43 results on '"Hjorth SA"'
Search Results
2. Protection of beta cells against cytokine-induced apoptosis by the gut microbial metabolite butyrate.
- Author
-
Jørgensen KS, Pedersen SS, Hjorth SA, Billestrup N, and Prause M
- Abstract
Type 1 diabetes (T1D) is characterized by immune cell infiltration in the islets of Langerhans, leading to the destruction of insulin-producing beta cells. This destruction is driven by secreted cytokines and cytotoxic T cells inducing apoptosis in beta cells. Butyrate, a metabolite produced by the gut microbiota, has been shown to have various health benefits, including anti-inflammatory and anti-diabetic effects. In this study, we investigated the potential protective effects of butyrate on cytokine-induced apoptosis in beta cells and explored the underlying mechanisms. Insulin-secreting INS-1E cells and isolated mouse islets were treated with interleukin-1beta (IL-1β) or a combination of IL-1β and interferon-gamma (IFN-γ) in the presence or absence of butyrate. We analyzed apoptosis, nitric oxide (NO) levels, expression of stress-related genes, and immune cell migration. Our results demonstrated that butyrate significantly attenuated cytokine-induced apoptosis in both INS-1E cells and mouse islets, accompanied by a reduction in NO levels. Butyrate also decreased the expression of endoplasmic reticulum (ER) stress markers such as Chop, phosphorylated eIF2α and Atf4, as well as some pro-apoptotic genes including Dp5 and Puma. Butyrate reduced the cytokine-induced expression of the chemokine genes Cxcl1 and Cxcl10 in mouse islets, as well as the chemotactic activity of THP-1 monocytes toward conditioned media from IL-1β-exposed islets. In conclusion, these findings indicate that butyrate protects beta cells from cytokine-induced apoptosis and ER stress, suggesting its potential as a therapeutic agent to prevent beta cell destruction in T1D., (© 2024 Federation of European Biochemical Societies.)
- Published
- 2024
- Full Text
- View/download PDF
3. Migration mediated by the oxysterol receptor GPR183 depends on arrestin coupling but not receptor internalization.
- Author
-
Kjær VMS, Daugvilaite V, Stepniewski TM, Madsen CM, Jørgensen AS, Bhuskute KR, Inoue A, Ulven T, Benned-Jensen T, Hjorth SA, Hjortø GM, Moo EV, Selent J, and Rosenkilde MM
- Subjects
- Ligands, beta-Arrestins metabolism, beta-Arrestin 1 genetics, beta-Arrestin 1 metabolism, Endocytosis, Arrestin metabolism, Arrestins genetics, Arrestins metabolism
- Abstract
The chemotactic G protein-coupled receptor GPR183 and its most potent endogenous oxysterol ligand 7α,25-dihydroxycholesterol (7α,25-OHC) are important for immune cell positioning in secondary lymphoid tissues. This receptor-ligand pair is associated with various diseases, in some cases contributing favorably and in other cases adversely, making GPR183 an attractive target for therapeutic intervention. We investigated the mechanisms underlying GPR183 internalization and the role of internalization in the main biological function of the receptor, chemotaxis. We found that the C terminus of the receptor was important for ligand-induced internalization but less so for constitutive (ligand-independent) internalization. β-arrestin potentiated ligand-induced internalization but was not required for ligand-induced or constitutive internalization. Caveolin and dynamin were the main mediators of both constitutive and ligand-induced receptor internalization in a mechanism independent of G protein activation. Clathrin-mediated endocytosis also contributed to constitutive GPR183 internalization in a β-arrestin-independent manner, suggesting the existence of different pools of surface-localized GPR183. Chemotaxis mediated by GPR183 depended on receptor desensitization by β-arrestins but could be uncoupled from internalization, highlighting an important biological role for the recruitment of β-arrestin to GPR183. The role of distinct pathways in internalization and chemotaxis may aid in the development of GPR183-targeting drugs for specific disease contexts.
- Published
- 2023
- Full Text
- View/download PDF
4. Self-organized metabotyping of obese individuals identifies clusters responding differently to bariatric surgery.
- Author
-
Lappa D, Meijnikman AS, Krautkramer KA, Olsson LM, Aydin Ö, Van Rijswijk AS, Acherman YIZ, De Brauw ML, Tremaroli V, Olofsson LE, Lundqvist A, Hjorth SA, Ji B, Gerdes VEA, Groen AK, Schwartz TW, Nieuwdorp M, Bäckhed F, and Nielsen J
- Subjects
- Humans, Obesity surgery, Adipose Tissue, Algorithms, Diabetes Mellitus, Type 2 surgery, Bariatric Surgery
- Abstract
Weight loss through bariatric surgery is efficient for treatment or prevention of obesity related diseases such as type 2 diabetes and cardiovascular disease. Long term weight loss response does, however, vary among patients undergoing surgery. Thus, it is difficult to identify predictive markers while most obese individuals have one or more comorbidities. To overcome such challenges, an in-depth multiple omics analyses including fasting peripheral plasma metabolome, fecal metagenome as well as liver, jejunum, and adipose tissue transcriptome were performed for 106 individuals undergoing bariatric surgery. Machine leaning was applied to explore the metabolic differences in individuals and evaluate if metabolism-based patients' stratification is related to their weight loss responses to bariatric surgery. Using Self-Organizing Maps (SOMs) to analyze the plasma metabolome, we identified five distinct metabotypes, which were differentially enriched for KEGG pathways related to immune functions, fatty acid metabolism, protein-signaling, and obesity pathogenesis. The gut metagenome of the most heavily medicated metabotypes, treated simultaneously for multiple cardiometabolic comorbidities, was significantly enriched in Prevotella and Lactobacillus species. This unbiased stratification into SOM-defined metabotypes identified signatures for each metabolic phenotype and we found that the different metabotypes respond differently to bariatric surgery in terms of weight loss after 12 months. An integrative framework that utilizes SOMs and omics integration was developed for stratifying a heterogeneous bariatric surgery cohort. The multiple omics datasets described in this study reveal that the metabotypes are characterized by a concrete metabolic status and different responses in weight loss and adipose tissue reduction over time. Our study thus opens a path to enable patient stratification and hereby allow for improved clinical treatments., Competing Interests: The authors declare no conflict of interest., (Copyright: © 2023 Lappa et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
5. Type 2 diabetes is associated with increased circulating levels of 3-hydroxydecanoate activating GPR84 and neutrophil migration.
- Author
-
Mikkelsen RB, Arora T, Trošt K, Dmytriyeva O, Jensen SK, Meijnikman AS, Olofsson LE, Lappa D, Aydin Ö, Nielsen J, Gerdes V, Moritz T, van de Laar A, de Brauw M, Nieuwdorp M, Hjorth SA, Schwartz TW, and Bäckhed F
- Abstract
Obesity and diabetes are associated with inflammation and altered plasma levels of several metabolites, which may be involved in disease progression. Some metabolites can activate G protein-coupled receptors (GPCRs) expressed on immune cells where they can modulate metabolic inflammation. Here, we find that 3-hydroxydecanoate is enriched in the circulation of obese individuals with type 2 diabetes (T2D) compared with nondiabetic controls. Administration of 3-hydroxydecanoate to mice promotes immune cell recruitment to adipose tissue, which was associated with adipose inflammation and increased fasting insulin levels. Furthermore, we demonstrate that 3-hydroxydecanoate stimulates migration of primary human and mouse neutrophils, but not monocytes, through GPR84 and Gα
i signaling in vitro . Our findings indicate that 3-hydroxydecanoate is a T2D-associated metabolite that increases inflammatory responses and may contribute to the chronic inflammation observed in diabetes., Competing Interests: M.N. is in the SAB of Caelus Health and Kaleido Biosciences; however, none of these are relevant for the current manuscript. F.B. is founder and holds equity of Implexion Pharma AB and receives research funding from Biogaia AB, both of which are unrelated to this study., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
6. Selective release of gastrointestinal hormones induced by an orally active GPR39 agonist.
- Author
-
Grunddal KV, Diep TA, Petersen N, Tough IR, Skov LJ, Liu L, Buijink JA, Mende F, Jin C, Jepsen SL, Sørensen LME, Achiam MP, Strandby RB, Bach A, Hartmann B, Frimurer TM, Hjorth SA, Bouvier M, Cox H, and Holst B
- Subjects
- Animals, Appetite Regulation, Bariatric Surgery, Body Weight, Eating, Enteroendocrine Cells, Gastric Inhibitory Polypeptide pharmacology, Ghrelin metabolism, Glucagon-Like Peptide 1 metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Obesity metabolism, Peptide YY metabolism, Receptors, G-Protein-Coupled genetics, Receptors, Gastrointestinal Hormone, Weight Loss, Gastrointestinal Hormones metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled metabolism
- Abstract
Objectives: Obesity is a complex disease associated with a high risk of comorbidities. Gastric bypass surgery, an invasive procedure with low patient eligibility, is currently the most effective intervention that achieves sustained weight loss. This beneficial effect is attributed to alterations in gut hormone signaling. An attractive alternative is to pharmacologically mimic the effects of bariatric surgery by targeting several gut hormonal axes. The G protein-coupled receptor 39 (GPR39) expressed in the gastrointestinal tract has been shown to mediate ghrelin signaling and control appetite, food intake, and energy homeostasis, but the broader effect on gut hormones is largely unknown. A potent and efficacious GPR39 agonist (Cpd1324) was recently discovered, but the in vivo function was not addressed. Herein we studied the efficacy of the GPR39 agonist, Cpd1324, on metabolism and gut hormone secretion., Methods: Body weight, food intake, and energy expenditure in GPR39 agonist-treated mice and GPR39 KO mice were studied in calorimetric cages. Plasma ghrelin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) levels were measured. Organoids generated from murine and human small intestine and mouse colon were used to study GLP-1 and PYY release. Upon GPR39 agonist administration, dynamic changes in intracellular GLP-1 content were studied via immunostaining and changes in ion transport across colonic mucosa were monitored in Ussing chambers. The G protein activation underlying GPR39-mediated selective release of gut hormones was studied using bioluminescence resonance energy transfer biosensors., Results: The GPR39 KO mice displayed a significantly increased food intake without corresponding increases in respiratory exchange ratios or energy expenditure. Oral administration of a GPR39 agonist induced an acute decrease in food intake and subsequent weight loss in high-fat diet (HFD)-fed mice without affecting their energy expenditure. The tool compound, Cpd1324, increased GLP-1 secretion in the mice as well as in mouse and human intestinal organoids, but not in GPR39 KO mouse organoids. In contrast, the GPR39 agonist had no effect on PYY or GIP secretion. Transepithelial ion transport was acutely affected by GPR39 agonism in a GLP-1- and calcitonin gene-related peptide (CGRP)-dependent manner. Analysis of Cpd1324 signaling properties showed activation of Gα
q and Gαi/o signaling pathways in L cells, but not Gαs signaling., Conclusions: The GPR39 agonist described in this study can potentially be used by oral administration as a weight-lowering agent due to its stimulatory effect on GLP-1 secretion, which is most likely mediated through a unique activation of Gα subunits. Thus, GPR39 agonism may represent a novel approach to effectively treat obesity through selective modulation of gastrointestinal hormonal axes., Competing Interests: Conflicts of interest M.B. is the chairman of the scientific advisory board of Domain Therapeutics to which some of the biosensors used in this study are licensed for commercial use. The biosensors are freely available for academic research from M.B. The rest of the authors declare no conflicts of interest., (Copyright © 2021. Published by Elsevier GmbH.)- Published
- 2021
- Full Text
- View/download PDF
7. Disruption of GPR35 Signaling in Bone Marrow-Derived Cells Does Not Influence Vascular Inflammation and Atherosclerosis in Hyperlipidemic Mice.
- Author
-
Baumgartner R, Casagrande FB, Mikkelsen RB, Berg M, Polyzos KA, Forteza MJ, Arora A, Schwartz TW, Hjorth SA, and Ketelhuth DFJ
- Abstract
G-protein-coupled receptor-35 (GPR35) has been identified as a receptor for the tryptophan metabolite kynurenic acid (KynA) and suggested to modulate macrophage polarization in metabolic tissues. Whether GPR35 can influence vascular inflammation and atherosclerosis has however never been tested. Lethally irradiated LdlrKO mice were randomized to receive GPR35KO or wild type (WT) bone marrow transplants and fed a high cholesterol diet for eight weeks to develop atherosclerosis. GPR35KO and WT chimeric mice presented no difference in the size of atherosclerotic lesions in the aortic arch (2.37 ± 0.58% vs. 1.95 ± 0.46%, respectively) or in the aortic roots (14.77 ± 3.33% vs. 11.57 ± 2.49%, respectively). In line with these data, no changes in the percentage of VCAM-1+, IA
b + cells, and CD3+ T cells, as well as alpha smooth muscle cell actin expression, was observed between groups. Interestingly, the GPR35KO group presented a small but significant increase in CD68+ macrophage infiltration in the plaque. However, in vitro culture experiments using bone marrow-derived macrophages from both groups indicated that GPR35 plays no role in modulating the secretion of major inflammatory cytokines. Our study indicates that GPR35 expression does not play a direct role in macrophage activation, vascular inflammation, and the development of atherosclerosis.- Published
- 2021
- Full Text
- View/download PDF
8. Extracellular succinate hyperpolarizes M2 macrophages through SUCNR1/GPR91-mediated Gq signaling.
- Author
-
Trauelsen M, Hiron TK, Lin D, Petersen JE, Breton B, Husted AS, Hjorth SA, Inoue A, Frimurer TM, Bouvier M, O'Callaghan CA, and Schwartz TW
- Subjects
- Arrestins metabolism, Female, GTP-Binding Protein alpha Subunits, Gi-Go metabolism, Gene Expression Regulation, Gene Ontology, HEK293 Cells, Humans, Ligands, Macrophages immunology, Male, Models, Biological, Protein Subunits metabolism, Receptors, G-Protein-Coupled agonists, Receptors, G-Protein-Coupled genetics, Transcriptional Activation genetics, Type C Phospholipases metabolism, Extracellular Space chemistry, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Macrophages metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Succinic Acid metabolism
- Abstract
Succinate functions both as a classical TCA cycle metabolite and an extracellular metabolic stress signal sensed by the mainly Gi-coupled succinate receptor SUCNR1. In the present study, we characterize and compare effects and signaling pathways activated by succinate and both classes of non-metabolite SUCNR1 agonists. By use of specific receptor and pathway inhibitors, rescue in G-protein-depleted cells and monitoring of receptor G protein activation by BRET, we identify Gq rather than Gi signaling to be responsible for SUCNR1-mediated effects on basic transcriptional regulation. Importantly, in primary human M2 macrophages, in which SUCNR1 is highly expressed, we demonstrate that physiological concentrations of extracellular succinate act through SUCNR1-activated Gq signaling to efficiently regulate transcription of immune function genes in a manner that hyperpolarizes their M2 versus M1 phenotype. Thus, sensing of stress-induced extracellular succinate by SUCNR1 is an important transcriptional regulator in human M2 macrophages through Gq signaling., Competing Interests: Declaration of interests M.B. is the chair of the scientific advisory board for Domain Therapeutics (DT), and some of the BRET-based biosensors used in the present study were licensed to DT for commercial use. The other authors declare no competing interests., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
9. Microbial regulation of enteroendocrine cells.
- Author
-
Arora T, Vanslette AM, Hjorth SA, and Bäckhed F
- Subjects
- Enteroendocrine Cells metabolism, Glucagon-Like Peptide 1 metabolism, Humans, Receptors, G-Protein-Coupled metabolism, Diabetes Mellitus, Type 2 drug therapy, Gastrointestinal Microbiome
- Abstract
There has been an enormous interest to investigate impact of gut microbiota on host physiology over the past decade. To further understand its role at organismal level, it is important to delineate host-microbiota interaction at tissue and cell level. Diet, antibiotics, disease, or surgery produce shifts in composition of the gut microbiota that further alter levels of microbial-derived metabolites. Enteroendocrine cells (EECs) are specialized hormone-producing cells in the gut epithelium that sense changes in the intestinal milieu through chemosensing G protein-coupled receptors. Accordingly, microbial metabolites interact with the EECs to stimulate or suppress hormone secretion, which act through endocrine and paracrine signaling to regulate local intestinal and diverse physiological functions and impact overall host metabolism. The remarkable success of glucagon-like peptide-1-based drugs for treatment of type 2 diabetes and obesity highlights the relevance to investigate microbial regulation of EECs to tackle metabolic diseases through novel microbiota-based therapies., Competing Interests: Declaration of interests F.B. receives research support from Biogaia AB and is founder and shareholder in Implexion pharma AB. All other authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
10. Evidence that a deviation in the kynurenine pathway aggravates atherosclerotic disease in humans.
- Author
-
Baumgartner R, Berg M, Matic L, Polyzos KP, Forteza MJ, Hjorth SA, Schwartz TW, Paulsson-Berne G, Hansson GK, Hedin U, and Ketelhuth DFJ
- Subjects
- Down-Regulation, Humans, Inflammation metabolism, Kynurenic Acid metabolism, Kynurenine blood, Macrophages metabolism, Plaque, Atherosclerotic metabolism, Tryptophan blood, Up-Regulation, Carotid Artery Diseases metabolism, Kynurenine metabolism, Tryptophan metabolism
- Abstract
Background: The metabolism of tryptophan (Trp) along the kynurenine pathway has been shown to carry strong immunoregulatory properties. Several experimental studies indicate that this pathway is a major regulator of vascular inflammation and influences atherogenesis. Knowledge of the role of this pathway in human atherosclerosis remains incomplete., Objectives: In this study, we performed a multiplatform analysis of tissue samples, in vitro and in vivo functional assays to elucidate the potential role of the kynurenine pathway in human atherosclerosis., Methods and Results: Comparison of transcriptomic data from carotid plaques and control arteries revealed an upregulation of enzymes within the quinolinic branch of the kynurenine pathway in the disease state, whilst the branch leading to the formation of kynurenic acid (KynA) was downregulated. Further analyses indicated that local inflammatory responses are closely tied to the deviation of the kynurenine pathway in the vascular wall. Analysis of cerebrovascular symptomatic and asymptomatic carotid stenosis data showed that the downregulation of KynA branch enzymes and reduced KynA production were associated with an increased probability of patients to undergo surgery due to an unstable disease. In vitro, we showed that KynA-mediated signalling through aryl hydrocarbon receptor (AhR) is a major regulator of human macrophage activation. Using a mouse model of peritoneal inflammation, we showed that KynA inhibits leukocyte recruitment., Conclusions: We have found that a deviation in the kynurenine pathway is associated with an increased probability of developing symptomatic unstable atherosclerotic disease. Our study suggests that KynA-mediated signalling through AhR is an important mechanism involved in the regulation of vascular inflammation., (© 2020 The Authors. Journal of Internal Medicine published by John Wiley & Sons Ltd on behalf of Association for Publication of The Journal of Internal Medicine.)
- Published
- 2021
- Full Text
- View/download PDF
11. The aromatic amino acid sensor GPR142 controls metabolism through balanced regulation of pancreatic and gut hormones.
- Author
-
Rudenko O, Shang J, Munk A, Ekberg JP, Petersen N, Engelstoft MS, Egerod KL, Hjorth SA, Wu M, Feng Y, Zhou YP, Mokrosinski J, Thams P, Reimann F, Gribble F, Rehfeld JF, Holst JJ, Treebak JT, Howard AD, and Schwartz TW
- Subjects
- Amino Acids, Aromatic metabolism, Animals, Blood Glucose metabolism, Glucagon metabolism, Glucagon-Like Peptide 1 metabolism, Glucagon-Like Peptide-1 Receptor metabolism, Glucagon-Secreting Cells metabolism, Glucose metabolism, Insulin metabolism, Insulin-Secreting Cells metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, Rats, Rats, Zucker, Receptor-Interacting Protein Serine-Threonine Kinase 2 metabolism, Receptors, G-Protein-Coupled biosynthesis, Receptors, Gastrointestinal Hormone metabolism, Receptors, Glucagon metabolism, Tryptophan metabolism, Islets of Langerhans metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Objectives: GPR142, which is highly expressed in pancreatic islets, has recently been deorphanized as a receptor for aromatic amino acids; however, its physiological role and pharmacological potential is unclear., Methods and Results: We find that GPR142 is expressed not only in β- but also in α-cells of the islets as well as in enteroendocrine cells, and we confirm that GPR142 is a highly selective sensor of essential aromatic amino acids, in particular Trp and oligopeptides with N-terminal Trp. GPR142 knock-out mice displayed a very limited metabolic phenotype but demonstrated that L-Trp induced secretion of pancreatic and gut hormones is mediated through GPR142 but that the receptor is not required for protein-induced hormone secretion. A synthetic GPR142 agonist stimulated insulin and glucagon as well as GIP, CCK, and GLP-1 secretion. In particular, GIP secretion was sensitive to oral administration of the GPR142 agonist an effect which in contrast to the other hormones was blocked by protein load. Oral administration of the GPR142 agonist increased [
3 H]-2-deoxyglucose uptake in muscle and fat depots mediated through insulin action while it lowered liver glycogen conceivably mediated through glucagon, and, consequently, it did not lower total blood glucose. Nevertheless, acute administration of the GPR142 agonist strongly improved oral glucose tolerance in both lean and obese mice as well as Zucker fatty rat. Six weeks in-feed chronic treatment with the GPR142 agonist did not affect body weight in DIO mice, but increased energy expenditure and carbohydrate utilization, lowered basal glucose, and improved insulin sensitivity., Conclusions: GPR142 functions as a sensor of aromatic amino acids, controlling GIP but also CCK and GLP-1 as well as insulin and glucagon in the pancreas. GPR142 agonists could have novel interesting potential in modifying metabolism through a balanced action of gut hormones as well as both insulin and glucagon., (Copyright © 2018. Published by Elsevier GmbH.)- Published
- 2019
- Full Text
- View/download PDF
12. Receptor structure-based discovery of non-metabolite agonists for the succinate receptor GPR91.
- Author
-
Trauelsen M, Rexen Ulven E, Hjorth SA, Brvar M, Monaco C, Frimurer TM, and Schwartz TW
- Subjects
- Cells, Cultured, Drug Discovery methods, HEK293 Cells, Humans, Protein Binding, Quantitative Structure-Activity Relationship, Receptors, G-Protein-Coupled chemistry, Receptors, G-Protein-Coupled metabolism, Small Molecule Libraries chemistry, Molecular Docking Simulation, Receptors, G-Protein-Coupled agonists, Small Molecule Libraries pharmacology
- Abstract
Objective: Besides functioning as an intracellular metabolite, succinate acts as a stress-induced extracellular signal through activation of GPR91 (SUCNR1) for which we lack suitable pharmacological tools., Methods and Results: Here we first determined that the cis conformation of the succinate backbone is preferred and that certain backbone modifications are allowed for GPR91 activation. Through receptor modeling over the X-ray structure of the closely related P2Y1 receptor, we discovered that the binding pocket is partly occupied by a segment of an extracellular loop and that succinate therefore binds in a very different mode than generally believed. Importantly, an empty side-pocket is identified next to the succinate binding site. All this information formed the basis for a substructure-based search query, which, combined with molecular docking, was used in virtual screening of the ZINC database to pick two serial mini-libraries of a total of only 245 compounds from which sub-micromolar, selective GPR91 agonists of unique structures were identified. The best compounds were backbone-modified succinate analogs in which an amide-linked hydrophobic moiety docked into the side-pocket next to succinate as shown by both loss- and gain-of-function mutagenesis. These compounds displayed GPR91-dependent activity in altering cytokine expression in human M2 macrophages similar to succinate, and importantly were devoid of any effect on the major intracellular target, succinate dehydrogenase., Conclusions: These novel, synthetic non-metabolite GPR91 agonists will be valuable both as pharmacological tools to delineate the GPR91-mediated functions of succinate and as leads for the development of GPR91-targeted drugs to potentially treat low grade metabolic inflammation and diabetic complications such as retinopathy and nephropathy., (Copyright © 2017 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
13. GPCR-Mediated Signaling of Metabolites.
- Author
-
Husted AS, Trauelsen M, Rudenko O, Hjorth SA, and Schwartz TW
- Subjects
- Animals, Enteroendocrine Cells metabolism, Gastrointestinal Microbiome, Humans, Ligands, Metabolome, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
In addition to their bioenergetic intracellular function, several classical metabolites act as extracellular signaling molecules activating cell-surface G-protein-coupled receptors (GPCRs), similar to hormones and neurotransmitters. "Signaling metabolites" generated from nutrients or by gut microbiota target primarily enteroendocrine, neuronal, and immune cells in the lamina propria of the gut mucosa and the liver and, through these tissues, the rest of the body. In contrast, metabolites from the intermediary metabolism act mainly as metabolic stress-induced autocrine and paracrine signals in adipose tissue, the liver, and the endocrine pancreas. Importantly, distinct metabolite GPCRs act as efficient pro- and anti-inflammatory regulators of key immune cells, and signaling metabolites may thus function as important drivers of the low-grade inflammation associated with insulin resistance and obesity. The concept of key metabolites as ligands for specific GPCRs has broadened our understanding of metabolic signaling significantly and provides a number of novel potential drug targets., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
14. Cutting Edge: identification of neutrophil PGLYRP1 as a ligand for TREM-1.
- Author
-
Read CB, Kuijper JL, Hjorth SA, Heipel MD, Tang X, Fleetwood AJ, Dantzler JL, Grell SN, Kastrup J, Wang C, Brandt CS, Hansen AJ, Wagtmann NR, Xu W, and Stennicke VW
- Subjects
- Humans, Immunoprecipitation, Ligands, Surface Plasmon Resonance, Triggering Receptor Expressed on Myeloid Cells-1, Cytokines immunology, Immunity, Innate immunology, Membrane Glycoproteins immunology, Neutrophils immunology, Receptors, Immunologic immunology
- Abstract
Triggering receptor expressed on myeloid cells (TREM)-1 is an orphan receptor implicated in innate immune activation. Inhibition of TREM-1 reduces sepsis in mouse models, suggesting a role for it in immune responses triggered by bacteria. However, the absence of an identified ligand has hampered a full understanding of TREM-1 function. We identified complexes between peptidoglycan recognition protein 1 (PGLYRP1) and bacterially derived peptidoglycan that constitute a potent ligand capable of binding TREM-1 and inducing known TREM-1 functions. Interestingly, multimerization of PGLYRP1 bypassed the need for peptidoglycan in TREM-1 activation, demonstrating that the PGLYRP1/TREM-1 axis can be activated in the absence of bacterial products. The role for PGLYRP1 as a TREM-1 activator provides a new mechanism by which bacteria can trigger myeloid cells, linking two known, but previously unrelated, pathways in innate immunity., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
15. Crystal structure of interleukin-21 receptor (IL-21R) bound to IL-21 reveals that sugar chain interacting with WSXWS motif is integral part of IL-21R.
- Author
-
Hamming OJ, Kang L, Svensson A, Karlsen JL, Rahbek-Nielsen H, Paludan SR, Hjorth SA, Bondensgaard K, and Hartmann R
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Binding Sites, Crystallography, X-Ray, Glycosylation, Humans, Interleukins genetics, Mannose metabolism, Models, Molecular, Molecular Sequence Data, Protein Binding, Protein Structure, Secondary, Receptors, Interleukin-21 genetics, Interleukins chemistry, Interleukins metabolism, Receptors, Interleukin-21 chemistry, Receptors, Interleukin-21 metabolism
- Abstract
IL-21 is a class I cytokine that exerts pleiotropic effects on both innate and adaptive immune responses. It signals through a heterodimeric receptor complex consisting of the IL-21 receptor (IL-21R) and the common γ-chain. A hallmark of the class I cytokine receptors is the class I cytokine receptor signature motif (WSXWS). The exact role of this motif has not been determined yet; however, it has been implicated in diverse functions, including ligand binding, receptor internalization, proper folding, and export, as well as signal transduction. Furthermore, the WXXW motif is known to be a consensus sequence for C-mannosylation. Here, we present the crystal structure of IL-21 bound to IL-21R and reveal that the WSXWS motif of IL-21R is C-mannosylated at the first tryptophan. We furthermore demonstrate that a sugar chain bridges the two fibronectin domains that constitute the extracellular domain of IL-21R and anchors at the WSXWS motif through an extensive hydrogen bonding network, including mannosylation. The glycan thus transforms the V-shaped receptor into an A-frame. This finding offers a novel structural explanation of the role of the class I cytokine signature motif.
- Published
- 2012
- Full Text
- View/download PDF
16. Rational design of interleukin-21 antagonist through selective elimination of the gammaC binding epitope.
- Author
-
Kang L, Bondensgaard K, Li T, Hartmann R, and Hjorth SA
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Binding Sites, Cell Line, Epitopes chemistry, Epitopes genetics, Epitopes metabolism, Humans, In Vitro Techniques, Interleukin-2 chemistry, Interleukin-2 genetics, Interleukin-4 chemistry, Interleukin-4 genetics, Interleukins chemistry, Interleukins genetics, Interleukins metabolism, Models, Molecular, Molecular Sequence Data, Multiprotein Complexes, Mutagenesis, Site-Directed, Protein Interaction Domains and Motifs, Receptors, Interleukin-21 chemistry, Receptors, Interleukin-21 genetics, Receptors, Interleukin-21 metabolism, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Structural Homology, Protein, Drug Design, Interleukins antagonists & inhibitors
- Abstract
The cytokine interleukin (IL)-21 exerts pleiotropic effects acting through innate as well as adaptive immune responses. The activities of IL-21 are mediated through binding to its cognate receptor complex composed of the IL-21 receptor private chain (IL-21Ralpha) and the common gamma-chain (gammaC), the latter being shared by IL-2, IL-4, IL-7, IL-9, and IL-15. The binding energy of the IL-21 ternary complex is predominantly provided by the high affinity interaction between IL-21 and IL-21Ralpha, whereas the interaction between IL-21 and gammaC, albeit essential for signaling, is rather weak. The design of IL-21 analogues, which have lost most or all affinity toward the signaling gammaC chain, while simultaneously maintaining a tight interaction with the private chain, would in theory represent candidates for IL-21 antagonists. We predicted the IL-21 residues, which compose the gammaC binding epitope using homology modeling and alignment with the related cytokines, IL-2 and IL-4. Next we systematically analyzed the predicted binding epitope by a mutagenesis study. Indeed two mutants, which have significantly impaired gammaC affinity with undiminished IL-21Ralpha affinity, were successfully identified. Functional studies confirmed that these two novel hIL-21 double mutants do act as hIL-21 antagonists.
- Published
- 2010
- Full Text
- View/download PDF
17. The existence of multiple conformers of interleukin-21 directs engineering of a superpotent analogue.
- Author
-
Bondensgaard K, Breinholt J, Madsen D, Omkvist DH, Kang L, Worsaae A, Becker P, Schiødt CB, and Hjorth SA
- Subjects
- Amino Acid Sequence, Cell Line, Crystallography, X-Ray, Humans, Magnetic Resonance Spectroscopy methods, Models, Molecular, Molecular Conformation, Molecular Sequence Data, Protein Conformation, Protein Engineering, Protein Structure, Secondary, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Temperature, Interleukins chemistry
- Abstract
The high resolution three-dimensional structure of human interleukin (hIL)-21 has been resolved by heteronuclear NMR spectroscopy. Overall, the hIL-21 structure is dominated by a well defined central four-helical bundle, arranged in an up-up-down-down topology, as observed for other cytokines. A segment of the hIL-21 molecule that includes the third helical segment, helix C, is observed to exist in two distinct and interchangeable states. In one conformer, the helix C segment is presented in a regular, alpha-helical conformation, whereas in the other conformer, this segment is largely disordered. A structure-based sequence alignment of hIL-21 with receptor complexes of the related cytokines, interleukin-2 and -4, implied that this particular segment is involved in receptor binding. An hIL-21 analog was designed to stabilize the region around helix C through the introduction of a segment grafted from hIL-4. This novel hIL-21 analog was demonstrated to exhibit a 10-fold increase in potency in a cellular assay.
- Published
- 2007
- Full Text
- View/download PDF
18. Typical Danish Caucasian type 2 diabetic patients do not commonly carry genetic variants in GIP and GLP-1 encoding regions of the proGIP and proglucagon genes.
- Author
-
Ørskov C, Hjorth SA, Jensen AD, and Vilsbøll T
- Subjects
- Aged, Base Sequence, DNA analysis, Denmark, Diabetes Mellitus, Type 2 metabolism, Female, Gastric Inhibitory Polypeptide metabolism, Genetic Predisposition to Disease, Glucagon metabolism, Glucagon-Like Peptide 1, Glucagon-Like Peptide-1 Receptor, Humans, Male, Middle Aged, Molecular Sequence Data, Peptide Fragments metabolism, Polymerase Chain Reaction, Predictive Value of Tests, Protein Precursors metabolism, Receptors, Gastrointestinal Hormone metabolism, Receptors, Glucagon metabolism, Sequence Analysis, DNA, White People, Diabetes Mellitus, Type 2 genetics, Gastric Inhibitory Polypeptide genetics, Glucagon genetics, Peptide Fragments genetics, Phenotype, Protein Precursors genetics
- Abstract
Background: The enteroinsular-axis is abnormal in type 2 diabetics, which contributes to the diabetic phenotype. The effect of the incretin hormone gastric inhibitory polypeptide (GIP) and the secretion of the incretin hormone glucagon-like peptide-1 (GLP-1) are thus greatly diminished. The explanation for these changes could be changes in the structure of either of the hormones or their receptors. Thus, the aim of this study was to study the occurrence of genetic variants in the GIP and GLP-1 encoding regions of the proGIP and proglucagon genes in type 2 diabetic patients and matched control subjects., Methods and Results: Genomic DNA was extracted from buffy coats from 12 Caucasian type 2 diabetics and 12 healthy subjects, matched with respect to sex, age and BMI. The GIP and GLP-1 sequences were amplified using specific primers using the polymerase chain reaction (PCR). The amplified products were then sequenced. No germ-line mutations were identified in the GIP and the GLP-1 encoding regions of the proGIP and proglucagon genes in either the type 2 diabetic or the control subjects., Conclusions: The perturbed incretin effect in type 2 diabetics is not commonly caused by genetic variants in either the GIP or the GLP-1 encoding genes in type 2 diabetics.
- Published
- 2004
- Full Text
- View/download PDF
19. Interaction of a non-peptide agonist with angiotensin II AT1 receptor mutants.
- Author
-
Costa-Neto CM, Miyakawa AA, Pesquero JB, Oliveira L, Hjorth SA, Schwartz TW, and Paiva AC
- Subjects
- Amino Acid Sequence physiology, Angiotensin II metabolism, Angiotensin II pharmacology, Animals, Biphenyl Compounds pharmacology, COS Cells, Chlorocebus aethiops, Dose-Response Relationship, Drug, Imidazoles pharmacology, Molecular Sequence Data, Rats, Receptor, Angiotensin, Type 1, Receptors, Angiotensin chemistry, Receptors, Angiotensin genetics, Biphenyl Compounds metabolism, Imidazoles metabolism, Mutagenesis, Site-Directed, Receptors, Angiotensin agonists, Receptors, Angiotensin metabolism
- Abstract
To identify residues of the rat AT1A angiotensin II receptor involved with signal transduction and binding of the non-peptide agonist L-162,313 (5,7-dimethyl-2-ethyl-3-[[4-[2(n-butyloxycarbonylsulfonamido)-5-isobutyl-3-thienyl]phenyl]methyl]imidazol[4,5,6]-pyridine) we have performed ligand binding and inositol phosphate turnover assays in COS-7 cells transiently transfected with the wild-type and mutant forms of the receptor. Mutant receptors bore modifications in the extracellular region: T88H, Y92H, G1961, G196W, and D278E. Compound L-162,313 displaced [125I]-Sar1,Leu8-AngII from the mutants G196I and G196W with IC50 values similar to that of the wild-type. The affinity was, however, slightly affected by the D278E mutation and more significantly by the T88H and Y92H mutations. In inositol phosphate turnover assays, the ability of L-162,313 to trigger the activation cascade was compared with that of angiotensin II. These assays showed that the G196W mutant reached a relative maximum activation exceeding that of the wild-type receptor; the efficacy was slightly reduced in the G1961 mutant and further reduced in the T88H, Y92H, and D278E mutants. Our data suggest that residues of the extracellular domain of the AT1 receptor are involved in the binding of the non-peptide ligand, or in a general receptor activation phenomenon that involves conformational modifications for a preferential binding of agonists or antagonists.
- Published
- 2002
- Full Text
- View/download PDF
20. Constitutive activation of tethered-peptide/corticotropin-releasing factor receptor chimeras.
- Author
-
Nielsen SM, Nielsen LZ, Hjorth SA, Perrin MH, and Vale WW
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Corticotropin-Releasing Hormone antagonists & inhibitors, Corticotropin-Releasing Hormone pharmacology, Humans, Ligands, Models, Molecular, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments metabolism, Peptide Fragments pharmacology, Protein Structure, Secondary, Receptors, Corticotropin-Releasing Hormone chemistry, Receptors, Corticotropin-Releasing Hormone genetics, Receptors, Thrombin chemistry, Receptors, Thrombin genetics, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Transfection, Urocortins, Receptors, Corticotropin-Releasing Hormone physiology, Receptors, Thrombin physiology
- Abstract
Constitutive activity, or ligand-independent activity, of mutant G protein-coupled receptors (GPCRs) has been described extensively and implicated in the pathology of many diseases. Using the corticotropin-releasing factor (CRF) receptor and the thrombin receptor as a model, we present a ligand-dependent constitutive activation of a GPCR. A chimera in which the N-terminal domain of the CRF receptor is replaced by the amino-terminal 16 residues of CRF displays significant levels of constitutive activation. The activity, as measured by intracellular levels of cAMP, is blocked in a dose-dependent manner by the nonpeptide antagonist antalarmin. These results support a propinquity effect in CRF receptor activation, in which the amino-terminal portion of the CRF peptide is presented to the body of the receptor in the proper proximity for activation. This form of ligand-dependent constitutive activation may be of general applicability for the creation of constitutively activated GPCRs that are regulated by peptide ligands such as CRF. These chimeras may prove useful in analyzing mechanisms of receptor regulation and in the structural analysis of ligand activated receptors.
- Published
- 2000
- Full Text
- View/download PDF
21. Mutational analysis of the interaction of the N- and C-terminal ends of angiotensin II with the rat AT(1A) receptor.
- Author
-
Costa-Neto CM, Miyakawa AA, Oliveira L, Hjorth SA, Schwartz TW, and Paiva AC
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Angiotensin II chemistry, Angiotensin II pharmacology, Animals, Binding, Competitive drug effects, COS Cells, Dose-Response Relationship, Drug, Inositol Phosphates metabolism, Iodine Radioisotopes, Molecular Sequence Data, Mutagenesis, Mutation, Protein Binding drug effects, Radioligand Assay, Rats, Receptor, Angiotensin, Type 1, Receptors, Angiotensin genetics, Angiotensin II metabolism, Receptors, Angiotensin metabolism
- Abstract
1. The role of different residues of the rat AT(1A) receptor in the interaction with the N- and C-terminal ends of angiotensin II (AngII) was studied by determining ligand binding and production of inositol phosphates (IP) in COS-7 cells transiently expressing the following AT(1A) mutants: T88H, Y92H, G196I, G196W and D278E. 2. G196W and G196I retained significant binding and IP-production properties, indicating that bulky substituents in position 196 did not affect the interaction of AngII's C-terminal carboxyl with Lys(199) located three residues below. 3. Although the T88A mutation did not affect binding, the T88H mutant had greatly decreased affinity for AngII, suggesting that substitution of Thr(88) by His might hinder binding through an indirect effect. 4. The Y92H mutation caused loss of affinity for AngII that was much less pronounced than that reported for Y92A, indicating that His in that position can fulfil part of the requirements for binding. 5. Replacing Asp(278) by Glu caused a much smaller reduction in affinity than replacing it by Ala, indicating the importance of Asp's beta-carboxyl group for AngII binding. 6. Mutations in residues Thr(88), Tyr(92) and Asp(278) greatly reduced affinity for AngII but not for Sar(1) Leu(8)-AngII, suggesting unfavourable interactions between these residues and AngII's aspartic acid side-chain or N-terminal amino group, which might account for the proposed role of the N-terminal amino group of AngII in the agonist-induced desensitization (tachyphylaxis) of smooth muscles.
- Published
- 2000
- Full Text
- View/download PDF
22. Binding of norbinaltorphimine (norBNI) congeners to wild-type and mutant mu and kappa opioid receptors: molecular recognition loci for the pharmacophore and address components of kappa antagonists.
- Author
-
Larson DL, Jones RM, Hjorth SA, Schwartz TW, and Portoghese PS
- Subjects
- Amino Acid Substitution, Animals, COS Cells, Cloning, Molecular, Glutamic Acid chemistry, Ligands, Models, Molecular, Mutagenesis, Site-Directed, Naltrexone chemistry, Naltrexone metabolism, Narcotic Antagonists chemistry, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, kappa genetics, Receptors, Opioid, mu genetics, Structure-Activity Relationship, Transfection, Naltrexone analogs & derivatives, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu metabolism
- Abstract
Molecular modifications of both the kappa opioid antagonist norbinaltorphimine (norBNI, 1) and the kappa receptor have provided evidence that the selectivity of this ligand is conferred through ionic interaction if its N17' protonated amine group (an "address") with a nonconserved acidic residue (Glu297) on the kappa receptor. In the present study, we have examined the effect of structural modifications on the affinity of norBNI analogues for wild-type and mutant kappa and mu opioid receptors expressed in COS-7 cells. Compounds 2, 3, and 7, which have an antagonist pharmacophore and basic N17' group in common with norBNI, retained high affinity for the wild-type kappa but exhibited greatly reduced affinity for mutant kappa receptors (E297K and E297A). Modification of the phenolic or N-substituent groups of the antagonist pharmacophore (4 and 5) or removal of basicity at the address N17' center (6) led to greatly reduced affinity for the wild-type and mutant receptors. The reduced affinity upon modification of the kappa receptor is consistent with the ionic interaction of the protonated N17' group of kappa antagonists (1-3, 7) with the carboxylate group of E297 at the top of TM6. This was supported by the greatly enhanced affinity of compounds 1-3 for the mutant mu receptor (K303E), as compared to the wild-type mu receptor, given that residue K303 occupies a position equivalent to that of E297 in the kappa receptor. In view of the high degree of homology of the seven TM domains of the kappa and mu opioid receptors, it is suggested that the antagonist pharmacophore is bound within this highly conserved region of the kappa or mutant mu receptor and that an anionic residue at the top of TM6 (E297 or K303E, respectively) provides additional binding affinity.
- Published
- 2000
- Full Text
- View/download PDF
23. Mutational evidence for a common kappa antagonist binding pocket in the wild-type kappa and mutant mu[K303E] opioid receptors.
- Author
-
Jones RM, Hjorth SA, Schwartz TW, and Portoghese PS
- Subjects
- Amino Acid Sequence, Animals, Binding, Competitive, COS Cells, Guanidines, Guinea Pigs, Ileum drug effects, In Vitro Techniques, Male, Mice, Models, Molecular, Molecular Conformation, Molecular Sequence Data, Morphinans, Muscle, Smooth drug effects, Mutation, Naltrexone chemical synthesis, Naltrexone metabolism, Naltrexone pharmacology, Narcotic Antagonists metabolism, Narcotic Antagonists pharmacology, Rats, Receptors, Opioid, kappa metabolism, Vas Deferens drug effects, Naltrexone analogs & derivatives, Narcotic Antagonists chemical synthesis, Receptors, Opioid, kappa antagonists & inhibitors, Receptors, Opioid, mu genetics, Receptors, Opioid, mu metabolism
- Published
- 1998
- Full Text
- View/download PDF
24. Steric hindrance mutagenesis versus alanine scan in mapping of ligand binding sites in the tachykinin NK1 receptor.
- Author
-
Holst B, Zoffmann S, Elling CE, Hjorth SA, and Schwartz TW
- Subjects
- Binding Sites, Ligands, Mutagenesis, Protein Conformation, Receptors, Neurokinin-1 physiology, Signal Transduction physiology, Structure-Activity Relationship, Alanine chemistry, Alanine metabolism, Peptide Mapping methods, Protein Structure, Secondary, Receptors, Neurokinin-1 chemistry, Receptors, Neurokinin-1 metabolism
- Abstract
Residues in transmembrane domain (TM)-III, TM-V, TM-VI, and TM-VII believed to be facing the deep part of the presumed main ligand-binding pocket of the NK1 receptor were probed by alanine substitution and introduction of residues with larger and/or chemically distinct side chains. Unaltered or even improved binding affinity for four peptide agonists, substance P, substance P-O-methyl ester, eledoisin, and neurokinin A, as well as normal EC50 values for substance P in stimulating phosphatidylinositol turnover indicated that these mutations did not alter the overall functional integrity of the receptor. The alanine substitutions in general had only minor effects on nonpeptide antagonist binding. However, the introduction of the larger and polar aspartic acid and histidine residues at positions corresponding to the monoamine binding aspartic acid in TM-III of the beta 2-adrenoceptor (ProIII:08, Pro112 in the NK1 receptor) and to the presumed monoamine interacting "two serines" in TM-V (ThrV:09, Thr201; and IleV:12, Ile204) impaired by > 100-fold the binding of a group of nonpeptide antagonists, including CP96,345, CP99,994, RP67,580, RPR100,893, and CAM4092. In contrast, another group of nonpeptide antagonists, LY303,870, FK888, and SR140,333, were little or not at all affected by the space-filling substitutions. Two of these compounds, FK888 and LY303,870, were those most seriously affected (75-89-fold) by alanine substitution of PheVI:20 located in the upper part of the main ligand-binding crevice. Surprisingly, substitution of AlaIII:11 (Ala115), which is located in the middle of TM-III, conceivably pointing toward TM-VII, with a larger valine residue increased the affinity for all 13 ligands tested, presumably by creating a closer interhelical packing. It is concluded that the introduction of larger side chains at positions at which molecular models indicate that this is structurally allowed can be a powerful method of locating ligand-binding sites due to the considerable difference between positive and negative results. Such steric hindrance mutagenesis strongly indicates that one population of nonpeptide antagonists bind in the deep pocket of the main ligand-binding crevice of the NK1 receptor, whereas another group of nonpeptide antagonists, especially SR140,333, was surprisingly resistant to mutational mapping in this pocket.
- Published
- 1998
- Full Text
- View/download PDF
25. Constitutive activity of glucagon receptor mutants.
- Author
-
Hjorth SA, Orskov C, and Schwartz TW
- Subjects
- Adenylyl Cyclases metabolism, Amino Acid Sequence, Amino Acid Substitution genetics, Animals, COS Cells, Histidine genetics, Histidine physiology, Molecular Sequence Data, Radioligand Assay, Rats, Receptors, Glucagon chemistry, Recombinant Proteins chemical synthesis, Threonine genetics, Mutagenesis, Site-Directed, Receptors, Glucagon genetics, Receptors, Glucagon physiology
- Abstract
Increased constitutive activity has been observed in the PTH receptor in association with naturally occurring mutations of two residues that are conserved between members of the glucagon/vasoactive intestinal peptide/calcitonin 7TM receptor family. Here, the corresponding residues of the glucagon receptor, His178 and Thr352, were probed by mutagenesis. An elevated level of basal cAMP production was observed after the exchange of His178 into Arg, but not for the exchange into Lys, Ala, or Glu. However, for all of these His178 substitutions, an increased binding affinity for glucagon was observed [dissociation constant (Kd) ranging from 1.1-6.4 nM, wild type: Kd = 12.0 nM]. A further increase in cAMP production was observed for the [H178R] construct upon stimulation with glucagon, albeit the EC50 surprisingly was increased approximately 10-fold relative to the wild-type receptor. Substitution of Thr352, located at the intracellular end of transmembrane segment VI, with Ala led to a slightly elevated basal cAMP level, while the introduction of Pro or Ser at this position affected rather the binding affinity of glucagon or the EC50 for stimulation of cAMP production. The large extracellular segment, which is essential for glucagon binding, was not required for constitutive activation of the glucagon receptor as the introduction of the [H178R] mutation into an N-terminally truncated construct exhibited an elevated basal level of cAMP production. The analog des-His1-[Glu9]glucagon amide, which in vivo is a glucagon antagonist, was an agonist on both the wild-type and the [H178R] receptor and did not display any activity as an inverse agonist. It is concluded that the various phenotypes displayed by the constitutively active glucagon receptor mutants reflect the existence of multiple agonist-preferring receptor conformers, which include functionally active as well as inactive states. This view agrees with a recent multi-state extension of the ternary complex model for 7TM receptor activation.
- Published
- 1998
- Full Text
- View/download PDF
26. Ligand binding pocket of the human somatostatin receptor 5: mutational analysis of the extracellular domains.
- Author
-
Greenwood MT, Hukovic N, Kumar U, Panetta R, Hjorth SA, Srikant CB, and Patel YC
- Subjects
- Amino Acid Sequence, Animals, Binding Sites genetics, Binding, Competitive, CHO Cells, Cricetinae, Humans, Immunohistochemistry, Ligands, Membrane Proteins chemistry, Molecular Sequence Data, Mutagenesis genetics, Octreotide analogs & derivatives, Octreotide metabolism, Protein Conformation, Receptors, Somatostatin agonists, Receptors, Somatostatin metabolism, Sequence Alignment, Receptors, Somatostatin genetics
- Abstract
The ligand binding domain of G protein-coupled receptors for peptide ligands consists of a pocket formed by extracellular and transmembrane domain (TM) residues. In the case of somatostatin (SRIF), however, previous studies have suggested that the binding cavity of the octapeptide analog SMS201-995 (SMS) is lined by residues in TMs III-VII. The additional involvement of the extracellular domains for binding SMS or the natural SRIF ligands (SRIF-14, SRIF-28) has not been clarified. Using a cassette construct cDNA for the human somatostatin 5 receptor (sst5R), we systematically examined the role of exofacial structures in ligand binding by creating a series of mutants in which the extracellular portions have been altered by conservative segment exchange (CSE) mutagenesis for the extracellular loops (ECLs) and by deletion (for the NH2-terminal segment) or truncation analysis (ECL3). CHO-K1 cells were stably transfected with wild type or mutant human sst5R constructs, and agonist binding was assessed using membrane binding assays with 125I-LTT SRIF-28 ligand. Deletion of the NH2 terminus or CSE mutagenesis of ECL1 and ECL3 produced minor 2-8-fold decreases in affinity for SRIF-14, SRIF-28, and SMS ligands. Truncation of ECL3 to mimic the size of this loop in sst1R and sst4R (the two subtypes that do not bind SMS) did not interfere with the binding of SMS, SRIF-14, or SRIF-28. In contrast, both ECL2 mutants failed to bind 125I-LTT SRIF-28. Immunocytochemical analysis of nonpermeabilized cells with a human sst5R antibody revealed that the mutant receptors were targeted to the plasma membrane. Labeled SMS (125I-Tyr3 SMS) also failed to bind to the mutant ECL2 receptors. These results suggest a potential contribution of ECL2 (in addition to the previously identified residues in TMs III-VII) to the SRIF ligand binding pocket.
- Published
- 1997
- Full Text
- View/download PDF
27. How receptor mutagenesis may confirm or confuse receptor classification.
- Author
-
Schwartz TW, Perlman S, Rosenkilde MM, and Hjorth SA
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Humans, Ligands, Models, Molecular, Molecular Sequence Data, Mutagenesis, Protein Binding, Protein Folding, Receptors, Cell Surface chemistry, Receptors, Cell Surface classification, Receptors, Cell Surface genetics, Terminology as Topic
- Published
- 1997
- Full Text
- View/download PDF
28. Metal-ion sites as structural and functional probes of helix-helix interactions in 7TM receptors.
- Author
-
Elling CE, Thirstrup K, Nielsen SM, Hjorth SA, and Schwartz TW
- Subjects
- Animals, Binding Sites, Kinetics, Ligands, Models, Molecular, Molecular Probes, Molecular Structure, Protein Structure, Secondary, Receptors, Neurokinin-1 chemistry, Receptors, Neurokinin-1 metabolism, Zinc metabolism, GTP-Binding Proteins metabolism, Metals metabolism, Receptors, Cell Surface chemistry, Receptors, Cell Surface metabolism
- Published
- 1997
- Full Text
- View/download PDF
29. Dual agonistic and antagonistic property of nonpeptide angiotensin AT1 ligands: susceptibility to receptor mutations.
- Author
-
Perlman S, Costa-Neto CM, Miyakawa AA, Schambye HT, Hjorth SA, Paiva AC, Rivero RA, Greenlee WJ, and Schwartz TW
- Subjects
- Animals, Dose-Response Relationship, Drug, Humans, Ligands, Rats, Receptors, Angiotensin genetics, Angiotensin II pharmacology, Antihypertensive Agents pharmacology, Imidazoles pharmacology, Mutation genetics, Receptors, Angiotensin drug effects, Tetrazoles pharmacology
- Abstract
Two nonpeptide ligands that differ chemically by only a single methyl group but have agonistic (L-162,782) and antagonistic (L-162,389) properties in vivo were characterized on the cloned angiotensin AT1 receptor. Both compounds bound with high affinity (K(I) = 8 and 28 nM, respectively) to the AT1 receptor expressed transiently in COS-7 cells as determined in radioligand competition assays. L-162,782 acted as a powerful partial agonist, stimulating phosphatidylinositol turnover with a bell-shaped dose-response curve to 64% of the maximal level reached in response to angiotensin II. Surprisingly, L-162,389 also stimulated phosphatidylinositol turnover, albeit only to a small percentage of the angiotensin response. The prototype nonpeptide AT1 agonist L-162,313 gave a response of approximately 50%. The apparent EC50 values for all three compounds in stimulating phosphatidylinositol turnover were similar, approximately 30 nM, corresponding to their binding affinity. Each of the three compounds also acted as angiotensin antagonists, yet in this capacity the compounds differed markedly, with IC50 values ranging from 1.05 x 10(-7) M for L-162,389 to 6.5 x 10(-6) for L-162,782. A series of point mutations in the transmembrane segments (TMs) of the AT1 receptor had only minor effect on the binding affinity of the nonpeptide compounds, with the exception of A104V at the top of TM III, which selectively impaired the binding of L-162,782 and L-162,389. Substitutions in the middle of TM III, VI, or VII, which did not affect the binding affinity of the compounds, impaired or eliminated the agonistic efficacy of the nonpeptides but with only minor or no effect on the angiotensin potency or efficacy. Thus, in the N295D rat AT1 construct, L-162,782, L-162,313, and L-162,389 all antagonized the angiotensin-induced phosphatidylinositol turnover with surprisingly similar IC50 values (90-180 nM), and they all bound with unaltered, high affinity (22-36 nM). However, L-162,313 and L-162,782 could stimulate phosphatidylinositol turnover to only 20% of that of angiotensin. It is concluded that minor chemical modifications of either the compound or the receptor can dramatically alter the agonistic efficacy of biphenyl imidazole compounds on the AT1 receptor without affecting their affinity, as determined in binding assays, and that a number of substitutions in the middle of the TM segments affect the efficacy of nonpeptide agonists as opposed to angiotensin.
- Published
- 1997
- Full Text
- View/download PDF
30. Engineering of metal-ion sites as distance constraints in structural and functional analysis of 7TM receptors.
- Author
-
Elling CE, Thirstrup K, Nielsen SM, Hjorth SA, and Schwartz TW
- Subjects
- Binding Sites, Cations, Divalent, GTP-Binding Proteins metabolism, Receptors, Cell Surface metabolism, Structure-Activity Relationship, Zinc metabolism, Receptors, Cell Surface chemistry
- Abstract
G-protein-coupled receptors with their seven transmembrane (7TM) segments constitute the largest superfamily of proteins known. Unfortunately, still only relatively low resolution structures derived from electron cryo-microscopy analysis of 2D crystals are available for these proteins. We have used artificially designed Zn(II) metal-ion binding sites to probe 7TM receptors structurally and functionally and to define some basic distance constraints for molecular modeling. In this way, the relative helical rotation and vertical translocation of transmembrane helices TM-II, TM-III, TM-V, and TM-VI of the tachykinin NK-1 receptor have been restricted. Collectively, these zinc sites constitute a basic network of distance constraints that limit the degrees of freedom of the interhelical contact faces in molecular models of 7TM receptors. The construction of artificially designed metal-ion sites is discussed also in the context of probes for conformational changes occurring during receptor activation.
- Published
- 1997
- Full Text
- View/download PDF
31. Radioligand-dependent discrepancy in agonist affinities enhanced by mutations in the kappa-opioid receptor.
- Author
-
Hjorth SA, Thirstrup K, and Schwartz TW
- Subjects
- Animals, Benzofurans metabolism, Benzofurans pharmacology, COS Cells drug effects, COS Cells metabolism, Diprenorphine metabolism, Diprenorphine pharmacology, Dynorphins metabolism, Dynorphins pharmacology, Kinetics, Narcotic Antagonists metabolism, Narcotic Antagonists pharmacology, Peptide Fragments metabolism, Peptide Fragments pharmacology, Pyrrolidines metabolism, Pyrrolidines pharmacology, Radioligand Assay, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu metabolism, Recombinant Fusion Proteins metabolism, Tritium, Mutation, Receptors, Opioid, kappa agonists, Receptors, Opioid, kappa genetics
- Abstract
A series of kappa/mu receptor chimeras and a number of kappa receptors substituted in the second transmembrane segment (TM-II) were investigated using as radioligands, respectively, the kappa-selective agonist [3H]C1977 and the nonselective opioid antagonist [3H]diprenorphine (DIP). All of the receptor constructs bound [3H]DIP with similar and high affinity, whereas the apparent affinity of the nonpeptide agonist C1977, when estimated in competition binding with the antagonist [3H]DIP, was impaired between 42- and > 500-fold in the kappa/mu chimeras and between 64- and 153-fold in three of the kappa receptor mutants that had been substituted in the TM-II segment. However, homologous competition binding experiments, using [3H]C1977 as radioligand, showed that the high affinity binding of this nonpeptide agonist was in fact not impaired in four of the kappa/mu chimeras and in three TM-II substituted kappa receptors compared with the wild-type kappa receptor. In all cases in which mutations decreased the apparent affinity of C1977 without affecting its actual affinity, as determined in homologous assays using [3H]C1977, the calculated number of receptor sites (Bmax) was decreased. In three of the kappa/mu constructs, binding of [3H]C1977 was undetectable, indicating that in these chimeras the affinity of the nonpeptide agonist had actually been affected. Also, for the kappa-selective peptide agonist dynorphin A(1-8), the measured affinity for the receptor mutants was strongly dependent on whether it was determined using the antagonist [3H]DIP or the agonist [3H]C1977 in that < or = 800-fold higher Ki values were determined in competition with the antagonist. It is concluded that mutations in the kappa-opioid receptor can cause large discrepancies between the affinity determined for agonists in homologous versus heterologous competition binding assays and that this pattern, which is compatible with a partial uncoupling of receptors, is observed in surprisingly many types of receptor mutations.
- Published
- 1996
32. Glucagon and GLP-1 receptors: lessons from chimeric ligands and receptors.
- Author
-
Hjorth SA and Schwartz TW
- Subjects
- Amino Acid Sequence, Glucagon-Like Peptide 1, Ligands, Molecular Sequence Data, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Glucagon genetics, Glucagon metabolism, Peptide Fragments genetics, Peptide Fragments metabolism, Protein Precursors genetics, Protein Precursors metabolism, Receptors, Glucagon genetics, Receptors, Glucagon metabolism
- Published
- 1996
- Full Text
- View/download PDF
33. Construction of a high affinity zinc switch in the kappa-opioid receptor.
- Author
-
Thirstrup K, Elling CE, Hjorth SA, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Histidine chemistry, Models, Chemical, Molecular Sequence Data, Mutagenesis, Site-Directed, Protein Structure, Tertiary, Rats, Receptors, Neurokinin-1 chemistry, Structure-Activity Relationship, Zinc chemistry, Receptors, Opioid, kappa chemistry
- Abstract
Very limited structural information is available concerning the superfamily of G-protein-coupled receptors with their seven-transmembrane segments. Recently a non-peptide antagonist site was structurally and functionally replaced by a metal ion site in the tachykinin NK-1 receptor. Here, this Zn(II) site is transferred to the kappa-opioid receptor by substituting two residues at the outer portion of transmembrane V (TM-V), Asp223 and Lys227, and one residue at the top of TM-VI, Ala298, with histidyl residues. The histidyl residues had no direct effect on the binding of either the non-peptide antagonist [3H]diprenorphine or the non-peptide agonist, [3H]CI977, just as these mutations/substitutions did not affect the apparent affinity of a series of other peptide and non-peptide ligands when tested in competition binding experiments. However, zinc ions in a dose-dependent manner prevented binding of both agonist and antagonist ligands with an apparent affinity for the metal ion, which gradually was built up to 10(-6) M. This represents an increase in affinity for the metal ion of about 1000-fold as compared with the wild-type kappa receptor and is specific for Zn(II) as the affinity for e.g. Cu(II) was almost unaffected. The direct transfer of this high affinity metal ion switch between two only distantly related receptors indicates a common overall arrangement of the seven-helix bundle among receptors of the rhodopsin family.
- Published
- 1996
- Full Text
- View/download PDF
34. Analysis of selective binding epitopes for the kappa-opioid receptor antagonist nor-binaltorphimine.
- Author
-
Hjorth SA, Thirstrup K, Grandy DK, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Glutamine genetics, Molecular Sequence Data, Naltrexone metabolism, Naltrexone pharmacology, Rats, Receptors, Opioid, kappa genetics, Receptors, Opioid, kappa immunology, Receptors, Opioid, kappa metabolism, Receptors, Opioid, mu genetics, Recombinant Fusion Proteins, Epitopes analysis, Naltrexone analogs & derivatives, Receptors, Opioid, kappa antagonists & inhibitors
- Abstract
The structural determinants for the selective binding of the nonpeptide opioid receptor antagonist nor-binaltorphimine (nor-BNI) to the kappa-opioid receptor were characterized using a systematic series of chimeras between the kappa receptor and the homologous mu-opioid receptor. All 10 chimeric constructs bound the nonselective antagonists (-)-naloxone and diprenorphine with similar affinities, as did the two wild-type receptors. Introduction of amino-terminal segments of increasing length, extending to and including transmembrane segment VI, from the mu receptor into the kappa receptor did not impair the high affinity binding of nor-BNI, and neither did introduction of the intracellular carboxyl-terminal extension of the mu receptor. In contrast, nor-BNI binding was impaired > or = 600-fold in constructs in which extracellular loop 3 and transmembrane segment VII originated from the mu receptor. The exchange of a single residue within this region, Glu297, for lysine, the corresponding residue from the mu receptor, reduced the binding affinity of nor-BNI 142-fold, without affecting the binding the nonselective compounds (-)-naloxone and diprenorphine. It is concluded that the selective binding of nor-BNI to the kappa-opioid receptor is determined by nonconserved residues located in extracellular loop 3 and transmembrane segment VII and that Glu297, located just outside transmembrane segment VI, plays a major role in the kappa-selective binding characteristics of nor-BNI.
- Published
- 1995
35. Interaction between the nonpeptide angiotensin antagonist SKF-108,566 and histidine 256 (HisVI:16) of the angiotensin type 1 receptor.
- Author
-
Schambye HT, Hjorth SA, Weinstock J, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Cells, Cultured, Histidine genetics, Humans, Molecular Sequence Data, Mutagenesis, Rats, Receptors, Angiotensin genetics, Acrylates metabolism, Acrylates pharmacology, Angiotensin I metabolism, Angiotensin Receptor Antagonists, Histidine metabolism, Imidazoles metabolism, Imidazoles pharmacology, Receptors, Angiotensin metabolism, Thiophenes
- Abstract
His256 (HisVI:16) of transmembrane segment (TM)-VI of the rat angiotensin type 1 (AT1) receptor was targeted for mutagenesis to investigate its potential involvement in ligand binding. Substitution of His256 with alanine, phenylalanine, glutamine, or isoleucine did not affect the binding of either angiotensin II or nine different biphenylimidazole AT1 antagonists. In contrast, the binding affinity of the prototype imidazoleacrylic acid antagonist SKF-108,566 was reduced 15-fold by the exchange of His256 with alanine. Substitution of His256 with either isoleucine or phenylalanine yielded similar results, whereas a glutamine residue was able to substitute for His256, suggesting that the epsilon-nitrogen of His256 could be involved in the interaction with the imidazoleacrylic acid. To identify the chemical groups on SKF-108,566 that interact with His256 and with Asn295, a previously identified interaction point for nonpeptide antagonists located in TM-VII, we tested the binding of 15 analogs of SKF-108,566 in which different chemical moieties were systematically exchanged. The results indicated that the carboxyphenyl group of SKF-108,566 interacts with the imidazole side chain of His256. The data did not point to any particular contact group on the antagonist for Asn295. It is concluded that the imidazoleacrylic acid antagonists share some interactions in TM-VII of the AT1 receptor with the biphenylimidazole antagonists, but the binding of the imidazoleacrylic acid compounds is uniquely dependent on His256 in TM-VI, possibly through the carboxyphenyl moiety.
- Published
- 1995
36. Non-peptide angiotensin agonist. Functional and molecular interaction with the AT1 receptor.
- Author
-
Perlman S, Schambye HT, Rivero RA, Greenlee WJ, Hjorth SA, and Schwartz TW
- Subjects
- Amino Acid Sequence, Angiotensin II metabolism, Angiotensin II pharmacology, Angiotensin Receptor Antagonists, Animals, Binding, Competitive, Biphenyl Compounds metabolism, CHO Cells, Cell Line, Chlorocebus aethiops, Cricetinae, Humans, Imidazoles metabolism, Kidney, Kinetics, Ligands, Molecular Sequence Data, Mutagenesis, Insertional, Point Mutation, Polymerase Chain Reaction, Protein Structure, Secondary, Rats, Receptors, Angiotensin chemistry, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Tetrazoles metabolism, Tetrazoles pharmacology, Transfection, Xenopus laevis, Angiotensins agonists, Biphenyl Compounds pharmacology, Imidazoles pharmacology, Inositol Phosphates metabolism, Phosphatidylinositols metabolism, Receptors, Angiotensin metabolism
- Abstract
Non-peptide ligands for peptide receptors for the G-protein-coupled type are generally antagonists, except in the opiate system. Recently, it was observed that a subset of biphenylimidazole derivatives surprisingly possessed angiotensin-like activity in vivo. In COS-7 cells transfected with the rat AT1 receptor a prototype of these compounds, L-162,313 stimulated phosphoinositide hydrolysis with an EC50 of 33 +/- 11 nM. The maximal response to the compound was 50% of that of angiotensin II in COS-7 cells but only 3% in stably transfected Chinese hamster ovary cells. The agonistic effect of L-162,313 was blocked by the AT1-specific antagonist L-158,809 and was not observed in untransfected cells. In Chinese hamster ovary cells, L-162,313 also acted as an insurmountable antagonist of the angiotensin stimulated phosphoinositide hydrolysis. In contrast to previously tested non-peptide ligands, L-162,313 bound with reasonably high affinity to the Xenopus laevis AT1 receptor. In the human receptor, the binding of L-162,313 was found to be unaffected by point mutations in transmembrane segments III and VII, which impaired the binding of biphenylimidazole antagonists. Substitutions in the extracellular domains of the human and rat receptor, which impaired the binding of angiotensin II, did not affect the binding of L-162,313. It is concluded that a subset of biphenylimidazole compounds can act as high affinity partial agonists on the AT1 receptor. These compounds have molecular interactions with the receptor which appear to differ both from that of the structurally similar non-peptide antagonists and from that of their functional counterpart, the peptide agonist.
- Published
- 1995
- Full Text
- View/download PDF
37. Cloning and expression in insect cells of two pancreatic lipases and a procolipase from Myocastor coypus.
- Author
-
Thirstrup K, Carrière F, Hjorth SA, Rasmussen PB, Nielsen PF, Ladefoged C, Thim L, and Boel E
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cloning, Molecular, DNA, Enzyme Precursors, Molecular Sequence Data, Nucleopolyhedroviruses genetics, Phylogeny, Rodentia, Spodoptera, Colipases genetics, Lipase genetics, Pancreas enzymology, Protein Precursors genetics
- Abstract
The physiological role of pancreatic lipases has traditionally been assigned solely to triacylglyceride metabolism, while the digestion of phospholipids requires the presence of the pancreatic phospholipase A2, a 14-kDa enzyme unrelated to pancreatic lipases. However, in the guinea pig, it was observed that the pancreatic phospholipase A2 was absent and that a guinea pig pancreatic-lipase-related protein 2 (GPL-RP2) was responsible for phospholipase activity, in contrast to the situation observed in other mammalian species. As the guinea pig is a member of the hystricomorph rodents, it was of interest to investigate if other species within this evolutionary suborder display similar characteristics. The coypu (Myocastor coypus) also a member of the hystricomorph rodents, was chosen for further investigations. The cDNAs encoding two pancreatic lipases and a procolipase from the coypu were cloned, expressed and characterized. One lipase, CoPL-RP2, was identified as belonging to the RP2 subfamily, while the second, CoPL, was found to belong to the classical pancreatic lipase subfamily. Enzymic characterization and sequence data suggest a role for coypu colipase as a specific cofactor for CoPL, while this coypu colipase cannot be an important cofactor for CoPL-RP2 in vivo. Also, the new lipase cDNA sequences were used in a phylogentic analysis to reinvestigate the taxonomical position of the hystricomorph rodents (e.g. coypu and guinea pig) with respect to the myomorph rodents (e.g. rat and mouse).
- Published
- 1995
- Full Text
- View/download PDF
38. Identification of peptide binding residues in the extracellular domains of the AT1 receptor.
- Author
-
Hjorth SA, Schambye HT, Greenlee WJ, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Biological Evolution, Cells, Cultured, Conserved Sequence, Humans, Molecular Sequence Data, Mutagenesis, Protein Binding, Protein Structure, Secondary, Rats, Receptors, Angiotensin chemistry, Angiotensin I metabolism, Peptides metabolism, Receptors, Angiotensin metabolism
- Abstract
To locate essential determinants for angiotensin II binding, we have performed a systematic mutational analysis of the exterior domain of the AT1 receptor. Receptor mutants, deficient in peptide binding, were analyzed using radiolabeled nonpeptide ligand as an important tool. Two independent strategies for mutagenesis were employed: conservative segment exchange and point mutagenesis of evolutionarily conserved residues. Results from the conservative segment exchange in which 6-17 residues were replaced with chemically similar, yet different, amino acid sequences of the same length suggested that important peptide ligand binding epitopes are located in the N-terminal extension of the AT1 receptor, in particular adjacent to the top of transmembrane segment I (TM-I), and in the third extracellular loop, close to the top of TM-VII. The substitution of residues from either of these regions resulted in a 5,000-20,000-fold decrease in affinity for the peptide agonist angiotensin II (AII) and the peptide antagonist [Sar1,Leu8]AII without affecting the binding of nonpeptide antagonists. Alanine substitution of evolutionarily conserved residues demonstrated that peptide binding was dependent on several residues in the N-terminal extension, near the top of TM-I, a tyrosine residue located in extracellular loop 1, close to TM-II, and 2 aspartate residues positioned in extracellular loop 3 on the same face of an alpha-helical extension of TM-VII. In all cases the binding of nonpeptide antagonist was unaffected by these substitutions. It is concluded that important epitopes involved in angiotensin II binding are located around the top of transmembrane segments I, II, and VII which conceivably are in close spatial proximity in the folded receptor structure.
- Published
- 1994
39. Glucagon and glucagon-like peptide 1: selective receptor recognition via distinct peptide epitopes.
- Author
-
Hjorth SA, Adelhorst K, Pedersen BB, Kirk O, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Binding, Competitive, Cell Membrane metabolism, Epitopes chemistry, Glucagon analogs & derivatives, Glucagon chemistry, Glucagon immunology, Glucagon-Like Peptide 1, Kinetics, Molecular Sequence Data, Peptide Fragments chemistry, Peptide Fragments immunology, Peptides chemical synthesis, Peptides chemistry, Peptides metabolism, Protein Precursors chemistry, Protein Precursors immunology, Swine, Epitopes metabolism, Glucagon metabolism, Liver metabolism, Peptide Fragments metabolism, Protein Precursors metabolism, Receptors, Glucagon metabolism
- Abstract
Glucagon and glucagon-like peptide 1 (GLP-1) are homologous peptide hormones that are recognized by likewise homologous, but highly selective receptors. Analogs of glucagon and GLP-1, in which the divergent residues were systematically exchanged, were employed to identify the structural requirements for their selective receptor recognition. Substitutions in the NH2-terminal part of the glucagon molecule with the corresponding GLP-1 residues, as for example in [Ala2,Glu3]-glucagon and [Val10,Ser12]glucagon, reduced the binding affinity for the glucagon receptor several hundred-fold without increasing the affinity for the GLP-1 receptor. In contrast, introduction of GLP-1 residues into the far COOH-terminal part of the glucagon molecule, e.g. [Val27,Lys28,Gly29,Arg30]glucagon, had a minimal effect on recognition of the glucagon receptor, but improved the affinity of the analog for the GLP-1 receptor up to 200-fold. Similarly, substitutions in especially the far COOH-terminal part of the GLP-1 molecule with the corresponding glucagon residues, e.g. des-Arg30-[Met27,Asn28,Thr29]GLP-1, decreased the affinity for the GLP-1 receptor several hundred-fold (IC50 = 0.4-190 nM) without increasing the affinity for the glucagon receptor. Conversely, substitutions in the NH2-terminal part of the GLP-1 molecule impaired the affinity for the GLP-1 receptor only moderately. We conclude that the selective recognition of the glucagon and GLP-1 receptors is determined by residues located at opposite ends of the homologous peptide ligands. This conclusion is supported by the observation that a "chimeric" peptide consisting of the NH2-terminal part of the glucagon molecule joined to the COOH-terminal part of the GLP-1 molecule was recognized with high affinity by both receptors.
- Published
- 1994
40. Mutations along transmembrane segment II of the NK-1 receptor affect substance P competition with non-peptide antagonists but not substance P binding.
- Author
-
Rosenkilde MM, Cahir M, Gether U, Hjorth SA, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Binding, Competitive, Cell Line, Chlorocebus aethiops, DNA Mutational Analysis, Humans, Kidney, Kinetics, Models, Structural, Molecular Sequence Data, Neurokinin A metabolism, Neurokinin-1 Receptor Antagonists, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Substance P analogs & derivatives, Transfection, Protein Structure, Secondary, Receptors, Neurokinin-1 chemistry, Receptors, Neurokinin-1 metabolism, Substance P metabolism, Substance P pharmacology
- Abstract
Mutational analysis of the NK-1 receptor indicates that residues involved in non-peptide antagonist binding cluster around the outer portion of transmembrane segments (TM) V and VI. In contrast mutations affecting the binding of the natural peptide agonist, substance P, are scattered in the exterior part of the receptor. Recently it was reported that a number of mutations in TM-II also seriously impair substance P binding. Here we confirm that Ala substitutions for these residues located on a hydrophilic helical face of TM-II basically eliminate substance P binding to the NK-1 receptor, provided that a radiolabeled non-peptide antagonist is used as radioligand. Surprisingly, radiolabeled substance P bound well to all these mutant receptors and was displaced with only slightly reduced affinity by the unlabeled peptide and by the non-peptide antagonists. The wild-type homologous NK-2 receptor displayed properties similar to those observed in the mutated NK-1 receptors, i.e. concomitant high affinity binding of radiolabeled agonist peptide (in this case neurokinin A), yet low affinity, G-protein independent competition of unlabeled peptide with radiolabeled non-peptide antagonist. It is concluded that substitutions in TM-II of the NK-1 receptor do not affect the high affinity binding of substance P but instead block the ability of the peptides to compete for non-peptide antagonist binding. It is suggested that certain mutations can impair interchange between receptor conformations that each bind different ligands with high affinity.
- Published
- 1994
41. Mutations in transmembrane segment VII of the AT1 receptor differentiate between closely related insurmountable and competitive angiotensin antagonists.
- Author
-
Schambye HT, von Wijk B, Hjorth SA, Wienen W, Entzeroth M, Bergsma DJ, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Binding, Competitive physiology, Humans, In Vitro Techniques, Ligands, Membranes metabolism, Molecular Sequence Data, Polymerase Chain Reaction, Rabbits, Recombinant Fusion Proteins metabolism, Xenopus, Angiotensin I antagonists & inhibitors, Angiotensin I metabolism, Angiotensin Receptor Antagonists, Mutation genetics, Receptors, Angiotensin genetics
- Abstract
Chimeric constructs between the human and the Xenopus laevis AT1 receptor have demonstrated, that the binding of non-peptide angiotensin antagonists is dependent on non-conserved residues located deep in transmembrane segment VII of the AT1 receptor. Here we have studied four pairs of closely related antagonists each consisting of a competitive and an insurmountable compound differentiated by one out of three different types of minor chemical modifications. None of the antagonists bound to the Xenopus receptor and the binding of all of the compounds to the human receptor was severely impaired by the introduction of non-conserved residues from transmembrane segment VII of the Xenopus receptor. In all four pairs of antagonists the competitive compound was affected more by these substitutions than the corresponding insurmountable one (209 vs. 22, 281 vs. 29, 290 vs. 29 and 992 vs. 325-fold increase in Ki values). A similar pattern was observed in response to substitution of a single non-conserved residue in transmembrane segment VII, Asn295 to Ser. These results indicate that a common molecular mechanism distinguishes the interaction of insurmountable and competitive antagonists with the AT1 receptor.
- Published
- 1994
- Full Text
- View/download PDF
42. Differentiation between binding sites for angiotensin II and nonpeptide antagonists on the angiotensin II type 1 receptors.
- Author
-
Schambye HT, Hjorth SA, Bergsma DJ, Sathe G, and Schwartz TW
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Humans, Molecular Sequence Data, Molecular Structure, Mutation, Peptides metabolism, Receptors, Angiotensin metabolism, Xenopus laevis, Angiotensin II metabolism, Angiotensin Receptor Antagonists
- Abstract
To characterize binding sites for nonpeptide angiotensin antagonists on the human angiotensin II receptor type 1 (AT1 receptor) we have systematically exchanged segments of the human receptor with corresponding segments from a homologous Xenopus laevis receptor, which does not bind the nonpeptide compounds. Substitution of transmembrane segment VII of the human AT1 receptor dramatically reduced the binding affinity of all of the 11 nonpeptide antagonists tested (55- to > 2000-fold) with no effect on the binding of angiotensin. The affinity for the nonpeptide compounds decreased additionally one order of magnitude when transmembrane segment VI and the connecting extracellular loop 3 from the Xenopus receptor were also introduced into the human AT1 receptor. Exchanges of smaller segments and single residues in transmembrane segments VI and VII and extracellular loop 3 revealed that the binding of nonpeptide antagonists was dependent on nonconserved residues located deep within the transmembrane segments VI and VII, in particular Asn295 in transmembrane segment VII. Surprisingly, all exchanges in transmembrane segment VII, including the Asn295 to Ser substitution, had a more pronounced effect on the binding of the competitive antagonists relative to the insurmountable antagonists. It is concluded that the binding mode for peptide and nonpeptide ligands on the AT1 receptor is rather different and that competitive and insurmountable antagonists presumably bind to overlapping but distinct sites located in transmembrane segments VI and VII.
- Published
- 1994
- Full Text
- View/download PDF
43. [Non-peptide antagonists to angiotensin II receptors. A review].
- Author
-
Schambye HT, Hjorth SA, and Schwartz TW
- Subjects
- Angiotensin II chemistry, Angiotensin Receptor Antagonists, Antihypertensive Agents therapeutic use, Biphenyl Compounds chemistry, Biphenyl Compounds therapeutic use, Humans, Imidazoles chemistry, Imidazoles therapeutic use, Losartan, Receptors, Angiotensin chemistry, Tetrazoles chemistry, Tetrazoles therapeutic use, Angiotensin II antagonists & inhibitors, Receptors, Angiotensin drug effects, Renin-Angiotensin System drug effects
- Abstract
The renin-angiotensin system is the most important hormone system in the control of blood pressure and electrolyte homeostasis. Pharmacological blockade of the system by means of beta-blockers or ACE-inhibitors is a major tool in the treatment of hypertension and congestive heart failure. Inhibition of the binding of angiotensin to its receptor is, however, theoretically a more direct and selective blocking method. Recently, a series of potent non-peptide antagonists have been developed, which are active when given orally and appear to be promising drug candidates. The clinical and theoretical implications of this discovery are reviewed based upon the present knowledge of the renin-angiotensin system and the available methods for therapeutic intervention in the system.
- Published
- 1993
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.