flexibility and stereo specificity so that no other amino acids can fit into its place. Leu(583) is less specific. The improvement in hormone binding by substitutions was offset by the severe impairment of signal generation of cAMP and/or inositol phosphate. For example, the Phe or Tyr substitution of Leu(583) improved the hormone binding and cAMP induction but impaired IP induction. On the other hand, the substitutions for Ile(584) and Lys(590) abolished the cAMP and IP induction. Our results open a logical question whether Leu(583), Ile(584), and Lys(590) interact with the exodomain and/or the hormone. The answers will provide new insights into the mechanisms of hormone binding and signal generation.


Subject(s)
Follicle Stimulating Hormone/chemistry , Receptors, FSH/chemistry , Alanine/chemistry , Amino Acid Sequence , Animals , Binding, Competitive , Cell Line , Cyclic AMP/metabolism , DNA Mutational Analysis , Dose-Response Relationship, Drug , Follicle Stimulating Hormone/metabolism , Humans , Inositol Phosphates/chemistry , Inositol Phosphates/metabolism , Kinetics , Leucine/chemistry , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Proline/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, FSH/metabolism , Signal Transduction , Software
13.
Methods ; 27(4): 318-23, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12217647

ABSTRACT

This article describes a novel method to access functional interactions of two defective mutant receptors. As a model, luteinizing hormone receptor, a G-protein-coupled receptor, was used by coexpressing two different mutants, one defective in hormone binding and the other defective in signal generation. When these two mutants were coexpressed in a cell, the cell responded to the hormone and induced the hormone action, indicating the interaction of the two receptors and rescue of the activity. The luteinizing hormone receptor consists of a 350-amino-acid extracellular N-terminal domain (exodomain), followed by seven transmembrane domains and connecting loops (endodomain). Hormone binds to the exodomain, whereas hormone signals are generated in the endodomain. Here, we show that binding of hormone to one receptor can activate adenylyl cyclase through its transmembrane bundle, intramolecular activation (cis-activation), as well as intermolecular activation (trans-activation) through the transmembrane bundle of an adjacent receptor, without forming a stable receptor dimer. Our observations provide new insights into the mechanism of receptor activation mechanisms, and have implications for the treatment of inherited disorders of glycoprotein hormone receptors.


Subject(s)
Mutagenesis/physiology , Receptors, LH/genetics , Receptors, LH/metabolism , Amino Acid Sequence , Binding Sites/physiology , Cyclic AMP/metabolism , Dimerization , Genetic Complementation Test , Luteinizing Hormone/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Receptors, LH/chemistry , Signal Transduction/physiology , Transfection
14.
Mol Endocrinol ; 16(6): 1299-308, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040016

ABSTRACT

G protein-coupled receptors (GPCRs) accommodate a wide spectrum of activators from ions to glycoprotein hormones. The mechanism of activation for this large and clinically important family of receptors is poorly understood. Although initially thought to function as monomers, there is a growing body of evidence that GPCR dimers form, and in some cases that these dimers are essential for signal transduction. Here we describe a novel mechanism of intermolecular GPCR activation, which we refer to as trans-activation, in the LH receptor, a GPCR that does not form stable dimers. The LH receptor consists of a 350-amino acid amino-terminal domain, which is responsible for high-affinity binding to human CG, followed by seven-transmembrane domains and connecting loops. This seven-transmembrane domain bundle transmits the signal from the extracellular amino terminus to intracellular G proteins and adenylyl cyclase. Here, we show that binding of hormone to one receptor can activate adenylyl cyclase through its transmembrane bundle, intramolecular activation (cis-activation), as well as trans-activation through the transmembrane bundle of an adjacent receptor, without forming a stable receptor dimer. Coexpression of a mutant receptor defective in hormone binding and another mutant defective in signal generation rescues hormone-activated cAMP production. Our observations provide new insights into the mechanism of receptor activation mechanisms and have implications for the treatment of inherited disorders of glycoprotein hormone receptors.


Subject(s)
Receptors, LH/chemistry , Receptors, LH/metabolism , Transcriptional Activation , Cell Line , Chorionic Gonadotropin/metabolism , Cyclic AMP/metabolism , Humans , Kinetics , Molecular Conformation , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, LH/genetics
15.
Endocrinology ; 143(4): 1310-7, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11897687

ABSTRACT

Regulators of G protein signaling (RGS) play a pivotal role in cellular signal transduction. RGS3 or RGS10 were overexpressed in GGH(3) cells [GH(3) cells stably expressing the GnRH receptor (GnRHR)]. Responsiveness to a GnRH agonist was assessed because RGS proteins attenuate production of inositol phosphates (IP) and/or cAMP, molecules believed to be involved in GnRH signaling. In addition, site-directed mutagenesis of a potentially palmitoylated Cys(60) residue of RGS10 was used to assess the significance of this site. We observed maximum inhibition of GnRH-stimulated IP responses by RGS3 and by the conserved domain of RGS10 at both 48 and 72 h after transfection, indicating their involvement in G(q)alpha mediated signaling. Significantly diminished cAMP production was observed at all times when cells overexpressed the conserved domain of RGS10; no effect was observed with RGS3 on G(s)alpha-mediated signaling. Palmitic acid incorporation into RGS3 was dependent on agonist occupancy of GnRHR, whereas palmitoylation of RGS10 was constitutive. Mutation of the conserved Cys(60) residue of RGS10 obviated its negative regulatory action on GnRH-stimulated responses, indicating that this site is crucial for its activity on this system. This study is the first demonstration of a role for palmitoylation of this conserved Cys(60) in mammalian G protein signaling.


Subject(s)
GTP-Binding Proteins , Palmitic Acid/metabolism , RGS Proteins/genetics , Receptors, LHRH/biosynthesis , Repressor Proteins , Animals , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Buserelin/pharmacology , Cysteine/metabolism , Electrophoresis, Polyacrylamide Gel , GTPase-Activating Proteins/metabolism , Gene Expression Regulation/genetics , Humans , Kinetics , Mutagenesis, Site-Directed/genetics , Rats , Receptors, LHRH/agonists , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Sodium Fluoride
16.
J Biol Chem ; 277(18): 15795-800, 2002 May 03.
Article in English | MEDLINE | ID: mdl-11859079

ABSTRACT

Luteinizing hormone receptor is a G protein-coupled receptor and consists of two halves: the N-terminal extracellular half (exodomain) and C-terminal membrane-associated half (endodomain). Hormone binds to the exodomain, and the resulting hormone-exodomain complex modulates the endodomain to generate signals. There are mutations that impair either hormone binding or signal generation. We report that the coexpression of a binding defective mutant and a signal-defective mutant rescues signal generation to produce cAMP. This rescue requires both types of mutant receptors and is dependent on the human chorionic gonadotropin dose, the surface concentration of mutant receptors, and the amino acid position of mutations. Furthermore, random collisions among mutant receptors are not involved in the rescue. Our observations provide new insights into the mechanisms of the functional and structural relationship of the exo- and endodomain, signal transduction, and receptor genetics, in particular for defective heterozygotes.


Subject(s)
Chorionic Gonadotropin/metabolism , Mutation , Receptors, LH/genetics , Animals , Binding Sites , Cell Line , Cyclic AMP/metabolism , DNA, Complementary , Follicle Stimulating Hormone/metabolism , GTP-Binding Proteins/metabolism , Heterozygote , Humans , Kinetics , Rats , Receptors, LH/physiology , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...