• Title/Summary/Keyword: GPCR signaling

Search Result 38, Processing Time 0.028 seconds

Biased G Protein-Coupled Receptor Signaling: New Player in Modulating Physiology and Pathology

  • Bologna, Zuzana;Teoh, Jian-peng;Bayoumi, Ahmed S.;Tang, Yaoliang;Kim, Il-man
    • Biomolecules & Therapeutics
    • /
    • v.25 no.1
    • /
    • pp.12-25
    • /
    • 2017
  • G protein-coupled receptors (GPCRs) are a family of cell-surface proteins that play critical roles in regulating a variety of pathophysiological processes and thus are targeted by almost a third of currently available therapeutics. It was originally thought that GPCRs convert extracellular stimuli into intracellular signals through activating G proteins, whereas ${\beta}$-arrestins have important roles in internalization and desensitization of the receptor. Over the past decade, several novel functional aspects of ${\beta}$-arrestins in regulating GPCR signaling have been discovered. These previously unanticipated roles of ${\beta}$-arrestins to act as signal transducers and mediators of G protein-independent signaling have led to the concept of biased agonism. Biased GPCR ligands are able to engage with their target receptors in a manner that preferentially activates only G protein- or ${\beta}$-arrestin-mediated downstream signaling. This offers the potential for next generation drugs with high selectivity to therapeutically relevant GPCR signaling pathways. In this review, we provide a summary of the recent studies highlighting G protein- or ${\beta}$-arrestin-biased GPCR signaling and the effects of biased ligands on disease pathogenesis and regulation.

US28, a Virally-Encoded GPCR as an Antiviral Target for Human Cytomegalovirus Infection

  • Lee, Sungjin;Chung, Yoon Hee;Lee, Choongho
    • Biomolecules & Therapeutics
    • /
    • v.25 no.1
    • /
    • pp.69-79
    • /
    • 2017
  • Viruses continue to evolve a new strategy to take advantage of every aspect of host cells in order to maximize their survival. Due to their central roles in transducing a variety of transmembrane signals, GPCRs seem to be a prime target for viruses to pirate for their own use. Incorporation of GPCR functionality into the genome of herpesviruses has been demonstrated to be essential for pathogenesis of many herpesviruses-induced diseases. Here, we introduce US28 of human cytomegalovirus (HCMV) as the best-studied example of virally-encoded GPCRs to manipulate host GPCR signaling. In this review, we wish to summarize a number of US28-related topics including its regulation of host signaling pathways, its constitutive internalization, its structural and functional analysis, its roles in HCMV biology and pathogenesis, its proliferative activities and role in oncogenesis, and pharmacological modulation of its biological activities. This review will aid in our understanding of how pathogenic viruses usurp the host GPCR signaling for successful viral infection. This kind of knowledge will enable us to build a better strategy to control viral infection by normalizing the virally-dysregulated host GPCR signaling.

A Study on the Detection of Similarity GPCRs by using protein Secondary structure (단백질 2차 구조를 이용한 유사 GPCR 검출에 관한 연구)

  • Ku, Ja-Hyo;Han, Chan-Myung;Yoon, Young-Woo
    • Journal of the Korea Society of Computer and Information
    • /
    • v.14 no.1
    • /
    • pp.73-80
    • /
    • 2009
  • G protein-coupled receptors(GPCRs) family is a cell membrane protein, and plays an important role in a signaling mechanism which transmits external signals through cell membranes into cells. But, GPCRs each are known to have various complex control mechanisms and very unique signaling mechanisms. Structural features, and family and subfamily of GPCRs are well known by function. and accordingly, the most fundamental work in studies identifying the previous GPCRs is to classify the GPCRs with given protein sequences. Studies for classifying previously identified GPCRs more easily with mathematical models have been mainly going on. In this paper Considering that functions of proteins are determined by their stereoscopic structures, the present paper proposes a method to compare secondary structures of two GPCRs having different amino acid sequences, and then detect an unknown GPCRs assumed to have a same function in databases of previously identified GPCRs.

Calcium Signaling of Lysophosphatidylethanolamine through LPA1 in Human SH-SY5Y Neuroblastoma Cells

  • Lee, Jung-Min;Park, Soo-Jin;Im, Dong-Soon
    • Biomolecules & Therapeutics
    • /
    • v.25 no.2
    • /
    • pp.194-201
    • /
    • 2017
  • Lysophosphatidylethanolamine (LPE), a lyso-type metabolite of phosphatidylethanolamine, has been reported to be an intercellular signaling molecule. LPE mobilizes intracellular $Ca^{2+}$ through G-protein-coupled receptor (GPCR) in some cells types. However, GPCRs for lysophosphatidic acid (LPA) were not implicated in the LPE-mediated activities in LPA GPCR overexpression systems or in SK-OV3 ovarian cancer cells. In the present study, in human SH-SY5Y neuroblastoma cells, experiments with $LPA_1$ antagonists showed LPE induced intracellular $Ca^{2+}$ increases in an $LPA_1$ GPCR-dependent manner. Furthermore, LPE increased intracellular $Ca^{2+}$ through pertussis-sensitive G proteins, edelfosine-sensitive-phospholipase C, 2-APB-sensitive $IP_3$ receptors, $Ca^{2+}$ release from intracellular $Ca^{2+}$ stores, and subsequent $Ca^{2+}$ influx across plasma membranes, and LPA acted on $LPA_1$ and $LPA_2$ receptors to induce $Ca^{2+}$ response in a 2-APB-sensitive and insensitive manner. These findings suggest novel involvements for LPE and LPA in calcium signaling in human SH-SY5Y neuroblastoma cells.

Role of Regulators of G-Protein Signaling 4 in $Ca^{2+}$ Signaling in Mouse Pancreatic Acinar Cells

  • Park, Soon-Hong;Lee, Syng-Ill;Shin, Dong-Min
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.15 no.6
    • /
    • pp.383-388
    • /
    • 2011
  • Regulators of G-protein signaling (RGS) proteins are regulators of $Ca^{2+}$ signaling that accelerate the GTPase activity of the G-protein ${\alpha}$ -subunit. RGS1, RGS2, RGS4, and RGS16 are expressed in the pancreas, and RGS2 regulates G-protein coupled receptor (GPCR)-induced $Ca^{2+}$ oscillations. However, the role of RGS4 in $Ca^{2+}$ signaling in pancreatic acinar cells is unknown. In this study, we investigated the mechanism of GPCR-induced $Ca^{2+}$ signaling in pancreatic acinar cells derived from $RGS4^{-/-}$ mice. $RGS4^{-/-}$ acinar cells showed an enhanced stimulus intensity response to a muscarinic receptor agonist in pancreatic acinar cells. Moreover, deletion of RGS4 increased the frequency of $Ca^{2+}$ oscillations. $RGS4^{-/-}$ cells also showed increased expression of sarco/endoplasmic reticulum $Ca^{2+}$ ATPase type 2. However, there were no significant alterations, such as $Ca^{2+}$ signaling in treated high dose of agonist and its related amylase secretion activity, in acinar cells from $RGS4^{-/-}$ mice. These results indicate that RGS4 protein regulates $Ca^{2+}$ signaling in mouse pancreatic acinar cells.

A Machine Learning Based Method for the Prediction of G Protein-Coupled Receptor-Binding PDZ Domain Proteins

  • Eo, Hae-Seok;Kim, Sungmin;Koo, Hyeyoung;Kim, Won
    • Molecules and Cells
    • /
    • v.27 no.6
    • /
    • pp.629-634
    • /
    • 2009
  • G protein-coupled receptors (GPCRs) are part of multi-protein networks called 'receptosomes'. These GPCR interacting proteins (GIPs) in the receptosomes control the targeting, trafficking and signaling of GPCRs. PDZ domain proteins constitute the largest protein family among the GIPs, and the predominant function of the PDZ domain proteins is to assemble signaling pathway components into close proximity by recognition of the last four C-terminal amino acids of GPCRs. We present here a machine learning based approach for the identification of GPCR-binding PDZ domain proteins. In order to characterize the network of interactions between amino acid residues that contribute to the stability of the PDZ domain-ligand complex and to encode the complex into a feature vector, amino acid contact matrices and physicochemical distance matrix were constructed and adopted. This novel machine learning based method displayed high performance for the identification of PDZ domain-ligand interactions and allowed the identification of novel GPCR-PDZ domain protein interactions.

Comprehensive Analysis of Non-Synonymous Natural Variants of G Protein-Coupled Receptors

  • Kim, Hee Ryung;Duc, Nguyen Minh;Chung, Ka Young
    • Biomolecules & Therapeutics
    • /
    • v.26 no.2
    • /
    • pp.101-108
    • /
    • 2018
  • G protein-coupled receptors (GPCRs) are the largest superfamily of transmembrane receptors and have vital signaling functions in various organs. Because of their critical roles in physiology and pathology, GPCRs are the most commonly used therapeutic target. It has been suggested that GPCRs undergo massive genetic variations such as genetic polymorphisms and DNA insertions or deletions. Among these genetic variations, non-synonymous natural variations change the amino acid sequence and could thus alter GPCR functions such as expression, localization, signaling, and ligand binding, which may be involved in disease development and altered responses to GPCR-targeting drugs. Despite the clinical importance of GPCRs, studies on the genotype-phenotype relationship of GPCR natural variants have been limited to a few GPCRs such as b-adrenergic receptors and opioid receptors. Comprehensive understanding of non-synonymous natural variations within GPCRs would help to predict the unknown genotype-phenotype relationship and yet-to-be-discovered natural variants. Here, we analyzed the non-synonymous natural variants of all non-olfactory GPCRs available from a public database, UniProt. The results suggest that non-synonymous natural variations occur extensively within the GPCR superfamily especially in the N-terminus and transmembrane domains. Within the transmembrane domains, natural variations observed more frequently in the conserved residues, which leads to disruption of the receptor function. Our analysis also suggests that only few non-synonymous natural variations have been studied in efforts to link the variations with functional consequences.

Intercellular Lipid Mediators and GPCR Drug Discovery

  • Im, Dong-Soon
    • Biomolecules & Therapeutics
    • /
    • v.21 no.6
    • /
    • pp.411-422
    • /
    • 2013
  • G-protein-coupled receptors (GPCR) are the largest superfamily of receptors responsible for signaling between cells and tissues, and because they play important physiological roles in homeostasis, they are major drug targets. New technologies have been developed for the identification of new ligands, new GPCR functions, and for drug discovery purposes. In particular, intercellular lipid mediators, such as, lysophosphatidic acid and sphingosine 1-phosphate have attracted much attention for drug discovery and this has resulted in the development of fingolimod (FTY-720) and AM095. The discovery of new intercellular lipid mediators and their GPCRs are discussed from the perspective of drug development. Lipid GPCRs for lysophospholipids, including lysophosphatidylserine, lysophosphatidylinositol, lysophosphatidylcholine, free fatty acids, fatty acid derivatives, and other lipid mediators are reviewed.

Detection of similar GPCRs by using protein secondary structures

  • Ku, Ja-Hyo;Yoon, Young-Woo
    • 한국정보컨버전스학회:학술대회논문집
    • /
    • 2008.06a
    • /
    • pp.39-42
    • /
    • 2008
  • G protein-coupled receptor(GPCR) family is a cell membrane protein, and plays an important role in a signaling mechanism which transmits external signals through cell membranes into cells. Now, it is estimated that there may be about 800-1000 GPCRs in a human genome. But, GPCRs each are known to have various complex control mechanisms and very unique signaling mechanisms. GPCRs are involved in maintaining homeostasis of various human systems including an endocrine system or a neural system and thus, disorders in activity control of GPCRs are thought to be the major source of cardiovascular disorders, metabolic disorders, degenerative disorders, carcinogenesis and the like. As more than 60% of currently marketed therapeutic agents target GPCRs, the GPCR field has been actively explored in the pharmaceutical industry. Structural features, and class and subfamily of GPCRs are well known by function, and accordingly, the most fundamental work in studies identifying the previous GPCRs is to classify the GPCRs with given protein sequences. Studies for classifying previously identified GPCRs more easily with mathematical models have been mainly going on. Considering that secondary sequences of proteins, namely, secondary binding structures of amino acids constituting proteins are closely related to functions, the present paper does not place the focus on primary sequences of proteins as previously practiced, but instead, proposes a method to transform primary sequences into secondary structures and compare the secondary structures, and then detect an unknown GPCR assumed to have a same function in databases of previously identified GPCRs.

  • PDF

β-arrestin Promotes c-Jun N-terminal Kinase Mediated Apoptosis via a GABABR·β-arrestin·JNK Signaling Module

  • Wu, Jin-Xia;Shan, Feng-Xiao;Zheng, Jun-Nian;Pei, Dong-Sheng
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.2
    • /
    • pp.1041-1046
    • /
    • 2014
  • Evidence is growing that the $GABA_B$ receptor, which belongs to the G protein-coupled receptor (GPCR) superfamily, is involved in tumorigenesis. Recent studies have shown that ${\beta}$-arrestin can serve as a scaffold to recruit signaling protein c-Jun N-terminal knase (JNK) to GPCR. Here we investigated whether ${\beta}$-arrestin recruits JNK to the $GABA_B$ receptor and facilitates its activation to affect the growth of cancer cells. Our results showed that ${\beta}$-arrestin expression is decreased in breast cancer cells in comparison with controls. ${\beta}$-arrestin could enhance interactions of the $GABA_BR{\cdot}{\beta}-arrestin{\cdot}JNK$ signaling module in MCF-7 and T-47D cells. Further studies revealed that increased expression of ${\beta}$-arrestin enhances the phosphorylation of JNK and induces cancer cells apoptosis. Collectively, these results indicate that ${\beta}$-arrestin promotes JNK mediated apoptosis via a $GABA_BR{\cdot}{\beta}-arrestin{\cdot}JNK$ signaling module.