1
|
Song Y, Guo JF, Lan PS, Wang M, Du QY. Investigation of the pan-cancer property of FNDC1 and its molecular mechanism to promote lung adenocarcinoma metastasis. Transl Oncol 2024; 44:101953. [PMID: 38593585 PMCID: PMC11024379 DOI: 10.1016/j.tranon.2024.101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/07/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Fibronectin type III domain containing 1 (FNDC1) has been associated with the metastasis of many tumors, but its function in lung cancer remains uncertain. METHODS FNDC1 expression was analyzed in The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx), evaluate its prognostic value. Gene Set Enrichment Analysis (GSEA) enrichment analysis of differential expression of FNDC1 in lung cancer. The expression of FNDC1 was detected in five types of lung cancer cells, and screened to establish FNDC1 stable knockdown cell strains. To observe the migration and invasion ability of lung cancer cells after FNDC1 knockdown. Finally, we used rhIL-6 to interfere with the stable knockdown of FNDC1 in A549 cells and observed the recovery of migration and invasion. RESULT Our results showed that FNDC1 expression was increased in 21 tumor tissues, including lung cancer, and was associated with poor prognosis in five cancers, including lung adenocarcinoma (LUAD) (P < 0.05). GSEA enrichment analysis showed that FNDC1 was related to the pathways involved the JAK-STAT signaling pathway. Stable knockdown of FNDC1 in A549 and H292 cells resulted in decreased migration and invasion ability of both cells, accompanied by decreased expression of MMP-2 and Snail, and a significant decline in the expression of p-JAK2 and p-STAT3. The suppressive effect of FNDC1 knockdown on lung cancer cell metastasis counteracted by the JAK-STAT agonist rhIL-6 were presented in the nude mouse metastatic tumor model. CONCLUSION FNDC1 is implicated in poor prognosis of a diverse range of malignant tumors, which can promote metastasis and invasion of lung cancer through the JAK2-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yang Song
- Emergency Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, PR China
| | - Jun-Feng Guo
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, PR China
| | - Pei-Shu Lan
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, PR China
| | - Miao Wang
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, PR China
| | - Quan-Yu Du
- Endocrinology Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan 610072, PR China.
| |
Collapse
|
2
|
He X, Li X, Du X, Han J, Zhang H, Zhu Y, Ma H. Rs420137, rs386360 and rs7763726 polymorphisms in fibronectin type III domain containing 1 are associated with susceptibility to coronary heart disease: Analysis in the Han population. Front Cardiovasc Med 2022; 9:964978. [PMID: 36277792 PMCID: PMC9583258 DOI: 10.3389/fcvm.2022.964978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Numerous genetic studies have shown that genes are related to the pathogenesis of coronary heart disease (CHD). The main aim of this study was to confirm whether fibronectin type III domain containing 1 (FNDC1) polymorphisms correlate with the risk of CHD. Methods In this study, in order to assess the association between three FNDC1 single nucleotide polymorphisms (SNPs) and the risk of CHD, we conducted a case-control study involving 630 patients with CHD and 568 healthy controls using Agena MassARRAY (Agena Bioscience, San Diego, CA, USA). Genotype distribution in case and control groups was analyzed by Chi square test. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by logistic regression models adjusted for age, sex, smoking, and alcohol consumption to assess the correlation between SNPs and CHD risk. Results Our results indicated that FNDC1-rs420137, -rs386360, and -rs7763726 played important roles in enhancing the risk of CHD. Subgroup analysis revealed that rs420137 increased the susceptibility to CHD in males, smokers, and patients aged ≤62 years. Rs360 had an increased risk of CHD in males, patients at aged ≤62 years, smokers, and non-drinkers. Furthermore, the association of rs7763726 with increased CHD risk was also observed in males, patients aged ≤62 years, smokers, and drinkers. Last but not least, these three SNPs we selected were protective factors against hypertension in CHD individuals. Conclusion Our research suggest that FNDC1-rs420137, -rs386360, and -rs7763726 variants may be regarded as novel biomarkers for predicting CHD risk and other specific mechanisms of action of CHD need to be further studied.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Honghong Ma
- *Correspondence: Honghong Ma mhh1001@sohucom
| |
Collapse
|
3
|
Ferré S, Ciruela F, Dessauer CW, González-Maeso J, Hébert TE, Jockers R, Logothetis DE, Pardo L. G protein-coupled receptor-effector macromolecular membrane assemblies (GEMMAs). Pharmacol Ther 2022; 231:107977. [PMID: 34480967 PMCID: PMC9375844 DOI: 10.1016/j.pharmthera.2021.107977] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in cellular signaling across the plasma membrane and a major class of drug targets. The canonical model for GPCR signaling involves three components - the GPCR, a heterotrimeric G protein and a proximal plasma membrane effector - that have been generally thought to be freely mobile molecules able to interact by 'collision coupling'. Here, we synthesize evidence that supports the existence of GPCR-effector macromolecular membrane assemblies (GEMMAs) comprised of specific GPCRs, G proteins, plasma membrane effector molecules and other associated transmembrane proteins that are pre-assembled prior to receptor activation by agonists, which then leads to subsequent rearrangement of the GEMMA components. The GEMMA concept offers an alternative and complementary model to the canonical collision-coupling model, allowing more efficient interactions between specific signaling components, as well as the integration of the concept of GPCR oligomerization as well as GPCR interactions with orphan receptors, truncated GPCRs and other membrane-localized GPCR-associated proteins. Collision-coupling and pre-assembled mechanisms are not exclusive and likely both operate in the cell, providing a spectrum of signaling modalities which explains the differential properties of a multitude of GPCRs in their different cellular environments. Here, we explore the unique pharmacological characteristics of individual GEMMAs, which could provide new opportunities to therapeutically modulate GPCR signaling.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Addiction, Intramural Research Program, NIH, DHHS, Baltimore, MD, USA.
| | - Francisco Ciruela
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, IDIBELL, University of Barcelona, L’Hospitalet de Llobregat, Spain
| | - Carmen W. Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Terence E. Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec
| | - Ralf Jockers
- University of Paris, Institute Cochin, INSERM, CNRS, Paris, France
| | - Diomedes E. Logothetis
- Laboratory of Electrophysiology, Departments of Pharmaceutical Sciences, Chemistry and Chemical Biology and Center for Drug Discovery, School of Pharmacy at the Bouvé College of Health Sciences and College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Leonardo Pardo
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| |
Collapse
|
4
|
Mansour A, Nagi K, Dallaire P, Lukasheva V, Le Gouill C, Bouvier M, Pineyro G. Comprehensive Signaling Profiles Reveal Unsuspected Functional Selectivity of δ-Opioid Receptor Agonists and Allow the Identification of Ligands with the Greatest Potential for Inducing Cyclase Superactivation. ACS Pharmacol Transl Sci 2021; 4:1483-1498. [PMID: 34661070 PMCID: PMC8506601 DOI: 10.1021/acsptsci.1c00019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Indexed: 11/29/2022]
Abstract
![]()
Prolonged exposure
to opioid receptor agonists triggers adaptations
in the adenylyl cyclase (AC) pathway that lead to enhanced production
of cyclic adenosine monophosphate (cAMP) upon withdrawal. This cellular
phenomenon contributes to withdrawal symptoms, hyperalgesia and analgesic
tolerance that interfere with clinical management of chronic pain
syndromes. Since δ-opioid receptors (DOPrs) are a promising
target for chronic pain management, we were interested in finding
out if cell-based signaling profiles as generated for drug discovery
purposes could inform us of the ligand potential to induce sensitization
of the cyclase path. For this purpose, signaling of DOPr agonists
was monitored at multiple effectors. The resulting signaling profiles
revealed marked functional selectivity, particularly for Met-enkephalin
(Met-ENK) whose signaling bias profile differed from those of synthetic
ligands like SNC-80 and ARM390. Signaling diversity among ligands
was systematized by clustering agonists according to similarities
in Emax and Log(τ) values for the
different responses. The classification process revealed that the
similarity in Gα/Gβγ, but not in β-arrestin
(βarr), responses was correlated with the potential of Met-ENK,
deltorphin II, (d-penicillamine2,5)-enkephalin (DPDPE), ARM390,
and SNC-80 to enhance cAMP production, all of which required Ca2+ mobilization to produce this response. Moreover, superactivation
by Met-ENK, which was the most-effective Ca2+ mobilizing
agonist, required Gαi/o activation, availability of Gβγ
subunits at the membrane, and activation of Ca2+ effectors
such as calmodulin and protein kinase C (PKC). In contrast, superactivation by (N-(l-tyrosyl)-(3S)-1,2,3,4-tetrahydroisoquinoline-3-carbonyl)-l-phenylalanyl-l-phenylalanine (TIPP), which was set
in a distinct category through clustering, required activation of
Gαi/o subunits but was independent of the Gβγ dimer
and Ca2+ mobilization, relying instead on Src and Raf-1
to induce this cellular adaptation.
Collapse
Affiliation(s)
- Ahmed Mansour
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada.,CHU Sainte-Justine Research Center, Montréal, Quebec H3T 1C5, Canada
| | - Karim Nagi
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Paul Dallaire
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada.,CHU Sainte-Justine Research Center, Montréal, Quebec H3T 1C5, Canada
| | - Viktoriya Lukasheva
- Institute for Research in Immunology and Cancer, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Graciela Pineyro
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, Quebec H3T 1J4, Canada.,CHU Sainte-Justine Research Center, Montréal, Quebec H3T 1C5, Canada
| |
Collapse
|
5
|
Lin K, Zhang L, Wang Y, Li J, Xu Y, Che D, Mai H, Yu H, Fu L, Wei B, Jiang Z, Pi L, Gu X. FNDC1 Polymorphism (rs3003174 C > T) Increased the Incidence of Coronary Artery Aneurysm in Patients with Kawasaki Disease in a Southern Chinese Population. J Inflamm Res 2021; 14:2633-2640. [PMID: 34188513 PMCID: PMC8232870 DOI: 10.2147/jir.s311956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background A large number of studies demonstrated that the key to the occurrence and development of Kawasaki disease (KD) is the over-activation of immune cells and the generation of various inflammatory factors, leading to the imbalance of the immune system. Recently, mutations in the FNDC1 gene have been shown to be associated with inflammatory responses. However, there have been no reports on the relationship between FNDC1 gene and KD so far. Methods We enrolled 1611 controls and 1459 patients with KD, including 372 patients with coronary artery aneurysm (CAA) and 179 patients with coronary artery lesion (CAL). The relationship between FNDC1 rs3003174 polymorphism and KD with CAA or without CAA was investigated. Results This study showed no evidence that the association between FNDC1 rs3003174 C>T polymorphism and KD susceptibility was statistically significant (CT versus CC: adjusted odds ratio (OR) =0.897, 95% confidence interval (CI) =0.769–1.045, P=0.162; TT versus CC: adjusted OR=0.995, 95% CI=0.786–1.260, P=0.968; dominant model: adjusted OR=0.916, 95% CI=0.792–1.059, P=0.235; and recessive model: adjusted OR=1.055, 95% CI=0.845–1.316, P=0.638). However, our further stratified analysis in the control and KD group bore out that the incidence of TT genotype of FNDC1 rs3003174 C > T polymorphism was higher than that of CC/CT genotype in KD patients stratified by CAA (adjusted OR=1.437, 95% CI=1.034–1.996, P=0.031). Moreover, a stratified analysis of age and gender in KD patients indicated that the rs3003174 TT genotype increased the risk of CAA formation in aged ≦60 months (CC/CT vs TT: adjusted OR=1.580, 95% CI=1.106–2.259, P=0.012) and male (CC/CT vs TT: adjusted OR=1.653, 95% CI=1.101–2.481, P=0.015) KD patients. Conclusion The results of this study demonstrated that the FNDC1 rs3003174 C>T polymorphism may be a hazard factor in the formation of CAA in KD patients that was not disclosed before.
Collapse
Affiliation(s)
- Kun Lin
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China.,Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Linyuan Zhang
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China.,Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Yishuai Wang
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China.,Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China.,School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Jinqing Li
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Yufen Xu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Hanran Mai
- Department of Andrology, Guangzhou Women and Children's Medical Center. Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Hongyan Yu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Lanyan Fu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Bing Wei
- Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Zhiyong Jiang
- Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Lei Pi
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China.,Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People's Republic of China
| |
Collapse
|
6
|
Chung YK, Wong YH. Re‐examining the ‘Dissociation Model’ of G protein activation from the perspective of Gβγ signaling. FEBS J 2020; 288:2490-2501. [DOI: 10.1111/febs.15605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Yin Kwan Chung
- Division of Life Science and Biotechnology Research Institute Hong Kong University of Science and Technology Hong Kong China
| | - Yung Hou Wong
- Division of Life Science and Biotechnology Research Institute Hong Kong University of Science and Technology Hong Kong China
- State Key Laboratory of Molecular Neuroscience the Molecular Neuroscience Center Hong Kong University of Science and Technology Kowloon China
| |
Collapse
|
7
|
de Hoog E, Lukewich MK, Spencer GE. Retinoid receptor-based signaling plays a role in voltage-dependent inhibition of invertebrate voltage-gated Ca 2+ channels. J Biol Chem 2019; 294:10076-10093. [PMID: 31048374 DOI: 10.1074/jbc.ra118.006444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
The retinoic acid receptor (RAR) and retinoid X receptor (RXR) mediate the cellular effects of retinoids (derivatives of vitamin A). Both RAR and RXR signaling events are implicated in hippocampal synaptic plasticity. Furthermore, retinoids can interact with calcium signaling during homeostatic plasticity. We recently provided evidence that retinoids attenuate calcium current (I Ca) through neuronal voltage-gated calcium channels (VGCCs). We now examined the possibility that constitutive activity of neuronal RXR and/or RAR alters calcium influx via the VGCCs. We found that in neurons of the mollusk Lymnaea stagnalis, two different RXR antagonists (PA452 and HX531) had independent and opposing effects on I Ca that were also time-dependent; whereas the RXR pan-antagonist PA452 enhanced I Ca, HX531 reduced I Ca Interestingly, this effect of HX531 occurred through voltage-dependent inhibition of VGCCs, a phenomenon known to influence neurotransmitter release from neurons. This inhibition appeared to be independent of G proteins and was largely restricted to Cav2 Ca2+ channels. Of note, an RAR pan-antagonist, LE540, also inhibited I Ca but produced G protein-dependent, voltage-dependent inhibition of VGCCs. These findings provide evidence that retinoid receptors interact with G proteins in neurons and suggest mechanisms by which retinoids might affect synaptic calcium signaling.
Collapse
Affiliation(s)
- Eric de Hoog
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Mark K Lukewich
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| | - Gaynor E Spencer
- From the Department of Biological Sciences, Brock University, St. Catharines, Ontario L2S 3A1, Canada
| |
Collapse
|
8
|
Senarath K, Kankanamge D, Samaradivakara S, Ratnayake K, Tennakoon M, Karunarathne A. Regulation of G Protein βγ Signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:133-191. [PMID: 29776603 DOI: 10.1016/bs.ircmb.2018.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) deliver external signals to the cell interior, upon activation by the external signal stimulated G protein-coupled receptors (GPCRs).While the activated GPCRs control several pathways independently, activated G proteins control the vast majority of cellular and physiological functions, ranging from vision to cardiovascular homeostasis. Activated GPCRs dissociate GαGDPβγ heterotrimer into GαGTP and free Gβγ. Earlier, GαGTP was recognized as the primary signal transducer of the pathway and Gβγ as a passive signaling modality that facilitates the activity of Gα. However, Gβγ later found to regulate more number of pathways than GαGTP does. Once liberated from the heterotrimer, free Gβγ interacts and activates a diverse range of signaling regulators including kinases, lipases, GTPases, and ion channels, and it does not require any posttranslation modifications. Gβγ family consists of 48 members, which show cell- and tissue-specific expressions, and recent reports show that cells employ the subtype diversity in Gβγ to achieve desired signaling outcomes. In addition to activated GPCRs, which induce free Gβγ generation and the rate of GTP hydrolysis in Gα, which sequester Gβγ in the heterotrimer, terminating Gβγ signaling, additional regulatory mechanisms exist to regulate Gβγ activity. In this chapter, we discuss structure and function, subtype diversity and its significance in signaling regulation, effector activation, regulatory mechanisms as well as the disease relevance of Gβγ in eukaryotes.
Collapse
|
9
|
Brand CS, Sadana R, Malik S, Smrcka AV, Dessauer CW. Adenylyl Cyclase 5 Regulation by Gβγ Involves Isoform-Specific Use of Multiple Interaction Sites. Mol Pharmacol 2015; 88:758-67. [PMID: 26206488 PMCID: PMC4576683 DOI: 10.1124/mol.115.099556] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/23/2015] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclase (AC) converts ATP into cyclic AMP (cAMP), an important second messenger in cell signaling. Heterotrimeric G proteins and other regulators are important for control of AC activity. Depending on the AC isoform, Gβγ subunits can either conditionally stimulate or inhibit cAMP synthesis. We previously showed that the Gαs-βγ heterotrimer binds to the N terminus (NT) of type 5 AC (AC5). We now show that Gβγ binds to the NT of a wide variety of AC isoforms. We hypothesized that Gβγ/AC5 interactions involving inactive heterotrimer and Gβγ stimulation of AC5 were separable events. Mutations of the Gβγ "hotspot" show that this site is necessary for AC5 stimulation but not for interactions with the first 198 aa of AC5NT, which is a G protein scaffolding site. This contrasts with AC6, where the Gβγ hotspot is required for both interactions with AC6NT and for stimulation of AC6. Additionally, the SIGK hotspot peptide disrupts Gβγ regulation of AC isoforms 1, 2, and 6, but not AC5. Gβγ also binds the C1/C2 catalytic domains of AC5 and AC6. Finally, cellular interactions with full-length AC5 depend on multiple sites on Gβγ. This suggests an isoform-specific mechanism in which bound Gβγ at the AC5NT is ideally situated for spatiotemporal control of AC5. We propose Gβγ regulation of AC involves multiple binding events, and the role of the AC NT for mechanisms of regulation by heterotrimeric G protein subunits is isoform-specific.
Collapse
Affiliation(s)
- Cameron S Brand
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas (C.S.B., C.W.D.); Department of Natural Sciences, University of Houston-Downtown, Houston, Texas (R.S.); and Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, New York (S.M., A.V.S.)
| | - Rachna Sadana
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas (C.S.B., C.W.D.); Department of Natural Sciences, University of Houston-Downtown, Houston, Texas (R.S.); and Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, New York (S.M., A.V.S.)
| | - Sundeep Malik
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas (C.S.B., C.W.D.); Department of Natural Sciences, University of Houston-Downtown, Houston, Texas (R.S.); and Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, New York (S.M., A.V.S.)
| | - Alan V Smrcka
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas (C.S.B., C.W.D.); Department of Natural Sciences, University of Houston-Downtown, Houston, Texas (R.S.); and Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, New York (S.M., A.V.S.)
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas (C.S.B., C.W.D.); Department of Natural Sciences, University of Houston-Downtown, Houston, Texas (R.S.); and Department of Pharmacology and Physiology, University of Rochester School of Medicine, Rochester, New York (S.M., A.V.S.)
| |
Collapse
|
10
|
Zhang P, Kofron CM, Mende U. Heterotrimeric G protein-mediated signaling and its non-canonical regulation in the heart. Life Sci 2015; 129:35-41. [PMID: 25818188 PMCID: PMC4415990 DOI: 10.1016/j.lfs.2015.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/31/2015] [Accepted: 02/11/2015] [Indexed: 11/20/2022]
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) regulate a multitude of signaling pathways in mammalian cells by transducing signals from G protein-coupled receptors (GPCRs) to effectors, which in turn regulate cellular function. In the myocardium, G protein signaling occurs in all cardiac cell types and is centrally involved in the regulation of heart rate, pump function, and vascular tone and in the response to hemodynamic stress and injury. Perturbations in G protein-mediated signaling are well known to contribute to cardiac hypertrophy, failure, and arrhythmias. Most of the currently used drugs for cardiac and other diseases target GPCR signaling. In the canonical G protein signaling paradigm, G proteins that are located at the cytoplasmic surface of the plasma membrane become activated after an agonist-induced conformational change of GPCRs, which then allows GTP-bound Gα and free Gβγ subunits to activate or inhibit effector proteins. Research over the past two decades has markedly broadened the original paradigm with a GPCR-G protein-effector at the cell surface at its core by revealing novel binding partners and additional subcellular localizations for heterotrimeric G proteins that facilitate many previously unrecognized functional effects. In this review, we focus on non-canonical and epigenetic-related mechanisms that regulate heterotrimeric G protein expression, activation, and localization and discuss functional consequences using cardiac examples where possible. Mechanisms reviewed involve microRNAs, histone deacetylases, chaperones, alternative modes of G protein activation, and posttranslational modifications. Some of these newly characterized mechanisms may be further developed into novel strategies for the treatment of cardiac disease and beyond.
Collapse
Affiliation(s)
- Peng Zhang
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA
| | - Celinda M Kofron
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
11
|
Stekhoven FMAHS, van der Velde G, Lee TH, Bottrill AR. Proteomic study of the brackish water mussel Mytilopsis leucophaeata. Zool Stud 2015; 54:e22. [PMID: 31966109 PMCID: PMC6661436 DOI: 10.1186/s40555-014-0081-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 12/10/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND We encountered the opportunity to study proteochemically a brackish water invertebrate animal, Mytilopsis leucophaeata, belonging to the bivalves which stem from the second half of the Cambrian Period (about 510 million years ago). This way, we were able to compare it with the vertebrate animal, the frilled shark (Chlamydoselachus anguineus) that stems from a much later period of geologic time (Permian: 245-286 MYA). RESULTS The mussel contains a well-adapted system of protein synthesis on the ER, protein folding on the ER, protein trafficking via COPI or clathrin-coated vesicles from endoplasmic reticulum (ER) to Golgi and plasmalemma, an equally well-developed system of actin filaments that with myosin forms the transport system for vesicular proteins and tubulin, which is also involved in ATP-driven vesicular protein transport via microtubules or transport of chromosomes in mitosis and meiosis. A few of the systems that we could not detect in M. leucophaeata in comparison with C. anguineus are the synaptic vesicle cycle components as synaptobrevin, cellubrevin (v-snare) and synaptosomal associated protein 25-A (t-snare), although one component: Ras-related protein (O-Rab1) could be involved in synaptic vesicle traffic. Another component that we did not find in M. leucophaeata was Rab11 that is involved in the tubulovesicular recycling process of H+/K+-ATPase in C. anguineus. We have not been able to trace the H+/K+-ATPase of M. leucophaeata, but Na+/K+-ATPase was present. Furthermore, we have studied the increase of percent protein expression between 1,070 MYA (the generation of the Amoeba Dictyostelium discoideum) and present (the generation of the mammal Sus scrofa = wild boar). In this time span, three proteomic uprises did occur: 600 to 500 MYA, 47.5 to 4.75 MYA, and 1.4 to 0 MYA. The first uprise covers the generation of bivalves, the second covers gold fish, chicken, brine shrimp, house mouse, rabbit, Japanese medaka and Rattus norvegicus, and the third covers cow, chimpanzee, Homo sapiens, dog, goat, Puccinia graminis and wild boar. We hypothesise that the latter two uprises are related to geological and climate changes and their compensation in protein function expression. CONCLUSIONS The proteomic and evolutionary data demonstrate that M. leucophaeata is a highly educatioanal animal to study.
Collapse
Affiliation(s)
- Feico MAH Schuurmans Stekhoven
- Department of Animal Ecology and Ecophysiology, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Gerard van der Velde
- Department of Animal Ecology and Ecophysiology, Faculty of Science, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
- Naturalis Biodiversity Center, P.O. Box 9517,2300RA Leiden, The Netherlands
| | - Tsung-Han Lee
- Department of Life Sciences, National Chung-Hsing University, Taichung 402, Taiwan
| | - Andrew R Bottrill
- Protein and Nucleic Acid Chemistry Laboratory, Proteomics Facility, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| |
Collapse
|
12
|
Protection of cardiomyocytes from the hypoxia-mediated injury by a peptide targeting the activator of G-protein signaling 8. PLoS One 2014; 9:e91980. [PMID: 24632710 PMCID: PMC3954831 DOI: 10.1371/journal.pone.0091980] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/16/2014] [Indexed: 11/19/2022] Open
Abstract
Signaling via heterotrimeric G-protein is involved in the development of human diseases including ischemia-reperfusion injury of the heart. We previously identified an ischemia-inducible G-protein activator, activator of G-protein signaling 8 (AGS8), which regulates Gβγ signaling and plays a key role in the hypoxia-induced apoptosis of cardiomyocytes. Here, we attempted to intervene in the AGS8-Gβγ signaling process and protect cardiomyocytes from hypoxia-induced apoptosis with a peptide that disrupted the AGS8-Gβγ interaction. Synthesized AGS8-peptides, with amino acid sequences based on those of the Gβγ-binding domain of AGS8, successfully inhibited the association of AGS8 with Gβγ. The AGS8-peptide effectively blocked hypoxia-induced apoptosis of cardiomyocytes, as determined by DNA end-labeling and an increase in cleaved caspase-3. AGS8-peptide also inhibited the change in localization/permeability of channel protein connexin 43, which was mediated by AGS8-Gβγ under hypoxia. Small compounds that inhibit a wide range of Gβγ signals caused deleterious effects in cardiomyocytes. In contrast, AGS8-peptide did not cause cell damage under normoxia, suggesting an advantage inherent in targeted disruption of the AGS8-Gβγ signaling pathway. These data indicate a pivotal role for the interaction of AGS8 with Gβγ in hypoxia-induced apoptosis of cardiomyocytes, and suggest that targeted disruption of the AGS8-Gβγ signal provides a novel approach for protecting the myocardium against ischemic injury.
Collapse
|
13
|
Blumer JB, Lanier SM. Activators of G protein signaling exhibit broad functionality and define a distinct core signaling triad. Mol Pharmacol 2014; 85:388-96. [PMID: 24302560 PMCID: PMC3935153 DOI: 10.1124/mol.113.090068] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/03/2013] [Indexed: 12/14/2022] Open
Abstract
Activators of G protein signaling (AGS), initially discovered in the search for receptor-independent activators of G protein signaling, define a broad panel of biologic regulators that influence signal transfer from receptor to G-protein, guanine nucleotide binding and hydrolysis, G protein subunit interactions, and/or serve as alternative binding partners for Gα and Gβγ independently of the classic heterotrimeric Gαβγ. AGS proteins generally fall into three groups based upon their interaction with and regulation of G protein subunits: group I, guanine nucleotide exchange factors (GEF); group II, guanine nucleotide dissociation inhibitors; and group III, entities that bind to Gβγ. Group I AGS proteins can engage all subclasses of G proteins, whereas group II AGS proteins primarily engage the Gi/Go/transducin family of G proteins. A fourth group of AGS proteins with selectivity for Gα16 may be defined by the Mitf-Tfe family of transcription factors. Groups I-III may act in concert, generating a core signaling triad analogous to the core triad for heterotrimeric G proteins (GEF + G proteins + effector). These two core triads may function independently of each other or actually cross-integrate for additional signal processing. AGS proteins have broad functional roles, and their discovery has advanced new concepts in signal processing, cell and tissue biology, receptor pharmacology, and system adaptation, providing unexpected platforms for therapeutic and diagnostic development.
Collapse
Affiliation(s)
- Joe B Blumer
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | | |
Collapse
|
14
|
Jayachandran R, Liu X, BoseDasgupta S, Müller P, Zhang CL, Moshous D, Studer V, Schneider J, Genoud C, Fossoud C, Gambino F, Khelfaoui M, Müller C, Bartholdi D, Rossez H, Stiess M, Houbaert X, Jaussi R, Frey D, Kammerer RA, Deupi X, de Villartay JP, Lüthi A, Humeau Y, Pieters J. Coronin 1 regulates cognition and behavior through modulation of cAMP/protein kinase A signaling. PLoS Biol 2014; 12:e1001820. [PMID: 24667537 PMCID: PMC3965382 DOI: 10.1371/journal.pbio.1001820] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 02/12/2014] [Indexed: 01/14/2023] Open
Abstract
Cognitive and behavioral disorders are thought to be a result of neuronal dysfunction, but the underlying molecular defects remain largely unknown. An important signaling pathway involved in the regulation of neuronal function is the cyclic AMP/Protein kinase A pathway. We here show an essential role for coronin 1, which is encoded in a genomic region associated with neurobehavioral dysfunction, in the modulation of cyclic AMP/PKA signaling. We found that coronin 1 is specifically expressed in excitatory but not inhibitory neurons and that coronin 1 deficiency results in loss of excitatory synapses and severe neurobehavioral disabilities, including reduced anxiety, social deficits, increased aggression, and learning defects. Electrophysiological analysis of excitatory synaptic transmission in amygdala revealed that coronin 1 was essential for cyclic-AMP-protein kinase A-dependent presynaptic plasticity. We further show that upon cell surface stimulation, coronin 1 interacted with the G protein subtype Gαs to stimulate the cAMP/PKA pathway. The absence of coronin 1 or expression of coronin 1 mutants unable to interact with Gαs resulted in a marked reduction in cAMP signaling. Strikingly, synaptic plasticity and behavioral defects of coronin 1-deficient mice were restored by in vivo infusion of a membrane-permeable cAMP analogue. Together these results identify coronin 1 as being important for cognition and behavior through its activity in promoting cAMP/PKA-dependent synaptic plasticity and may open novel avenues for the dissection of signal transduction pathways involved in neurobehavioral processes.
Collapse
Affiliation(s)
| | - Xiaolong Liu
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | - Chun-Lei Zhang
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | | | - Vera Studer
- Biozentrum, University of Basel, Basel, Switzerland
| | - Jacques Schneider
- Department of Radiology, University Children Hospital, UKBB, Basel, Switzerland
| | - Christel Genoud
- Center for Cellular Imaging and NanoAnalytics, University of Basel, Basel, Switzerland
- Friedrich Miescher Institute, Basel, Switzerland
| | | | | | - Malik Khelfaoui
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | | | | | | | | | - Xander Houbaert
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | - Rolf Jaussi
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Daniel Frey
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Richard A. Kammerer
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
| | - Xavier Deupi
- Biomolecular Research Laboratory, Paul Scherrer Institute, Villigen, Switzerland
- Condensed Matter Theory, Paul Scherrer Institute, Villigen, Switzerland
| | | | | | - Yann Humeau
- Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | - Jean Pieters
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
15
|
Bondar A, Lazar J. Dissociated GαGTP and Gβγ protein subunits are the major activated form of heterotrimeric Gi/o proteins. J Biol Chem 2013; 289:1271-81. [PMID: 24307173 DOI: 10.1074/jbc.m113.493643] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although most heterotrimeric G proteins are thought to dissociate into Gα and Gβγ subunits upon activation, the evidence in the Gi/o family has long been inconsistent and contradictory. The Gi/o protein family mediates inhibition of cAMP production and regulates the activity of ion channels. On the basis of experimental evidence, both heterotrimer dissociation and rearrangement have been postulated as crucial steps of Gi/o protein activation and signal transduction. We have now investigated the process of Gi/o activation in living cells directly by two-photon polarization microscopy and indirectly by observations of G protein-coupled receptor kinase-derived polypeptides. Our observations of existing fluorescently labeled and non-modified Gαi/o constructs indicate that the molecular mechanism of Gαi/o activation is affected by the presence and localization of the fluorescent label. All investigated non-labeled, non-modified Gi/o complexes dissociate extensively upon activation. The dissociated subunits can activate downstream effectors and are thus likely to be the major activated Gi/o form. Constructs of Gαi/o subunits fluorescently labeled at the N terminus (GAP43-CFP-Gαi/o) seem to faithfully reproduce the behavior of the non-modified Gαi/o subunits. Gαi constructs labeled within the helical domain (Gαi-L91-YFP) largely do not dissociate upon activation, yet still activate downstream effectors, suggesting that the dissociation seen in non-modified Gαi/o proteins is not required for downstream signaling. Our results appear to reconcile disparate published data and settle a long running dispute.
Collapse
Affiliation(s)
- Alexey Bondar
- From the Faculty of Science, University of South Bohemia, Branisovska 31a, 37005 Ceske Budejovice, Czech Republic and
| | | |
Collapse
|
16
|
Sun Z, Smrcka AV, Chen S. WDR26 functions as a scaffolding protein to promote Gβγ-mediated phospholipase C β2 (PLCβ2) activation in leukocytes. J Biol Chem 2013; 288:16715-16725. [PMID: 23625927 DOI: 10.1074/jbc.m113.462564] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have recently identified WDR26 as a novel WD40 repeat protein that binds Gβγ and promotes Gβγ signaling during leukocyte migration. Here, we have determined the mechanism by which WDR26 enhances Gβγ-mediated phospholipase C β2 (PLCβ2) activation in leukocytes. We show that WDR26 not only directly bound Gβγ but also PLCβ2. The binding sites of WDR26 and PLCβ2 on Gβ1γ2 were overlapping but not identical. WDR26 used the same domains for binding Gβγ and PLCβ but still formed a signaling complex with Gβγ and PLCβ2 probably due to the fact that WDR26 formed a higher order oligomer through its Lis homology and C-terminal to LisH (LisH-CTLH) and WD40 domains. Additional studies indicated that the formation of higher order oligomers was required for WDR26 to promote PLCβ2 interaction with and activation by Gβγ. Moreover, WDR26 was required for PLCβ2 translocation from the cytosol to the membrane in polarized leukocytes, and the translocation of PLCβ2 was sufficient to cause partial activation of PLCβ2. Collectively, our data indicate that WDR26 functions as a scaffolding protein to promote PLCβ2 membrane translocation and interaction with Gβγ, thereby enhancing PLCβ2 activation in leukocytes. These findings have identified a novel mechanism of regulating Gβγ signaling through a scaffolding protein.
Collapse
Affiliation(s)
- Zhizeng Sun
- Departments of Pharmacology, Iowa City, Iowa 52242
| | - Alan V Smrcka
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Songhai Chen
- Departments of Pharmacology, Iowa City, Iowa 52242; Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242.
| |
Collapse
|
17
|
Differential regulation of chemotaxis: Role of Gβγ in chemokine receptor-induced cell migration. Cell Signal 2013; 25:729-35. [DOI: 10.1016/j.cellsig.2012.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/17/2012] [Accepted: 12/21/2012] [Indexed: 01/17/2023]
|
18
|
Runne C, Chen S. WD40-repeat proteins control the flow of Gβγ signaling for directional cell migration. Cell Adh Migr 2013; 7:214-8. [PMID: 23302952 DOI: 10.4161/cam.22940] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The ability of cells to generate a highly polarized intracellular signal through G protein-coupled receptors (GPCRs) is essential for their migration toward chemoattractants. The Gβγ subunits of heterotrimeric G proteins play a critical role in transmitting chemotactic signals from GPCRs via the activation of diverse effectors, including PLCβ and PI3K, primarily at the leading edge of cells. Although Gβγ can directly activate many of these effectors through protein-protein interactions in vitro, it remains unclear how Gβγ spatially and temporally orchestrates the activation of these effectors in vivo. A yeast two-hybrid screen for Gβ interacting proteins identified two WD40-repeat domain containing proteins, RACK1 and WDR26, which are predicted to serve as scaffolding/adaptor proteins. Previous data indicates that RACK1 negatively regulates Gβγ-mediated leukocyte migration by inhibiting Gβγ-stimulated PLCβ and PI3K activities. In contrast, recently published work by Sun et al. indicates that WDR26 promotes leukocyte migration by enhancing Gβγ-mediated signal transduction. These findings reveal a novel mechanism regulating Gβγ signaling during chemotaxis, namely through the positive and negative regulation of WDR26 and RACK1 on Gβγ to promote and fine tune Gβγ-mediated effector activation, ultimately governing the ability of cells to polarize and migrate toward a chemoattractant gradient.
Collapse
Affiliation(s)
- Caitlin Runne
- Department of Pharmacology; Roy J. and Lucille A. Carver College of Medicine; University of Iowa; Iowa City, IA USA
| | | |
Collapse
|
19
|
San Martin L, Cerda F, Jimenez V, Fuentealba J, Muñoz B, Aguayo LG, Guzman L. Inhibition of the ethanol-induced potentiation of α1 glycine receptor by a small peptide that interferes with Gβγ binding. J Biol Chem 2012; 287:40713-21. [PMID: 23035114 DOI: 10.1074/jbc.m112.393603] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gβγ interaction with GlyR is an important determinant in ethanol potentiation of this channel. RESULTS A small peptide, RQH(C7), can inhibit ethanol potentiation of GlyR currents. CONCLUSION Results with RQH(C7) indicate that ethanol mediated potentiation of GlyR is in part by Gβγ activation. SIGNIFICANCE Molecular interaction between Gβγ and GlyR could be used as a target for pharmacological modification of ethanol effects. Previous studies indicate that ethanol can modulate glycine receptors (GlyR), in part, through Gβγ interaction with basic residues in the intracellular loop. In this study, we show that a seven-amino acid peptide (RQH(C7)), which has the primary structure of a motif in the large intracellular loop of GlyR (GlyR-IL), was able to inhibit the ethanol-elicited potentiation of this channel from 47 ± 2 to 16 ± 4%, without interfering with the effect of Gβγ on GIRK (G protein activated inwardly rectifying potassium channel) activation. RQH(C7) displayed a concentration-dependent effect on ethanol action in evoked and synaptic currents. A fragment of GlyR-IL without the basic amino acids did not interact with Gβγ or inhibit ethanol potentiation of GlyR. In silico analysis using docking and molecular dynamics allowed to identify a region of ~350Å(2) involving aspartic acids 186, 228, and 246 in Gβγ where we propose that RQH(C7) binds and exerts its blocking action on the effect of ethanol in GlyR.
Collapse
Affiliation(s)
- Loreto San Martin
- Department of Physiology, Faculty of Biological Sciences, University of Concepcion, 403901 Concepcion, Chile
| | | | | | | | | | | | | |
Collapse
|
20
|
Blumer JB, Oner SS, Lanier SM. Group II activators of G-protein signalling and proteins containing a G-protein regulatory motif. Acta Physiol (Oxf) 2012; 204:202-18. [PMID: 21615707 DOI: 10.1111/j.1748-1716.2011.02327.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Beyond the core triad of receptor, Gαβγ and effector, there are multiple accessory proteins that provide alternative modes of signal input and regulatory adaptability to G-protein signalling systems. Such accessory proteins may segregate a signalling complex to microdomains of the cell, regulate the basal activity, efficiency and specificity of signal propagation and/or serve as alternative binding partners for Gα or Gβγ independent of the classical heterotrimeric Gαβγ complex. The latter concept led to the postulate that Gα and Gβγ regulate intracellular events distinct from their role as transducers for cell surface seven-transmembrane span receptors. One general class of such accessory proteins is defined by AGS proteins or activators of G-protein signalling that refer to mammalian cDNAs identified in a specific yeast-based functional screen. The discovery of AGS proteins and related entities revealed a number of unexpected mechanisms for regulation of G-protein signalling systems and expanded functional roles for this important signalling system.
Collapse
Affiliation(s)
- J B Blumer
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, 29425, USA
| | | | | |
Collapse
|
21
|
Oner SS, An N, Vural A, Breton B, Bouvier M, Blumer JB, Lanier SM. Regulation of the AGS3·G{alpha}i signaling complex by a seven-transmembrane span receptor. J Biol Chem 2010; 285:33949-58. [PMID: 20716524 DOI: 10.1074/jbc.m110.138073] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
G-protein signaling modulators (GPSM) play diverse functional roles through their interaction with G-protein subunits. AGS3 (GPSM1) contains four G-protein regulatory motifs (GPR) that directly bind Gα(i) free of Gβγ providing an unusual scaffold for the "G-switch" and signaling complexes, but the mechanism by which signals track into this scaffold are not well understood. We report the regulation of the AGS3·Gα(i) signaling module by a cell surface, seven-transmembrane receptor. AGS3 and Gα(i1) tagged with Renilla luciferase or yellow fluorescent protein expressed in mammalian cells exhibited saturable, specific bioluminescence resonance energy transfer indicating complex formation in the cell. Activation of α(2)-adrenergic receptors or μ-opioid receptors reduced AGS3-RLuc·Gα(i1)-YFP energy transfer by over 30%. The agonist-mediated effects were inhibited by pertussis toxin and co-expression of RGS4, but were not altered by Gβγ sequestration with the carboxyl terminus of GRK2. Gα(i)-dependent and agonist-sensitive bioluminescence resonance energy transfer was also observed between AGS3 and cell-surface receptors typically coupled to Gα(i) and/or Gα(o) indicating that AGS3 is part of a larger signaling complex. Upon receptor activation, AGS3 reversibly dissociates from this complex at the cell cortex. Receptor coupling to both Gαβγ and GPR-Gα(i) offer additional flexibility for systems to respond and adapt to challenges and orchestrate complex behaviors.
Collapse
Affiliation(s)
- Sukru Sadik Oner
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Sato M, Ishikawa Y. Accessory proteins for heterotrimeric G-protein: Implication in the cardiovascular system. PATHOPHYSIOLOGY 2010; 17:89-99. [DOI: 10.1016/j.pathophys.2009.03.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 03/11/2009] [Accepted: 03/20/2009] [Indexed: 01/19/2023] Open
|
23
|
Jensen JB, Lyssand JS, Hague C, Hille B. Fluorescence changes reveal kinetic steps of muscarinic receptor-mediated modulation of phosphoinositides and Kv7.2/7.3 K+ channels. ACTA ACUST UNITED AC 2010; 133:347-59. [PMID: 19332618 PMCID: PMC2699104 DOI: 10.1085/jgp.200810075] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
G protein–coupled receptors initiate signaling cascades. M1 muscarinic receptor (M1R) activation couples through Gαq to stimulate phospholipase C (PLC), which cleaves phosphatidylinositol 4,5-bisphosphate (PIP2). Depletion of PIP2 closes PIP2-requiring Kv7.2/7.3 potassium channels (M current), thereby increasing neuronal excitability. This modulation of M current is relatively slow (6.4 s to reach within 1/e of the steady-state value). To identify the rate-limiting steps, we investigated the kinetics of each step using pairwise optical interactions likely to represent fluorescence resonance energy transfer for M1R activation, M1R/Gβ interaction, Gαq/Gβ separation, Gαq/PLC interaction, and PIP2 hydrolysis. Electrophysiology was used to monitor channel closure. Time constants for M1R activation (<100 ms) and M1R/Gβ interaction (200 ms) are both fast, suggesting that neither of them is rate limiting during muscarinic suppression of M current. Gαq/Gβ separation and Gαq/PLC interaction have intermediate 1/e times (2.9 and 1.7 s, respectively), and PIP2 hydrolysis (6.7 s) occurs on the timescale of M current suppression. Overexpression of PLC accelerates the rate of M current suppression threefold (to 2.0 s) to become nearly contemporaneous with Gαq/PLC interaction. Evidently, channel release of PIP2 and closure are rapid, and the availability of active PLC limits the rate of M current suppression.
Collapse
Affiliation(s)
- Jill B Jensen
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | | |
Collapse
|
24
|
NMR analysis of G-protein betagamma subunit complexes reveals a dynamic G(alpha)-Gbetagamma subunit interface and multiple protein recognition modes. Proc Natl Acad Sci U S A 2009; 107:639-44. [PMID: 20018744 DOI: 10.1073/pnas.0909503107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
G-protein betagamma (Gbetagamma) subunits interact with a wide range of molecular partners including: G(alpha) subunits, effectors, peptides, and small molecule inhibitors. The molecular mechanisms underlying the ability to accommodate this wide range of structurally distinct binding partners are not well understood. To uncover the role of protein flexibility and alterations in protein conformation in molecular recognition by Gbetagamma, a method for site-specific (15)N-labeling of Gbeta-Trp residue backbone and indole amines in insect cells was developed. Transverse Relaxation Optimized Spectroscopy-Heteronuclear Single-Quantum Coherence Nuclear Magnetic Resonance (TROSY-HSQC NMR) analysis of (15)N-Trp Gbetagamma identified well-dispersed signals for the individual Trp residue side chain and amide positions. Surprisingly, a wide range of signal intensities was observed in the spectrum, likely representing a range of backbone and side chain mobilities. The signal for GbetaW99 indole was very intense, suggesting a high level of mobility on the protein surface and molecular dynamics simulations indicate that GbetaW99 is highly mobile on the nanosecond timescale in comparison with other Gbeta tryptophans. Binding of peptides and phosducin dramatically altered the mobility of GbetaW99 and GbetaW332 in the binding site and the chemical shifts at sites distant from the direct binding surface in distinct ways. In contrast, binding of G(alpha)(i1)-GDP to Gbetagamma had relatively little effect on the spectrum and, most surprisingly, did not significantly alter Trp mobility at the subunit interface. This suggests the inactive heterotrimer in solution adopts a conformation with an open subunit interface a large percentage of the time. Overall, these data show that Gbetagamma subunits explore a range of conformations that can be exploited during molecular recognition by diverse binding partners.
Collapse
|
25
|
Sadana R, Dascal N, Dessauer CW. N terminus of type 5 adenylyl cyclase scaffolds Gs heterotrimer. Mol Pharmacol 2009; 76:1256-64. [PMID: 19783621 PMCID: PMC2784731 DOI: 10.1124/mol.109.058370] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Accepted: 09/24/2009] [Indexed: 11/22/2022] Open
Abstract
According to accepted doctrine, agonist-bound G protein-coupled receptors catalyze the exchange of GDP for GTP and facilitate the dissociation of Galpha and Gbetagamma, which in turn regulate their respective effectors. More recently, the existence of preformed signaling complexes, which may include receptors, heterotrimeric G proteins, and/or effectors, is gaining acceptance. We show herein the existence of a preformed complex of inactive heterotrimer (Galpha(s) x betagamma) and the effector type 5 adenylyl cyclase (AC5), localized by the N terminus of AC5. GST fusions of AC5 N terminus (5NT) bind to purified G protein subunits (GDP-Galpha(s) and Gbetagamma) with apparent affinities of 270 +/- 21 and 190 +/- 7 nM, respectively. GDP-bound Galpha(s) and Gbetagamma did not compete, but rather facilitated their interaction with 5NT, consistent with the isolation of a ternary complex (5NT, Galpha(s), and Gbetagamma) by gel filtration. The AC5/Gbetagamma interaction was also demonstrated by immunoprecipitation and fluorescence resonance energy transfer (FRET) and the binding site of heterotrimer Galpha(s) x betagamma mapped to amino acids 60 to 129 of 5NT. Deletion of this region in full-length AC5 resulted in significant reduction of FRET between Gbetagamma and AC. 5NT also interacts with the catalytic core of AC, mainly via the C1 domain, to enhance Galpha(s)--and forskolin-stimulated activity of C1/C2 domains. The N terminus also serves to constrain Galpha(i)-mediated inhibition of AC5, which is relieved in the presence of Gbetagamma. These results reveal that 5NT plays a key regulatory role by interacting with the catalytic core and scaffolding inactive heterotrimeric G proteins, forming a preassembled complex that is potentially braced for GPCR activation.
Collapse
Affiliation(s)
- Rachna Sadana
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
26
|
Sato M, Jiao Q, Honda T, Kurotani R, Toyota E, Okumura S, Takeya T, Minamisawa S, Lanier SM, Ishikawa Y. Activator of G protein signaling 8 (AGS8) is required for hypoxia-induced apoptosis of cardiomyocytes: role of G betagamma and connexin 43 (CX43). J Biol Chem 2009; 284:31431-40. [PMID: 19723622 DOI: 10.1074/jbc.m109.014068] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ischemic injury of the heart is associated with activation of multiple signal transduction systems including the heterotrimeric G-protein system. Here, we report a role of the ischemia-inducible regulator of G betagamma subunit, AGS8, in survival of cardiomyocytes under hypoxia. Cultured rat neonatal cardiomyocytes (NCM) were exposed to hypoxia or hypoxia/reoxygenation following transfection of AGS8siRNA or pcDNA::AGS8. Hypoxia-induced apoptosis of NCM was completely blocked by AGS8siRNA, whereas overexpression of AGS8 increased apoptosis. AGS8 formed complexes with G-proteins and channel protein connexin 43 (CX43), which regulates the permeability of small molecules under hypoxic stress. AGS8 initiated CX43 phosphorylation in a G betagamma-dependent manner by providing a scaffold composed of G betagamma and CX43. AGS8siRNA blocked internalization of CX43 following exposure of NCM to repetitive hypoxia; however it did not influence epidermal growth factor-mediated internalization of CX43. The decreased dye flux through CX43 that occurred with hypoxic stress was also prevented by AGS8siRNA. Interestingly, the G betagamma inhibitor Gallein mimicked the effect of AGS8 knockdown on both the CX43 internalization and the changes in cell permeability elicited by hypoxic stress. These data indicate that AGS8 is required for hypoxia-induced apoptosis of NCM, and that AGS8-G betagamma signal input increased the sensitivity of cells to hypoxic stress by influencing CX43 regulation and associated cell permeability. Under hypoxic stress, this unrecognized response program plays a critical role in the fate of NCM.
Collapse
Affiliation(s)
- Motohiko Sato
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Fukuura, Yokohama 236-0004, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Dupré DJ, Robitaille M, Rebois RV, Hébert TE. The role of Gbetagamma subunits in the organization, assembly, and function of GPCR signaling complexes. Annu Rev Pharmacol Toxicol 2009; 49:31-56. [PMID: 18834311 DOI: 10.1146/annurev-pharmtox-061008-103038] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of Gbetagamma subunits in cellular signaling has become well established in the past 20 years. Not only do they regulate effectors once thought to be the sole targets of Galpha subunits, but it has become clear that they also have a unique set of binding partners and regulate signaling pathways that are not always localized to the plasma membrane. However, this may be only the beginning of the story. Gbetagamma subunits interact with G protein-coupled receptors, Galpha subunits, and several different effector molecules during assembly and trafficking of receptor-based signaling complexes and not simply in response to ligand stimulation at sites of receptor cellular activity. Gbetagamma assembly itself seems to be tightly regulated via the action of molecular chaperones and in turn may serve a similar role in the assembly of specific signaling complexes. We propose that specific Gbetagamma subunits have a broader role in controlling the architecture, assembly, and activity of cellular signaling pathways.
Collapse
Affiliation(s)
- Denis J Dupré
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | |
Collapse
|
28
|
McIntire WE. Structural determinants involved in the formation and activation of G protein betagamma dimers. Neurosignals 2009; 17:82-99. [PMID: 19212142 DOI: 10.1159/000186692] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 05/13/2008] [Indexed: 01/08/2023] Open
Abstract
Heterotrimeric G proteins, composed of an alpha, beta and gamma subunit, represent one of the most important and dynamic families of signaling proteins. As a testament to the significance of G protein signaling, the hundreds of seven-transmembrane-spanning receptors that interact with G proteins are estimated to occupy 1-2% of the human genome. This broad diversity of receptors is echoed in the number of potential heterotrimer combinations that can arise from the 23 alpha subunit, 7 beta subunit and 12 gamma subunit isoforms that have been identified. The potential for such vast complexity implies that the receptor G protein interface is the site of much regulation. The historical model for the activation of a G protein holds that activated receptor catalyzes the exchange of GDP for GTP on the alpha subunit, inducing a conformational change that substantially lowers the affinity of alpha for betagamma. This decreased affinity enables dissociation of betagamma from alpha and receptor. The free form of betagamma is thought to activate effectors, until the hydrolysis of GTP by G alpha (aided by RGS proteins) allows the subunits to re-associate, effectively deactivating the G protein until another interaction with activated receptor.
Collapse
Affiliation(s)
- William E McIntire
- Department of Pharmacology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| |
Collapse
|
29
|
Layden BT, Saengsawang W, Donati RJ, Yang S, Mulhearn DC, Johnson ME, Rasenick MM. Structural model of a complex between the heterotrimeric G protein, Gsalpha, and tubulin. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:964-73. [PMID: 18373982 DOI: 10.1016/j.bbamcr.2008.02.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 02/11/2008] [Accepted: 02/13/2008] [Indexed: 11/17/2022]
Abstract
A number of studies have demonstrated interplay between the cytoskeleton and G protein signaling. Many of these studies have determined a specific interaction between tubulin, the building block of microtubules, and G proteins. The alpha subunits of some heterotrimeric G proteins, including Gsalpha, have been shown to interact strongly with tubulin. Binding of Galpha to tubulin results in increased dynamicity of microtubules due to activation of GTPase of tubulin. Tubulin also activates Gsalpha via a direct transfer of GTP between these molecules. Structural insight into the interaction between tubulin and Gsalpha was required, and was determined, in this report, through biochemical and molecular docking techniques. Solid phase peptide arrays suggested that a portion of the amino terminus, alpha2-beta4 (the region between switch II and switch III) and alpha3-beta5 (just distal to the switch III region) domains of Gsalpha are important for interaction with tubulin. Molecular docking studies revealed the best-fit models based on the biochemical data, showing an interface between the two molecules that includes the adenylyl cyclase/Gbetagamma interaction regions of Gsalpha and the exchangeable nucleotide-binding site of tubulin. These structural models explain the ability of tubulin to facilitate GTP exchange on Galpha and the ability of Galpha to activate tubulin GTPase.
Collapse
Affiliation(s)
- Brian T Layden
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Optical techniques to analyze real-time activation and signaling of G-protein-coupled receptors. Trends Pharmacol Sci 2008; 29:159-65. [PMID: 18262662 DOI: 10.1016/j.tips.2007.12.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2007] [Revised: 12/14/2007] [Accepted: 12/17/2007] [Indexed: 12/29/2022]
Abstract
The activation of G-protein-coupled receptors (GPCRs) is traditionally measured either by monitoring downstream physiological events or by membrane-based biochemical assays. Neither of these approaches permits detailed kinetic or spatial analysis of receptor activation and signaling. Recently, several optical techniques have been developed to monitor receptor activation either by using purified reconstituted GPCRs or by observing GPCRs, G proteins and second messengers in intact cells. These techniques are providing, literally, new views on both the mechanistic basis of the signaling process and the kinetic and spatial properties of GPCR-mediated signals. They suggest that agonists can activate GPCRs within milliseconds, that different compounds can induce distinct active conformations of GPCRs, that G-protein activation is the rate-limiting step in GPCR signaling, and that cellular signals can be temporally and spatially confined. They are also raising controversial issues, such as whether or not receptors and G proteins are pre-coupled and whether G proteins dissociate during activation.
Collapse
|
31
|
GTPase acceleration as the rate-limiting step in Arabidopsis G protein-coupled sugar signaling. Proc Natl Acad Sci U S A 2007; 104:17317-22. [PMID: 17951432 DOI: 10.1073/pnas.0704751104] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heterotrimeric G protein signaling is important for cell-proliferative and glucose-sensing signal transduction pathways in the model plant organism Arabidopsis thaliana. AtRGS1 is a seven-transmembrane, RGS domain-containing protein that is a putative membrane receptor for d-glucose. Here we show, by using FRET, that d-glucose alters the interaction between the AtGPA1 and AtRGS1 in vivo. AtGPA1 is a unique heterotrimeric G protein alpha subunit that is constitutively GTP-bound given its high spontaneous nucleotide exchange coupled with slow GTP hydrolysis. Analysis of a point mutation in AtRGS1 that abrogates GTPase-accelerating activity demonstrates that the regulation of AtGPA1 GTP hydrolysis mediates sugar signal transduction during Arabidopsis development, in contrast to animals where nucleotide exchange is the limiting step in the heterotrimeric G protein nucleotide cycle.
Collapse
|