Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 290(9): 5696-706, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25561730

ABSTRACT

Glucagon-like peptide-1 (GLP-1) plays a pivotal role in glucose homeostasis through its receptor GLP1R. Due to its multiple beneficial effects, GLP-1 has gained great attention for treatment of type 2 diabetes and obesity. However, little is known about the molecular mechanism underlying the interaction of GLP-1 with the heptahelical core domain of GLP1R conferring high affinity ligand binding and ligand-induced receptor activation. Here, using chimeric and point-mutated GLP1R, we determined that the evolutionarily conserved amino acid residue Arg(380) flanked by hydrophobic Leu(379) and Phe(381) in extracellular loop 3 (ECL3) may have an interaction with Asp(9) and Gly(4) of the GLP-1 peptide. The molecular modeling study showed that Ile(196) at transmembrane helix 2, Met(233) at ECL1, and Asn(302) at ECL2 of GLP1R have contacts with His(1) and Thr(7) of GLP-1. This study may shed light on the mechanism underlying high affinity interaction between the ligand and the binding pocket that is formed by these conserved residues in the GLP1R core domain.


Subject(s)
Amino Acids/chemistry , Glucagon-Like Peptide 1/chemistry , Protein Structure, Tertiary , Receptors, Glucagon/chemistry , Amino Acid Sequence , Amino Acids/genetics , Amino Acids/metabolism , Binding Sites/genetics , Conserved Sequence/genetics , Evolution, Molecular , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , HEK293 Cells , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Point Mutation , Protein Binding , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Sequence Homology, Amino Acid
2.
Endocrinol Metab (Seoul) ; 29(3): 320-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25309791

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 (GLP-1) is an incretin hormone produced by cleavage of proglucagon in intestinal L-cells. In the pancreas, GLP-1 stimulates post-prandial insulin secretion, promotes insulin biosynthesis, and improves insulin sensitivity. Because of its insulinotropic activity, GLP-1 has been considered a good candidate drug for treatment of diabetes mellitus. However, clinical use of GLP-1 has been limited by its short half-life, as a result of rapid degradation by dipeptidyl peptidase-IV (DPP-IV). METHODS: We designed a novel GLP-1 analog, Xenopus GLP-1 (xGLP)-E4. The Ala residue in the second position of xGLP was replaced with a Ser residue to increase the half-life in the body. The C-terminal tail of exendin-4 was added to enhance the binding affinity for the GLP-1 receptor (GLP1R). The potency of GLP-1 and its analogs was determined by luciferase assay. The stability of GLP1R agonists was evaluated by determining the activity of agonists that had been preincubated in the presence of fetal bovine serum, which contains innate DPP-IV activity. The effects of xGLP-E4 on insulin secretion and ß-cell growth were investigated using insulin enzyme-linked immunosorbent assay and cell counting. RESULTS: xGLP-E4 exhibited improved stability against DPP-IV activity and increased potency to GLP1R, compared with GLP-1. An increase in glucose-dependent insulin secretion was observed in xGLP-E4-treated pancreatic ß-cells. The effect of xGLP-E4 on ß-cell growth was greater than that of GLP-1. CONCLUSION: We developed a novel GLP-1 analog, xGLP-E4, that shows prolonged longevity and improved efficacy. This analog is a potential candidate for treatment of type 2 diabetes.

3.
J Biol Chem ; 289(25): 17709-20, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24811176

ABSTRACT

IKKß functions as a principal upstream activator of the canonical NF-κB pathway by phosphorylating IκB, leading to its proteasomal degradation. Because IKKß is considered a therapeutic target, understanding its regulation may facilitate the design of efficient regulators of this molecule. Here, we report a novel IKKß-interacting molecule, NME1L, a splicing variant of the NME1 protein. NME1 has attracted attention in cancer research because of its antimetastatic activity and reduced expression in multiple aggressive types of cancer. However, the effect was just moderate but not dramatic in anti-cancer activities. We found that only NME1L interacts with IKKß. Exogenous expression of NME1L resulted in a potent decrease in TNFα-stimulated NF-κB activation, whereas knockdown of NME1/NME1L with shRNA enhanced activity of NF-κB. NME1L down-regulates IKKß signaling by blocking IKKß-mediated IκB degradation. When NME1L was introduced into highly metastatic HT1080 cells, the mobility was efficiently inhibited. Furthermore, in a metastasis assay, NME1L-expressing cells did not colonize the lung. Based on these results, NME1L is a potent antimetastatic protein and may be a useful weapon in the fight against cancers.


Subject(s)
Alternative Splicing , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , NM23 Nucleoside Diphosphate Kinases/biosynthesis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , I-kappa B Kinase/genetics , NF-kappa B/genetics , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology
4.
J Mol Endocrinol ; 52(3): T15-27, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24598200

ABSTRACT

Glucagon-like peptide 1 (GLP1) is an intestinal incretin that regulates glucose homeostasis through stimulation of insulin secretion from pancreatic ß-cells and inhibits appetite by acting on the brain. Thus, it is a promising therapeutic agent for the treatment of type 2 diabetes mellitus and obesity. Studies using synteny and reconstructed ancestral chromosomes suggest that families for GLP1 and its receptor (GLP1R) have emerged through two rounds (2R) of whole genome duplication and local gene duplications before and after 2R. Exon duplications have also contributed to the expansion of the peptide family members. Specific changes in the amino acid sequence following exon/gene/genome duplications have established distinct yet related peptide and receptor families. These specific changes also confer selective interactions between GLP1 and GLP1R. In this review, we present a possible macro (genome level)- and micro (gene/exon level)-evolution mechanisms of GLP1 and GLP1R, which allows them to acquire selective interactions between this ligand-receptor pair. This information may provide critical insight for the development of potent therapeutic agents targeting GLP1R.


Subject(s)
Evolution, Molecular , Glucagon-Like Peptide 1/genetics , Glucose/metabolism , Receptors, Glucagon/genetics , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 2 , Gene Duplication , Glucagon-Like Peptide-1 Receptor , Humans , Insulin/metabolism , Insulin Secretion , Obesity , Vertebrates
5.
PLoS One ; 9(2): e87901, 2014.
Article in English | MEDLINE | ID: mdl-24498396

ABSTRACT

Gonadotropin-releasing hormone (GnRH) and the GnRH receptor (GnRHR) play an important role in vertebrate reproduction. Although many GnRHR genes have been identified in a large variety of vertebrate species, the evolutionary history of GnRHR in vertebrates is unclear. To trace the evolutionary origin of GnRHR we examined the conserved synteny of chromosomes harboring GnRHR genes and matched the genes to linkage groups of reconstructed vertebrate ancestor chromosomes. Consistent with the phylogenetic tree, three pairs of GnRHR subtypes were identified in three paralogous linkage groups, indicating that an ancestral pair emerged through local duplication before two rounds of whole genome duplication (2R). The 2R then led to the generation of six subtypes of GnRHR. Some subtypes were lost during vertebrate evolution after the divergence of teleosts and tetrapods. One subtype includes mammalian GnRHR and a coelacanth GnRHR that showed the greatest response to GnRH1 among the three types of GnRH. This study provides new insight into the evolutionary relationship of vertebrate GnRHRs.


Subject(s)
Evolution, Molecular , Gene Duplication , Genome , Gonadotropin-Releasing Hormone/genetics , Mammals/genetics , Receptors, LHRH/genetics , Amino Acid Sequence , Animals , Molecular Sequence Data , Phylogeny , Sequence Homology, Amino Acid , Synteny
6.
PLoS One ; 8(6): e65420, 2013.
Article in English | MEDLINE | ID: mdl-23776481

ABSTRACT

The glucagon (GCG) peptide family consists of GCG, glucagon-like peptide 1 (GLP1), and GLP2, which are derived from a common GCG precursor, and the glucose-dependent insulinotropic polypeptide (GIP). These peptides interact with cognate receptors, GCGR, GLP1R, GLP2R, and GIPR, which belong to the secretin-like G protein-coupled receptor (GPCR) family. We used bioinformatics to identify genes encoding a novel GCG-related peptide (GCRP) and its cognate receptor, GCRPR. The GCRP and GCRPR genes were found in representative tetrapod taxa such as anole lizard, chicken, and Xenopus, and in teleosts including medaka, fugu, tetraodon, and stickleback. However, they were not present in mammals and zebrafish. Phylogenetic and genome synteny analyses showed that GCRP emerged through two rounds of whole genome duplication (2R) during early vertebrate evolution. GCRPR appears to have arisen by local tandem gene duplications from a common ancestor of GCRPR, GCGR, and GLP2R after 2R. Biochemical ligand-receptor interaction analyses revealed that GCRP had the highest affinity for GCRPR in comparison to other GCGR family members. Stimulation of chicken, Xenopus, and medaka GCRPRs activated Gαs-mediated signaling. In contrast to chicken and Xenopus GCRPRs, medaka GCRPR also induced Gαq/11-mediated signaling. Chimeric peptides and receptors showed that the K(16)M(17)K(18) and G(16)Q(17)A(18) motifs in GCRP and GLP1, respectively, may at least in part contribute to specific recognition of their cognate receptors through interaction with the receptor core domain. In conclusion, we present novel data demonstrating that GCRP and GCRPR evolved through gene/genome duplications followed by specific modifications that conferred selective recognition to this ligand-receptor pair.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Vertebrates/classification , Vertebrates/metabolism , Animals , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/genetics , Phylogeny , Protein Binding , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Vertebrates/genetics
7.
Mol Biol Evol ; 30(5): 1119-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23427277

ABSTRACT

In humans, the secretin-like G protein-coupled receptor (GPCR) family comprises 15 members with 18 corresponding peptide ligand genes. Although members have been identified in a large variety of vertebrate and nonvertebrate species, the origin and relationship of these proteins remain unresolved. To address this issue, we employed large-scale genome comparisons to identify genome fragments with conserved synteny and matched these fragments to linkage groups in reconstructed early gnathostome ancestral chromosomes (GAC). This genome comparison revealed that most receptor and peptide genes were clustered in three GAC linkage groups and suggested that the ancestral forms of five peptide subfamilies (corticotropin-releasing hormone-like, calcitonin-like, parathyroid hormone-like, glucagon-like, and growth hormone-releasing hormone-like) and their cognate receptor families emerged through tandem local gene duplications before two rounds (2R) of whole-genome duplication. These subfamily genes have, then, been amplified by 2R whole-genome duplication, followed by additional local duplications and gene loss prior to the divergence of land vertebrates and teleosts. This study delineates a possible evolutionary scenario for whole secretin-like peptide and receptor family members and may shed light on evolutionary mechanisms for expansion of a gene family with a large number of paralogs.


Subject(s)
Gene Duplication/genetics , Genomics/methods , Peptide Hormones/genetics , Receptors, G-Protein-Coupled/genetics , Secretin/genetics , Evolution, Molecular , Humans
8.
J Cell Biochem ; 114(5): 1084-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23161284

ABSTRACT

CXCL14 is a chemokine family member that is involved in various cellular responses in addition to immune cell activation. Although constitutive CXCL14 expression in normal epithelial cells may help protect against infection by activating immune systems, its expression in cancer cells has raised controversy regarding its possible role in tumorigenesis. However, the underlying mechanisms for this disparity remain unknown. Investigation of cellular CXCL14 binding properties might increase our understanding of the peptide's roles in tumorigenesis. In the present study, we found that CXCL14 binds to various cell types. Interestingly, binding to NCI-H460 cells was prevented by heparan sulfate and N-acetyl neuraminic acid. Next, we examined effect of CXCL14 binding in NCI-H460 and NCI-H23. CXCL14 enhanced proliferation and migration in NCI-H460 but had no effect on NCI-H23. A reporter gene assay with various transcription factor response elements revealed that only nuclear factor-κB (NF-κB) signaling was activated by CXCL14 in NCI-H460 cells, which was blocked by BAPTA-AM, TPCA-1, and brefeldin A. Exogenous expression of some glycoproteins such as syndecan-4, podoplanin, and CD43 in these cells enhanced CXCL14 binding and NF-κB activity. Collectively, these results demonstrate that CXCL14 binding to glycoproteins harboring heparan sulfate proteoglycans and sialic acids leads proliferation and migration of some cancer cells.


Subject(s)
Cell Movement/drug effects , Chemokines, CXC/pharmacology , Glycoproteins/metabolism , Heparitin Sulfate/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , N-Acetylneuraminic Acid/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Endocytosis/drug effects , HEK293 Cells , Humans , NF-kappa B/metabolism , Protein Binding/drug effects , Receptors, Fc/metabolism
9.
Article in English | MEDLINE | ID: mdl-23181056

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a major player in the regulation of glucose homeostasis. It acts on pancreatic beta cells to stimulate insulin secretion and on the brain to inhibit appetite. Thus, it may be a promising therapeutic agent for the treatment of type 2 diabetes mellitus and obesity. Despite the physiological and clinical importance of GLP-1, molecular interaction with the GLP-1 receptor (GLP1R) is not well understood. Particularly, the specific amino acid residues within the transmembrane helices and extracellular loops of the receptor that may confer ligand-induced receptor activation have been poorly investigated. Amino acid sequence comparisons of GLP-1 and GLP1R with their orthologs and paralogs in vertebrates, combined with biochemical approaches, are useful to determine which amino acid residues in the peptide and the receptor confer selective ligand-receptor interaction. This article reviews how the molecular evolution of GLP-1 and GLP1R contributes to the selective interaction between this ligand-receptor pair, providing critical clues for the development of potent agonists for the treatment of diabetes mellitus and obesity.

10.
Front Neurosci ; 6: 3, 2012.
Article in English | MEDLINE | ID: mdl-22291614

ABSTRACT

The neuropeptides gonadotropin-releasing hormone (GnRH) and kisspeptin (KiSS), and their receptors gonadotropin-releasing hormone receptor (GnRHR) and kisspeptin receptor (KiSSR) play key roles in vertebrate reproduction. Multiple paralogous isoforms of these genes have been identified in various vertebrate species. Two rounds of genome duplication in early vertebrates likely contributed to the generation of these paralogous genes. Genome synteny and phylogenetic analyses in a variety of vertebrate species have provided insights into the evolutionary origin of and relationship between paralogous genes. The paralogous forms of these neuropeptides and their receptors have coevolved to retain high selectivity of the ligand-receptor interaction. These paralogous forms have become subfunctionalized, neofunctionalized, or dysfunctionalized during evolution. This article reviews the evolutionary mechanism of GnRH/GnRHR and KiSS/KiSSR, and the fate of the duplicated paralogs in vertebrates.

11.
J Biol Chem ; 287(6): 3873-84, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22105074

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) play important roles in insulin secretion through their receptors, GLP1R and GIPR. Although GLP-1 and GIP are attractive candidates for treatment of type 2 diabetes and obesity, little is known regarding the molecular interaction of these peptides with the heptahelical core domain of their receptors. These core domains are important not only for specific ligand binding but also for ligand-induced receptor activation. Here, using chimeric and point-mutated GLP1R/GIPR, we determined that evolutionarily conserved amino acid residues such as Ile(196) at transmembrane helix 2, Leu(232) and Met(233) at extracellular loop 1, and Asn(302) at extracellular loop 2 of GLP1R are responsible for interaction with ligand and receptor activation. Application of chimeric GLP-1/GIP peptides together with molecular modeling suggests that His(1) of GLP-1 interacts with Asn(302) of GLP1R and that Thr(7) of GLP-1 has close contact with a binding pocket formed by Ile(196), Leu(232), and Met(233) of GLP1R. This study may provide critical clues for the development of peptide and/or nonpeptide agonists acting at GLP1R.


Subject(s)
Evolution, Molecular , Models, Molecular , Receptors, Glucagon/chemistry , Animals , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , HEK293 Cells , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
12.
Auris Nasus Larynx ; 39(4): 425-7, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21862254

ABSTRACT

Sclerosing sweat duct carcinoma (SSDC) in the nose, a rare occurrence with no specific symptoms, is frequently confused with rhinophyma or other inflammatory disease. In this report, we describe a 64-year-old woman who presented with a mass on her nasal dorsum for 1 year. Initially, the tumor had been misdiagnosed as a rhinophyma and followed-up for 1 year. She was referred due to a persistent nasal and neck mass. Deep incisional biopsy was performed to produce sufficient tissue and the histopathologic results showed SSDC. We performed wide local excision and reconstruction using radial forearm musculocutaneous free flaps with a costal cartilage framework followed by bilateral neck dissection. SSDC is seldom recognized and is frequently clinically confused with benign syringoma prior to pathologic diagnosis. Wide and deep incisional biopsy is necessary for the correct diagnosis and successful treatment of SSDC.


Subject(s)
Carcinoma, Ductal/diagnosis , Carcinoma, Skin Appendage/diagnosis , Lymph Nodes/pathology , Nose/pathology , Sweat Gland Neoplasms/diagnosis , Carcinoma, Skin Appendage/pathology , Carcinoma, Skin Appendage/surgery , Diagnostic Errors , Female , Humans , Lymphatic Metastasis , Middle Aged , Neck , Nose/surgery , Rhinophyma/diagnosis , Rhinoplasty/methods , Sweat Gland Neoplasms/pathology , Sweat Gland Neoplasms/surgery
13.
Mol Cells ; 32(4): 389-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21904878

ABSTRACT

Glucagon-like peptide-1 (GLP-1) stimulates insulin secretion from pancreatic ß-cells in a glucose-dependent manner. However, factors other than glucose that regulate the ß-cell response to GLP-1 remain poorly understood. In this study, we examined the possible involvement of insulin and receptor tyrosine kinase signaling in regulation of the GLP-1 responsiveness of ß-cells. Pretreatment of ß-cells with HNMPA, an insulin receptor inhibitor, and AG1478, an epidermal growth factor receptor inhibitor, further increased the cAMP level and Erk phosphorylation in the presence of exendin-4 (exe-4), a GLP-1 agonist. When ß-cells were exposed to a high concentration of glucose (25 mM), which stimulates insulin secretion, exe-4-induced cAMP formation declined gradually as exposure time was increased. This decreased cAMP formation was not observed in the presence of HNMPA. HNMPA was able to further increase the exe-4-induced insulin secretion when ß-cells were exposed to high glucose for 18 h. Treatment of ß-cells with insulin significantly decreased exe-4-induced cAMP formation in a dose-dependent manner. Lowering the phospho-Akt level by HNMPA or LY294002, a PI3K inhibitor, further augmented exe-4-induced cAMP formation and Erk phosphorylation. These results suggest that insulin contributes to fine-tuning of the ß-cell response to GLP-1.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Line , ErbB Receptors/antagonists & inhibitors , Glucose/metabolism , Insulin-Secreting Cells/pathology , Isoquinolines/pharmacology , Naphthalenes/pharmacology , Oncogene Protein v-akt/metabolism , Organophosphonates/pharmacology , Phosphorylation , Quinazolines/pharmacology , Rats , Receptor Cross-Talk , Receptors, AMPA/antagonists & inhibitors , Tetrazoles/pharmacology , Tyrphostins/pharmacology
14.
Gen Comp Endocrinol ; 170(1): 68-78, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21036176

ABSTRACT

Gonadotropin-releasing hormone (GnRH) and its G protein-coupled receptor, GnRHR, play a pivotal role in the control of reproduction in vertebrates. To date, many GnRH and GnRHR genes have been identified in a large variety of vertebrate species using conventional biochemical and molecular biological tools in combination with bioinformatic tools. Phylogenetic approaches, primarily based on amino acid sequence identity, make it possible to classify these multiple GnRHs and GnRHRs into several lineages. Four vertebrate GnRH lineages GnRH1, GnRH2, GnRH3, and GnRH4 (for lamprey) are well established. Four vertebrate GnRHR lineages have also been proposed-three for nonmammalian GnRHRs and mammalian GnRHR2 as well as one for mammalian GnRHR1. However, these phylogenetic analyses cannot fully explain the evolutionary origins of each lineage and the relationships among the lineages. Rapid and vast accumulation of genome sequence information for many vertebrate species, together with advances in bioinformatic tools, has allowed large-scale genome comparison to explore the origin and relationship of gene families of interest. The present review discusses the evolutionary mechanism of vertebrate GnRHs and GnRHRs based on extensive genome comparison. In this article, we focus only on vertebrate genomes because of the difficulty in comparing invertebrate and vertebrate genomes due to their marked divergence.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Receptors, LHRH/metabolism , Vertebrates/metabolism , Animals , Evolution, Molecular , Gonadotropin-Releasing Hormone/classification , Humans , Vertebrates/genetics
15.
Mol Cells ; 30(2): 149-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20799012

ABSTRACT

Glucagon like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretin hormones released in response to food intake and potentiate insulin secretion from pancreatic beta cells through their distinct yet related G protein-coupled receptors, GLP1R and GIPR. While GLP-1 and GIP exhibit similarity in their N-terminal sequence and overall alpha-helical structure, GLP-1 does not bind to GIPR and vice versa. To determine which amino acid residues of these peptide ligands are responsible for specific interaction with their respective receptors, we generated mutant GIP in which several GLP-1-specific amino acid residues were substituted for the original amino acids. The potency of the mutant ligands was examined using HEK293 cells transfected with GLP1R or GIPR expression plasmids together with a cAMP-responsive element-driven luciferase (CRE-luc) reporter plasmid. A mutated GIP peptide in which Tyr(1), Ile(7), Asp(15), and His(18) were replaced by His, Thr, Glu, and Ala, respectively, was able to activate both GLP1R and GIPR with moderate potency. Replacing the original Tyr(1) and/or Ile(7) in the N-terminal moiety of this mutant peptide allowed full activation of GIPR but not of GLP1R. However, reintroducing Asp(15) and/or His(18) in the central alpha-helical region did not significantly alter the ligand potency. These results suggest that Tyr/His(1) and Ile/Thr(7) of GIP/GLP-1 peptides confer differential ligand selectivity toward GIPR and GLP1R.


Subject(s)
Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/metabolism , Isoleucine/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Exenatide , Glucagon-Like Peptide-1 Receptor , Humans , Ligands , Molecular Sequence Data , Peptides/chemistry , Rats , Receptors, Glucagon/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Structure-Activity Relationship , Venoms/chemistry
16.
Endocrinology ; 150(6): 2837-46, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19164475

ABSTRACT

Kisspeptin and its receptor GPR54 play important roles in mammalian reproduction and cancer metastasis. Because the KiSS and GPR54 genes have been identified in a limited number of vertebrate species, mainly in mammals, the evolutionary history of these genes is poorly understood. In the present study, we have cloned multiple forms of kisspeptin and GPR54 cDNAs from a variety of vertebrate species. We found that fish have two forms of kisspeptin genes, KiSS-1 and KiSS-2, whereas Xenopus possesses three forms of kisspeptin genes, KiSS-1a, KiSS-1b, and KiSS-2. The nonmammalian KiSS-1 gene was found to be the ortholog of the mammalian KiSS-1 gene, whereas the KiSS-2 gene is a novel form, encoding a C-terminally amidated dodecapeptide in the Xenopus brain. This study is the first to identify a mature form of KiSS-2 product in the brain of any vertebrate. Likewise, fish possess two receptors, GPR54-1 and GPR54-2, whereas Xenopus carry three receptors, GPR54-1a, GPR54-1b, and GPR54-2. Sequence identity and genome synteny analyses indicate that Xenopus GPR54-1a is a human GPR54 ortholog, whereas Xenopus GPR54-1b is a fish GPR54-1 ortholog. Both kisspeptins and GPR54s were abundantly expressed in the Xenopus brain, notably in the hypothalamus, suggesting that these ligand-receptor pairs have neuroendocrine and neuromodulatory roles. Synthetic KiSS-1 and KiSS-2 peptides activated GPR54s expressed in CV-1 cells with different potencies, indicating differential ligand selectivity. These data shed new light on the molecular evolution of the kisspeptin-GPR54 system in vertebrates.


Subject(s)
Evolution, Molecular , Receptors, G-Protein-Coupled/analysis , Receptors, G-Protein-Coupled/genetics , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/genetics , Vertebrates , Amino Acid Sequence , Animals , DNA, Complementary/genetics , Female , Humans , Hypothalamus/metabolism , Kisspeptins , Lampreys , Lizards , Male , Mice , Molecular Sequence Data , Oryzias , Phylogeny , Platypus , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Sharks , Tumor Suppressor Proteins/metabolism , Xenopus , Zebrafish
17.
J Biol Chem ; 283(30): 21054-64, 2008 Jul 25.
Article in English | MEDLINE | ID: mdl-18499677

ABSTRACT

A series of small compounds acting at the orphan G protein-coupled receptor GPR92 were screened using a signaling pathway-specific reporter assay system. Lipid-derived molecules including farnesyl pyrophosphate (FPP), N-arachidonylglycine (NAG), and lysophosphatidic acid were found to activate GPR92. FPP and lysophosphatidic acid were able to activate both G(q/11)- and G(s)-mediated signaling pathways, whereas NAG activated only the G(q/11)-mediated signaling pathway. Computer-simulated modeling combined with site-directed mutagenesis of GPR92 indicated that Thr(97), Gly(98), Phe(101), and Arg(267) of GPR92 are responsible for the interaction of GPR92 with FPP and NAG. Reverse transcription-PCR analysis revealed that GPR92 mRNA is highly expressed in the dorsal root ganglia (DRG) but faint in other brain regions. Peripheral tissues including, spleen, stomach, small intestine, and kidney also expressed GPR92 mRNA. Immunohistochemical analysis revealed that GPR92 is largely co-localized with TRPV1, a nonspecific cation channel that responds to noxious heat, in mouse and human DRG. FPP and NAG increased intracellular Ca(2+) levels in cultured DRG neurons. These results suggest that FPP and NAG play a role in the sensory nervous system through activation of GPR92.


Subject(s)
Arachidonic Acids/chemistry , Glycine/analogs & derivatives , Polyisoprenyl Phosphates/chemistry , Receptors, Lysophosphatidic Acid/chemistry , Sesquiterpenes/chemistry , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Glycine/chemistry , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Inositol Phosphates/metabolism , Ligands , Lysophospholipids/chemistry , Models, Biological , Neurons/metabolism , Signal Transduction , Tissue Distribution
18.
Gen Comp Endocrinol ; 156(1): 71-82, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18158152

ABSTRACT

Mesotocin (MT) and vasotocin (VT) are the nonmammalian orthologs of mammalian oxytocin (OT) and arginine vasopressin (AVP), respectively. The OT/AVP family of peptides has arisen from gene duplication but has evolved to possess high selectivity toward their cognate receptors. The process of molecular evolution of receptors to confer high selectivity to their cognate ligands, however, is poorly understood. We constructed a series of reciprocal chimeras using a pair of bullfrog MT receptor (MTR) and VT1 receptor (VT1R) DNA fragments. Among the MTR/VT1R chimeras, the MTR chimera containing a region from transmembrane domain (TMD) VI to the carboxyl-terminal tail (C-tail) of VT1R showed an increased sensitivity to VT, while a chimeric VT1R containing TMD VI to C-tail of MTR showed an increased sensitivity to MT. Further dissection of domains using additional chimeras demonstrated that the receptor with the fragment containing extracellular loop 3 (ECL3) and ECL3-proximal TMDs VI and VII of MTR increased MT selectivity. This fragment is also important for receptor conformation that permits the signaling ability of the receptor. Particularly, the amino acids Val/Ile(6.54) in TMD VI and Pro/Glu(7.29) in ECL3 appear to be involved in this activity, since double mutation of these amino acids completely blocked signaling activity while maintaining ligand binding activity. Mutations at these residues in human OT and AVP 1a receptors markedly decreased receptor signaling activity. This study provides clues for understanding molecular coevolution of the OT/AVP peptides and their receptors with regard to receptor-ligand binding and receptor signaling activity.


Subject(s)
Protein Structure, Tertiary/physiology , Receptors, Pituitary Hormone/metabolism , Receptors, Vasopressin/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Cell Line , Chimera , Chlorocebus aethiops , DNA Fragmentation , Ligands , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary/genetics , Rana catesbeiana , Rana esculenta , Receptors, Pituitary Hormone/genetics , Receptors, Vasopressin/analysis , Receptors, Vasopressin/genetics , Transfection
19.
Mol Cell Endocrinol ; 277(1-2): 51-60, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17825479

ABSTRACT

In mammals, apelin and its G protein-coupled receptor, APJ, regulate blood pressure, intake of food and water, and cardiac contractility. In this study, we report the cloning and functional characterization of APJ in the bullfrog, Rana catesbeiana. Bullfrog APJ (bfAPJ) cDNA contains an open reading frame of 1083 nucleotides encoding a protein of 360 amino acid residues. Sequence alignment reveals 75% amino acid identity with Xenopus, 63% identity with zebrafish and 40-42% identity with mammalian APJs. RT-PCR analysis and tissue binding assay reveal high expression of bfAPJ mRNA in the brain, particularly in the hypothalamus, and moderate expression in the pituitary, testis, adrenal gland and lung. Whereas [pGlu(1)]apelin-13 did not induce CRE-luc (protein kinase A-specific reporter) and SRE-luc (protein kinase C-specific reporter) activity in cells expressing bfAPJ, this apelin-13 decreased forskolin-induced CRE-luc activity and cAMP accumulation in a pertussis toxin-sensitive manner. This study indicates that bfAPJ may couple to G(i/o). [Pro(1)]apelin-13, a synthetic apelin based on the sequence of the putative apelin gene from many non-mammalian species, activates bfAPJ with 5-10-fold greater sensitivity/affinity than mammalian apelin-13. Collectively, this study expands our understanding of the physiological roles of this receptor system in non-mammalian species.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Proline/metabolism , Rana catesbeiana/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Ligands , Male , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction
20.
Gen Comp Endocrinol ; 153(1-3): 98-107, 2007.
Article in English | MEDLINE | ID: mdl-17286976

ABSTRACT

Recently, we cloned many of the bullfrog neuropeptide G protein-coupled receptors (GPCRs), including receptors for vasotocin (VT), mesotocin, gonadotropin-releasing hormone (GnRH), neurotensin, apelin, and metastin. Bullfrog GPCRs usually have high affinity for bullfrog ligands but relatively low affinity for mammalian ligands. Reciprocally, synthetic agonists and antagonists developed based upon mammalian ligands display lower affinity at bullfrog receptors. Studies using chimeric or domain-swapped receptors indicate that the motifs responsible for differential ligand selectivity usually reside within transmembrane domain 6 (TMD6)-extracellular loop 3 (ECL3)-transmembrane domain 7 (TMD7). Triple mutation of mammalian V1aR (Phe(6.51) to Tyr, Ile(6.53) to Thr, and Pro(7.33) to Thr) increases VT affinity but greatly reduces arginine vasopressin affinity. This binding profile is similar to that of bullfrog VT1R. Changing just three amino acids in the bullfrog GnRH receptor-1 (i.e. Ser-Gln-Ser in the ECL3) to those found in the type-I mammalian GnRH receptor (i.e. Ser-Glu-Pro) reverses GnRH selectivity. In conclusion, specific receptor motifs that govern ligand selectivity can be determined by comparative molecular analyses of GPCRs and their ligands. Such analysis provides clues for understanding how GPCRs maintain high affinity to their authentic ligands.


Subject(s)
Evolution, Molecular , Ligands , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Animals , Binding Sites/genetics , Humans , Models, Biological , Neuropeptides/genetics , Receptors, LHRH/genetics , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Oxytocin/genetics , Receptors, Vasopressin/genetics , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...