51
|
Hammad MM, Kuang YQ, Yan R, Allen H, Dupré DJ. Na+/H+ exchanger regulatory factor-1 is involved in chemokine receptor homodimer CCR5 internalization and signal transduction but does not affect CXCR4 homodimer or CXCR4-CCR5 heterodimer. J Biol Chem 2010; 285:34653-64. [PMID: 20801883 PMCID: PMC2966081 DOI: 10.1074/jbc.m110.106591] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 08/25/2010] [Indexed: 11/06/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor (GPCR) family. CCR5 is also the principal co-receptor for macrophage-tropic strains of human immunodeficiency virus, type 1 (HIV-1), and efforts have been made to develop ligands to inhibit HIV-1 infection by promoting CCR5 receptor endocytosis. Given the nature of GPCRs and their propensity to form oligomers, one can consider ligand-based therapies as unselective in terms of the oligomeric composition of complexes. For example, a ligand targeting a CCR5 homomer could likely induce signal transduction on a heteromeric CCR5-CXCR4. Other avenues could therefore be explored. We identified a receptor adaptor interacting specifically with one receptor complex but not others. NHERF1, an adaptor known for its role in desensitization, internalization, and regulation of the ERK signaling cascade for several GPCRs, interacts via its PDZ2 domain with the CCR5 homodimer but not with the CXCR4-CCR5 heterodimer or CXCR4 homodimer. To further characterize this interaction, we also show that NHERF1 increases the CCR5 recruitment of arrestin2 following stimulation. NHERF1 is also involved in CCR5 internalization, as we demonstrate that co-expression of constructs bearing the PDZ2 domain can block CCR5 internalization. We also show that NHERF1 potentiates RANTES (regulated on activation normal T cell expressed and secreted)-induced ERK1/2 phosphorylation via CCR5 activation and that this activation requires NHERF1 but not arrestin2. Taken together, our results suggest that oligomeric receptor complexes can associate specifically with partners and that in this case NHERF1 could represent an interesting new target for the regulation of CCR5 internalization and potentially HIV infection.
Collapse
Affiliation(s)
- Maha M. Hammad
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Yi-Qun Kuang
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Ronald Yan
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Heather Allen
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | - Denis J. Dupré
- From the Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| |
Collapse
|
52
|
Abstract
We studied the axons of the pyloric dilator neurons in the stomatogastric nervous system of the lobster. The several-centimeters-long portions of these axons in the motor nerves depolarize in response to low concentrations of dopamine (DA) and exhibit peripheral spike initiation in the absence of centrally generated activity. This effect is inhibited by blockers of hyperpolarization-activated inward current (I(h)). We show here that peripheral spike initiation was also elicited by D(1)-type receptor agonists and drugs that increase cAMP. This suggests that DA acts via a D(1)-type receptor mechanism to modulate hyperpolarization-activated cyclic nucleotide-gated channels. We used two-electrode voltage clamp of the axon to directly study the effect of DA on I(h). Surprisingly, DA decreased the maximal conductance. However, because of a shift of the activation curve to more depolarized potentials, and a change in the slope, conductance was increased at biologically relevant membrane potentials. These changes were solely caused by modulation of I(h), as DA had no discernible effect when I(h) was blocked. In addition, they were not induced by repeated activation and could be mimicked by application of drugs that increase cAMP concentration. DA modulation of I(h) persisted in the presence of a protein kinase A inhibitor and is therefore potentially mediated by a phosphorylation-independent direct effect of cAMP on the ion channel. A computer model of the axon showed that the changes in maximal conductance and voltage dependence were not qualitatively affected by space-clamp problems.
Collapse
|
53
|
Iegorova O, Fisyunov A, Krishtal O. G-protein-independent modulation of P-type calcium channels by mu-opioids in Purkinje neurons of rat. Neurosci Lett 2010; 480:106-11. [PMID: 20541588 DOI: 10.1016/j.neulet.2010.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 05/26/2010] [Accepted: 06/03/2010] [Indexed: 11/29/2022]
Abstract
P-type calcium channels play a key role in the synaptic transmission between mammalian central neurons since a major part of calcium entering pre-synaptic terminals is delivered via these channels. Using conventional whole-cell patch clamp techniques we have studied the effect of mu-opioids on P-type calcium channels in acutely isolated Purkinje neurons from rat cerebellum. The selective mu-opioid agonist DAMGO (10nM) produced a small, but consistent facilitation of current through P-type calcium channels (10+/-1%, n=27, p<0.001). The effect of DAMGO was rapid (less than 10s) and fully reversible. This effect was both concentration and voltage-dependent. The EC(50) for the effect of DAMGO was 1.3+/-0.4nM and the saturating concentration was 100nM. The endogenous selective agonist of mu-opioid receptors, endomorphin-1 demonstrated similar action. Intracellular perfusion of Purkinje neurons with GTPgammaS (0.5mM) or GDPbetaS (0.5mM), as well as strong depolarizing pre-pulses (+50mV), did not eliminate facilitatory action of DAMGO on P-channels indicating that this effect is not mediated by G-proteins. Furthermore, the effect of DAMGO was preserved in the presence of a non-specific inhibitor of PKA and PKC (H7, 10microM) inside the cell. DAMGO-induced facilitation of P-current was almost completely abolished by the selective mu-opioid antagonist CTOP (100nM). These observations indicate that mu-type opioid receptors modulate P-type calcium channels in Purkinje neurons via G-protein-independent mechanism.
Collapse
Affiliation(s)
- Olena Iegorova
- Bogomoletz Institute of Physiology, Department of Cellular Membranology, 4 Bogomoletz Street, Kiev 01024, Ukraine
| | | | | |
Collapse
|
54
|
Rondou P, Haegeman G, Van Craenenbroeck K. The dopamine D4 receptor: biochemical and signalling properties. Cell Mol Life Sci 2010; 67:1971-86. [PMID: 20165900 PMCID: PMC11115718 DOI: 10.1007/s00018-010-0293-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/19/2010] [Accepted: 01/26/2010] [Indexed: 01/20/2023]
Abstract
Dopamine is an important neurotransmitter that regulates several key functions in the brain, such as motor output, motivation and reward, learning and memory, and endocrine regulation. Dopamine does not mediate fast synaptic transmission, but rather modulates it by triggering slow-acting effects through the activation of dopamine receptors, which belong to the G-protein-coupled receptor superfamily. Besides activating different effectors through G-protein coupling, dopamine receptors also signal through interaction with a variety of proteins, collectively termed dopamine receptor-interacting proteins. We focus on the dopamine D4 receptor, which contains an important polymorphism in its third intracellular loop. This polymorphism has been the subject of numerous studies investigating links with several brain disorders, such as attention-deficit hyperactivity disorder and schizophrenia. We provide an overview of the structure, signalling properties and regulation of dopamine D4 receptors, and briefly discuss their physiological and pathophysiological role in the brain.
Collapse
Affiliation(s)
- Pieter Rondou
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
- Present Address: Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Medical Research Building, De Pintelaan 185, 9000 Ghent, Belgium
| | - Guy Haegeman
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
| | - Kathleen Van Craenenbroeck
- Laboratory of Eukaryotic Gene Expression and Signal Transduction (LEGEST), Ghent University (UGent), K.L. Ledeganckstraat 35, 9000 Ghent, Belgium
| |
Collapse
|
55
|
Weinman EJ, Biswas R, Steplock D, Douglass TS, Cunningham R, Shenolikar S. Sodium-hydrogen exchanger regulatory factor 1 (NHERF-1) transduces signals that mediate dopamine inhibition of sodium-phosphate co-transport in mouse kidney. J Biol Chem 2010; 285:13454-60. [PMID: 20200151 DOI: 10.1074/jbc.m109.094359] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dopamine inhibited phosphate transport in isolated renal brush border membrane vesicles and in cultured renal proximal tubule cells from wild-type but not from NHERF-1 null mice. Co-immunoprecipitation experiments established that NHERF-1 associated with D1-like receptors. In wild-type mice, dopamine stimulated cAMP accumulation and protein kinase C (PKC) activity in renal proximal tubule cells, an effect that was abolished by SCH-23390, a D1-like receptor antagonist. In NHERF-1 null kidney tissue; however, dopamine failed to stimulate either cAMP accumulation or PKC activity. Infection of proximal tubule cells from NHERF-1 null mice with adenovirus-green fluorescent protein-NHERF-1 restored the ability of dopamine to stimulate cAMP and PKC. Finally, in (32)P-labeled wild-type proximal tubule cells and in opossum kidney cells, dopamine increased NHERF-1 phosphorylation at serine 77 of the PDZ I domain of NHERF-1, a site previously shown to attenuate binding of cellular targets including the Npt2a (sodium-dependent phosphate transporter 2a). Together, these studies establish that NHERF-1 plays a key role in dopamine signaling and is also a downstream target of D1-like receptors in the mouse kidney. These studies suggest a novel role for the PDZ adapter protein NHERF-1 in coordinating dopamine signals that inhibit renal phosphate transport.
Collapse
Affiliation(s)
- Edward J Weinman
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA.
| | | | | | | | | | | |
Collapse
|
56
|
McCoy KL, Hepler JR. Regulators of G protein signaling proteins as central components of G protein-coupled receptor signaling complexes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 86:49-74. [PMID: 20374713 DOI: 10.1016/s1877-1173(09)86003-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The regulators of G protein signaling (RGS) proteins bind directly to G protein alpha (Gα) subunits to regulate the signaling functions of Gα and their linked G protein-coupled receptors (GPCRs). Recent studies indicate that RGS proteins also interact with GPCRs, not just G proteins, to form preferred functional pairs. Interactions between GPCRs and RGS proteins may be direct or indirect (via a linker protein) and are dictated by the receptors, rather than the linked G proteins. Emerging models suggest that GPCRs serve as platforms for assembling an overlapping and distinct constellation of signaling proteins that perform receptor-specific signaling tasks. Compelling evidence now indicates that RGS proteins are central components of these GPCR signaling complexes. This review will outline recent discoveries of GPCR/RGS pairs as well as new data in support of the idea that GPCRs serve as platforms for the formation of multiprotein signaling complexes.
Collapse
Affiliation(s)
- Kelly L McCoy
- Department of Pharmacology, G205 Rollins Research Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
57
|
Lin WZ, Xiao X, Chou KC. GPCR-GIA: a web-server for identifying G-protein coupled receptors and their families with grey incidence analysis. Protein Eng Des Sel 2009; 22:699-705. [PMID: 19776029 DOI: 10.1093/protein/gzp057] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) play fundamental roles in regulating various physiological processes as well as the activity of virtually all cells. Different GPCR families are responsible for different functions. With the avalanche of protein sequences generated in the postgenomic age, it is highly desired to develop an automated method to address the two problems: given the sequence of a query protein, can we identify whether it is a GPCR? If it is, what family class does it belong to? Here, a two-layer ensemble classifier called GPCR-GIA was proposed by introducing a novel scale called 'grey incident degree'. The overall success rate by GPCR-GIA in identifying GPCR and non-GPCR was about 95%, and that in identifying the GPCRs among their nine family classes was about 80%. These rates were obtained by the jackknife cross-validation tests on the stringent benchmark data sets where none of the proteins has > or = 50% pairwise sequence identity to any other in a same class. Moreover, a user-friendly web-server was established at http://218.65.61.89:8080/bioinfo/GPCR-GIA. For user's convenience, a step-by-step guide on how to use the GPCR-GIA web server is provided. Generally speaking, one can get the desired two-level results in around 10 s for a query protein sequence of 300-400 amino acids; the longer the sequence is, the more time that is needed.
Collapse
Affiliation(s)
- Wei-Zhong Lin
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen 333001, China
| | | | | |
Collapse
|
58
|
Kennedy DC, Tay LL, Lyn RK, Rouleau Y, Hulse J, Pezacki JP. Nanoscale aggregation of cellular beta2-adrenergic receptors measured by plasmonic interactions of functionalized nanoparticles. ACS NANO 2009; 3:2329-39. [PMID: 19702324 DOI: 10.1021/nn900488u] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Adrenergic signaling that controls the contraction of cardiac myocyte cells and the beating of the mammalian heart is initiated by ligand binding to adrenergic receptors contained in nanoscale multiprotein complexes at the cellular membrane. Here we demonstrate that the surface-enhanced Raman scattering (SERS) of functionalized silver nanoparticles can be used to report on the receptor aggregation state of specifically label beta(2)-adrenergic receptors on mouse cardiac myocyte cells. Furthermore, multimodal imaging including Raman, Rayleigh scattering, scanning electron microscopy, and luminescence imaging was combined to fully characterize the beta(2)-adrenergic receptor-mediated aggregation of silver nanoparticles on the membrane of cardiac myocytes. Scanning electron microscopy analysis reveals distinct SERS active clusters of between 10 and 70 nanoparticles per signaling domain from ultra-high-resolution images of beta(2)-adrenergic receptor clusters on the cellular membrane. These techniques can be generally applied to study the aggregation of other cell surface receptors and explore their distribution on cell surfaces.
Collapse
MESH Headings
- Animals
- Cell Line
- Cell Membrane/chemistry
- Metal Nanoparticles/analysis
- Metal Nanoparticles/chemistry
- Metal Nanoparticles/ultrastructure
- Mice
- Microscopy, Electron, Scanning
- Microscopy, Fluorescence
- Models, Molecular
- Molecular Structure
- Muscle Cells/chemistry
- Muscle Cells/metabolism
- Receptors, Adrenergic, beta-2/analysis
- Receptors, Adrenergic, beta-2/chemistry
- Receptors, Adrenergic, beta-2/metabolism
- Silver/chemistry
- Spectrum Analysis, Raman/methods
Collapse
Affiliation(s)
- David C Kennedy
- Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Canada
| | | | | | | | | | | |
Collapse
|
59
|
Dunham JH, Hall RA. Enhancement of the surface expression of G protein-coupled receptors. Trends Biotechnol 2009; 27:541-5. [PMID: 19679364 DOI: 10.1016/j.tibtech.2009.06.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 06/18/2009] [Accepted: 06/19/2009] [Indexed: 11/27/2022]
Abstract
G protein-coupled receptors (GPCRs) mediate physiological responses to a diverse array of stimuli and are the molecular targets for numerous therapeutic drugs. GPCRs primarily signal from the plasma membrane, but when expressed in heterologous cells many GPCRs exhibit poor trafficking to the cell surface. Multiple approaches have been taken to enhance GPCR surface expression in heterologous cells, including addition/deletion of receptor sequences, co-expression with interacting proteins, and treatment with pharmacological chaperones. In addition to providing enhanced surface expression of certain GPCRs in heterologous cells, these approaches have also shed light on the control of GPCR trafficking in vivo and in some cases have led to new therapeutic approaches for treating human diseases that result from defects in GPCR trafficking.
Collapse
Affiliation(s)
- Jill H Dunham
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA
| | | |
Collapse
|
60
|
Casadó V, Cortés A, Mallol J, Pérez-Capote K, Ferré S, Lluis C, Franco R, Canela EI. GPCR homomers and heteromers: a better choice as targets for drug development than GPCR monomers? Pharmacol Ther 2009; 124:248-57. [PMID: 19664655 DOI: 10.1016/j.pharmthera.2009.07.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 07/21/2009] [Indexed: 10/20/2022]
Abstract
G protein-coupled receptors (GPCR) are targeted by many therapeutic drugs marketed to fight against a variety of diseases. Selection of novel lead compounds are based on pharmacological parameters obtained assuming that GPCR are monomers. However, many GPCR are expressed as dimers/oligomers. Therefore, drug development may consider GPCR as homo- and hetero-oligomers. A two-state dimer receptor model is now available to understand GPCR operation and to interpret data obtained from drugs interacting with dimers, and even from mixtures of monomers and dimers. Heteromers are distinct entities and therefore a given drug is expected to have different affinities and different efficacies depending on the heteromer. All these concepts would lead to broaden the therapeutic potential of drugs targeting GPCRs, including receptor heteromer-selective drugs with a lower incidence of side effects, or to identify novel pharmacological profiles using cell models expressing receptor heteromers.
Collapse
Affiliation(s)
- Vicent Casadó
- Departament de Bioquímica i Biologia Molecular, CIBERNED (Centro de Investigación en Red de Enfermedades Neurodegenerativas) and Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Xiao X, Wang P, Chou KC. GPCR-CA: A cellular automaton image approach for predicting G-protein-coupled receptor functional classes. J Comput Chem 2009; 30:1414-23. [PMID: 19037861 DOI: 10.1002/jcc.21163] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Given an uncharacterized protein sequence, how can we identify whether it is a G-protein-coupled receptor (GPCR) or not? If it is, which functional family class does it belong to? It is important to address these questions because GPCRs are among the most frequent targets of therapeutic drugs and the information thus obtained is very useful for "comparative and evolutionary pharmacology," a technique often used for drug development. Here, we present a web-server predictor called "GPCR-CA," where "CA" stands for "Cellular Automaton" (Wolfram, S. Nature 1984, 311, 419), meaning that the CA images have been utilized to reveal the pattern features hidden in piles of long and complicated protein sequences. Meanwhile, the gray-level co-occurrence matrix factors extracted from the CA images are used to represent the samples of proteins through their pseudo amino acid composition (Chou, K.C. Proteins 2001, 43, 246). GPCR-CA is a two-layer predictor: the first layer prediction engine is for identifying a query protein as GPCR on non-GPCR; if it is a GPCR protein, the process will be automatically continued with the second-layer prediction engine to further identify its type among the following six functional classes: (a) rhodopsin-like, (b) secretin-like, (c) metabotrophic/glutamate/pheromone; (d) fungal pheromone, (e) cAMP receptor, and (f) frizzled/smoothened family. The overall success rates by the predictor for the first and second layers are over 91% and 83%, respectively, that were obtained through rigorous jackknife cross-validation tests on a new-constructed stringent benchmark dataset in which none of proteins has >or=40% pairwise sequence identity to any other in a same subset. GPCR-CA is freely accessible at http://218.65.61.89:8080/bioinfo/GPCR-CA, by which one can get the desired two-layer results for a query protein sequence within about 20 seconds.
Collapse
Affiliation(s)
- Xuan Xiao
- Computer Department, Jing-De-Zhen Ceramic Institute, Jing-De-Zhen 33300, China.
| | | | | |
Collapse
|
62
|
Cagliani R, Fumagalli M, Pozzoli U, Riva S, Cereda M, Comi GP, Pattini L, Bresolin N, Sironi M. A complex selection signature at the human AVPR1B gene. BMC Evol Biol 2009; 9:123. [PMID: 19486526 PMCID: PMC2700802 DOI: 10.1186/1471-2148-9-123] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 06/01/2009] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The vasopressin receptor type 1b (AVPR1B) is mainly expressed by pituitary corticotropes and it mediates the stimulatory effects of AVP on ACTH release; common AVPR1B haplotypes have been involved in mood and anxiety disorders in humans, while rodents lacking a functional receptor gene display behavioral defects and altered stress responses. RESULTS Here we have analyzed the two exons of the gene and the data we present suggest that AVPR1B has been subjected to natural selection in humans. In particular, analysis of exon 2 strongly suggests the action of balancing selection in African populations and Europeans: the region displays high nucleotide diversity, an excess of intermediate-frequency alleles, a higher level of within-species diversity compared to interspecific divergence and a genealogy with common haplotypes separated by deep branches. This relatively unambiguous situation coexists with unusual features across exon 1, raising the possibility that a nonsynonymous variant (Gly191Arg) in this region has been subjected to directional selection. CONCLUSION Although the underlying selective pressure(s) remains to be identified, we consider this to be among the first documented examples of a gene involved in mood disorders and subjected to natural selection in humans; this observation might add support to the long-debated idea that depression/low mood might have played an adaptive role during human evolution.
Collapse
Affiliation(s)
- Rachele Cagliani
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| | - Matteo Fumagalli
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
- Bioengineering Department, Politecnico di Milano, P.zza L. da Vinci, 32, 20133 Milan, Italy
| | - Uberto Pozzoli
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| | - Stefania Riva
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| | - Matteo Cereda
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| | - Giacomo P Comi
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Ospedale Maggiore Policlinico, Mangiagalli and Regina Elena Foundation, Via F. Sforza 35, 20100 Milan, Italy
| | - Linda Pattini
- Bioengineering Department, Politecnico di Milano, P.zza L. da Vinci, 32, 20133 Milan, Italy
| | - Nereo Bresolin
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
- Dino Ferrari Centre, Department of Neurological Sciences, University of Milan, IRCCS Ospedale Maggiore Policlinico, Mangiagalli and Regina Elena Foundation, Via F. Sforza 35, 20100 Milan, Italy
| | - Manuela Sironi
- Scientific Institute IRCCS E. Medea, Bioinformatic Lab, Via don L. Monza 20, 23842 Bosisio Parini (LC), Italy
| |
Collapse
|
63
|
The deubiquitinases USP33 and USP20 coordinate beta2 adrenergic receptor recycling and resensitization. EMBO J 2009; 28:1684-96. [PMID: 19424180 DOI: 10.1038/emboj.2009.128] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 04/09/2009] [Indexed: 12/30/2022] Open
Abstract
Agonist-induced ubiquitination of the beta(2) adrenergic receptor (beta(2)AR) functions as an important post-translational modification to sort internalized receptors to the lysosomes for degradation. We now show that this ubiquitination is reversed by two deubiquitinating enzymes, ubiquitin-specific proteases (USPs) 20 and 33, thus, inhibiting lysosomal trafficking when concomitantly promoting receptor recycling from the late-endosomal compartments as well as resensitization of recycled receptors at the cell surface. Dissociation of constitutively bound endogenously expressed USPs 20 and 33 from the beta(2)AR immediately after agonist stimulation and reassociation on prolonged agonist treatment allows receptors to first become ubiquitinated and then deubiquitinated, thus, providing a 'trip switch' between degradative and recycling pathways at the late-endosomal compartments. Thus, USPs 20 and 33 serve as novel regulators that dictate both post-endocytic sorting as well as the intensity and extent of beta(2)AR signalling from the cell surface.
Collapse
|
64
|
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
|
65
|
Milligan G. G protein-coupled receptor hetero-dimerization: contribution to pharmacology and function. Br J Pharmacol 2009; 158:5-14. [PMID: 19309353 DOI: 10.1111/j.1476-5381.2009.00169.x] [Citation(s) in RCA: 267] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The concept that G protein-coupled receptors (GPCRs) can form hetero-dimers or hetero-oligomers continues to gain experimental support. However, with the exception of the GABA(B) receptor and the sweet and umami taste receptors few reported examples meet all of the criteria suggested in a recent International Union of Basic and Clinical Pharmacology sponsored review (Pin et al., 2007) that should be required to define distinct and physiologically relevant receptor species. Despite this, there are many examples in which pairs of co-expressed GPCRs reciprocally modulate their function, trafficking and/or ligand pharmacology. Such data are at least consistent with physical interactions between the receptor pairs. In recent times, it has been suggested that specific GPCR hetero-dimer or hetero-oligomer pairs may represent key molecular targets of certain clinically effective, small molecule drugs and there is growing interest in efforts to identify ligands that may modulate hetero-dimer function selectively. The current review summarizes key recent developments in these topics.
Collapse
Affiliation(s)
- Graeme Milligan
- Molecular Pharmacology Group, Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
66
|
Vobornik D, Rouleau Y, Haley J, Bani-Yaghoub M, Taylor R, Johnston LJ, Pezacki JP. Nanoscale organization of beta2-adrenergic receptor-Venus fusion protein domains on the surface of mammalian cells. Biochem Biophys Res Commun 2009; 382:85-90. [PMID: 19265675 DOI: 10.1016/j.bbrc.2009.02.144] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 02/24/2009] [Indexed: 01/08/2023]
Abstract
Adrenergic receptors are a key component of nanoscale multiprotein complexes that are responsible for controlling the beat rate in a mammalian heart. We demonstrate the ability of near-field scanning optical microscopy (NSOM) to visualize beta(2)-adrenergic receptors (beta(2)AR) fused to the GFP analogue Venus at the nanoscale on HEK293 cells. The expression of the beta(2)AR-Venus fusion protein was tightly controlled using a tetracycline-induced promoter. Both the size and density of the observed nanoscale domains are dependent on the level of induction and thus the level of protein expression. At concentrations between 100 and 700 ng/ml of inducer doxycycline, the size of domains containing the beta(2)AR-Venus fusion protein appears to remain roughly constant, but the number of domains per cell increase. At 700 ng/ml doxycycline the functional receptors are organized into domains with an average diameter of 150 nm with a density similar to that observed for the native protein on primary murine cells. By contrast, larger micron-sized domains of beta(2)AR are observed in the membrane of the HEK293 cells that stably overexpress beta(2)AR-GFP and beta(2)AR-eYFP. We conclude that precise chemical control of gene expression is highly advantageous for the use beta(2)AR-Venus fusion proteins as models for beta(2)AR function. These observations are critical for designing future cell models and assays based on beta(2)AR, since the receptor biology is consistent with a relatively low density of nanoscale receptor domains.
Collapse
Affiliation(s)
- Dusan Vobornik
- Steacie Institute for Molecular Sciences, National Research Council Canada, Ottawa, ON, Canada K1A 0R6
| | | | | | | | | | | | | |
Collapse
|
67
|
Ward RJ, Jenkins L, Milligan G. Selectivity and functional consequences of interactions of family A G protein-coupled receptors with neurochondrin and periplakin. J Neurochem 2009; 109:182-92. [PMID: 19166508 DOI: 10.1111/j.1471-4159.2009.05918.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A wide range of intracellular proteins have been demonstrated to interact with individual G protein-coupled receptors (GPCRs) and, in certain cases, to modulate their function or trafficking. However, in only a few cases have the GPCR selectivity of such interactions been investigated. Interactions between the intracellular C-terminal tails of 44 GPCRs and both neurochondrin and periplakin were assessed in pull-down studies. 23 of these interacted with neurochondrin and periplakin, 10 interacted with neither whilst nine interacted with only neurochondrin and two with only periplakin. When appropriate GIP-interacting G(q)/G(11)-coupled GPCRs were expressed in cells inducibly expressing neurochondrin or periplakin this resulted in a reduction in the increase in intracellular [Ca(2+)] in response to agonist. However, induction of neurochondrin or periplakin was without functional consequences for GPCRs with which they did not interact. Unlike intracellular [Ca(2+)] signals, induction of expression of either interacting protein did not inhibit agonist-mediated ERK1/2 MAPK phosphorylation. These data indicate that both periplakin and neurochondrin can interact with a wide range of GPCRs and modulate function selectively. Details of the structure of the intracellular C-terminal tail of individual receptors will be required to fully understand the basis of such selectivity.
Collapse
Affiliation(s)
- Richard J Ward
- Molecular Pharmacology Group, Neuroscience and Molecular Pharmacology, Faculty of Biomedical and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
68
|
Aplin M, Bonde MM, Hansen JL. Molecular determinants of angiotensin II type 1 receptor functional selectivity. J Mol Cell Cardiol 2009; 46:15-24. [DOI: 10.1016/j.yjmcc.2008.09.123] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 09/09/2008] [Accepted: 09/18/2008] [Indexed: 01/14/2023]
|
69
|
Brinkerhoff CJ, Traynor JR, Linderman JJ. Collision coupling, crosstalk, and compartmentalization in G-protein coupled receptor systems: can a single model explain disparate results? J Theor Biol 2008; 255:278-86. [PMID: 18761019 PMCID: PMC2917770 DOI: 10.1016/j.jtbi.2008.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 08/01/2008] [Indexed: 02/04/2023]
Abstract
The collision coupling model describes interactions between receptors and G-proteins as first requiring the molecules to find each other by diffusion. A variety of experimental data on G-protein activation have been interpreted as suggesting (or not) the compartmentalization of receptors and/or G-proteins in addition to a collision coupling mechanism. In this work, we use a mathematical model of G-protein activation via collision coupling but without compartmentalization to demonstrate that these disparate observations do not imply the existence of such compartments. In experiments with GTP analogs (commonly GTPgammaS), the extent of G-protein activation is predicted to be a function of both receptor number and the rate of GTP analog hydrolysis. The sensitivity of G-protein activation to receptor number is shown to be dependent upon the assay used, with the sensitivity of phosphate production assays (GTPase) >GTPgammaS-binding assays >cAMP inhibition assays. Finally, the amount of competition or crosstalk between receptor species activating the same type of G-proteins is predicted to depend on receptor and G-protein number, but in some (common) experimental regimes this dependence is expected to be minimal. Taken together, these observations suggest that the collision coupling model, without compartments of receptors and/or G-proteins, is sufficient to explain a variety of observations in literature data.
Collapse
Affiliation(s)
- Christopher J. Brinkerhoff
- Department of Chemical Engineering, H.H. Dow Building, 2300 Hayward St, University of Michigan, Ann Arbor, MI 48109-2136
| | - John R. Traynor
- Department of Pharmacology, University of Michigan Medical School, 1301 MSRB II, 1150 W. Medical Center Drive, University of Michigan, Ann Arbor, MI 48109-0632
| | - Jennifer J. Linderman
- Department of Chemical Engineering, H.H. Dow Building, 2300 Hayward St, University of Michigan, Ann Arbor, MI 48109-2136
| |
Collapse
|
70
|
Schröder R, Merten N, Mathiesen JM, Martini L, Kruljac-Letunic A, Krop F, Blaukat A, Fang Y, Tran E, Ulven T, Drewke C, Whistler J, Pardo L, Gomeza J, Kostenis E. The C-terminal tail of CRTH2 is a key molecular determinant that constrains Galphai and downstream signaling cascade activation. J Biol Chem 2008; 284:1324-36. [PMID: 19010788 DOI: 10.1074/jbc.m806867200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Prostaglandin D(2) activation of the seven-transmembrane receptor CRTH2 regulates numerous cell functions that are important in inflammatory diseases, such as asthma. Despite its disease implication, no studies to date aimed at identifying receptor domains governing signaling and surface expression of human CRTH2. We tested the hypothesis that CRTH2 may take advantage of its C-tail to silence its own signaling and that this mechanism may explain the poor functional responses observed with CRTH2 in heterologous expression systems. Although the C terminus is a critical determinant for retention of CRTH2 at the plasma membrane, the presence of this domain confers a signaling-compromised conformation onto the receptor. Indeed, a mutant receptor lacking the major portion of its C-terminal tail displays paradoxically enhanced Galpha(i) and ERK1/2 activation despite enhanced constitutive and agonist-mediated internalization. Enhanced activation of Galpha(i) proteins and downstream signaling cascades is probably due to the inability of the tail-truncated receptor to recruit beta-arrestin2 and undergo homologous desensitization. Unexpectedly, CRTH2 is not phosphorylated upon agonist-stimulation, a primary mechanism by which GPCR activity is regulated. Dynamic mass redistribution assays, which allow label-free monitoring of all major G protein pathways in real time, confirm that the C terminus inhibits Galpha(i) signaling of CRTH2 but does not encode G protein specificity determinants. We propose that intrinsic CRTH2 inhibition by its C terminus may represent a rather unappreciated strategy employed by a GPCR to specify the extent of G protein activation and that this mechanism may compensate for the absence of the classical phosphorylation-dependent signal attenuation.
Collapse
Affiliation(s)
- Ralf Schröder
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Wacker JL, Feller DB, Tang XB, Defino MC, Namkung Y, Lyssand JS, Mhyre AJ, Tan X, Jensen JB, Hague C. Disease-causing mutation in GPR54 reveals the importance of the second intracellular loop for class A G-protein-coupled receptor function. J Biol Chem 2008; 283:31068-78. [PMID: 18772143 PMCID: PMC2576551 DOI: 10.1074/jbc.m805251200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/26/2008] [Indexed: 11/06/2022] Open
Abstract
The G-protein-coupled receptor (GPCR) GPR54 is essential for the development and maintenance of reproductive function in mammals. A point mutation (L148S) in the second intracellular loop (IL2) of GPR54 causes idiopathic hypogonadotropic hypogonadism, a disorder characterized by delayed puberty and infertility. Here, we characterize the molecular mechanism by which the L148S mutation causes disease and address the role of IL2 in Class A GPCR function. Biochemical, immunocytochemical, and pharmacological analysis demonstrates that the mutation does not affect the expression, ligand binding properties, or protein interaction network of GPR54. In contrast, diverse GPR54 functional responses are markedly inhibited by the L148S mutation. Importantly, the leucine residue at this position is highly conserved among class A GPCRs. Indeed, mutating the corresponding leucine of the alpha(1A)-AR recapitulates the effects observed with L148S GPR54, suggesting the critical importance of this hydrophobic IL2 residue for Class A GPCR functional coupling. Interestingly, co-immunoprecipitation studies indicate that L148S does not hinder the association of Galpha subunits with GPR54. However, fluorescence resonance energy transfer analysis strongly suggests that L148S impairs the ligand-induced catalytic activation of Galpha. Combining our data with a predictive Class A GPCR/Galpha model suggests that IL2 domains contain a conserved hydrophobic motif that, upon agonist stimulation, might stabilize the switch II region of Galpha. Such an interaction could promote opening of switch II of Galpha to facilitate GDP-GTP exchange and coupling to downstream signaling responses. Importantly, mutations that disrupt this key hydrophobic interface can manifest as human disease.
Collapse
Affiliation(s)
- Jennifer L Wacker
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Ullah H, Scappini EL, Moon AF, Williams LV, Armstrong DL, Pedersen LC. Structure of a signal transduction regulator, RACK1, from Arabidopsis thaliana. Protein Sci 2008; 17:1771-80. [PMID: 18715992 DOI: 10.1110/ps.035121.108] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The receptor for activated C-kinase 1 (RACK1) is a highly conserved WD40 repeat scaffold protein found in a wide range of eukaryotic species from Chlamydymonas to plants and humans. In tissues of higher mammals, RACK1 is ubiquitously expressed and has been implicated in diverse signaling pathways involving neuropathology, cellular stress, protein translation, and developmental processes. RACK1 has established itself as a scaffold protein through physical interaction with a myriad of signaling proteins ranging from kinases, phosphatases, ion channels, membrane receptors, G proteins, IP3 receptor, and with widely conserved structural proteins associated with the ribosome. In the plant Arabidopsis thaliana, RACK1A is implicated in diverse developmental and environmental stress pathways. Despite the functional conservation of RACK1-mediated protein-protein interaction-regulated signaling modes, the structural basis of such interactions is largely unknown. Here we present the first crystal structure of a RACK1 protein, RACK1 isoform A from Arabidopsis thaliana, at 2.4 A resolution, as a C-terminal fusion of the maltose binding protein. The structure implicates highly conserved surface residues that could play critical roles in protein-protein interactions and reveals the surface location of proposed post-transcriptionally modified residues. The availability of this structure provides a structural basis for dissecting RACK1-mediated cellular signaling mechanisms in both plants and animals.
Collapse
Affiliation(s)
- Hemayet Ullah
- Department of Biology, Howard University, Washington, DC 20059, USA
| | | | | | | | | | | |
Collapse
|
73
|
Turcotte M, Tang W, Ross EM. Coordinate regulation of G protein signaling via dynamic interactions of receptor and GAP. PLoS Comput Biol 2008; 4:e1000148. [PMID: 18716678 PMCID: PMC2518520 DOI: 10.1371/journal.pcbi.1000148] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/01/2008] [Indexed: 11/28/2022] Open
Abstract
Signal output from receptor–G-protein–effector modules is a dynamic function of the nucleotide exchange activity of the receptor, the GTPase-accelerating activity of GTPase-activating proteins (GAPs), and their interactions. GAPs may inhibit steady-state signaling but may also accelerate deactivation upon removal of stimulus without significantly inhibiting output when the receptor is active. Further, some effectors (e.g., phospholipase C-β) are themselves GAPs, and it is unclear how such effectors can be stimulated by G proteins at the same time as they accelerate G protein deactivation. The multiple combinations of protein–protein associations and interacting regulatory effects that allow such complex behaviors in this system do not permit the usual simplifying assumptions of traditional enzyme kinetics and are uniquely subject to systems-level analysis. We developed a kinetic model for G protein signaling that permits analysis of both interactive and independent G protein binding and regulation by receptor and GAP. We evaluated parameters of the model (all forward and reverse rate constants) by global least-squares fitting to a diverse set of steady-state GTPase measurements in an m1 muscarinic receptor–Gq–phospholipase C-β1 module in which GTPase activities were varied by ∼104-fold. We provide multiple tests to validate the fitted parameter set, which is consistent with results from the few previous pre-steady-state kinetic measurements. Results indicate that (1) GAP potentiates the GDP/GTP exchange activity of the receptor, an activity never before reported; (2) exchange activity of the receptor is biased toward replacement of GDP by GTP; (3) receptor and GAP bind G protein with negative cooperativity when G protein is bound to either GTP or GDP, promoting rapid GAP binding and dissociation; (4) GAP indirectly stabilizes the continuous binding of receptor to G protein during steady-state GTPase hydrolysis, thus further enhancing receptor activity; and (5) receptor accelerates GDP/GTP exchange primarily by opening an otherwise closed nucleotide binding site on the G protein but has minimal effect on affinity (Kassoc = kassoc/kdissoc) of G protein for nucleotide. Model-based simulation explains how GAP activity can accelerate deactivation >10-fold upon removal of agonist but still allow high signal output while the receptor is active. Analysis of GTPase flux through distinct reaction pathways and consequent accumulation of specific GTPase cycle intermediates indicate that, in the presence of a GAP, the receptor remains bound to G protein throughout the GTPase cycle and that GAP binds primarily during the GTP-bound phase. The analysis explains these behaviors and relates them to the specific regulatory phenomena described above. The work also demonstrates the applicability of appropriately data-constrained system-level analysis to signaling networks of this scale. Throughout the eukaryotes, G proteins convey information from receptors for diverse stimuli—neurotransmitters, hormones, light, odors, and pheromones—to intracellular regulatory proteins collectively known as effectors. G proteins function by transiting a dynamic cycle of activation and deactivation. Receptors accelerate activation, which allows G proteins to regulate effectors, and receptors thus increase signal output. GTPase-activating proteins, GAPs, accelerate deactivation. GAPs can thus attenuate signaling, but GAPs can also accelerate signal termination when stimulus is removed without inhibiting signal output while stimulus is present. Surprisingly, some effectors are also GAPs for the G proteins that activate them, essentially turning off their activator. We developed a mathematical model that describes control of G protein signaling by receptor and GAP and used experimental data to determine its important parameters. We show that GAPs actually potentiate G protein activation by receptor, a previously unsuspected effect. Further, GAPs indirectly stabilize receptor–G protein binding during stimulation, which we had previously predicted based on inconsistencies among other experimental results. The present results elucidate how GAPs can independently control signaling kinetics and amplitude and thus clarify how effectors can both respond to G proteins and act as G protein GAPs.
Collapse
Affiliation(s)
- Marc Turcotte
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | |
Collapse
|
74
|
Gesty-Palmer D, Luttrell LM. Heptahelical terpsichory. Who calls the tune? J Recept Signal Transduct Res 2008; 28:39-58. [PMID: 18437629 DOI: 10.1080/10799890801941921] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The discovery that arrestins can function as ligand-regulated signaling scaffolds has revealed a previously unappreciated level of complexity in G protein-coupled receptor (GPCR) signal transduction. Because arrestin-bound GPCRs are uncoupled from G proteins, arrestin binding can be viewed as switching receptors between two temporally and spatially distinct signaling modes. Recent work has established two factors that underscore this duality of GPCR signaling and suggest it may ultimately have therapeutic significance. The first is that signaling by receptor-arrestin "signalsomes" does not require heterotrimeric G protein activation. The second is that arrestin-dependent signals can be initiated by pathway-specific "biased agonists," creating the potential for drugs that selectively modulate different aspects of GPCR function. Currently, however, little is known about the physiological relevance of G protein-independent signals at the cellular or whole animal levels, and additional work is needed to determine whether arrestin pathway-selective drugs will find clinical application.
Collapse
Affiliation(s)
- Diane Gesty-Palmer
- Department of Medicine Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
75
|
Thirunavukkarasu M, Han Z, Zhan L, Penumathsa SV, Menon VP, Maulik N. Adeno-sh-beta-catenin abolishes ischemic preconditioning-mediated cardioprotection by downregulation of its target genes VEGF, Bcl-2, and survivin in ischemic rat myocardium. Antioxid Redox Signal 2008; 10:1475-84. [PMID: 18407748 PMCID: PMC2932533 DOI: 10.1089/ars.2008.2042] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
beta-Catenin, the downstream target of glycogen synthase kinase-3beta (GSK-3beta), plays a vital role in ischemic preconditioning (IP)-mediated cardioprotection. In the present study, we investigated the mechanism of IP-mediated cardioprotection through suppression of beta-catenin expression by intramyocardial injection of adeno-sh-RNA against beta-catenin (BCT) (4 x 10(8) pfu). Adeno-LacZ (LZ) was used as control. The rats were randomized into (a) LZ + ischemia-reperfusion (IR); (b) LZIPIR; (c) BCTIR; and (d) BCTIPIR. Isolated hearts from each group were subjected to 30 min of I followed by 2 h of R. Both IPIR group hearts were subjected to IP (5 min I + 10 min R; four cycles) before IR. Significant reduction in left ventricular functional recovery (78 vs. 88 mm Hg), dp/dt(max) (1,802 vs. 2,189 mm Hg/sec), and aortic flow (4 vs. 9 ml/min) was observed in BCTIPIR compared with LZIPIR at 120 min of reperfusion. Increased infarct size (42 vs. 24%) and apoptotic cardiomyocytes (122 vs. 58 counts/60 HPF) were observed in BCTIPIR compared with LZIPIR. Realtime PCR and Western blot analysis showed significant downregulation in mRNA and protein expression of VEGF, Bcl-2, and survivin in BCTIPIR compared with LZIPIR. These findings indicated for the first time that silencing beta-catenin abolished IP-mediated cardioprotection, probably through inhibition of VEGF-Bcl-2 and survivin.
Collapse
Affiliation(s)
- Mahesh Thirunavukkarasu
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery, University of Connecticut Medical Center, Farmington, Connecticut 06030-1110, USA
| | | | | | | | | | | |
Collapse
|
76
|
Detection of heteromerization of more than two proteins by sequential BRET-FRET. Nat Methods 2008; 5:727-33. [DOI: 10.1038/nmeth.1229] [Citation(s) in RCA: 226] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Accepted: 06/05/2008] [Indexed: 11/08/2022]
|
77
|
Amber-Cicek F, Ugur O, Sayar K, Ugur M. Cell adhesion modulates 5-HT(1D) and P2Y receptor signal trafficking differentially in LTK-8 cells. Eur J Pharmacol 2008; 590:12-9. [PMID: 18582865 DOI: 10.1016/j.ejphar.2008.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 04/15/2008] [Accepted: 05/13/2008] [Indexed: 12/31/2022]
Abstract
In this study, we investigated adhesion-induced changes in cellular responses to serotonin 5-HT(1D) and purinergic P2Y receptor stimulation. We demonstrated that detachment of LTK-8 cells increased 5-HT(1D) receptor-mediated intracellular Ca(2+) and extracellular signal regulated kinase (ERK) phosphorylation responses without affecting the adenylate cyclase response. Additionally, detachment enabled 5-HT(1D) receptor stimulation to inhibit P2Y receptor-induced [Ca(2+)](i) mobilization. Such a cross talk between the two receptor systems was not observed in attached cells. P2Y receptor-induced Ca(2+) response was insensitive to adhesion state of the cells, while ERK phosphorylation response was enhanced upon detachment. Integrity of the actin cytoskeleton did not appear to play a role in adhesion sensitivity of 5-HT(1D)-mediated responses, as treatment of attached cells with cytochalasin D did not mimic detachment-induced effects. Effects of detachment were reversed immediately after re-attachment of the suspended cells on poly-l-lysine coated cover slips, suggesting that the involvement of integrins or focal adhesion complexes is unlikely. Taken collectively, our results demonstrate that not only cellular responses induced by different G protein-coupled receptors, but also different responses induced by a particular G protein-coupled receptor, can be affected differentially by the adhesion status of cells. This suggests an important role for cell adhesion in controlling the coupling of a single G protein-coupled receptor to different intracellular responses.
Collapse
Affiliation(s)
- Figen Amber-Cicek
- Ankara University, Faculty of Medicine, Department of Biophysics, Ankara, Turkey
| | | | | | | |
Collapse
|
78
|
Lyssand JS, DeFino MC, Tang XB, Hertz AL, Feller DB, Wacker JL, Adams ME, Hague C. Blood pressure is regulated by an alpha1D-adrenergic receptor/dystrophin signalosome. J Biol Chem 2008; 283:18792-800. [PMID: 18468998 DOI: 10.1074/jbc.m801860200] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypertension is a cardiovascular disease associated with increased plasma catecholamines, overactivation of the sympathetic nervous system, and increased vascular tone and total peripheral resistance. A key regulator of sympathetic nervous system function is the alpha(1D)-adrenergic receptor (AR), which belongs to the adrenergic family of G-protein-coupled receptors (GPCRs). Endogenous catecholamines norepinephrine and epinephrine activate alpha(1D)-ARs on vascular smooth muscle to stimulate vasoconstriction, which increases total peripheral resistance and mean arterial pressure. Indeed, alpha(1D)-AR KO mice display a hypotensive phenotype and are resistant to salt-induced hypertension. Unfortunately, little information exists about how this important GPCR functions because of an inability to obtain functional expression in vitro. Here, we identified the dystrophin proteins, syntrophin, dystrobrevin, and utrophin as essential GPCR-interacting proteins for alpha(1D)-ARs. We found that dystrophins complex with alpha(1D)-AR both in vitro and in vivo to ensure proper functional expression. More importantly, we demonstrate that knock-out of multiple syntrophin isoforms results in the complete loss of alpha(1D)-AR function in mouse aortic smooth muscle cells and abrogation of alpha(1D)-AR-mediated increases in blood pressure. Our findings demonstrate that syntrophin and utrophin associate with alpha(1D)-ARs to create a functional signalosome, which is essential for alpha(1D)-AR regulation of vascular tone and blood pressure.
Collapse
Affiliation(s)
- John S Lyssand
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Hendriks-Balk MC, Peters SLM, Michel MC, Alewijnse AE. Regulation of G protein-coupled receptor signalling: focus on the cardiovascular system and regulator of G protein signalling proteins. Eur J Pharmacol 2008; 585:278-91. [PMID: 18410914 DOI: 10.1016/j.ejphar.2008.02.088] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 01/18/2008] [Accepted: 02/06/2008] [Indexed: 11/17/2022]
Abstract
G protein-coupled receptors (GPCRs) are involved in many biological processes. Therefore, GPCR function is tightly controlled both at receptor level and at the level of signalling components. Well-known mechanisms by which GPCR function can be regulated comprise desensitization/resensitization processes and GPCR up- and downregulation. GPCR function can also be regulated by several proteins that directly interact with the receptor and thereby modulate receptor activity. An additional mechanism by which receptor signalling is regulated involves an emerging class of proteins, the so-called regulators of G protein signalling (RGS). In this review we will describe some of these control mechanisms in more detail with some specific examples in the cardiovascular system. In addition, we will provide an overview on RGS proteins and the involvement of RGS proteins in cardiovascular function.
Collapse
Affiliation(s)
- Mariëlle C Hendriks-Balk
- Department Pharmacology and Pharmacotherapy, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
80
|
ER B, VV G, JN J, JL B, EV G. Arrestins and two receptor kinases are upregulated in Parkinson's disease with dementia. Neurobiol Aging 2008; 29:379-396. [PMID: 17125886 PMCID: PMC2275668 DOI: 10.1016/j.neurobiolaging.2006.10.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2006] [Revised: 09/04/2006] [Accepted: 10/11/2006] [Indexed: 10/23/2022]
Abstract
Arrestins and G proteins-coupled receptor kinases (GRKs) regulate signaling and trafficking of G protein-coupled receptors. We investigated changes in the expression of arrestins and GRKs in the striatum of patients with Parkinson's disease without (PD) or with dementia (PDD) at postmortem using Western blotting and ribonuclease protection assay. Both PD and PDD groups had similar degree of dopamine depletion in all striatal regions. Arrestin proteins and mRNAs were increased in the PDD group throughout striatum. Protein and mRNA of GRK5, the major subtype in the human striatum, and GRK3 were also upregulated, whereas GRK2 and 6 were mostly unchanged. The PD group had lower concentration of arrestins and GRKs than the PDD group. There was no statistical link between the load of Alzheimer's pathology and the expression of these signaling proteins. Upregulation of arrestins and GRK in PDD may confer resistance to the therapeutic effects of levodopa often observed in these patients. In addition, increased arrestin and GRK concentrations may lead to dementia via perturbation of multiple signaling mechanisms.
Collapse
Affiliation(s)
- Bychkov ER
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Laboratory of Neuroimmunology, Institute of Human Brain, St. Petersburg 197376, Russia
| | - Gurevich VV
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Joyce JN
- Parkinson's Disease Research Center, Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Benovic JL
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Gurevich EV
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
81
|
Extracellular calcium-sensing receptors in fishes. Comp Biochem Physiol A Mol Integr Physiol 2008; 149:225-45. [DOI: 10.1016/j.cbpa.2008.01.037] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 01/23/2008] [Accepted: 01/23/2008] [Indexed: 11/19/2022]
|
82
|
The multifunctional protein GC1q-R interacts specifically with the i3 loop arginine cluster of the vasopressin V2 receptor. ACTA ACUST UNITED AC 2008; 148:76-87. [PMID: 18358546 DOI: 10.1016/j.regpep.2008.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 01/16/2008] [Accepted: 01/17/2008] [Indexed: 11/22/2022]
Abstract
In this study, we identified the multifunctional protein GC1q-R as a novel vasopressin V(2) receptor (V(2)R) interacting protein. For this purpose, we have developed a proteomic approach combining pull-down assays using a cyclic peptide mimicking the third intracellular loop of V(2)R as a bait and mass spectrometry analyses of proteins isolated from either rat or human kidney tissues or the HEK 293 cell line. Co-immunoprecipitation of GC1q-R with the c-Myc-tagged h-V(2)R expressed in a HEK cell line confirmed the existence of a specific interaction between GC1q-R and the V(2) receptor. Then, construction of a mutant receptor in i3 loop allowed us to identify the i3 loop arginine cluster of the vasopressin V(2) receptor as the interacting determinant for GC1q-R interaction. Using purified receptor as a bait and recombinant (74-282) GC1q-R, we demonstrated a direct and specific interaction between these two proteins via the arginine cluster.
Collapse
|
83
|
Thompson MD, Cole DEC, Jose PA. Pharmacogenomics of G protein-coupled receptor signaling: insights from health and disease. Methods Mol Biol 2008; 448:77-107. [PMID: 18370232 DOI: 10.1007/978-1-59745-205-2_6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The identification and characterization of the processes of G protein-coupled receptor (GPCR) activation and inactivation have refined not only the study of the GPCRs but also the genomics of many accessory proteins necessary for these processes. This has accelerated progress in understanding the fundamental mechanisms involved in GPCR structure and function, including receptor transport to the membrane, ligand binding, activation and inactivation by GRK-mediated (and other) phosphorylation. The catalog of G(s)alpha and Gbeta subunit polymorphisms that result in complex phenotypes has complemented the effort to catalog the GPCRs and their variants. The study of the genomics of GPCR accessory proteins has also provided insight into pathways of disease, such as the contributions of regulator of G protein signaling (RGS) protein to hypertension and activator of G protein signaling (AGS) proteins to the response to hypoxia. In the case of the G protein-coupled receptor kinases (GRKs), identified originally in the retinal tissues that converge on rhodopsin, proteins such as GRK4 have been identified that have been subsequently associated with hypertension. Here, we review the structure and function of GPCR and associated proteins in the context of the gene families that encode them and the genetic disorders associated with their altered function. An understanding of the pharmacogenomics of GPCR signaling provides the basis for examining the GPCRs disrupted in monogenic disease and the pharmacogenetics of a given receptor system.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Laboratory Medicine and Pathobiology, Banting Institute, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
84
|
Andreeva AV, Kutuzov MA, Voyno-Yasenetskaya TA. Scaffolding proteins in G-protein signaling. J Mol Signal 2007; 2:13. [PMID: 17971232 PMCID: PMC2211295 DOI: 10.1186/1750-2187-2-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Accepted: 10/30/2007] [Indexed: 11/18/2022] Open
Abstract
Heterotrimeric G proteins are ubiquitous signaling partners of seven transmembrane-domain G-protein-coupled receptors (GPCRs), the largest (and most important pharmacologically) receptor family in mammals. A number of scaffolding proteins have been identified that regulate various facets of GPCR signaling. In this review, we summarize current knowledge concerning those scaffolding proteins that are known to directly bind heterotrimeric G proteins, and discuss the composition of the protein complexes they assemble and their effects on signal transduction. Emerging evidence about possible ways of regulation of activity of these scaffolding proteins is also discussed.
Collapse
Affiliation(s)
- Alexandra V Andreeva
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, 909 S, Wolcott Ave, Chicago, Illinois 60612, USA.
| | | | | |
Collapse
|
85
|
Jackson SW, Hoshi T, Wu Y, Sun X, Enjyoji K, Cszimadia E, Sundberg C, Robson SC. Disordered purinergic signaling inhibits pathological angiogenesis in cd39/Entpd1-null mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1395-404. [PMID: 17823293 PMCID: PMC1988887 DOI: 10.2353/ajpath.2007.070190] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CD39/ecto-nucleoside triphosphate diphosphohydrolase-type-1 (ENTPD1) is the dominant vascular ecto-nucleotidase that catalyzes the phosphohydrolysis of extracellular nucleotides in the blood and extracellular space. This ecto-enzymatic process modulates endothelial cell, leukocyte, and platelet purinergic receptor-mediated responses to extracellular nucleotides in the setting of thrombosis and vascular inflammation. We show here that deletion of Cd39/Entpd1 results in abrogation of angiogenesis, causing decreased growth of implanted tumors and inhibiting development of pulmonary metastases. Qualitative abnormalities of Cd39-null endothelial cell adhesion and integrin dysfunction were demonstrated in vitro. These changes were associated with decreased activation of focal adhesion kinase and extracellular signaling-regulated kinase-1 and -2 in endothelial cells. Our data indicate novel links between CD39/ENTPD1, extracellular nucleotide-mediated signaling, and vascular endothelial cell integrin function that impact on angiogenesis and tumor growth.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Apyrase/genetics
- Apyrase/metabolism
- Cell Adhesion
- Cell Proliferation
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Deletion
- Integrin alphaVbeta3/metabolism
- Integrins/metabolism
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/pathology
- Mice
- Mice, Mutant Strains
- Neoplasm Transplantation
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Protein Kinases/metabolism
- Purines/metabolism
- Receptors, Purinergic P2/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Shaun W Jackson
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Ogawa M, Miyakawa T, Nakamura K, Kitano J, Furushima K, Kiyonari H, Nakayama R, Nakao K, Moriyoshi K, Nakanishi S. Altered sensitivities to morphine and cocaine in scaffold protein tamalin knockout mice. Proc Natl Acad Sci U S A 2007; 104:14789-94. [PMID: 17766434 PMCID: PMC1976212 DOI: 10.1073/pnas.0706945104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tamalin is a scaffold protein that interacts with metabotropic glutamate receptors and the kinase-deficient neurotrophin TrkCT1 receptor and forms a protein complex with multiple protein-trafficking and intracellular signaling molecules. In culture, tamalin promotes intracellular trafficking of group 1 metabotropic glutamate receptors through its interaction with guanine nucleotide exchange factor cytohesins and causes actin reorganization and membrane ruffling via the TrkCT1/cytohesin-2 signaling mechanism. However, how tamalin serves its physiological function in vivo has remained elusive. In this study, we generated tamalin knockout (Tam(-/-) KO) mice and investigated behavioral alterations resulting from their deficiency in functional tamalin. Targeted deletion of functional tamalin altered neither the overall brain architecture nor the general behavior of the mice under ordinary conditions. However, Tam(-/-) KO mice showed a decrease in sensitivity to acute morphine-induced hyperlocomotion and morphine analgesic effects in the hot-plate test. Furthermore, tamalin deficiency impaired the ability of the animals to show conditioned place preference after repeated morphine administration and to display locomotor sensitization by chronic cocaine treatment. Upon in vivo microdialysis analysis of the nucleus accumbens, Tam(-/-) KO and wild-type mice showed no genotypic differences in their response patterns of extracellular dopamine and glutamate before or after morphine administration. These results demonstrate that the tamalin scaffold protein plays a unique role in both acute and adaptive behavioral responses to morphine and cocaine and could regulate common neural substrates implicated in drugs of abuse.
Collapse
Affiliation(s)
- Masaaki Ogawa
- *Department of Systems Biology, Osaka Bioscience Institute, Suita, Osaka 565-0874, Japan
- Department of Biological Sciences and
| | | | - Kenji Nakamura
- Mouse Genome Technology Laboratory, Mitsubishi Kagaku Institute of Life Sciences, Machida, Tokyo 194-8511, Japan; and
| | | | - Kenryo Furushima
- **Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Hyogo 650-0046, Japan
| | - Hiroshi Kiyonari
- **Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Hyogo 650-0046, Japan
| | - Rika Nakayama
- **Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Hyogo 650-0046, Japan
| | - Kazuki Nakao
- **Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Chuo-ku, Kobe, Hyogo 650-0046, Japan
| | - Koki Moriyoshi
- Department of Molecular and System Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shigetada Nakanishi
- *Department of Systems Biology, Osaka Bioscience Institute, Suita, Osaka 565-0874, Japan
- Department of Biological Sciences and
- Department of Molecular and System Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
87
|
Abstract
beta(2)-adrenergic receptors are present throughout the lung, including the alveolar airspace, where they play an important role for regulation of the active Na(+) transport needed for clearance of excess fluid out of alveolar airspace. beta(2)-adrenergic receptor signaling is required for up-regulation of alveolar epithelial active ion transport in the setting of excess alveolar edema. The positive, protective effects of beta(2)-adrenergic receptor signaling on alveolar active Na(+) transport in normal and injured lungs provide substantial support for the use of beta-adrenergic agonists to accelerate alveolar fluid clearance in patients with cardiogenic and noncardiogenic pulmonary edema. In this review, we summarize the role of beta(2)-adrenergic receptors in the alveolar epithelium with emphasis on their role in the regulation of alveolar active Na(+) transport in normal and injured lungs.
Collapse
Affiliation(s)
- Gökhan M Mutlu
- Northwestern University Feinberg School of Medicine, Pulmonary and Critical Care Medicine, 240 E. Huron Street, McGaw M-300, Chicago, IL 60611, USA.
| | | |
Collapse
|
88
|
Minezaki Y, Homma K, Nishikawa K. Intrinsically Disordered Regions of Human Plasma Membrane Proteins Preferentially Occur in the Cytoplasmic Segment. J Mol Biol 2007; 368:902-13. [PMID: 17368479 DOI: 10.1016/j.jmb.2007.02.033] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 01/29/2007] [Accepted: 02/09/2007] [Indexed: 12/30/2022]
Abstract
A systematic survey of intrinsically disordered (ID) regions was carried out in 2109 human plasma membrane proteins with full assignment of the transmembrane topology with respect to the lipid bilayer. ID regions with 30 consecutive residues or more were detected in 41.0% of the human proteins, a much higher percentage than the corresponding figure (4.7%) for inner membrane proteins of Escherichia coli. The domain organization of each of the membrane protein in terms of transmembrane helices, structural domains, ID, and unassigned regions as well as the distinction of inside or outside of the cell was determined. Long ID regions constitute 13.3 and 3.5% of the human plasma membrane proteins on the inside and outside of the cell, respectively, showing that they preferentially occur on the cytoplasmic side. We interpret this phenomenon as a reflection of the general scarcity of ID regions on the extracellular side and their relative abundance on the cytoplasmic side in multicellular eukaryotic organisms.
Collapse
Affiliation(s)
- Yoshiaki Minezaki
- Laboratory of Gene-Product Informatics, Center for Information Biology and DNA Data Bank of Japan, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 311-8540, Japan
| | | | | |
Collapse
|
89
|
Navratilova E, Waite S, Stropova D, Eaton MC, Alves ID, Hruby VJ, Roeske WR, Yamamura HI, Varga EV. Quantitative evaluation of human delta opioid receptor desensitization using the operational model of drug action. Mol Pharmacol 2007; 71:1416-26. [PMID: 17322005 DOI: 10.1124/mol.106.030023] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Agonist-mediated desensitization of the opioid receptors is thought to function as a protective mechanism against sustained opioid signaling and therefore may prevent the development of opioid tolerance. However, the exact molecular mechanism of opioid receptor desensitization remains unresolved because of difficulties in measuring and interpreting receptor desensitization. In the present study, we investigated deltorphin II-mediated rapid desensitization of the human delta opioid receptors (hDOR) by measuring guanosine 5'-O-(3-[(35)S]thio)-triphosphate binding and inhibition of cAMP accumulation. We developed a mathematical analysis based on the operational model of agonist action (Black et al., 1985) to calculate the proportion of desensitized receptors. This approach permits a correct analysis of the complex process of functional desensitization by taking into account receptor-effector coupling and the time dependence of agonist pretreatment. Finally, we compared hDOR desensitization with receptor phosphorylation at Ser363, the translocation of beta-arrestin2, and hDOR internalization. We found that in Chinese hamster ovary cells expressing the hDOR, deltorphin II treatment leads to phosphorylation of Ser363, translocation of beta-arrestin2 to the plasma membrane, receptor internalization, and uncoupling from G proteins. It is noteworthy that mutation of the primary phosphorylation site Ser363 to alanine had virtually no effect on agonist-induced beta-arrestin2 translocation and receptor internalization yet significantly attenuated receptor desensitization. These results strongly indicate that phosphorylation of Ser363 is the primary mechanism of hDOR desensitization.
Collapse
Affiliation(s)
- Edita Navratilova
- Department of Medical Pharmacology, The University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Sugi T, Oyama T, Muto T, Nakanishi S, Morikawa K, Jingami H. Crystal structures of autoinhibitory PDZ domain of Tamalin: implications for metabotropic glutamate receptor trafficking regulation. EMBO J 2007; 26:2192-205. [PMID: 17396155 PMCID: PMC1852777 DOI: 10.1038/sj.emboj.7601651] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 02/23/2007] [Indexed: 11/09/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) function as neuronal G-protein-coupled receptors and this requires efficient membrane targeting through associations with cytoplasmic proteins. However, the molecular mechanism regulating mGluR cell-surface trafficking remains unknown. We report here that mGluR trafficking is controlled by the autoregulatory assembly of a scaffold protein Tamalin. In the absence of mGluR, Tamalin self-assembles into autoinhibited conformations, through its PDZ domain and C-terminal intrinsic ligand motif. X-ray crystallographic analyses visualized integral parts of the oligomeric self-assemblies of Tamalin, which require not only the novel hydrophobic dimerization interface but also canonical and noncanonical PDZ/ligand autoinhibitory interactions. The mGluR cytoplasmic region can competitively bind to Tamalin at a higher concentration, disrupting weak inhibitory interactions. The atomic view of mGluR association suggests that this rearrangement is dominated by electrostatic attraction and repulsion. We also observed in mammalian cells that the association liberates the intrinsic ligand toward a motor protein receptor, thereby facilitating mGluR cell-surface trafficking. Our study suggests a novel regulatory mechanism of the PDZ domain, by which Tamalin switches between the trafficking-inhibited and -active forms depending on mGluR association.
Collapse
Affiliation(s)
- Takuma Sugi
- Department of Molecular Biology, Biomolecular Engineering Research Institute (BERI), Suita, Osaka, Japan
- Present address: Institute for Protein Research, Open Laboratories of Advanced Bioscience and Biotechnology (OLABB), Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan
| | - Takuji Oyama
- Department of Structural Biology, Biomolecular Engineering Research Institute (BERI), Suita, Osaka, Japan
- Present address: Institute for Protein Research, Open Laboratories of Advanced Bioscience and Biotechnology (OLABB), Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan
| | - Takanori Muto
- Department of Molecular Biology, Biomolecular Engineering Research Institute (BERI), Suita, Osaka, Japan
- Present address: Fuji Gotemba Research Laboratories, Chugai Pharmaceutical Co., Ltd, 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | | | - Kosuke Morikawa
- Department of Structural Biology, Biomolecular Engineering Research Institute (BERI), Suita, Osaka, Japan
- Present address: Institute for Protein Research, Open Laboratories of Advanced Bioscience and Biotechnology (OLABB), Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan
- Institute for Protein Research, Open Laboratories of Advanced Bioscience and Biotechnology (OLABB), Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan. Tel.: +81 6 6872 8201; Fax: +81 6 6872 19. E-mail:
| | - Hisato Jingami
- Department of Molecular Biology, Biomolecular Engineering Research Institute (BERI), Suita, Osaka, Japan
- Present address: Office of Graduate Courses for Integrated Research Training, Kyoto University Faculty of Medicine, Yoshida, Sakyo-Ku, Kyoto 606-8501, Japan
- Office of Graduate Courses for Integrated Research Training, Kyoto University, Yoshida-Konoe, Sakyo-Ku, Kyoto 606-8501, Japan. Tel.: +81 75 753 9493; Fax: +81 75 753 9495. E-mail:
| |
Collapse
|
91
|
Brum PC, Rolim NPL, Bacurau AVN, Medeiros A. Neurohumoral activation in heart failure: the role of adrenergic receptors. AN ACAD BRAS CIENC 2007; 78:485-503. [PMID: 16936938 DOI: 10.1590/s0001-37652006000300009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2005] [Accepted: 11/04/2005] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is a common endpoint for many forms of cardiovascular disease and a significant cause of morbidity and mortality. The development of end-stage HF often involves an initial insult to the myocardium that reduces cardiac output and leads to a compensatory increase in sympathetic nervous system activity. Acutely, the sympathetic hyperactivity through the activation of beta-adrenergic receptors increases heart rate and cardiac contractility, which compensate for decreased cardiac output. However, chronic exposure of the heart to elevated levels of catecholamines released from sympathetic nerve terminals and the adrenal gland may lead to further pathologic changes in the heart, resulting in continued elevation of sympathetic tone and a progressive deterioration in cardiac function. On a molecular level, altered beta-adrenergic receptor signaling plays a pivotal role in the genesis and progression of HF. beta-adrenergic receptor number and function are decreased, and downstream mechanisms are altered. In this review we will present an overview of the normal beta-adrenergic receptor pathway in the heart and the consequences of sustained adrenergic activation in HF. The myopathic potential of individual components of the adrenergic signaling will be discussed through the results of research performed in genetic modified animals. Finally, we will discuss the potential clinical impact of beta-adrenergic receptor gene polymorphisms for better understanding the progression of HF.
Collapse
MESH Headings
- Animals
- Cardiac Output, Low/physiopathology
- Disease Models, Animal
- Disease Progression
- Humans
- Mice
- Polymorphism, Genetic
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/physiology
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/physiology
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Patricia C Brum
- Laboratório de Fisiologia Cel. e Mol. do Exercício, Departamento de Biodinâmica do Movimento Humano, Escola de Educação Física e Esporte, Universidade de São Paulo, São Paulo, SP, Brasil.
| | | | | | | |
Collapse
|
92
|
Baker JG, Hill SJ. A comparison of the antagonist affinities for the Gi- and Gs-coupled states of the human adenosine A1-receptor. J Pharmacol Exp Ther 2007; 320:218-28. [PMID: 17018691 DOI: 10.1124/jpet.106.113589] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The antagonist affinity for a given receptor is traditionally considered to be constant, reflecting the chemical nature of the specific ligand-receptor interaction. However, recent observations with all three beta-adrenoceptors have cast doubt on this basic pharmacological principle. The extent to which this finding applies to other G protein-coupled receptors and their interaction with different G proteins is unknown. Therefore, we studied the influence of different agonists on antagonist affinity measurements for Gi- and Gs-coupled conformations of the adenosine A1-receptor in Chinese hamster ovary cells stably expressing the human adenosine A1-receptor and a cAMP-response element (CRE)-secreted placental alkaline phosphatase reporter gene. Gi-coupled inhibition of [3H]cAMP accumulation via the A1-receptor was observed at low concentrations of agonist; however, a small increase in [3H]cAMP accumulation was also seen at higher agonist concentrations. This biphasic response was more evident for A1-stimulated CRE-gene transcription. The inhibitory component was abolished by pretreatment with pertussis toxin, whereas the stimulatory component was augmented, suggesting that the responses were due to an A1-Gi-coupled inhibition followed by an A1-Gs-coupled stimulation. However, the antagonist affinity values measured at the Gi-coupled and Gs-coupled conformations of the receptor were the same in both functional responses and whole-cell binding. Thus, in marked contrast to the beta-adrenoceptors, the A1-receptor conforms to the long-held principle of pharmacology that antagonist affinity measurements are constant regardless of the response being measured and the competing agonist used to stimulate that response. This was true even when the receptor was shown, in the same assay, to exist in two different conformational states coupled to two different G proteins.
Collapse
Affiliation(s)
- Jillian G Baker
- Institute of Cell Signaling, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | | |
Collapse
|
93
|
Self TJ, Oakley SM, Hill SJ. Clathrin-independent internalization of the human histamine H1-receptor in CHO-K1 cells. Br J Pharmacol 2006; 146:612-24. [PMID: 16086035 PMCID: PMC1751180 DOI: 10.1038/sj.bjp.0706337] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The aim of the present study was to investigate the cellular pathway involved in histamine-stimulated internalization of the human H1-receptor in CHO-K1 cells expressing N-terminal myc-tagged H1-receptor (Myc-H1) or N-terminal myc-C-terminal green fluorescent protein (Myc-GFP H1) versions of the receptor. Studies of 3H-mepyramine binding and histamine-stimulated 3H-inositol phosphate accumulation in these cells showed that the Myc-H1 and Myc-GFP H1-receptors had identical pharmacology to the wild-type H1-receptor. The Myc-H1-receptor was rapidly internalized in CHO-K1 cells following stimulation with histamine (0.1 mM). This response occurred within 15 min, and could be prevented by the quaternary H1-receptor antagonist alpha-pirdonium. Similar data were obtained with the Myc-GFP H1-receptors. Internalization of the Myc-GFP H1-receptor was maintained in the absence of extracellular calcium and was not inhibited by the CAM kinase II inhibitor KN-62 (10 microM). Phorbol dibutyrate, an activator of protein kinase C, was also able to stimulate internalization of the H1-receptor. However, inhibition or downregulation of protein kinase C (which significantly modified histamine-stimulated inositol phosphate responses) was without effect on the internalization of the H1-receptor stimulated by histamine. Hypertonic sucrose did not prevent histamine-induced internalization of the Myc-GFP H1-receptor, but was able to attenuate internalization of transferrin via clathrin-mediated endocytosis in the same cells. In contrast, preincubation of cells with filipin or nystatin, which disrupts caveolae and lipid rafts, completely inhibited the histamine-induced internalization of the Myc-GFP H1-receptor, but was without effect on the sequestration of transferrin. The H1-receptor and cholera toxin subunit B were colocalized under resting conditions at the cell surface. Immunohistochemical studies with an antibody to caveolin-1 confirmed that this protein was also localized predominantly to the plasma membrane. However, following stimulation of CHO-Myc-GFP H1 cells with histamine, there was no evidence for internalization of caveolin-1 in parallel with the H1-receptor. These data provide strong evidence that the H1-receptor is internalized via a clathrin-independent mechanism and most likely involves lipid rafts.
Collapse
Affiliation(s)
- Timothy J Self
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
| | - Sarah M Oakley
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
| | - Stephen J Hill
- Institute of Cell Signalling, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH
- Author for correspondence:
| |
Collapse
|
94
|
Ni Y, Zhao X, Bao G, Zou L, Teng L, Wang Z, Song M, Xiong J, Bai Y, Pei G. Activation of beta2-adrenergic receptor stimulates gamma-secretase activity and accelerates amyloid plaque formation. Nat Med 2006; 12:1390-6. [PMID: 17115048 DOI: 10.1038/nm1485] [Citation(s) in RCA: 174] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Accepted: 09/22/2006] [Indexed: 01/03/2023]
Abstract
Amyloid plaque is the hallmark and primary cause of Alzheimer disease. Mutations of presenilin-1, the gamma-secretase catalytic subunit, can affect amyloid-beta (Abeta) production and Alzheimer disease pathogenesis. However, it is largely unknown whether and how gamma-secretase activity and amyloid plaque formation are regulated by environmental factors such as stress, which is mediated by receptors including beta(2)-adrenergic receptor (beta(2)-AR). Here we report that activation of beta(2)-AR enhanced gamma-secretase activity and thus Abeta production. This enhancement involved the association of beta(2)-AR with presenilin-1 and required agonist-induced endocytosis of beta(2)-AR and subsequent trafficking of gamma-secretase to late endosomes and lysosomes, where Abeta production was elevated. Similar effects were observed after activation of delta-opioid receptor. Furthermore, chronic treatment with beta(2)-AR agonists increased cerebral amyloid plaques in an Alzheimer disease mouse model. Thus, beta(2)-AR activation can stimulate gamma-secretase activity and amyloid plaque formation, which suggests that abnormal activation of beta(2)-AR might contribute to Abeta accumulation in Alzheimer disease pathogenesis.
Collapse
Affiliation(s)
- Yanxiang Ni
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Caunt CJ, Finch AR, Sedgley KR, McArdle CA. Seven-transmembrane receptor signalling and ERK compartmentalization. Trends Endocrinol Metab 2006; 17:276-83. [PMID: 16890451 DOI: 10.1016/j.tem.2006.07.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Revised: 06/30/2006] [Accepted: 07/17/2006] [Indexed: 12/01/2022]
Abstract
Vast numbers of extracellular signalling molecules exert effects on their target cells by activation of a relatively limited number of mitogen-activated protein kinase (MAPK) cascades, raising the question of how specificity is achieved. To a large extent, this appears to be attributable to differences in kinetics and compartmentalization of MAPK protein activation that are dictated by MAPK-associated proteins serving as scaffolds, anchors, activators or effectors. Here, we review spatiotemporal aspects of signalling via the Ras-Raf-extracellular signal-regulated kinase pathway, emphasizing recent work on roles of arrestins as scaffolds and transducers for seven transmembrane receptor signalling.
Collapse
Affiliation(s)
- Christopher J Caunt
- University of Bristol, Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Whitson Street, Bristol, BS1 3NY, UK
| | | | | | | |
Collapse
|
96
|
Xiao H, Hapiak VM, Smith KA, Lin L, Hobson RJ, Plenefisch J, Komuniecki R. SER-1, a Caenorhabditis elegans 5-HT2-like receptor, and a multi-PDZ domain containing protein (MPZ-1) interact in vulval muscle to facilitate serotonin-stimulated egg-laying. Dev Biol 2006; 298:379-91. [PMID: 16890216 DOI: 10.1016/j.ydbio.2006.06.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Revised: 06/22/2006] [Accepted: 06/26/2006] [Indexed: 02/07/2023]
Abstract
Serotonin (5-HT) stimulation of egg-laying in Caenorhabditis elegans is abolished in ser-1 (ok345) animals and is rescued by ser-1 expression in vulval muscle. A PDZ binding motif (ETFL) at the SER-1 C-terminus is not essential for rescue, but facilitates SER-1 signaling. SER-1 binds specifically to PDZ domain 10 of the multi-PDZ domain protein, MPZ-1, based on GST pulldown and co-immunoprecipitation. mpz-1 is expressed in about 60 neurons and body wall and vulval muscles. In neurons, GFP-tagged MPZ-1 is punctate and colocalizes with the synaptic marker, synaptobrevin. The expression patterns of ser-1 and mpz-1 overlap in 3 pairs of neurons and vulval muscle. In addition, MPZ-1 also interacts with other GPCRs with acidic amino acids in the -3 position of their PDZ binding motifs. mpz-1 RNAi reduces 5-HT stimulated egg-laying in wild type animals and in ser-1 mutants rescued by muscle expression of SER-1. In contrast, mpz-1 RNAi has no effect on 5-HT stimulated egg-laying in ser-1 mutants rescued by expression of a truncated SER-1 that lacks the C-terminal PDZ binding motif. The overexpression of MPZ-1 PDZ domain 10 also inhibits 5-HT stimulated egg-laying. These studies suggest that the SER-1/MPZ-1 interaction facilitates SER-1 mediated signaling.
Collapse
Affiliation(s)
- Hong Xiao
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA
| | | | | | | | | | | | | |
Collapse
|
97
|
Schaub MC, Hefti MA, Zaugg M. Integration of calcium with the signaling network in cardiac myocytes. J Mol Cell Cardiol 2006; 41:183-214. [PMID: 16765984 DOI: 10.1016/j.yjmcc.2006.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 03/07/2006] [Accepted: 04/04/2006] [Indexed: 12/23/2022]
Abstract
Calcium has evolved as global intracellular messenger for signal transduction in the millisecond time range by reversibly binding to calcium-sensing proteins. In the cardiomyocyte, ion pumps, ion exchangers and channels keep the cytoplasmic calcium level at rest around approximately 100 nM which is more than 10,000-fold lower than outside the cell. Intracellularly, calcium is mainly stored in the sarcoplasmic reticulum, which comprises the bulk of calcium available for the heartbeat. Regulation of cardiac function including contractility and energy production relies on a three-tiered control system, (i) immediate and fast feedback in response to mechanical load on a beat-to-beat basis (Frank-Starling relation), (ii) more sustained regulation involving transmitters and hormones as primary messengers, and (iii) long-term adaptation by changes in the gene expression profile. Calcium signaling over largely different time scales requires its integration with the protein kinase signaling network which is governed by G-protein-coupled receptors, growth factor and cytokine receptors at the surface membrane. Short-term regulation is dominated by the beta-adrenergic system, while long-term regulation with phenotypic remodeling depends on sustained signaling by growth factors, cytokines and calcium. Mechanisms and new developments in intracellular calcium handling and its interrelation with the MAPK signaling pathways are discussed in detail.
Collapse
Affiliation(s)
- Marcus C Schaub
- Institute of Pharmacology and Toxicology, University of Zurich, Switzerland.
| | | | | |
Collapse
|
98
|
Davidov T, Weiss HR, Tse J, Scholz PM. Chronic nitric oxide synthase blockade desensitizes the heart to the negative metabolic effects of nitric oxide. Life Sci 2006; 79:1674-80. [PMID: 16831448 DOI: 10.1016/j.lfs.2006.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 05/16/2006] [Accepted: 06/02/2006] [Indexed: 11/28/2022]
Abstract
The consequences of chronic nitric oxide synthase (NOS) blockade on the myocardial metabolic and guanylyl cyclase stimulatory effects of exogenous nitric oxide (NO) were determined. Thirty-three anesthetized open-chest rabbits were randomized into four groups: control, NO donor S-nitroso-N-acetyl-penicillamine (SNAP, 10(-4 )M), NOS blocking agent N(G)-nitro-L-arginine methyl ester (L-NAME, 20 mg/kg/day) for 10 days followed by a 24 hour washout and L-NAME for 10 days followed by a 24 hour washout plus SNAP. Myocardial O(2) consumption was determined from coronary flow (microspheres) and O(2) extraction (microspectrophotometry). Cyclic GMP and guanylyl cyclase activity were determined by radioimmunoassay. There were no baseline metabolic, functional or hemodynamic differences between control and L-NAME treated rabbits. SNAP in controls caused a reduction in O(2) consumption (SNAP 5.9+/-0.6 vs. control 8.4+/-0.8 ml O(2)/min/100 g) and a rise in cyclic GMP (SNAP 18.3+/-3.8 vs. control 10.4+/-0.9 pmol/g). After chronic L-NAME treatment, SNAP caused no significant changes in O(2) consumption (SNAP 7.1+/-0.8 vs. control 6.4+/-0.7) or cyclic GMP (SNAP 14.2+/-1.8 vs. control 12.1+/-1.3). In controls, guanylyl cyclase activity was significantly stimulated by SNAP (216.7+/-20.0 SNAP vs. 34.4+/-2.5 pmol/mg/min base), while this increase was blunted after L-NAME (115.9+/-24.5 SNAP vs. 24.9+/-4.7 base). These results demonstrated that chronic NOS blockade followed by washout blunts the response to exogenous NO, with little effect on cyclic GMP or myocardial O(2) consumption. This was related to reduced guanylyl cyclase activity after chronic L-NAME. These results suggest that, unlike many receptor systems, the NO-cyclic GMP signal transduction system becomes downregulated upon chronic inhibition.
Collapse
Affiliation(s)
- Tomer Davidov
- Heart and Brain Circulation Laboratory, Department of Surgery, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ 08854-5635, USA
| | | | | | | |
Collapse
|
99
|
Neitzel KL, Hepler JR. Cellular mechanisms that determine selective RGS protein regulation of G protein-coupled receptor signaling. Semin Cell Dev Biol 2006; 17:383-9. [PMID: 16647283 DOI: 10.1016/j.semcdb.2006.03.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regulators of G protein signaling (RGS proteins) bind directly to activated Galpha subunits to inhibit their signaling. However, recent findings show that RGS proteins selectively regulate signaling by certain G protein-coupled receptors (GPCRs) in cells, irrespective of the coupled G protein. New studies support an emerging model that suggests RGS proteins utilize both direct and indirect mechanisms to form stable functional pairs with preferred GPCRs to selectively modulate the signaling functions of those receptors and linked G proteins. Here, we discuss these findings and their implications for established models of GPCR signaling.
Collapse
Affiliation(s)
- Karen L Neitzel
- Department of Pharmacology, Emory University School of Medicine, G205 Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA
| | | |
Collapse
|
100
|
Jafri F, El-Shewy HM, Lee MH, Kelly M, Luttrell DK, Luttrell LM. Constitutive ERK1/2 activation by a chimeric neurokinin 1 receptor-beta-arrestin1 fusion protein. Probing the composition and function of the G protein-coupled receptor "signalsome". J Biol Chem 2006; 281:19346-57. [PMID: 16670094 DOI: 10.1074/jbc.m512643200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The beta-arrestins, a small family of G protein-coupled receptor (GPCR)-binding proteins involved in receptor desensitization, have been shown to bind extracellular signal-regulated kinases 1 and 2 (ERK1/2) and function as scaffolds for GPCR-stimulated ERK1/2 activation. To better understand the mechanism of beta-arrestin-mediated ERK1/2 activation, we compared ERK1/2 activation by the wild-type neurokinin 1 (NK1) receptor with a chimeric NK1 receptor having beta-arrestin1 fused to the receptor C terminus (NK1-betaArr1). The NK1 receptor couples to both G(s) and G(q/11), resides on the plasma membrane, and mediates rapid ERK1/2 activation and nuclear translocation in response to neurokinin A. In contrast, NK1-betaArr1 is a G protein-uncoupled "constitutively desensitized" receptor that resides almost entirely in an intracellular endosomal compartment. Despite its inability to respond to neurokinin A, we found that NK1-betaArr1 expression caused robust constitutive activation of cytosolic ERK1/2 and that endogenous Raf, MEK1/2, and ERK1/2 coprecipitated in a complex with NK1-betaArr1. While agonist-dependent ERK1/2 activation by the NK1 receptor was independent of protein kinase A (PKA) or PKC activity, NK1-betaArr1-mediated ERK1/2 activation was completely inhibited when basal PKA and PKC activity were blocked. In addition, the rate of ERK1/2 dephosphorylation was slowed in NK1-betaArr1-expressing cells, suggesting that beta-arrestin-bound ERK1/2 is protected from mitogen-activated protein kinase phosphatase activity. These data suggest that beta-arrestin binding to GPCRs nucleates the formation of a stable "signalsome" that functions as a passive scaffold for the ERK1/2 cascade while confining ERK1/2 activity to an extranuclear compartment.
Collapse
Affiliation(s)
- Farahdiba Jafri
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|