1
|
Sun S, Wang W. Mechanosensitive adhesion G protein-coupled receptors: Insights from health and disease. Genes Dis 2025; 12:101267. [PMID: 39935605 PMCID: PMC11810715 DOI: 10.1016/j.gendis.2024.101267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 01/15/2024] [Accepted: 02/28/2024] [Indexed: 02/13/2025] Open
Abstract
Ontogeny cannot be separated from mechanical forces. Cells are continuously subjected to different types of mechanical stimuli that convert into intracellular signals through mechanotransduction. As a member of the G protein-coupled receptor superfamily, adhesion G protein-coupled receptors (aGPCRs) have attracted extensive attention due to their unique extracellular domain and adhesion properties. In the past few decades, increasing evidence has indicated that sensing mechanical stimuli may be one of the main physiological activities of aGPCRs. Here, we review the general structure and activation mechanisms of these receptors and highlight the lesion manifestations relevant to each mechanosensitive aGPCR.
Collapse
Affiliation(s)
- Shiying Sun
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
- Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
- Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Wen Wang
- Department of Orthodontics, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
- Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
- Hebei Clinical Research Center for Oral Diseases, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| |
Collapse
|
2
|
Dietzsch AN, Al-Hasani H, Altschmied J, Bottermann K, Brendler J, Haendeler J, Horn S, Kaczmarek I, Körner A, Krause K, Landgraf K, Le Duc D, Lehmann L, Lehr S, Pick S, Ricken A, Schnorr R, Schulz A, Strnadová M, Velluva A, Zabri H, Schöneberg T, Thor D, Prömel S. Dysfunction of the adhesion G protein-coupled receptor latrophilin 1 (ADGRL1/LPHN1) increases the risk of obesity. Signal Transduct Target Ther 2024; 9:103. [PMID: 38664368 PMCID: PMC11045723 DOI: 10.1038/s41392-024-01810-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Obesity is one of the diseases with severe health consequences and rapidly increasing worldwide prevalence. Understanding the complex network of food intake and energy balance regulation is an essential prerequisite for pharmacological intervention with obesity. G protein-coupled receptors (GPCRs) are among the main modulators of metabolism and energy balance. They, for instance, regulate appetite and satiety in certain hypothalamic neurons, as well as glucose and lipid metabolism and hormone secretion from adipocytes. Mutations in some GPCRs, such as the melanocortin receptor type 4 (MC4R), have been associated with early-onset obesity. Here, we identified the adhesion GPCR latrophilin 1 (ADGRL1/LPHN1) as a member of the regulating network governing food intake and the maintenance of energy balance. Deficiency of the highly conserved receptor in mice results in increased food consumption and severe obesity, accompanied by dysregulation of glucose homeostasis. Consistently, we identified a partially inactivating mutation in human ADGRL1/LPHN1 in a patient suffering from obesity. Therefore, we propose that LPHN1 dysfunction is a risk factor for obesity development.
Collapse
Affiliation(s)
- André Nguyen Dietzsch
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munich-Neuherberg, Germany
| | - Joachim Altschmied
- Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katharina Bottermann
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Pharmacology, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jana Brendler
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Judith Haendeler
- Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute (CARID), Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Susanne Horn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, Hospital for Children and Adolescents, Medical Faculty, Leipzig University, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, Leipzig University Medical Center, Leipzig, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, Hospital for Children and Adolescents, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany
| | - Laura Lehmann
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, Medical Faculty, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munich-Neuherberg, Germany
| | - Stephanie Pick
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Albert Ricken
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Rene Schnorr
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Angela Schulz
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Martina Strnadová
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Akhil Velluva
- Institute of Human Genetics, Leipzig University Medical Center, Leipzig, Germany
| | - Heba Zabri
- Institute of Pharmacology, Medical Faculty, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany.
| | - Simone Prömel
- Institute of Cell Biology, Department of Biology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
3
|
Lala T, Hall RA. Adhesion G protein-coupled receptors: structure, signaling, physiology, and pathophysiology. Physiol Rev 2022; 102:1587-1624. [PMID: 35468004 PMCID: PMC9255715 DOI: 10.1152/physrev.00027.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/11/2022] [Accepted: 04/16/2022] [Indexed: 01/17/2023] Open
Abstract
Adhesion G protein-coupled receptors (AGPCRs) are a family of 33 receptors in humans exhibiting a conserved general structure but diverse expression patterns and physiological functions. The large NH2 termini characteristic of AGPCRs confer unique properties to each receptor and possess a variety of distinct domains that can bind to a diverse array of extracellular proteins and components of the extracellular matrix. The traditional view of AGPCRs, as implied by their name, is that their core function is the mediation of adhesion. In recent years, though, many surprising advances have been made regarding AGPCR signaling mechanisms, activation by mechanosensory forces, and stimulation by small-molecule ligands such as steroid hormones and bioactive lipids. Thus, a new view of AGPCRs has begun to emerge in which these receptors are seen as massive signaling platforms that are crucial for the integration of adhesive, mechanosensory, and chemical stimuli. This review article describes the recent advances that have led to this new understanding of AGPCR function and also discusses new insights into the physiological actions of these receptors as well as their roles in human disease.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
4
|
ADGRL1 haploinsufficiency causes a variable spectrum of neurodevelopmental disorders in humans and alters synaptic activity and behavior in a mouse model. Am J Hum Genet 2022; 109:1436-1457. [PMID: 35907405 PMCID: PMC9388395 DOI: 10.1016/j.ajhg.2022.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/22/2022] [Indexed: 02/06/2023] Open
Abstract
ADGRL1 (latrophilin 1), a well-characterized adhesion G protein-coupled receptor, has been implicated in synaptic development, maturation, and activity. However, the role of ADGRL1 in human disease has been elusive. Here, we describe ten individuals with variable neurodevelopmental features including developmental delay, intellectual disability, attention deficit hyperactivity and autism spectrum disorders, and epilepsy, all heterozygous for variants in ADGRL1. In vitro, human ADGRL1 variants expressed in neuroblastoma cells showed faulty ligand-induced regulation of intracellular Ca2+ influx, consistent with haploinsufficiency. In vivo, Adgrl1 was knocked out in mice and studied on two genetic backgrounds. On a non-permissive background, mice carrying a heterozygous Adgrl1 null allele exhibited neurological and developmental abnormalities, while homozygous mice were non-viable. On a permissive background, knockout animals were also born at sub-Mendelian ratios, but many Adgrl1 null mice survived gestation and reached adulthood. Adgrl1-/- mice demonstrated stereotypic behaviors, sexual dysfunction, bimodal extremes of locomotion, augmented startle reflex, and attenuated pre-pulse inhibition, which responded to risperidone. Ex vivo synaptic preparations displayed increased spontaneous exocytosis of dopamine, acetylcholine, and glutamate, but Adgrl1-/- neurons formed synapses in vitro poorly. Overall, our findings demonstrate that ADGRL1 haploinsufficiency leads to consistent developmental, neurological, and behavioral abnormalities in mice and humans.
Collapse
|
5
|
Thwarting of Lphn3 Functions in Cell Motility and Signaling by Cancer-Related GAIN Domain Somatic Mutations. Cells 2022; 11:cells11121913. [PMID: 35741042 PMCID: PMC9221416 DOI: 10.3390/cells11121913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/21/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer progression relies on cellular transition states accompanied by changes in the functionality of adhesion molecules. The gene for adhesion G protein-coupled receptor latrophilin-3 (aGPCR Lphn3 or ADGRL3) is targeted by tumor-specific somatic mutations predominantly affecting the conserved GAIN domain where most aGPCRs are cleaved. However, it is unclear how these GAIN domain-altering mutations impact Lphn3 function. Here, we studied Lphn3 cancer-related mutations as a proxy for revealing unknown GAIN domain functions. We found that while intra-GAIN cleavage efficiency was unaltered, most mutations produced a ligand-specific impairment of Lphn3 intercellular adhesion profile paralleled by an increase in cell-matrix actin-dependent contact structures for cells expressing the select S810L mutation. Aberrant remodeling of the intermediate filament vimentin, which was found to coincide with Lphn3-induced modification of nuclear morphology, had less impact on the nuclei of S810L expressing cells. Notoriously, receptor signaling through G13 protein was deficient for all variants bearing non-homologous amino acid substitutions, including the S810L variant. Analysis of cell migration paradigms revealed a non-cell-autonomous impairment in collective cell migration indistinctly of Lphn3 or its cancer-related variants expression, while cell-autonomous motility was potentiated in the presence of Lphn3, but this effect was abolished in S810L GAIN mutant-expressing cells. These data identify the GAIN domain as an important regulator of Lphn3-dependent cell motility, thus furthering our understanding of cellular and molecular events linking Lphn3 genetic somatic mutations to cancer-relevant pathogenesis mechanisms.
Collapse
|
6
|
Petschner P, Baksa D, Hullam G, Torok D, Millinghoffer A, Deakin JFW, Bagdy G, Juhasz G. A replication study separates polymorphisms behind migraine with and without depression. PLoS One 2021; 16:e0261477. [PMID: 34972135 PMCID: PMC8719675 DOI: 10.1371/journal.pone.0261477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022] Open
Abstract
The largest migraine genome-wide association study identified 38 candidate loci. In this study we assessed whether these results replicate on a gene level in our European cohort and whether effects are altered by lifetime depression. We tested SNPs of the loci and their vicinity with or without interaction with depression in regression models. Advanced analysis methods such as Bayesian relevance analysis and a neural network based classifier were used to confirm findings. Main effects were found for rs2455107 of PRDM16 (OR = 1.304, p = 0.007) and five intergenic polymorphisms in 1p31.1 region: two of them showed risk effect (OR = 1.277, p = 0.003 for both rs11209657 and rs6686879), while the other three variants were protective factors (OR = 0.4956, p = 0.006 for both rs12090642 and rs72948266; OR = 0.4756, p = 0.005 for rs77864828). Additionally, 26 polymorphisms within ADGRL2, 2 in REST, 1 in HPSE2 and 33 mostly intergenic SNPs from 1p31.1 showed interaction effects. Among clumped results representing these significant regions, only rs11163394 of ADGRL2 showed a protective effect (OR = 0.607, p = 0.002), all other variants were risk factors (rs1043215 of REST with the strongest effect: OR = 6.596, p = 0.003). Bayesian relevance analysis confirmed the relevance of intergenic rs6660757 and rs12128399 (p31.1), rs1043215 (REST), rs1889974 (HPSE2) and rs11163394 (ADGRL2) from depression interaction results, and the moderate relevance of rs77864828 and rs2455107 of PRDM16 from main effect analysis. Both main and interaction effect SNPs could enhance predictive power with the neural network based classifier. In summary, we replicated p31.1, PRDM16, REST, HPSE2 and ADGRL2 genes with classic genetic and advanced analysis methods. While the p31.1 region and PRDM16 are worthy of further investigations in migraine in general, REST, HPSE2 and ADGRL2 may be prime candidates behind migraine pathophysiology in patients with comorbid depression.
Collapse
Affiliation(s)
- Peter Petschner
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Gokasho, Uji, Kyoto, Japan
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Daniel Baksa
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- SE-NAP2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabor Hullam
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
| | - Andras Millinghoffer
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, Budapest, Hungary
- NAP-2-SE New Antidepressant Target Research Group, Semmelweis University, Budapest, Hungary
| | - J. F. William Deakin
- Neuroscience and Psychiatry Unit, Division of Neuroscience and Experimental Psychology, The University of Manchester and Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- NAP-2-SE New Antidepressant Target Research Group, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- SE-NAP2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Regan SL, Pitzer EM, Hufgard JR, Sugimoto C, Williams MT, Vorhees CV. A novel role for the ADHD risk gene latrophilin-3 in learning and memory in Lphn3 knockout rats. Neurobiol Dis 2021; 158:105456. [PMID: 34352385 PMCID: PMC8440465 DOI: 10.1016/j.nbd.2021.105456] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/21/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Latrophilins (LPHNs) are adhesion G protein-coupled receptors with three isoforms but only LPHN3 is brain specific (caudate, prefrontal cortex, dentate, amygdala, and cerebellum). Variants of LPHN3 are associated with ADHD. Null mutations of Lphn3 in rat, mouse, zebrafish, and Drosophila result in hyperactivity, but its role in learning and memory (L&M) is largely unknown. Using our Lphn3 knockout (KO) rats we examined the cognitive abilities, long-term potentiation (LTP) in CA1, NMDA receptor expression, and neurohistology from heterozygous breeding pairs. KO rats were impaired in egocentric L&M in the Cincinnati water maze, spatial L&M and cognitive flexibility in the Morris water maze (MWM), with no effects on conditioned freezing, novel object recognition, or temporal order recognition. KO-associated locomotor hyperactivity had no effect on swim speed. KO rats had reduced early-LTP but not late-LTP and had reduced hippocampal NMDA-NR1 expression. In a second experiment, KO rats responded to a light prepulse prior to an acoustic startle pulse, reflecting visual signal detection. In a third experiment, KO rats given extra MWM pretraining and hidden platform overtraining showed no evidence of reaching WT rats' levels of learning. Nissl histology revealed no structural abnormalities in KO rats. LPHN3 has a selective effect on egocentric and allocentric L&M without effects on conditioned freezing or recognition memory.
Collapse
Affiliation(s)
- Samantha L Regan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| | - Emily M Pitzer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| | - Jillian R Hufgard
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| |
Collapse
|
8
|
Rosa M, Noel T, Harris M, Ladds G. Emerging roles of adhesion G protein-coupled receptors. Biochem Soc Trans 2021; 49:1695-1709. [PMID: 34282836 PMCID: PMC8421042 DOI: 10.1042/bst20201144] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022]
Abstract
Adhesion G protein-coupled receptors (aGPCRs) form a sub-group within the GPCR superfamily. Their distinctive structure contains an abnormally large N-terminal, extracellular region with a GPCR autoproteolysis-inducing (GAIN) domain. In most aGPCRs, the GAIN domain constitutively cleaves the receptor into two fragments. This process is often required for aGPCR signalling. Over the last two decades, much research has focussed on aGPCR-ligand interactions, in an attempt to deorphanize the family. Most ligands have been found to bind to regions N-terminal to the GAIN domain. These receptors may bind a variety of ligands, ranging across membrane-bound proteins and extracellular matrix components. Recent advancements have revealed a conserved method of aGPCR activation involving a tethered ligand within the GAIN domain. Evidence for this comes from increased activity in receptor mutants exposing the tethered ligand. As a result, G protein-coupling partners of aGPCRs have been more extensively characterised, making use of their tethered ligand to create constitutively active mutants. This has led to demonstrations of aGPCR function in, for example, neurodevelopment and tumour growth. However, questions remain around the ligands that may bind many aGPCRs, how this binding is translated into changes in the GAIN domain, and the exact mechanism of aGPCR activation following GAIN domain conformational changes. This review aims to examine the current knowledge around aGPCR activation, including ligand binding sites, the mechanism of GAIN domain-mediated receptor activation and how aGPCR transmembrane domains may relate to activation. Other aspects of aGPCR signalling will be touched upon, such as downstream effectors and physiological roles.
Collapse
Affiliation(s)
- Matthew Rosa
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Timothy Noel
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| |
Collapse
|
9
|
Regan SL, Williams MT, Vorhees CV. Latrophilin-3 disruption: Effects on brain and behavior. Neurosci Biobehav Rev 2021; 127:619-629. [PMID: 34022279 DOI: 10.1016/j.neubiorev.2021.04.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/22/2022]
Abstract
Latrophilin-3 (LPHN3), a G-protein-coupled receptor belonging to the adhesion subfamily, is a regulator of synaptic function and maintenance in brain regions that mediate locomotor activity, attention, and memory for location and path. Variants of LPHN3 are associated with increased risk for attention deficit hyperactivity disorder (ADHD) in some patients. Here we review the role of LPHN3 in the central nervous system (CNS). We describe synaptic localization of LPHN3, its trans-synaptic binding partners, links to neurodevelopmental disorders, animal models of Lphn3 disruption in different species, and evidence that LPHN3 is involved in cognition as well as activity and attention. The evidence shows that LPHN3 plays a more significant role in neuroplasticity than previously appreciated.
Collapse
Affiliation(s)
- Samantha L Regan
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Michael T Williams
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Charles V Vorhees
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| |
Collapse
|
10
|
Morgan RK, Anderson GR, Araç D, Aust G, Balenga N, Boucard A, Bridges JP, Engel FB, Formstone CJ, Glitsch MD, Gray RS, Hall RA, Hsiao CC, Kim HY, Knierim AB, Kusuluri DK, Leon K, Liebscher I, Piao X, Prömel S, Scholz N, Srivastava S, Thor D, Tolias KF, Ushkaryov YA, Vallon M, Van Meir EG, Vanhollebeke B, Wolfrum U, Wright KM, Monk KR, Mogha A. The expanding functional roles and signaling mechanisms of adhesion G protein-coupled receptors. Ann N Y Acad Sci 2019; 1456:5-25. [PMID: 31168816 PMCID: PMC7891679 DOI: 10.1111/nyas.14094] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022]
Abstract
The adhesion class of G protein-coupled receptors (GPCRs) is the second largest family of GPCRs (33 members in humans). Adhesion GPCRs (aGPCRs) are defined by a large extracellular N-terminal region that is linked to a C-terminal seven transmembrane (7TM) domain via a GPCR-autoproteolysis inducing (GAIN) domain containing a GPCR proteolytic site (GPS). Most aGPCRs undergo autoproteolysis at the GPS motif, but the cleaved fragments stay closely associated, with the N-terminal fragment (NTF) bound to the 7TM of the C-terminal fragment (CTF). The NTFs of most aGPCRs contain domains known to be involved in cell-cell adhesion, while the CTFs are involved in classical G protein signaling, as well as other intracellular signaling. In this workshop report, we review the most recent findings on the biology, signaling mechanisms, and physiological functions of aGPCRs.
Collapse
Affiliation(s)
- Rory K. Morgan
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - Garret R. Anderson
- Department of Molecular, Cell and Systems Biology, University of California – Riverside, Riverside, California
| | - Demet Araç
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - Gabriela Aust
- Research Laboratories, Department of Surgery, Leipzig University, Leipzig, Germany
| | - Nariman Balenga
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
- Program in Molecular and Structural Biology, Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, Baltimore, Maryland
| | - Antony Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, México
| | - James P. Bridges
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, Ohio
- Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Felix B. Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Caroline J. Formstone
- Centre for Developmental Neurobiology, Guys Campus, Kings College London, London, UK
- Department of Biological and Environmental Sciences, College Lane Campus, University of Hertfordshire, Hatfield, UK
| | - Maike D. Glitsch
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Ryan S. Gray
- Department of Pediatrics, University of Texas at Austin, Dell Medical School, Austin, Texas
| | - Randy A. Hall
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Alexander B. Knierim
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Deva Krupakar Kusuluri
- Institute of Molecular Physiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Katherine Leon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, Illinois
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Xianhua Piao
- Newborn Brain Research Institute, Department of Pediatrics, University of California – San Francisco, San Francisco, California
| | - Simone Prömel
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Nicole Scholz
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Leipzig University, Leipzig, Germany
| | - Swati Srivastava
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | | | | | - Mario Vallon
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California
| | - Erwin G. Van Meir
- Laboratory of Molecular Neuro-Oncology, Departments of Neurosurgery and Hematology & Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wallonia, Belgium
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Kevin M. Wright
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - Kelly R. Monk
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| | - Amit Mogha
- Vollum Institute, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
11
|
Scholz N, Langenhan T, Schöneberg T. Revisiting the classification of adhesion GPCRs. Ann N Y Acad Sci 2019; 1456:80-95. [PMID: 31365134 PMCID: PMC6900090 DOI: 10.1111/nyas.14192] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/19/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
Abstract
G protein–coupled receptors (GPCRs) are encoded by over 800 genes in the human genome. Motivated by different scientific rationales, the two classification systems that are mainly in use, the ABC and GRAFS systems, organize GPCRs according to their pharmacological features and phylogenetic relations, respectively. Within those systems, adhesion GPCRs (aGPCRs) constitute a group of over 30 mammalian homologs, most of which are still orphans with undefined activating signals and signal transduction properties. Previous efforts have further subdivided mammalian aGPCRs into nine subfamilies to indicate phylogenetic relationships. However, this subclassification scheme has shortcomings and inconsistencies that require attention. Here, we have reassessed the phylogenetic relationships of aGPCRs from vertebrate and invertebrate species. Our findings confirm that secretin receptor–like GPCRs most probably emerged from ancestral aGPCRs. We show that reassignment of several aGPCRs to families essentially requires input from functional data. Our analyses establish the need for introducing novel aGPCR subfamilies due to aGPCR sequences from invertebrate species that are not readily assignable to any existing subfamily. We conclude that the current classification systems ought to be updated to consider an unambiguous taxonomy of a hierarchically organized classification and pharmacological properties, and to accommodate phylogenetic affiliations between aGPCR genes within mammals and across the animal kingdom.
Collapse
Affiliation(s)
- Nicole Scholz
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Tobias Langenhan
- Division of General Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Torsten Schöneberg
- Division of Molecular Biochemistry, Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| |
Collapse
|
12
|
Genetic basis of functional variability in adhesion G protein-coupled receptors. Sci Rep 2019; 9:11036. [PMID: 31363148 PMCID: PMC6667449 DOI: 10.1038/s41598-019-46265-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
The enormous sizes of adhesion G protein-coupled receptors (aGPCRs) go along with complex genomic exon-intron architectures giving rise to multiple mRNA variants. There is a need for a comprehensive catalog of aGPCR variants for proper evaluation of the complex functions of aGPCRs found in structural, in vitro and animal model studies. We used an established bioinformatics pipeline to extract, quantify and visualize mRNA variants of aGPCRs from deeply sequenced transcriptomes. Data analysis showed that aGPCRs have multiple transcription start sites even within introns and that tissue-specific splicing is frequent. On average, 19 significantly expressed transcript variants are derived from a given aGPCR gene. The domain architecture of the N terminus encoded by transcript variants often differs and N termini without or with an incomplete seven-helix transmembrane anchor as well as separate seven-helix transmembrane domains are frequently derived from aGPCR genes. Experimental analyses of selected aGPCR transcript variants revealed marked functional differences. Our analysis has an impact on a rational design of aGPCR constructs for structural analyses and gene-deficient mouse lines and provides new support for independent functions of both, the large N terminus and the transmembrane domain of aGPCRs.
Collapse
|
13
|
Ovando-Zambrano JC, Arias-Montaño JA, Boucard AA. Alternative splicing event modifying ADGRL1/latrophilin-1 cytoplasmic tail promotes both opposing and dual cAMP signaling pathways. Ann N Y Acad Sci 2019; 1456:168-185. [PMID: 31339586 DOI: 10.1111/nyas.14198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/11/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Abstract
The adhesion G protein-coupled receptor ADGRL1/latrophilin-1 (LPHN1) stabilizes synapse formation through heterophilic interactions. A growing consensus is pointing to the role of LPHN1 in modulating intracellular levels of cAMP, although conflicting data exist. Variants of LPHN1 resulting from alternative splicing differ at multiple sites, two of which, designated as SSA and SSB, modify extracellular and intracellular receptor regions, respectively. While SSA splicing modulates receptor-ligand affinity, the function of SSB splicing remains elusive. Here, we explored the role of SSB in an attempt to unify current findings on LPHN1 signaling pathways by testing SSB-containing and SSB-deficient receptor variants in signaling paradigms involving interaction with their ligands neurexin and FLRT. cAMP competitive binding assays revealed that cells expressing either receptor variant exhibited a ligand-dependent decrease in the forskolin-induced cAMP accumulation. Surprisingly, the expression of SSB-containing LPHN1 promoted both constitutive and ligand-dependent cAMP production, whereas SSB-deficient LPHN1 did not. Pertussis toxin treatment unveiled a constitutive coupling to Gαi/o for SSB-containing LPHN1 while abrogating the ligand-mediated activation of Gαs . Importantly, neither receptor variant increased the intracellular concentration of Ca2+ nor MAP kinase activation in the presence of ligands. These results suggest that SSB splicing selectively affects the duality of LPHN1 signaling toward opposing cAMP pathways.
Collapse
Affiliation(s)
- José-Carlos Ovando-Zambrano
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - Antony A Boucard
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| |
Collapse
|
14
|
Moreno-Salinas AL, Avila-Zozaya M, Ugalde-Silva P, Hernández-Guzmán DA, Missirlis F, Boucard AA. Latrophilins: A Neuro-Centric View of an Evolutionary Conserved Adhesion G Protein-Coupled Receptor Subfamily. Front Neurosci 2019; 13:700. [PMID: 31354411 PMCID: PMC6629964 DOI: 10.3389/fnins.2019.00700] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
The adhesion G protein-coupled receptors latrophilins have been in the limelight for more than 20 years since their discovery as calcium-independent receptors for α-latrotoxin, a spider venom toxin with potent activity directed at neurotransmitter release from a variety of synapse types. Latrophilins are highly expressed in the nervous system. Although a substantial amount of studies has been conducted to describe the role of latrophilins in the toxin-mediated action, the recent identification of endogenous ligands for these receptors helped confirm their function as mediators of adhesion events. Here we hypothesize a role for latrophilins in inter-neuronal contacts and the formation of neuronal networks and we review the most recent information on their role in neurons. We explore molecular, cellular and behavioral aspects related to latrophilin adhesion function in mice, zebrafish, Drosophila melanogaster and Caenorhabditis elegans, in physiological and pathophysiological conditions, including autism spectrum, bipolar, attention deficit and hyperactivity and substance use disorders.
Collapse
Affiliation(s)
- Ana L. Moreno-Salinas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Monserrat Avila-Zozaya
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Paul Ugalde-Silva
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - David A. Hernández-Guzmán
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Fanis Missirlis
- Department of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| | - Antony A. Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
15
|
Sita LV, Diniz GB, Horta-Junior JAC, Casatti CA, Bittencourt JC. Nomenclature and Comparative Morphology of the Teneurin/TCAP/ADGRL Protein Families. Front Neurosci 2019; 13:425. [PMID: 31130838 PMCID: PMC6510184 DOI: 10.3389/fnins.2019.00425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/15/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Luciane V. Sita
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Giovanne B. Diniz
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - José A. C. Horta-Junior
- Department of Anatomy, Institute of Biosciences, São Paulo State University, São Paulo, Brazil
| | - Claudio A. Casatti
- Department of Basic Sciences, São Paulo State University, São Paulo, Brazil
| | - Jackson C. Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Center for Neuroscience and Behavior, Department of Experimental Psychology, Institute of Psychology, University of São Paulo, São Paulo, Brazil
- *Correspondence: Jackson C. Bittencourt,
| |
Collapse
|
16
|
Cruz-Ortega JS, Boucard AA. Actin cytoskeleton remodeling defines a distinct cellular function for adhesion G protein-coupled receptors ADGRL/latrophilins 1, 2 and 3. Biol Open 2019; 8:bio.039826. [PMID: 30926595 PMCID: PMC6503996 DOI: 10.1242/bio.039826] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Latrophilins represent a subgroup of the adhesion G protein-coupled receptor family, which bind to actin-associated scaffolding proteins. They are expressed in various tissues, suggesting that they might participate in biological processes that are ubiquitous. Here we focus on actin cytoskeleton dynamics to explore the role of latrophilins in mammalian cells. Individual overexpression of each latrophilin isoform comparably increased cell volume while modifying the net profile of F-actin-dependent cell extensions, as evaluated by confocal microscopy analysis. Latrophilin deletion mutants evidenced that direct coupling to the intracellular machinery was a requirement for modulating cell extensions. The association between latrophilins and the actin cytoskeleton was detected by co-immunoprecipitation assays and corroborated with immunocytochemistry analysis. Consistent with the destabilization of F-actin structures, latrophilin isoforms constitutively induced a prominent increase in the activity of actin-depolymerizing factor, cofilin. Intercellular adhesion events stabilized by heterophilic Teneurin-4 trans-interactions disrupted latrophilin colocalization with F-actin and led to an isoform-specific rescue of cell extensions. Thus, we find that the actin cytoskeleton machinery constitutes an important component of constitutive as well as ligand-induced signaling for latrophilins. This article has an associated First Person interview with the first author of the paper. Summary: Synapses involve the adhesion function of latrophilins within existing neuronal extensions. We show that latrophilins engage the actin cytoskeleton, both constitutively and upon ligand stimulation, to dictate cell extension patterns.
Collapse
Affiliation(s)
- Judith S Cruz-Ortega
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City 07360, México
| | - Antony A Boucard
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México City 07360, México
| |
Collapse
|
17
|
Vysokov NV, Silva JP, Lelianova VG, Suckling J, Cassidy J, Blackburn JK, Yankova N, Djamgoz MB, Kozlov SV, Tonevitsky AG, Ushkaryov YA. Proteolytically released Lasso/teneurin-2 induces axonal attraction by interacting with latrophilin-1 on axonal growth cones. eLife 2018; 7:37935. [PMID: 30457553 PMCID: PMC6245728 DOI: 10.7554/elife.37935] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 10/19/2018] [Indexed: 11/15/2022] Open
Abstract
A presynaptic adhesion G-protein-coupled receptor, latrophilin-1, and a postsynaptic transmembrane protein, Lasso/teneurin-2, are implicated in trans-synaptic interaction that contributes to synapse formation. Surprisingly, during neuronal development, a substantial proportion of Lasso is released into the intercellular space by regulated proteolysis, potentially precluding its function in synaptogenesis. We found that released Lasso binds to cell-surface latrophilin-1 on axonal growth cones. Using microfluidic devices to create stable gradients of soluble Lasso, we show that it induces axonal attraction, without increasing neurite outgrowth. Using latrophilin-1 knockout in mice, we demonstrate that latrophilin-1 is required for this effect. After binding latrophilin-1, Lasso causes downstream signaling, which leads to an increase in cytosolic calcium and enhanced exocytosis, processes that are known to mediate growth cone steering. These findings reveal a novel mechanism of axonal pathfinding, whereby latrophilin-1 and Lasso mediate both short-range interaction that supports synaptogenesis, and long-range signaling that induces axonal attraction. The brain is a complex mesh of interconnected neurons, with each cell making tens, hundreds, or even thousands of connections. These links can stretch over long distances, and establishing them correctly during development is essential. Developing neurons send out long and thin structures, called axons, to reach distant cells. To guide these growing axons, neurons release molecules that work as traffic signals: some attract axons whilst others repel them, helping the burgeoning structures to twist and turn along their travel paths. When an axon reaches its target cell, the two cells join to each other by forming a structure called a synapse. To make the connection, surface proteins on the axon latch onto matching proteins on the target cell, zipping up the synapse. There are many different types of synapses in the brain, but we only know a few of the surface molecules involved in their creation – not enough to explain synaptic variety. Two of these surface proteins are latrophilin-1, which is produced by the growing axon, and Lasso, which sits on the membrane of the target cell. The two proteins interact strongly, anchoring the axon to the target cell and allowing the synapse to form. However, a previous recent discovery by Vysokov et al. has revealed that an enzyme can also cut Lasso from the membrane of the target cell. The ‘free’ protein can still interact with latrophilin-1, but as it is shed by the target cell, it can no longer serve as an anchor for the synapse. Could it be that free Lasso acts as a traffic signal instead? Here, Vysokov et al. tried to answer this by growing neurons from a part of the brain called the hippocampus in a special labyrinth dish. When free Lasso was gradually introduced in the culture through microscopic channels, it interacted with latrophilin-1 on the surface of the axons. This triggered internal changes that led the axons to add more membrane where they had sensed Lasso, making them grow towards the source of the signal. The results demonstrate that a target cell can both carry and release Lasso, using this duplicitous protein to help attract growing axons as well as anchor them. The work by Vysokov et al. contributes to our knowledge of how neurons normally connect, which could shed light on how this process can go wrong. This may be relevant to understand conditions such as schizophrenia and ADHD, where patients’ brains often show incorrect wiring.
Collapse
Affiliation(s)
- Nickolai V Vysokov
- School of Pharmacy, University of Kent, Chatham, United Kingdom.,Department of Life Sciences, Imperial College London, London, United Kingdom.,Wolfson Centre for Age Related Diseases, King's College London, London, United Kingdom.,BrainPatch Ltd, London, United Kingdom
| | - John-Paul Silva
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Department of Bioanalytical Sciences, Non-clinical development, UCB-Pharma, Berkshire, United Kingdom
| | - Vera G Lelianova
- School of Pharmacy, University of Kent, Chatham, United Kingdom.,Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Jason Suckling
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Thomsons Online Benefits, London, United Kingdom
| | - John Cassidy
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Arix Bioscience, London, United Kingdom
| | - Jennifer K Blackburn
- School of Pharmacy, University of Kent, Chatham, United Kingdom.,Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, United States
| | - Natalia Yankova
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Mustafa Ba Djamgoz
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Serguei V Kozlov
- Center for Advanced Preclinical Research, National Cancer Institute, Frederick, United States
| | - Alexander G Tonevitsky
- Higher School of Economics, Moscow, Russia.,Scientific Research Centre Bioclinicum, Moscow, Russia
| | - Yuri A Ushkaryov
- School of Pharmacy, University of Kent, Chatham, United Kingdom.,Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Dark C, Homman-Ludiye J, Bryson-Richardson RJ. The role of ADHD associated genes in neurodevelopment. Dev Biol 2018; 438:69-83. [DOI: 10.1016/j.ydbio.2018.03.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/04/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
|
19
|
Gao S, Xiong W, Wei L, Liu J, Liu X, Xie J, Song X, Bi J, Li B. Transcriptome profiling analysis reveals the role of latrophilin in controlling development, reproduction and insecticide susceptibility in Tribolium castaneum. Genetica 2018; 146:287-302. [DOI: 10.1007/s10709-018-0020-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/30/2018] [Indexed: 12/23/2022]
|
20
|
Cazorla-Vázquez S, Engel FB. Adhesion GPCRs in Kidney Development and Disease. Front Cell Dev Biol 2018; 6:9. [PMID: 29468160 PMCID: PMC5808184 DOI: 10.3389/fcell.2018.00009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/23/2018] [Indexed: 12/25/2022] Open
Abstract
Chronic kidney disease (CKD) represents the fastest growing pathology worldwide with a prevalence of >10% in many countries. In addition, kidney cancer represents 5% of all new diagnosed cancers. As currently no effective therapies exist to restore kidney function after CKD- as well as cancer-induced renal damage, it is important to elucidate new regulators of kidney development and disease as new therapeutic targets. G protein-coupled receptors (GPCRs) represent the most successful class of pharmaceutical targets. In recent years adhesion GPCRs (aGPCRs), the second largest GPCR family, gained significant attention as they are present on almost all mammalian cells, are associated to a plethora of diseases and regulate important cellular processes. aGPCRs regulate for example cell polarity, mitotic spindle orientation, cell migration, and cell aggregation; all processes that play important roles in kidney development and/or disease. Moreover, polycystin-1, a major regulator of kidney development and disease, contains a GAIN domain, which is otherwise only found in aGPCRs. In this review, we assess the potential of aGPCRs as therapeutic targets for kidney disease. For this purpose we have summarized the available literature and analyzed data from the databases The Human Protein Atlas, EURExpress, Nephroseq, FireBrowse, cBioPortal for Cancer Genomics and the National Cancer Institute Genomic Data Commons data portal (NCIGDC). Our data indicate that most aGPCRs are expressed in different spatio-temporal patterns during kidney development and that altered aGPCR expression is associated with a variety of kidney diseases including CKD, diabetic nephropathy, lupus nephritis as well as renal cell carcinoma. We conclude that aGPCRs present a promising new class of therapeutic targets and/or might be useful as diagnostic markers in kidney disease.
Collapse
Affiliation(s)
- Salvador Cazorla-Vázquez
- Department of Nephropathology, Experimental Renal and Cardiovascular Research, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Felix B Engel
- Department of Nephropathology, Experimental Renal and Cardiovascular Research, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
21
|
Structure of a plant β-galactosidase C-terminal domain. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1411-8. [PMID: 27451952 DOI: 10.1016/j.bbapap.2016.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/17/2016] [Accepted: 07/19/2016] [Indexed: 02/02/2023]
Abstract
Most plant β-galactosidases, which belong to glycoside hydrolase family 35, have a C-terminal domain homologous to animal galactose and rhamnose-binding lectins. To investigate the structure and function of this domain, the C-terminal domain of the rice (Oryza sativa L.) β-galactosidase 1 (OsBGal1 Cter) was expressed in Escherichia coli and purified to homogeneity. The free OsBGal1 Cter is monomeric with a native molecular weight of 15kDa. NMR spectroscopy indicated that OsBGal1 Cter comprises five β-strands and one α-helix. The structure of this domain is similar to lectin domains from animals, but loops A and C of OsBGal1 Cter are longer than the corresponding loops from related animal lectins with known structures. In addition, loop A of OsBGal1 Cter was not well defined, suggesting it is flexible. Although OsBGal1 Cter was predicted to be a galactose/rhamnose-binding domain, binding with rhamnose, galactose, glucose, β-1,4-d-galactobiose and raffinose could not be observed in NMR experiments.
Collapse
|
22
|
Woelfle R, D’Aquila AL, Lovejoy DA. Teneurins, TCAP, and latrophilins: roles in the etiology of mood disorders. Transl Neurosci 2016; 7:17-23. [PMID: 28123817 PMCID: PMC5017594 DOI: 10.1515/tnsci-2016-0004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/11/2015] [Indexed: 12/26/2022] Open
Abstract
Mood disorders, including anxiety and depression, are thought to be characterized by disrupted neuronal synapses and altered brain plasticity. The etiology is complex, involving numerous regions of the brain, comprising a multitude of neurotransmitter and neuromodulator systems. Recently, new studies on the teneurins, an evolutionary ancient family of type II transmembrane proteins have been shown to interact with latrophilins (LPHN), a similarly phylogenetically old family of adhesion G protein-coupled receptors (GPCR) forming a transsynaptic adhesion and ligand-receptor pair. Each of the four teneurin proteins contains bioactive sequences termed the teneurin C-terminal associated peptides (TCAP-1-4), which possess a number of neuromodulatory effects. The primary structures of the TCAP are most closely similar to the corticotropin-releasing factor (CRF) family of peptides. CRF has been implicated in a number of diverse mood disorders. Via an association with dystroglycans, synthetic TCAP-1 administration to both embryonic and primary hippocampal cultures induces long-term changes in neuronal structure, specifically increased neurite outgrowth, dendritic branching, and axon growth. Rodent models treated with TCAP-1 show reduced anxiety responses in the elevated plus-maze, openfield test, and acoustic startle test and inhibited CRF-mediated cocaine-seeking behaviour. Thus the teneurin/TCAP-latrophilin interaction may play a major role in the origin, development and treatment of mood disorders.
Collapse
Affiliation(s)
- Rebecca Woelfle
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Andrea L. D’Aquila
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - David A. Lovejoy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
23
|
Krishnan A, Nijmeijer S, de Graaf C, Schiöth HB. Classification, Nomenclature, and Structural Aspects of Adhesion GPCRs. Handb Exp Pharmacol 2016; 234:15-41. [PMID: 27832482 DOI: 10.1007/978-3-319-41523-9_2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Representation of the nine distinct aGPCR subfamilies and their unique N-terminal domain architecture. The illustration also shows the extracellular structural feature shared by all aGPCRs (except ADGRA1), known as the GPCR autoproteolysis-inducing (GAIN) domain, that mediates autoproteolysis and subsequent attachment of the cleaved NTF and CTF fragments The adhesion family of G protein-coupled receptors (aGPCRs) is unique among all GPCR families with long N-termini and multiple domains that are implicated in cell-cell and cell-matrix interactions. Initially, aGPCRs in the human genome were phylogenetically classified into nine distinct subfamilies based on their 7TM sequence similarity. This phylogenetic grouping of genes into subfamilies was found to be in congruence in closely related mammals and other vertebrates as well. Over the years, aGPCR repertoires have been mapped in many species including model organisms, and, currently, there is a growing interest in exploring the pharmacological aspects of aGPCRs. Nonetheless, the aGPCR nomenclature has been highly diverse because experts in the field have used different names for different family members based on their characteristics (e.g., epidermal growth factor-seven-span transmembrane (EGF-TM7)), but without harmonization with regard to nomenclature efforts. In order to facilitate naming of orthologs and other genetic variants in different species in the future, the Adhesion-GPCR Consortium, together with the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification, proposed a unified nomenclature for aGPCRs. Here, we review the classification and the most recent/current nomenclature of aGPCRs and as well discuss the structural topology of the extracellular domain (ECD)/N-terminal fragment (NTF) that is comparable with this 7TM subfamily classification. Of note, we systematically describe the structural domains in the ECD of aGPCR subfamilies and highlight their role in aGPCR-protein interactions.
Collapse
Affiliation(s)
- Arunkumar Krishnan
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Biomedical Center, 593, Uppsala, 75 124, Sweden
| | - Saskia Nijmeijer
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, The Netherlands
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, Biomedical Center, 593, Uppsala, 75 124, Sweden.
| |
Collapse
|
24
|
Abstract
Epidermal growth factor, latrophilin and seven-transmembrane domain-containing 1 (ELTD1), an orphan G-protein-coupled receptor (GPCR) belonging to the adhesion GPCR family, has recently been identified as a potential cancer biomarker and a novel regulator of angiogenesis. In this mini-review, we present an overview of the current literature on ELTD1 and present bioinformatics data showing ELTD1's sequence conservation, its expression in cancer cell lines and its mutational frequency in human cancers. Additionally, we present sequence homology alignment results confirming ELTD1 to be a hybrid comprising motifs shared with individual members in both adhesion GPCR subfamilies 1 and 2. Finally, we discuss why tumour endothelial ELTD1 expression may confer a good prognosis yet still represent a therapeutic target.
Collapse
|
25
|
Woelfle R, D'Aquila AL, Pavlović T, Husić M, Lovejoy DA. Ancient interaction between the teneurin C-terminal associated peptides (TCAP) and latrophilin ligand-receptor coupling: a role in behavior. Front Neurosci 2015; 9:146. [PMID: 25964737 PMCID: PMC4408839 DOI: 10.3389/fnins.2015.00146] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/08/2015] [Indexed: 12/28/2022] Open
Abstract
Teneurins are multifunctional transmembrane proteins that are found in all multicellular animals and exist as four paralogous forms in vertebrates. They are highly expressed in the central nervous system, where they exert their effects, in part, by high-affinity binding to latrophilin (LPHN), a G-protein coupled receptor (GPCR) related to the adhesion and secretin GPCR families. The teneurin C-terminal associated peptides (TCAPs) are encoded by the terminal exon of all four teneurins, where TCAPs 1 and 3 are independently transcribed as soluble peptides, and TCAPs 2 and 4 remain tethered to their teneurin proprotein. Synthetic TCAP-1 interacts with LPHN, with an association with β-dystroglycan, to induce a tissue-dependent signal cascade to modulate cytoskeletal dynamics. TCAP-1 reduces stress-induced behaviors associated with anxiety, addiction and depression in a variety of models, in part, by regulating synaptic plasticity. Therefore, the TCAP-1-teneurin-LPHN interaction represents a novel receptor-ligand model and may represent a key mechanism underlying the association of behavior and neurological conditions.
Collapse
Affiliation(s)
- Rebecca Woelfle
- Department of Cell and Systems Biology, University of Toronto Toronto, ON, Canada
| | - Andrea L D'Aquila
- Department of Cell and Systems Biology, University of Toronto Toronto, ON, Canada
| | - Téa Pavlović
- Department of Cell and Systems Biology, University of Toronto Toronto, ON, Canada
| | - Mia Husić
- Department of Cell and Systems Biology, University of Toronto Toronto, ON, Canada
| | - David A Lovejoy
- Department of Cell and Systems Biology, University of Toronto Toronto, ON, Canada ; Protagenic Therapeutics Inc. New York, NY, USA
| |
Collapse
|
26
|
Casas E, Hessman BE, Keele JW, Ridpath JF. A genome-wide association study for the incidence of persistent bovine viral diarrhea virus infection in cattle. Anim Genet 2014; 46:8-15. [DOI: 10.1111/age.12239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2014] [Indexed: 01/19/2023]
Affiliation(s)
- E. Casas
- National Animal Disease Center; ARS; USDA; Ames IA USA
| | | | - J. W. Keele
- US Meat Animal Research Center; ARS; USDA; Clay Center NE USA
| | - J. F. Ridpath
- National Animal Disease Center; ARS; USDA; Ames IA USA
| |
Collapse
|
27
|
Fève M, Saliou JM, Zeniou M, Lennon S, Carapito C, Dong J, Van Dorsselaer A, Junier MP, Chneiweiss H, Cianférani S, Haiech J, Kilhoffer MC. Comparative expression study of the endo-G protein coupled receptor (GPCR) repertoire in human glioblastoma cancer stem-like cells, U87-MG cells and non malignant cells of neural origin unveils new potential therapeutic targets. PLoS One 2014; 9:e91519. [PMID: 24662753 PMCID: PMC3963860 DOI: 10.1371/journal.pone.0091519] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 02/10/2014] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas (GBMs) are highly aggressive, invasive brain tumors with bad prognosis and unmet medical need. These tumors are heterogeneous being constituted by a variety of cells in different states of differentiation. Among these, cells endowed with stem properties, tumor initiating/propagating properties and particularly resistant to chemo- and radiotherapies are designed as the real culprits for tumor maintenance and relapse after treatment. These cells, termed cancer stem-like cells, have been designed as prominent targets for new and more efficient cancer therapies. G-protein coupled receptors (GPCRs), a family of membrane receptors, play a prominent role in cell signaling, cell communication and crosstalk with the microenvironment. Their role in cancer has been highlighted but remains largely unexplored. Here, we report a descriptive study of the differential expression of the endo-GPCR repertoire in human glioblastoma cancer stem-like cells (GSCs), U-87 MG cells, human astrocytes and fetal neural stem cells (f-NSCs). The endo-GPCR transcriptome has been studied using Taqman Low Density Arrays. Of the 356 GPCRs investigated, 138 were retained for comparative studies between the different cell types. At the transcriptomic level, eight GPCRs were specifically expressed/overexpressed in GSCs. Seventeen GPCRs appeared specifically expressed in cells with stem properties (GSCs and f-NSCs). Results of GPCR expression at the protein level using mass spectrometry and proteomic analysis are also presented. The comparative GPCR expression study presented here gives clues for new pathways specifically used by GSCs and unveils novel potential therapeutic targets.
Collapse
Affiliation(s)
- Marie Fève
- Laboratoire d'Innovation Thérapeutique, UMR7200, Laboratoire d'Excellence Medalis, CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Jean-Michel Saliou
- Laboratoire de Spectrométrie de Masse BioOrganique, UMR7178, CNRS, Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Strasbourg, France
| | - Maria Zeniou
- Laboratoire d'Innovation Thérapeutique, UMR7200, Laboratoire d'Excellence Medalis, CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Sarah Lennon
- Laboratoire de Spectrométrie de Masse BioOrganique, UMR7178, CNRS, Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse BioOrganique, UMR7178, CNRS, Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Strasbourg, France
| | - Jihu Dong
- Laboratoire d'Innovation Thérapeutique, UMR7200, Laboratoire d'Excellence Medalis, CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Alain Van Dorsselaer
- Laboratoire de Spectrométrie de Masse BioOrganique, UMR7178, CNRS, Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Strasbourg, France
| | - Marie-Pierre Junier
- Neuroscience Paris Seine, UMR8246, Inserm U1130, Institut de Biologie Paris Seine, CNRS, Université Pierre et Marie Curie, Paris, France
| | - Hervé Chneiweiss
- Neuroscience Paris Seine, UMR8246, Inserm U1130, Institut de Biologie Paris Seine, CNRS, Université Pierre et Marie Curie, Paris, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, UMR7178, CNRS, Institut Pluridisciplinaire Hubert Curien, Université de Strasbourg, Strasbourg, France
| | - Jacques Haiech
- Laboratoire d'Innovation Thérapeutique, UMR7200, Laboratoire d'Excellence Medalis, CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Marie-Claude Kilhoffer
- Laboratoire d'Innovation Thérapeutique, UMR7200, Laboratoire d'Excellence Medalis, CNRS, Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| |
Collapse
|
28
|
Anholt RRH. Olfactomedin proteins: central players in development and disease. Front Cell Dev Biol 2014; 2:6. [PMID: 25364714 PMCID: PMC4206993 DOI: 10.3389/fcell.2014.00006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/07/2014] [Indexed: 12/14/2022] Open
Abstract
Olfactomedin proteins are characterized by a conserved domain of \texorpdfstring~\textasciitilde250 amino acids corresponding to the olfactomedin archetype first discovered in olfactory neuroepithelium. They arose early in evolution and occur throughout the animal kingdom. In mice and humans olfactomedin proteins comprise a diverse array of glycoproteins, many of which are critical for early development and functional organization of the nervous system as well as hematopoiesis. Olfactomedin domains appear to facilitate protein-protein interactions, intercellular interactions, and cell adhesion. Several members of the family have been implicated in various common diseases, notably myocilin in glaucoma and OLFM4 in cancer. This review highlights this important, hitherto understudied family of proteins.
Collapse
Affiliation(s)
- Robert R. H. Anholt
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, North Carolina State UniversityRaleigh, NC, USA
| |
Collapse
|
29
|
Patel MV, Hallal DA, Jones JW, Bronner DN, Zein R, Caravas J, Husain Z, Friedrich M, Vanberkum MFA. Dramatic expansion and developmental expression diversification of the methuselah gene family during recent Drosophila evolution. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2012; 318:368-87. [PMID: 22711569 DOI: 10.1002/jez.b.22453] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Functional studies of the methuselah/methuselah-like (mth/mthl) gene family have focused on the founding member mth, but little is known regarding the developmental functions of this receptor or any of its paralogs. We undertook a comprehensive analysis of developmental expression and sequence divergence in the mth/mthl gene family. Using in situ hybridization techniques, we detect expression of six genes (mthl1, 5, 9, 11, 13, and 14) in the embryo during gastrulation and development of the gut, heart, and lymph glands. Four receptors (mthl3, 4, 6, and 8) are expressed in the larval central nervous system, imaginal discs, or both, and two receptors (mthl10 and mth) are expressed in both embryos and larvae. Phylogenetic analysis of all mth/mthl genes in five Drosophila species, mosquito and flour beetle structured the mth/mthl family into several subclades. mthl1, 5, and 14 are present in most species, each forming a separate clade. A newly identified Drosophila mthl gene (CG31720; herein mthl15) formed another ancient clade. The remaining Drosophila receptors, including mth, are members of a large "superclade" that diversified relatively recently during dipteran evolution, in many cases within the melanogaster subgroup. Comparing the expression patterns of the mth/mthl "superclade" paralogs to the embryonic expression of the singleton ortholog in Tribolium suggests both subfunctionalization and acquisition of novel functionalities. Taken together, our findings shed novel light on mth as a young member of an adaptively evolving developmental gene family.
Collapse
Affiliation(s)
- Meghna V Patel
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wallis D, Hill DS, Mendez IA, Abbott LC, Finnell RH, Wellman PJ, Setlow B. Initial characterization of mice null for Lphn3, a gene implicated in ADHD and addiction. Brain Res 2012; 1463:85-92. [PMID: 22575564 DOI: 10.1016/j.brainres.2012.04.053] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/26/2012] [Accepted: 04/28/2012] [Indexed: 02/05/2023]
Abstract
The LPHN3 gene has been associated with both attention deficit-hyperactivity disorder (ADHD) and addiction, suggesting that it may play a role in the etiology of these disorders. Unfortunately, almost nothing is known about the normal functions of this gene, which has hampered understanding of its potential pathogenic role. To begin to characterize such normal functions, we utilized a gene-trap embryonic stem cell line to generate mice mutant for the Lphn3 gene. We evaluated differential gene expression in whole mouse brain between mutant and wild type male littermates at postnatal day 0 using TaqMan gene expression assays. Most notably, we found changes in dopamine and serotonin receptors and transporters (Dat1, Drd4, 5Htt, 5Ht2a), changes in neurotransmitter metabolism genes (Th, Gad1), as well as changes in neural developmental genes (Nurr, Ncam). When mice were examined at 4-6 weeks of age, null mutants showed increased levels of dopamine and serotonin in the dorsal striatum. Finally, null mutant mice had a hyperactive phenotype in the open field test, independent of sex, and were more sensitive to the locomotor stimulant effects of cocaine. Considered together, these results suggest that Lphn3 plays a role in development and/or regulation of monoamine signaling. Given the central role for monoamines in ADHD and addiction, it seems likely that the influence of LPHN3 genotype on these disorders is mediated through alterations in monoamine signaling.
Collapse
Affiliation(s)
- Deeann Wallis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station 77843-3474, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Näreoja K, Näsman J. Selective targeting of G-protein-coupled receptor subtypes with venom peptides. Acta Physiol (Oxf) 2012; 204:186-201. [PMID: 21481193 DOI: 10.1111/j.1748-1716.2011.02305.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The G-protein-coupled receptor (GPCR) family is one of the largest gene superfamilies with approx. 370 members responding to endogenous ligands in humans and a roughly equal amount of receptors sensitive to external stimuli from the surrounding. A number of receptors from this superfamily are well recognized targets for medical treatment of various disease conditions, whereas for many others the potential medical benefit of interference is still obscure. A general problem associated with GPCR research and therapeutics is the insufficient specificity of available ligands to differentiate between closely homologous receptor subtypes. In this context, venom peptides could make a significant contribution to the development of more specific drugs. Venoms from certain animals specialized in biochemical hunting contain a mixture of molecules that are directed towards a variety of membrane proteins. Peptide toxins isolated from these mixtures usually exhibit high specificity for their targets. Muscarinic toxins found from mamba snakes attracted much attention during the 1990s. These are 65-66 amino acid long peptides with a structural three-finger folding similar to the α-neurotoxins and they target the muscarinic acetylcholine receptors in a subtype-selective manner. Recently, several members of the three-finger toxins from mamba snakes as well as conotoxins from marine cone snails have been shown to selectively interact with subtypes of adrenergic receptors. In this review, we will discuss the GPCR-directed peptide toxins found from different venoms and how some of these can be useful in exploring specific roles of receptor subtypes.
Collapse
Affiliation(s)
- K Näreoja
- Department of Biosciences, Biochemistry, Åbo Akademi University, Turku, Finland
| | | |
Collapse
|
32
|
Latrophilin 1 and its endogenous ligand Lasso/teneurin-2 form a high-affinity transsynaptic receptor pair with signaling capabilities. Proc Natl Acad Sci U S A 2011; 108:12113-8. [PMID: 21724987 DOI: 10.1073/pnas.1019434108] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Latrophilin 1 (LPH1), a neuronal receptor of α-latrotoxin, is implicated in neurotransmitter release and control of presynaptic Ca(2+). As an "adhesion G-protein-coupled receptor," LPH1 can convert cell surface interactions into intracellular signaling. To examine the physiological functions of LPH1, we used LPH1's extracellular domain to purify its endogenous ligand. A single protein of ∼275 kDa was isolated from rat brain and termed Lasso. Peptide sequencing and molecular cloning have shown that Lasso is a splice variant of teneurin-2, a brain-specific orphan cell surface receptor with a function in neuronal pathfinding and synaptogenesis. We show that LPH1 and Lasso interact strongly and specifically. They are always copurified from rat brain extracts. Coculturing cells expressing LPH1 with cells expressing Lasso leads to their mutual attraction and formation of multiple junctions to which both proteins are recruited. Cells expressing LPH1 form chimerical synapses with hippocampal neurons in cocultures; LPH1 and postsynaptic neuronal protein PSD-95 accumulate on opposite sides of these structures. Immunoblotting and immunoelectron microscopy of purified synapses and immunostaining of cultured hippocampal neurons show that LPH1 and Lasso are enriched in synapses; in both systems, LPH1 is presynaptic, whereas Lasso is postsynaptic. A C-terminal fragment of Lasso interacts with LPH1 and induces Ca(2+) signals in presynaptic boutons of hippocampal neurons and in neuroblastoma cells expressing LPH1. Thus, LPH1 and Lasso can form transsynaptic complexes capable of inducing presynaptic Ca(2+) signals, which might affect synaptic functions.
Collapse
|
33
|
Li SQ, Luckins A, Lun ZR. Trypanosoma brucei brucei: A comparison of gene expression in the liver and spleen of infected mice utilizing cDNA microarray technology. Exp Parasitol 2011; 128:256-64. [PMID: 21376043 DOI: 10.1016/j.exppara.2011.02.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 02/15/2011] [Accepted: 02/24/2011] [Indexed: 11/18/2022]
Abstract
Trypanosoma brucei brucei, the infectious agent of the disease known as Nagana, is a pathogenic trypanosome occurring in Africa, where it causes significant economic loss to domesticated livestock. Although many studies on the histopathology of organs of mice infected with T. b. brucei have been reported, little work has been done regarding gene expression in these organs in infected mice. In this paper, we describe the use of cDNA microarray to determine gene expression profiles in the liver and spleen of mice infected with T. b. brucei (STIB 920) at peak parasitaemia (12 days after infection). Our results showed that a total of 123 genes in the liver and 389 genes in the spleen were expressed differentially in T. b. brucei infected mice. In contrast, however, in an acute infection in mice caused by Trypanosoma brucei evansi, a species genetically related to T. b. brucei, 336 genes in the liver and 190 genes in the spleen were expressed, differentially, indicating that the liver of mice was more affected by the acute T. b. evansi infection whilst the spleen was more affected by the subacute T. b. brucei infection. Our results provide a number of possible reasons why mice infected with T. b. evansi die sooner than those infected with T. b. brucei: (1) mice infected with T. b. evansi may need more stress response proteins to help them pass through the infection and these are probably excessively consumed; (2) proliferating cell nuclear antigen was more down-regulated in the liver of mice infected with T. b. evansi, which indicated that the inhibition of proliferation of hepatocytes in mice infected with T. b. evansi might be more severe than that in T. b. brucei infection; and (3) more hepatocyte apoptosis occurred in the mice infected with T. b. evansi and this might be probably the most important reason why mice died sooner than those infected with T. b. brucei. Studies of the changes in the gene expression profile in the liver and spleen of mice infected with T. b. brucei may be helpful in understanding the mechanisms of pathogenesis in Nagana disease at the molecular level. By comparing the gene profiles of the liver and spleen of mice infected with T. b. brucei with T. b. evansi, we have identified a number of factors that could explain the differences in pathogenesis in mice infected with these two African trypanosomes.
Collapse
Affiliation(s)
- San-Qiang Li
- Center for Parasitic Organisms, State Key Laboratory of Biocontrol, School of Life Sciences, Key Laboratory of Tropical Diseases Control, The Ministry of Education, Sun Yat-Sen (Zhongshan) University, Guangzhou 510275, PR China
| | | | | |
Collapse
|
34
|
Bioinformatic analyses identifies novel protein-coding pharmacogenomic markers associated with paclitaxel sensitivity in NCI60 cancer cell lines. BMC Med Genomics 2011; 4:18. [PMID: 21314952 PMCID: PMC3050680 DOI: 10.1186/1755-8794-4-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 02/11/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Paclitaxel is a microtubule-stabilizing drug that has been commonly used in treating cancer. Due to genetic heterogeneity within patient populations, therapeutic response rates often vary. Here we used the NCI60 panel to identify SNPs associated with paclitaxel sensitivity. Using the panel's GI50 response data available from Developmental Therapeutics Program, cell lines were categorized as either sensitive or resistant. PLINK software was used to perform a genome-wide association analysis of the cellular response to paclitaxel with the panel's SNP-genotype data on the Affymetrix 125 k SNP array. FastSNP software helped predict each SNP's potential impact on their gene product. mRNA expression differences between sensitive and resistant cell lines was examined using data from BioGPS. Using Haploview software, we investigated for haplotypes that were more strongly associated with the cellular response to paclitaxel. Ingenuity Pathway Analysis software helped us understand how our identified genes may alter the cellular response to paclitaxel. RESULTS 43 SNPs were found significantly associated (FDR<0.005) with paclitaxel response, with 10 belonging to protein-coding genes (CFTR, ROBO1, PTPRD, BTBD12, DCT, SNTG1, SGCD, LPHN2, GRIK1, ZNF607). SNPs in GRIK1, DCT, SGCD and CFTR were predicted to be intronic enhancers, altering gene expression, while SNPs in ZNF607 and BTBD12 cause conservative missense mutations. mRNA expression analysis supported these findings as GRIK1, DCT, SNTG1, SGCD and CFTR showed significantly (p<0.05) increased expression among sensitive cell lines. Haplotypes found in GRIK1, SGCD, ROBO1, LPHN2, and PTPRD were more strongly associated with response than their individual SNPs. CONCLUSIONS Our study has taken advantage of available genotypic data and its integration with drug response data obtained from the NCI60 panel. We identified 10 SNPs located within protein-coding genes that were not previously shown to be associated with paclitaxel response. As only five genes showed differential mRNA expression, the remainder would not have been detected solely based on expression data. The identified haplotypes highlight the role of utilizing SNP combinations within genomic loci of interest to improve the risk determination associated with drug response. These genetic variants represent promising biomarkers for predicting paclitaxel response and may play a significant role in the cellular response to paclitaxel.
Collapse
|
35
|
Kompass KS, Witte JS. Co-regulatory expression quantitative trait loci mapping: method and application to endometrial cancer. BMC Med Genomics 2011; 4:6. [PMID: 21226949 PMCID: PMC3032645 DOI: 10.1186/1755-8794-4-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 01/12/2011] [Indexed: 01/16/2023] Open
Abstract
Background Expression quantitative trait loci (eQTL) studies have helped identify the genetic determinants of gene expression. Understanding the potential interacting mechanisms underlying such findings, however, is challenging. Methods We describe a method to identify the trans-acting drivers of multiple gene co-expression, which reflects the action of regulatory molecules. This method-termed co-regulatory expression quantitative trait locus (creQTL) mapping-allows for evaluation of a more focused set of phenotypes within a clear biological context than conventional eQTL mapping. Results Applying this method to a study of endometrial cancer revealed regulatory mechanisms supported by the literature: a creQTL between a locus upstream of STARD13/DLC2 and a group of seven IFNβ-induced genes. This suggests that the Rho-GTPase encoded by STARD13 regulates IFNβ-induced genes and the DNA damage response. Conclusions Because of the importance of IFNβ in cancer, our results suggest that creQTL may provide a finer picture of gene regulation and may reveal additional molecular targets for intervention. An open source R implementation of the method is available at http://sites.google.com/site/kenkompass/.
Collapse
Affiliation(s)
- Kenneth S Kompass
- Department of Epidemiology and Biostatistics, Institute for Human Genetics, University of California, San Francisco, USA
| | | |
Collapse
|
36
|
Domené S, Stanescu H, Wallis D, Tinloy B, Pineda DE, Kleta R, Arcos-Burgos M, Roessler E, Muenke M. Screening of human LPHN3 for variants with a potential impact on ADHD susceptibility. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:11-8. [PMID: 21184580 DOI: 10.1002/ajmg.b.31141] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 10/18/2010] [Indexed: 11/06/2022]
Abstract
Attention deficit hyperactivity disorder (ADHD) is the most common behavioral disorder in childhood, and often has effects detectable into adulthood. Advances in genetic linkage and association analysis have begun to elucidate some of the genetic factors underlying this complex disorder. Recently, we identified LPHN3, a novel ADHD susceptibility gene harbored in 4q, and showed that a LPHN3 common haplotype confers susceptibility to ADHD and predicts effectiveness of stimulant medication. Here we present the mutational analysis of the entire coding region of LPHN3 in a cohort of 139 ADHD subjects and 52 controls from across the USA. We identified 21 variants, of which 14 have been reported and 7 are novel. These include 5 missense, 8 synonymous, and 8 intronic changes. Interestingly, neither susceptibility nor protective haplotype alleles are associated with obviously significant coding region changes, or canonical splice site alterations, suggesting that non-coding variations determining the quantity and/or quality of LPHN3 isoforms are the likely contributors to this common behavioral disorder.
Collapse
Affiliation(s)
- Sabina Domené
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892-3717, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Martinez AF, Muenke M, Arcos-Burgos M. From the black widow spider to human behavior: Latrophilins, a relatively unknown class of G protein-coupled receptors, are implicated in psychiatric disorders. Am J Med Genet B Neuropsychiatr Genet 2011; 156B:1-10. [PMID: 21184579 PMCID: PMC4101183 DOI: 10.1002/ajmg.b.31137] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 09/28/2010] [Indexed: 12/24/2022]
Abstract
The findings of a recent study associate LPHN3, a member of the latrophilin family, with an increased risk of developing attention deficit/hyperactivity disorder (ADHD), the most common psychiatric disorder in childhood and adolescence. Latrophilins comprise a new family of G protein-coupled receptors of unknown native physiological function that mediate the neurotoxic effects of α-latrotoxin, a potent toxin found in black widow spider venom. This receptor-toxin interaction has helped to elucidate the mechanistic aspects of neurotransmitter and hormone release in vertebrates. Such unprecedented discovery points to a new direction in the assessment of ADHD and suggest that further study of this receptor family may provide novel insights into the etiology and treatment of ADHD and other related psychiatric conditions.
Collapse
Affiliation(s)
| | | | - Mauricio Arcos-Burgos
- Correspondence to: Dr. Mauricio Arcos-Burgos, M.D., Ph.D., National Human Genome Research Institute, National Institutes of Health, 35 Convent Drive, MSC 3717, Building 35, Room 1B209, Bethesda, MD 20892.
| |
Collapse
|
38
|
Arcos-Burgos M, Muenke M. Toward a better understanding of ADHD: LPHN3 gene variants and the susceptibility to develop ADHD. ACTA ACUST UNITED AC 2010; 2:139-47. [PMID: 21432600 DOI: 10.1007/s12402-010-0030-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 07/29/2010] [Indexed: 12/14/2022]
Abstract
During the past 15 years, an impressive amount of genetic information has become available in the research field of psychiatry, particularly as it relates to attention-deficit/hyperactivity disorder (ADHD). However, the classical clinical approach to ADHD has minimally affected and not significantly been improved by this genetic revolution. It is difficult to predict how long it will take for genetic findings to alter the way clinicians treat patients with ADHD. New medications or treatment protocols may take years to become routine clinical practice. However, when taken together, recent successes in genomics, pharmacogenomics, and genetic epidemiology have the potential (1) to prevent comorbid consequences of ADHD, (2) to individualize therapies for patients with ADHD, and (3) to define new epidemiological policies to aid with the impact of ADHD on society. Here, we present an overview of how genetic research may affect and improve the quality of life of patients with ADHD: as an example, we use the discovery of LPHN3, a new gene in which variants have recently been shown to be associated with ADHD.
Collapse
Affiliation(s)
- Mauricio Arcos-Burgos
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
39
|
Abstract
There are a total of 33 members of adhesion G protein-coupled receptors (GPCRs) in humans and 30 members in mice and rats. More than half of these receptors are expressed in the central nervous system (CNS), indicating their possible roles in the development and function of the CNS. Indeed, it has been shown that adhesion-GPCRs are involved in the regulation of neurulation, cortical development and neurite growth. Among the few adhesion-GPCRs being studied, GPR56 is so far the only member associated with a human brain malformation called bilateral frontoparietal polymicrogyria (BFPP). The histopathology of BFPP is a cobblestone-like brain malformation characterized by neuronal overmigration through a breached pial basement membrane (BM). Further studies in the Gpr56 knockout mouse model revealed that GPR56 is expressed in radial glial cells and regulates the integrity of the pial BM by binding a putative ligand in the extracellular matrix of the developing brain.
Collapse
|
40
|
Abstract
Tumor growth is a highly complex, multistep process that involves tumor cell detachment, migration, invasion and metastasis accompanied by angiogenesis and extracellular matrix turn-over. Each of the steps is influenced by tumor cell interaction and interaction of the tumor cell with its microenvironment that consists of different cell types as tumor-associated fibroblasts, endothelial cells and leukocytes as well as the extracellular matrix produced by the tumor cells themselves or by the fibroblasts. Cellular communication takes place by the regulated expression of adhesion receptors. Adhesion-GPCRs are characterized by very long extracellular N-termini that have multiple domains. When considering this complex structure it is only logical that adhesion-GPCRs are involved in tumor cell interactions. Moreover, these receptors function in cell guidance and/or trafficking, which, in addition to their structure, makes them interesting for tumorigenesis. The aberrant expression of several adhesion-GPCRs on tumor cells and their involvement in tumor growth have been shown for some of the family members. This overview summarizes expression database data as well as data from original research articles of adhesion-GPCRs in tumors.
Collapse
|
41
|
Silva JP, Ushkaryov YA. The latrophilins, "split-personality" receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 706:59-75. [PMID: 21618826 DOI: 10.1007/978-1-4419-7913-1_5] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Latrophilin, a neuronal "adhesion-G protein-coupled receptor", is the major brain receptor for alpha-latrotoxin, a black widow spidertoxin which stimulates strong neuronal exocytosis in vertebrates. Latrophilin has an unusual structure consisting of two fragments that are produced by the proteolytic cleavage of the parental molecule and that behave independently in the plasma membrane. On binding an agonist, the fragments reassociate and send an intracellular signal. This signal, transduced by a heterotrimeric G protein, causes release of calcium from intracellular stores and massive release of neurotransmitters. Latrophilin represents a phylogenetically conserved family of receptors, with orthologues found in all animals and up to three homologues present in most chordate species. From mammalian homologues, latrophilins 1 and 3 are expressed in neurons, while latrophilin 2 is ubiquitous. Latrophilin 1 may control synapse maturation and exocytosis, whereas latrophilin 2 may be involved in breast cancer. Latrophilins may play different roles during development and in adult animals: thus, LAT-1 determines cell fate in early embryogenesis in Caenorhabditis elegans and controls neurotransmitter release in adult nematodes. This diversity suggests that the functions of latrophilins may be determined by their interactions with respective ligands. The finding of the ligand of latrophilin 1, the large postsynaptic protein lasso, is the first step in the quest for the physiological functions of latrophilins.
Collapse
|
42
|
|
43
|
Abstract
For more than three decades, the venom of the black widow spider and its principal active components, latrotoxins, have been used to induce release of neurotransmitters and hormones and to study the mechanisms of exocytosis. Given the complex nature of alpha--latrotoxin (alpha-LTX) actions, this research has been continuously overshadowed by many enigmas, misconceptions and perpetual changes of the underlying hypotheses. Some of the toxin's mechanisms of action are still not completely understood. Despite all these difficulties, the extensive work of several generations of neurobiologists has brought about a great deal of fascinating insights into pre-synaptic processes and has led to the discovery of several novel proteins and synaptic systems. For example, alpha-LTX studies have contributed to the widespread acceptance of the vesicular theory of transmitter release. Pre-synaptic receptors for alpha-LTX--neurexins, latrophilins and protein tyrosine phosphatase sigma--and their endogenous ligands have now become centrepieces of their own areas of research, with a potential of uncovering new mechanisms of synapse formation and regulation that may have medical implications. However, any future success of alpha-LTX research will require a better understanding of this unusual natural tool and a more precise dissection of its multiple mechanisms.
Collapse
Affiliation(s)
- John-Paul Silva
- Division of Cell and Molecular Biology, Imperial College London, Exhibition Road, London, UK
| | | | | |
Collapse
|
44
|
The putative cyclooctadepsipeptide receptor depsiphilin of the canine hookworm Ancylostoma caninum. Parasitol Res 2009; 105 Suppl 1:S91-100. [PMID: 19575230 DOI: 10.1007/s00436-009-1500-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The G-Protein-coupled receptor Hc110-R of Haemonchus contortus and its orthologue in Caenorhabditis elegans, the latrophilin-like protein 1 (LAT-1), were shown to play a role in the mode of action of the new anthelmintic compound emodepside. C. elegans LAT-1 knockout mutants showed a decreased paralysing effect of emodepside on the pharyngeal muscle. In the present study, the LAT-1 orthologue in the canine hookworm Ancylostoma caninum was identified and named depsiphilin. To obtain more information about the regulation of this receptor and to facilitate phylogenetic and evolutionary analyses of parasitic nematode genes, the genomic structure of A. caninum depsiphilin was investigated. High consistency regarding the position of introns in comparison to C. elegans LAT-1 was observed, providing indication of the same origin of the genes. With a view to possible differences in efficacy of emodepside on different developmental stages, we analysed the transcript level of A. caninum depsiphilin in eggs, L1, L3, male and female adult worms using quantitative real-time PCR. Depsiphilin is transcribed in all five examined stages, but we found a significantly lower transcript level in third-stage larvae. A correlation between these findings and a reduced emodepside activity remains to be investigated.
Collapse
|
45
|
Xing Y, Nakamura Y, Rainey WE. G protein-coupled receptor expression in the adult and fetal adrenal glands. Mol Cell Endocrinol 2009; 300:43-50. [PMID: 19027826 PMCID: PMC2679220 DOI: 10.1016/j.mce.2008.10.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 10/22/2008] [Accepted: 10/22/2008] [Indexed: 10/21/2022]
Abstract
Hormonal regulation of adrenal function occurs primarily through G protein-coupled receptors (GPCR), which may play different roles in fetal vs. adult adrenal glands. In this study, we compared the transcript levels of GPCR between fetal and adult adrenal and found that gonadotropin-releasing hormone receptor (GnRHR), latrophilin 3 receptor, G protein-coupled receptor 37, angiotensin II receptor type 2, latrophilin 2 receptor and melanocortin receptor were expressed at significantly higher levels in fetal adrenal. High GnRHR protein expression was also detected in fetal adrenal using immunohistochemical analysis. To define potential ligand sources for fetal adrenal GnRHR, we demonstrated that GnRH1 mRNA was expressed at high levels in the placenta, while fetal adrenal had high expression of GnRH2. In summary, certain GPCR particularly GnRHR were highly expressed in fetal adrenal and the expression of GnRH mRNA in the placenta and the fetal adrenal raises the possibility of endocrine and/or paracrine/autocrine influences on fetal adrenal function. However, the exact function of GnRHR in fetal adrenal remains to be determined.
Collapse
MESH Headings
- Adrenal Glands/cytology
- Adrenal Glands/physiology
- Female
- Fetus/anatomy & histology
- Fetus/physiology
- Gene Expression Regulation, Developmental
- Humans
- Oligonucleotide Array Sequence Analysis
- Pregnancy
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, LHRH/genetics
- Receptors, LHRH/metabolism
- Receptors, Melanocortin/genetics
- Receptors, Melanocortin/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
Collapse
Affiliation(s)
| | | | - William E. Rainey
- Corresponding author: William E Rainey, Ph.D., Address: Department of Physiology, Medical College of Georgia, 1120 15th Street, CA Building – Room 3094, Augusta, GA 30912, Phone: 706-721-7665, Fax: 706-721-8360,
| |
Collapse
|
46
|
Yona S, Lin HH, Siu WO, Gordon S, Stacey M. Adhesion-GPCRs: emerging roles for novel receptors. Trends Biochem Sci 2008; 33:491-500. [PMID: 18789697 DOI: 10.1016/j.tibs.2008.07.005] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 07/21/2008] [Accepted: 07/24/2008] [Indexed: 01/08/2023]
Abstract
The G protein-coupled receptor (GPCR) family comprises the largest class of cell surface receptors found in metazoan proteomes. Within the novel GPCR subfamily of adhesion-GPCRs, approximately 150 distinct orthologues, from invertebrates to mammals, have been identified to date. All members of this family contain a large extracellular region, often containing common protein modules, coupled to a seven-transmembrane domain via a stalk region that seems to be crucial for functionality. Owing to their unique structure, restricted expression profile and involvement in several human diseases, adhesion-GPCRs have long been proposed to have vital dual roles in cellular adhesion and signalling. More recent studies have provided structural, evolutionary, developmental and immunological insights in relation to the adhesion-GPCR family.
Collapse
Affiliation(s)
- Simon Yona
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | | | | | | | | |
Collapse
|
47
|
Vakonakis I, Langenhan T, Prömel S, Russ A, Campbell ID. Solution structure and sugar-binding mechanism of mouse latrophilin-1 RBL: a 7TM receptor-attached lectin-like domain. Structure 2008; 16:944-53. [PMID: 18547526 PMCID: PMC2430599 DOI: 10.1016/j.str.2008.02.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 02/16/2008] [Accepted: 02/19/2008] [Indexed: 11/30/2022]
Abstract
Latrophilin-1 (Lat-1), a target receptor for α-Latrotoxin, is a putative G protein-coupled receptor implicated in synaptic function. The extracellular portion of Lat-1 contains a rhamnose binding lectin (RBL)-like domain of unknown structure. RBL domains, first isolated from the eggs of marine species, are also found in the ectodomains of other metazoan transmembrane proteins, including a recently discovered coreceptor of the neuronal axon guidance molecule SLT-1/Slit. Here, we describe a structure of this domain from the mouse Lat-1. RBL adopts a unique α/β fold with long structured loops important for monosaccharide recognition, as shown in the structure of a complex with L-rhamnose. Sequence alignments and mutagenesis show that residues important for carbohydrate binding are often absent in other receptor-attached examples of RBL, including the SLT-1/Slit coreceptor. We postulate that this domain class facilitates direct protein-protein interactions in many transmembrane receptors.
Collapse
Affiliation(s)
- Ioannis Vakonakis
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, United Kingdom.
| | | | | | | | | |
Collapse
|
48
|
Kamesh N, Aradhyam GK, Manoj N. The repertoire of G protein-coupled receptors in the sea squirt Ciona intestinalis. BMC Evol Biol 2008; 8:129. [PMID: 18452600 PMCID: PMC2396169 DOI: 10.1186/1471-2148-8-129] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Accepted: 05/01/2008] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND G protein-coupled receptors (GPCRs) constitute a large family of integral transmembrane receptor proteins that play a central role in signal transduction in eukaryotes. The genome of the protochordate Ciona intestinalis has a compact size with an ancestral complement of many diversified gene families of vertebrates and is a good model system for studying protochordate to vertebrate diversification. An analysis of the Ciona repertoire of GPCRs from a comparative genomic perspective provides insight into the evolutionary origins of the GPCR signalling system in vertebrates. RESULTS We have identified 169 gene products in the Ciona genome that code for putative GPCRs. Phylogenetic analyses reveal that Ciona GPCRs have homologous representatives from the five major GRAFS (Glutamate, Rhodopsin, Adhesion, Frizzled and Secretin) families concomitant with other vertebrate GPCR repertoires. Nearly 39% of Ciona GPCRs have unambiguous orthologs of vertebrate GPCR families, as defined for the human, mouse, puffer fish and chicken genomes. The Rhodopsin family accounts for ~68% of the Ciona GPCR repertoire wherein the LGR-like subfamily exhibits a lineage specific gene expansion of a group of receptors that possess a novel domain organisation hitherto unobserved in metazoan genomes. CONCLUSION Comparison of GPCRs in Ciona to that in human reveals a high level of orthology of a protochordate repertoire with that of vertebrate GPCRs. Our studies suggest that the ascidians contain the basic ancestral complement of vertebrate GPCR genes. This is evident at the subfamily level comparisons since Ciona GPCR sequences are significantly analogous to vertebrate GPCR subfamilies even while exhibiting Ciona specific genes. Our analysis provides a framework to perform future experimental and comparative studies to understand the roles of the ancestral chordate versions of GPCRs that predated the divergence of the urochordates and the vertebrates.
Collapse
Affiliation(s)
- N Kamesh
- Department of Biotechnology, Bhupat and Jyothi Mehta School of Biosciences Building, Indian Institute of Technology Madras, Chennai 600036, India.
| | | | | |
Collapse
|
49
|
Abstract
alpha-Latrotoxin (alpha-LTX) from black widow spider venom induces exhaustive release of neurotransmitters from vertebrate nerve terminals and endocrine cells. This 130-kDa protein has been employed for many years as a molecular tool to study exocytosis. However, its action is complex: in neurons, alpha-LTX induces massive secretion both in the presence of extracellular Ca(2+) (Ca(2+) (e)) and in its absence; in endocrine cells, it usually requires Ca(2+) (e). To use this toxin for further dissection of secretory mechanisms, one needs an in-depth understanding of its functions. One such function that explains some alpha-LTX effects is its ability to form cation-permeable channels in artificial lipid bilayers. The mechanism of alpha-LTX pore formation, revealed by cryo-electron microscopy, involves toxin assembly into homotetrameric complexes which harbor a central channel and can insert into lipid membranes. However, in biological membranes, alpha-LTX cannot exert its actions without binding to specific receptors of the plasma membrane. Three proteins with distinct structures have been found to bind alpha-LTX: neurexin Ialpha, latrophilin 1, and receptor-like protein tyrosine phosphatase sigma. Upon binding a receptor, alpha-LTX forms channels permeable to cations and small molecules; the toxin may also activate the receptor. To distinguish between the pore- and receptor-mediated effects, and to study structure-function relationships in the toxin, alpha-LTX mutants have been used.
Collapse
Affiliation(s)
- Yuri A Ushkaryov
- Division of Cell and Molecular Biology, Imperial College London, London, SW7 2AY, UK.
| | | | | |
Collapse
|
50
|
Doyle SE, Scholz MJ, Greer KA, Hubbard AD, Darnell DK, Antin PB, Klewer SE, Runyan RB. Latrophilin-2 is a novel component of the epithelial-mesenchymal transition within the atrioventricular canal of the embryonic chicken heart. Dev Dyn 2007; 235:3213-21. [PMID: 17016846 DOI: 10.1002/dvdy.20973] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Endothelial cells in the atrioventricular canal of the heart undergo an epithelial-mesenchymal transition (EMT) to form heart valves. We surveyed an on-line database (http://www.geisha.arizona.edu/) for clones expressed during gastrulation to identify novel EMT components. One gene, latrophilin-2, was identified as expressed in the heart and appeared to be functional in EMT. This molecule was chosen for further examination. In situ localization showed it to be expressed in both the myocardium and endothelium. Several antisense DNA probes and an siRNA for latrophilin-2 produced a loss of EMT in collagen gel cultures. Latrophilin-2 is a putative G-protein-coupled receptor and we previously identified a pertussis toxin-sensitive G-protein signal transduction pathway. Microarray experiments were performed to examine whether these molecules were related. After treatment with antisense DNA against latrophilin-2, expression of 1,385 genes and ESTs was altered. This represented approximately 12.5% of the microarray elements. In contrast, pertussis toxin altered only 103 (0.9%) elements of the array. There appears to be little overlap between the two signal transduction pathways. Latrophilin-2 is thus a novel component of EMT and provides a new avenue for investigation of this cellular process.
Collapse
Affiliation(s)
- Sally E Doyle
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona 85724-5044, USA
| | | | | | | | | | | | | | | |
Collapse
|