1
|
Colin M, Delaitre C, Foulquier S, Dupuis F. The AT 1/AT 2 Receptor Equilibrium Is a Cornerstone of the Regulation of the Renin Angiotensin System beyond the Cardiovascular System. Molecules 2023; 28:5481. [PMID: 37513355 PMCID: PMC10383525 DOI: 10.3390/molecules28145481] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The AT1 receptor has mainly been associated with the pathological effects of the renin-angiotensin system (RAS) (e.g., hypertension, heart and kidney diseases), and constitutes a major therapeutic target. In contrast, the AT2 receptor is presented as the protective arm of this RAS, and its targeting via specific agonists is mainly used to counteract the effects of the AT1 receptor. The discovery of a local RAS has highlighted the importance of the balance between AT1/AT2 receptors at the tissue level. Disruption of this balance is suggested to be detrimental. The fine tuning of this balance is not limited to the regulation of the level of expression of these two receptors. Other mechanisms still largely unexplored, such as S-nitrosation of the AT1 receptor, homo- and heterodimerization, and the use of AT1 receptor-biased agonists, may significantly contribute to and/or interfere with the settings of this AT1/AT2 equilibrium. This review will detail, through several examples (the brain, wound healing, and the cellular cycle), the importance of the functional balance between AT1 and AT2 receptors, and how new molecular pharmacological approaches may act on its regulation to open up new therapeutic perspectives.
Collapse
Affiliation(s)
- Mélissa Colin
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France
- Department of Pharmacology and Toxicology, MHeNS-School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, MHeNS-School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, 6200 MD Maastricht, The Netherlands
| | | |
Collapse
|
2
|
Endocytosis and signaling of angiotensin II type 1 receptor. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:141-157. [PMID: 36631190 DOI: 10.1016/bs.pmbts.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A vasoactive octapeptide angiotensin II (Ang II) hormone is the key regulator of the renin-angiotensin system (RAS). It binds with the two different plasma membrane receptors like angiotensin II type 1 (AT1) and type 2 (AT2) and consequence various biological responses occur. Further, AT1 has two subtypes such as AT1A and AT1B. These angiotensin receptors are classified to be G protein-coupled receptors (GPCRs). The main constituent of RAS is the AT1 receptor (AT1R), and its activation, signal transduction, and regulation have been extensively studied. After Ang II stimulation, the ligand-receptor complexes internalized and trafficked through the early endosome, recycling endosome, and some receptors skipped the recycling endosome and trafficked to the lysosome for metabolic degradation. Moreover, some short sequence motifs located in the carboxyl-terminus (CT) of the receptor play a vital role in the internalization, phosphorylation, subcellular trafficking, signaling, and desensitization. Furthermore, in endocytosis, the various proteins interact with the CT region of the receptor. This chapter highlights the basic mechanism of AT1 receptor internalization, trafficking and signaling in both physiological and pathophysiological conditions.
Collapse
|
3
|
Rainbow RD, Brennan S, Jackson R, Beech AJ, Bengreed A, Waldschmidt HV, Tesmer JJG, Challiss RAJ, Willets JM. Small-Molecule G Protein-Coupled Receptor Kinase Inhibitors Attenuate G Protein-Coupled Receptor Kinase 2-Mediated Desensitization of Vasoconstrictor-Induced Arterial Contractions. Mol Pharmacol 2018; 94:1079-1091. [PMID: 29980659 DOI: 10.1124/mol.118.112524] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/29/2018] [Indexed: 01/01/2023] Open
Abstract
Vasoconstrictor-driven G protein-coupled receptor (GPCR)/phospholipase C (PLC) signaling increases intracellular Ca2+ concentration to mediate arterial contraction. To counteract vasoconstrictor-induced contraction, GPCR/PLC signaling can be desensitized by G protein-coupled receptor kinases (GRKs), with GRK2 playing a predominant role in isolated arterial smooth muscle cells. In this study, we use an array of GRK2 inhibitors to assess their effects on the desensitization of UTP and angiotensin II (AngII)-mediated arterial contractions. The effects of GRK2 inhibitors on the desensitization of UTP- or AngII-stimulated mesenteric third-order arterial contractions, and PLC activity in isolated mesenteric smooth muscle cells (MSMC), were determined using wire myography and Ca2+ imaging, respectively. Applying a stimulation protocol to cause receptor desensitization resulted in reductions in UTP- and AngII-stimulated arterial contractions. Preincubation with the GRK2 inhibitor paroxetine almost completely prevented desensitization of UTP- and attenuated desensitization of AngII-stimulated arterial contractions. In contrast, fluoxetine was ineffective. Preincubation with alternative GRK2 inhibitors (Takeda compound 101 or CCG224063) also attenuated the desensitization of UTP-mediated arterial contractile responses. In isolated MSMC, paroxetine, Takeda compound 101, and CCG224063 also attenuated the desensitization of UTP- and AngII-stimulated increases in Ca2+, whereas fluoxetine did not. In human uterine smooth muscle cells, paroxetine reversed GRK2-mediated histamine H1 receptor desensitization, but not GRK6-mediated oxytocin receptor desensitization. Utilizing various small-molecule GRK2 inhibitors, we confirm that GRK2 plays a central role in regulating vasoconstrictor-mediated arterial tone, highlighting a potentially novel strategy for blood pressure regulation through targeting GRK2 function.
Collapse
Affiliation(s)
- Richard D Rainbow
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Sean Brennan
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Robert Jackson
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Alison J Beech
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Amal Bengreed
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Helen V Waldschmidt
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - John J G Tesmer
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - R A John Challiss
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| | - Jonathon M Willets
- Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom (A.B., R.A.J.C., J.M.W.); Department of Cardiovascular Sciences, University of Leicester, Clinical Sciences Wing, Glenfield General Hospital, Leicester, United Kingdom (R.D.R., S.B., R.J., A.J.B.); Life Sciences Institute and Departments of Pharmacology, Biological Sciences, and Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan (H.V.W., J.J.G.T.); and Department of Biological Sciences, Purdue University, West Lafayette, Indiana (J.J.G.T.)
| |
Collapse
|
4
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
5
|
Willets JM, Nash CA, Rainbow RD, Nelson CP, Challiss RAJ. Defining the roles of arrestin2 and arrestin3 in vasoconstrictor receptor desensitization in hypertension. Am J Physiol Cell Physiol 2015; 309:C179-89. [PMID: 25972452 PMCID: PMC4525080 DOI: 10.1152/ajpcell.00079.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/11/2015] [Indexed: 11/22/2022]
Abstract
Prolonged vasoconstrictor-stimulated phospholipase C activity can induce arterial constriction, hypertension, and smooth muscle hypertrophy/hyperplasia. Arrestin proteins are recruited by agonist-occupied G protein-coupled receptors to terminate signaling and counteract changes in vascular tone. Here we determine whether the development of hypertension affects arrestin expression in resistance arteries and how such changes alter arterial contractile signaling and function. Arrestin2/3 expression was increased in mesenteric arteries of 12-wk-old spontaneously hypertensive rats (SHR) compared with normotensive Wistar-Kyoto (WKY) controls, while no differences in arrestin expression were observed between 6-wk-old SHR and WKY animals. In mesenteric artery myography experiments, high extracellular K(+)-stimulated contractions were increased in both 6- and 12-wk-old SHR animals. Concentration-response experiments for uridine 5'-triphosphate (UTP) acting through P2Y receptors displayed a leftward shift in 12-wk, but not 6-wk-old animals. Desensitization of UTP-stimulated vessel contractions was increased in 12-wk-old (but not 6-wk-old) SHR animals. Dual IP3/Ca(2+) imaging in mesenteric arterial cells showed that desensitization of UTP and endothelin-1 (ET1) responses was enhanced in 12-wk-old (but not 6-wk-old) SHR compared with WKY rats. siRNA-mediated depletion of arrestin2 for UTP and arrestin3 for ET1, reversed the desensitization of PLC signaling. In conclusion, arrestin2 and 3 expression is elevated in resistance arteries during the emergence of the early hypertensive phenotype, which underlies an enhanced ability to desensitize vasoconstrictor signaling and vessel contraction. Such regulatory changes may act to compensate for increased vasoconstrictor-induced vessel contraction.
Collapse
Affiliation(s)
- Jonathon M Willets
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom; and
| | - Craig A Nash
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom; and
| | - Richard D Rainbow
- Department of Cardiovascular Sciences, University of Leicester, Glenfield General Hospital, Leicester, United Kingdom
| | - Carl P Nelson
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom; and
| | - R A John Challiss
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, United Kingdom; and
| |
Collapse
|
6
|
Bojjireddy N, Guzman-Hernandez ML, Reinhard NR, Jovic M, Balla T. EFR3s are palmitoylated plasma membrane proteins that control responsiveness to G-protein-coupled receptors. J Cell Sci 2014; 128:118-28. [PMID: 25380825 DOI: 10.1242/jcs.157495] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The yeast Efr3p protein is a main regulator of the Stt4p phosphatidylinositol 4-kinase at contact sites between the endoplasmic reticulum and the plasma membrane. A mutation in its fly homologue Rbo, leads to diminished light responses in the eye attributed to progressively impaired PLC signaling. Here, we find that Efr3s plays a role in maintaining responsiveness to the type-I angiotensin II (AngII) receptors. siRNA-mediated depletion of EFR3A and EFR3B impaired the sustained phase of cytosolic Ca(2+) response to high concentration of AngII in HEK293 cells that express wild type but not truncated AGTR1 (AT1a receptor), missing the phosphorylation sites. Efr3 depletion had minimal effect on the recovery of plasma membrane phosphoinositides during stimulation, and AT1 receptors still underwent ligand-induced internalization. A higher level of basal receptor phosphorylation and a larger response was observed after stimulation. Moreover, Gq activation more rapidly desensitized after AngII stimulation in Efr3 downregulated cells. A similar but less pronounced effect of EFR3 depletion was observed on the desensitization of the cAMP response after stimulation with isoproterenol. These data suggest that mammalian Efr3s contribute to the control of the phosphorylation state and, hence, desensitization of AT1a receptors, and could affect responsiveness of G-protein-coupled receptors in higher eukaryotes.
Collapse
Affiliation(s)
- Naveen Bojjireddy
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria Luisa Guzman-Hernandez
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nathalie Renée Reinhard
- Swammerdam Institute for Life Sciences, University of Amsterdam, 1012 WX Amsterdam, The Netherlands
| | - Marko Jovic
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver NICHD, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Balakumar P, Jagadeesh G. Structural determinants for binding, activation, and functional selectivity of the angiotensin AT1 receptor. J Mol Endocrinol 2014; 53:R71-92. [PMID: 25013233 DOI: 10.1530/jme-14-0125] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The renin-angiotensin system (RAS) plays an important role in the pathophysiology of cardiovascular disorders. Pharmacologic interventions targeting the RAS cascade have led to the discovery of renin inhibitors, angiotensin-converting enzyme inhibitors, and AT(1) receptor blockers (ARBs) to treat hypertension and some cardiovascular and renal disorders. Mutagenesis and modeling studies have revealed that differential functional outcomes are the results of multiple active states conformed by the AT(1) receptor upon interaction with angiotensin II (Ang II). The binding of agonist is dependent on both extracellular and intramembrane regions of the receptor molecule, and as a consequence occupies more extensive area of the receptor than a non-peptide antagonist. Both agonist and antagonist bind to the same intramembrane regions to interfere with each other's binding to exhibit competitive, surmountable interaction. The nature of interactions with the amino acids in the receptor is different for each of the ARBs given the small differences in the molecular structure between drugs. AT(1) receptors attain different conformation states after binding various Ang II analogues, resulting in variable responses through activation of multiple signaling pathways. These include both classical and non-classical pathways mediated through growth factor receptor transactivations, and provide cross-communication between downstream signaling molecules. The structural requirements for AT(1) receptors to activate extracellular signal-regulated kinases 1 and 2 through G proteins, or G protein-independently through β-arrestin, are different. We review the structural and functional characteristics of Ang II and its analogs and antagonists, and their interaction with amino acid residues in the AT(1) receptor.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Pharmacology UnitFaculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, MalaysiaDivision of Cardiovascular and Renal ProductsCenter for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Gowraganahalli Jagadeesh
- Pharmacology UnitFaculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, MalaysiaDivision of Cardiovascular and Renal ProductsCenter for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland 20993, USA
| |
Collapse
|
8
|
Huang LL, Pan C, Yu TT, Guo K, Wang XH, Zhang JY, Wang HZ, Gao S. Benefical therapeutic effect of Chinese Herbal Xinji'erkang formula on hypertension-induced renal injury in the 2-kidney-1-clip hypertensive rats. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES : AJTCAM 2014; 11:16-27. [PMID: 25395699 PMCID: PMC4202512 DOI: 10.4314/ajtcam.v11i5.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Increase in evidence shows that the role of kidney injury in hypertension is important. Xinji'erkang (XJEK), a Chinese herbal formula, has been identified as an effective preparation in the treatment of coronary heart disease and myocarditis. We have previously demonstrated that XJEK attenuate oxidative stress and hypertension target organ damage. The aim of this study was to assess the renal protective function of XJEK. MATERIALS AND METHODS Two Kidney One Clip (2K1C) model was adopted to induce hypertension in rats. We submitted male Sprague Dawley (150-180) g rats to either renal artery clipping or sham operation. Renal hypertension was established after four weeks of surgery. Rats were randomized divided into the four groups: sham-operated group (Sh-Op) (n=10), two-kidney, one-clip hypertension group (2K1C) (n=10), Xinji'erkang treatment group (XJEK) (n=10) and Fosinopril (n=10) treatment group. Drugs were administered orally daily for four weeks. Systolic pressures were measured every week using the tail-cuff apparatus. 24h before death, urine samples were collected for detect of urinary proteins. The kidney weight (KW) index was expressed as kidney weight/body weight (KW/BW). The histological changes were investigated by hematoxylin and eosin and Van Gieson staining. Immunohistochemical assay was employed to observe the intra-renal transforming growth factor-β1 (TGF-β1) protein expression. Serum creatinine (SCR) and blood urea nitrogen (BUN) were assayed by automatic biochemical analyzer. ELISA kit was used to assay Angiotensin II (Ang II) and TGF-β1 content in serum. RESULTS Administration of XJEK markedly alleviated the rise in blood pressure and declined LKW/BW ratio. Histo-pathological injuries including hypertrophic glomerular, glomerular sclerosis, glomerular and interstitial fibrosis were attenuated. XJEK also decreased SCR, BUN, urinary proteins in 24h urine, serum Ang II and TGF-β1 concentrations and the intra-renal TGF-β1 protein expression. CONCLUSION XJEK therapy in the 2K1C hypertensive rats affects the rise in blood pressure and ameliorates the severity of kidney injury. The protective effect is most likely due to the ability of XJEK to affect the Renin-Angiotensin-Aldosterone System (RAAS) and the TGF-β systems.
Collapse
Affiliation(s)
- Ling-Ling Huang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Science, Hefei 230031, China
| | - Chen Pan
- Department of Clinical of Pharmacy, Lishui People's Hospital, Zhe Jiang 323000, China
| | - Ting-ting Yu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Kun Guo
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Xing-hui Wang
- Department of Pharmacy, the Second People's Hospital of Hefei, Hefei 230011, China
| | - Jun-Yan Zhang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Hong-zhi Wang
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Science, Hefei 230031, China
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
9
|
Scimia MC, Blass BE, Koch WJ. Apelin receptor: its responsiveness to stretch mechanisms and its potential for cardiovascular therapy. Expert Rev Cardiovasc Ther 2014; 12:733-41. [DOI: 10.1586/14779072.2014.911661] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
10
|
βArrestins in cardiac G protein-coupled receptor signaling and function: partners in crime or "good cop, bad cop"? Int J Mol Sci 2013; 14:24726-41. [PMID: 24351844 PMCID: PMC3876138 DOI: 10.3390/ijms141224726] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/12/2022] Open
Abstract
βarrestin (βarr)-1 and -2 (βarrs) (or Arrestin-2 and -3, respectively) are universal G protein-coupled receptor (GPCR) adapter proteins expressed abundantly in extra-retinal tissues, including the myocardium. Both were discovered in the lab of the 2012 Nobel Prize in Chemistry co-laureate Robert Lefkowitz, initially as terminators of signaling from the β-adrenergic receptor (βAR), a process known as functional desensitization. They are now known to switch GPCR signaling from G protein-dependent to G protein-independent, which, in the case of βARs and angiotensin II type 1 receptor (AT1R), might be beneficial, e.g., anti-apoptotic, for the heart. However, the specific role(s) of each βarr isoform in cardiac GPCR signaling and function (or dysfunction in disease), remain unknown. The current consensus is that, whereas both βarr isoforms can desensitize and internalize cardiac GPCRs, they play quite different (even opposing in certain instances) roles in the G protein-independent signaling pathways they initiate in the cardiovascular system, including in the myocardium. The present review will discuss the current knowledge in the field of βarrs and their roles in GPCR signaling and function in the heart, focusing on the three most important, for cardiac physiology, GPCR types (β1AR, β2AR & AT1R), and will also highlight important questions that currently remain unanswered.
Collapse
|
11
|
Arrestins in the cardiovascular system. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:297-334. [PMID: 23764059 DOI: 10.1016/b978-0-12-394440-5.00012-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Of the four mammalian arrestins, only the β-arrestins (βarrs; Arrestin2 and -3) are expressed throughout the cardiovascular system, where they regulate, as either desensitizers/internalizers or signal transducers, several G-protein-coupled receptors (GPCRs) critical for cardiovascular homeostasis. The cardiovascular roles of βarrs have been delineated at an accelerated pace via a variety of techniques and tools, such as knockout mice, siRNA knockdown, artificial or naturally occurring polymorphic GPCRs, and availability of new βarr "biased" GPCR ligands. This chapter summarizes the current knowledge of cardiovascular arrestin physiology and pharmacology, addressing the individual cardiovascular receptors affected by βarrs in vivo, as well as the individual cell types, tissues, and organs of the cardiovascular system in which βarr effects are exerted; for example, cardiac myocyte or fibroblast, vascular smooth muscle, adrenal gland and platelet. In the broader scope of cardiovascular βarr pharmacology, a discussion of the βarr "bias" of certain cardiovascular GPCR ligands is also included.
Collapse
|
12
|
On the segregation of protein ionic residues by charge type. Amino Acids 2012; 43:2231-47. [PMID: 23081700 DOI: 10.1007/s00726-012-1418-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 10/06/2012] [Indexed: 10/27/2022]
Abstract
Based on ubiquitous presence of large ionic motifs and clusters in proteins involved in gene transcription and protein synthesis, we analyzed the distribution of ionizable sidechains in a broad selection of proteins with regulatory, metabolic, structural and adhesive functions, in agonist, antagonist, toxin and antimicrobial peptides, and in self-excising inteins and intron-derived proteins and sequence constructs. All tested groups, regardless of taxa or sequence size, show considerable segregation of ionizable sidechains into same type charge (homoionic) tracts. These segments in most cases exceed half of the sequence length and comprise more than two-thirds of all ionizable sidechains. This distribution of ionic residues apparently reflects a fundamental advantage of sorted electrostatic contacts in association of sequence elements within and between polypeptides, as well as in interaction with polynucleotides. While large ionic densities are encountered in highly interactive proteins, the average ionic density in most sets does not change appreciably with size of the homoionic segments, which supports the segregation as a modular feature favoring association.
Collapse
|
13
|
Abstract
Drug discovery efforts targeting G-protein-coupled receptors (GPCR) have been immensely successful in creating new cardiovascular medicines. Currently marketed GPCR drugs are broadly classified as either agonists that activate receptors or antagonists that prevent receptor activation by endogenous stimuli. However, GPCR couple to a multitude of intracellular signaling pathways beyond classical G-protein signals, and these signals can be independently activated by biased ligands to vastly expand the potential for new drugs at these classic targets. By selectively engaging only a subset of a receptor's potential intracellular partners, biased ligands may deliver more precise therapeutic benefit with fewer side effects than current GPCR-targeted drugs. In this review, we discuss the history of biased ligand research, the current understanding of how biased ligands exert their unique pharmacology, and how research into GPCR signaling has uncovered previously unappreciated capabilities of receptor pharmacology. We focus on several receptors to illustrate the approaches taken and discoveries made, and how these are steadily illuminating the intricacies of GPCR pharmacology. Discoveries of biased ligands targeting the angiotensin II type 1 receptor and of separable pharmacology suggesting the potential value of biased ligands targeting the β-adrenergic receptors and nicotinic acid receptor GPR109a highlight the powerful clinical promise of this new category of potential therapeutics.
Collapse
|
14
|
Evidence of an intracellular angiotensin-generating system and non-AT1, non-AT2 binding site in a human pancreatic cell line. Pancreas 2011; 40:701-7. [PMID: 21602736 DOI: 10.1097/mpa.0b013e318215a891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To assess the presence of a local angiotensin-generating systems (LAGS) and its participation in tumor growth in the human pancreatic cancer derived cell line Capan-1. METHODS Capan-1 cells were cultured in Dulbecco modified Eagle medium, and angiotensin I was assayed by radioimmunoassay and angiotensin II and vascular endothelial growth factor were assayed by enzyme-linked immunosorbent assay in the supernatant. Immunohistochemistry and reverse transcription-polymerase chain reaction were performed for the expression of AT1 and AT2 receptors. Angiotensin II binding assays and blockade were studied. RESULTS High levels of both angiotensins I and II were found in Capan-1 cells, although neither angiotensin I nor angiotensin II was detected in the cell culture supernatant. Reverse transcription-polymerase chain reaction and immunocytochemistry revealed that Capan-1 cells do not express AT1 and AT2 receptors; however, specific binding to the cell membrane was identified for angiotensin II. Neither exogenous angiotensin II nor Dup753 (specific AT1 receptor blocker) affected Capan-1 cells' proliferation or vascular endothelial growth factor secretion. CONCLUSIONS Detection of both angiotensin I and angiotensin II along with specific binding of angiotensin II in Capan-1 cells provides evidence of the existence of a LAGS that operates in an intracrine manner. Intracellular angiotensin II may play a role in the aggressiveness of pancreatic cancer and is a possible target for therapeutic agents.
Collapse
|
15
|
Zhang X, Dong C, Wu QJ, Balch WE, Wu G. Di-acidic motifs in the membrane-distal C termini modulate the transport of angiotensin II receptors from the endoplasmic reticulum to the cell surface. J Biol Chem 2011; 286:20525-35. [PMID: 21507945 DOI: 10.1074/jbc.m111.222034] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The molecular mechanisms underlying the endoplasmic reticulum (ER) export and cell surface transport of nascent G protein-coupled receptors (GPCRs) have just begun to be revealed and previous studies have shown that hydrophobic motifs in the putative amphipathic 8(th) α-helical region within the membrane-proximal C termini play an important role. In this study, we demonstrate that di-acidic motifs in the membrane-distal, nonstructural C-terminal portions are required for the exit from the ER and transport to the plasma membrane of angiotensin II receptors, but not adrenergic receptors. More interestingly, distinct di-acidic motifs dictate optimal export trafficking of different angiotensin II receptors and export ability of each acidic residue in the di-acidic motifs cannot be fully substituted by other acidic residue. Moreover, the function of the di-acidic motifs is likely mediated through facilitating the recruitment of the receptors onto the ER-derived COPII transport vesicles. Therefore, the di-acidic motifs located in the membrane-distal C termini may represent the first linear motifs which recruit selective GPCRs onto the COPII vesicles to control their export from the ER.
Collapse
Affiliation(s)
- Xiaoping Zhang
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | |
Collapse
|
16
|
Balla A, Erdélyi LS, Soltész-Katona E, Balla T, Várnai P, Hunyady L. Demonstration of angiotensin II-induced Ras activation in the trans-Golgi network and endoplasmic reticulum using bioluminescence resonance energy transfer-based biosensors. J Biol Chem 2010; 286:5319-27. [PMID: 21062747 DOI: 10.1074/jbc.m110.176933] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Previous studies have demonstrated that molecules of the Ras signaling pathway are present in intracellular compartments, including early endosomes, the endoplasmic reticulum (ER), and the Golgi, and suggested that mitogens can regulate Ras activity in these endomembranes. In this study, we investigated the effect of angiotensin II (AngII) on intracellular Ras activity in living HEK293 cells expressing angiotensin type 1 receptors (AT(1)-Rs) using newly developed bioluminescence resonance energy transfer biosensors. To investigate the subcellular localization of AngII-induced Ras activation, we targeted our probes to various intracellular compartments, such as the trans-Golgi network (TGN), the ER, and early endosomes. Using these biosensors, we detected AngII-induced Ras activation in the TGN and ER, but not in early endosomes. In cells expressing a cytoplasmic tail deletion AT(1)-R mutant, the AngII-induced response was enhanced, suggesting that receptor internalization and β-arrestin binding are not required for AngII-induced Ras activation in endomembranes. Although we were able to demonstrate EGF-induced Ras activation in the plasma membrane and TGN, but not in other endomembranes, AG1478, an EGF receptor inhibitor, did not affect the AngII-induced response, suggesting that the latter is independent of EGF receptor transactivation. AngII was unable to stimulate Ras activity in the studied compartments in cells expressing a G protein coupling-deficient AT(1)-R mutant ((125)DRY(127) to (125)AAY(127)). These data suggest that AngII can stimulate Ras activity in the TGN and ER with a G protein-dependent mechanism, which does not require β-arrestin-mediated signaling, receptor internalization, and EGF receptor transactivation.
Collapse
Affiliation(s)
- András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, H-1444 Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
17
|
Esseltine JL, Dale LB, Ferguson SSG. Rab GTPases bind at a common site within the angiotensin II type I receptor carboxyl-terminal tail: evidence that Rab4 regulates receptor phosphorylation, desensitization, and resensitization. Mol Pharmacol 2010; 79:175-84. [PMID: 20943774 DOI: 10.1124/mol.110.068379] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The human angiotensin II type 1 receptor (AT₁R) is a member of the G protein-coupled receptor (GPCR) superfamily and represents an important target for cardiovascular therapeutic intervention. Agonist-activation of the AT₁R induces β-arrestin-dependent endocytosis to early endosomes in which the receptor resides as a protein complex with the Rab GTPase Rab5. In the present study, we examined whether other Rab GTPases that regulate receptor trafficking through endosomal compartments also bind to the AT₁R. We find that Rab4, Rab7, and Rab11 all bind to the last 10 amino acid residues of the AT₁R carboxyl-terminal tail. Rab11 binds AT₁R more effectively than Rab5, whereas Rab4 binds less effectively than Rab5. Alanine scanning mutagenesis reveals that proline 354 and cysteine 355 contribute to Rab protein binding, and mutation of these residues does not affect G protein coupling. We find that the Rab GTPases each compete with one another for receptor binding and that although Rab4 interacts poorly with the AT₁R, it effectively displaces Rab11 from the receptor. In contrast, Rab11 overexpression does not prevent Rab4 binding to the AT₁R. Overexpression of wild-type Rab4, but not Rab11, facilitates AT₁R dephosphorylation, and a constitutively active Rab4-Q67L mutant reduces AT₁R desensitization and promotes AT₁R resensitization. Taken together, our data indicate that multiple Rab GTPases bind to a motif localized to the distal end of the AT₁R tail and that increased Rab4 activity may contribute to the regulation AT₁R desensitization and dephosphorylation.
Collapse
Affiliation(s)
- Jessica L Esseltine
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | | | | |
Collapse
|
18
|
Min LJ, Mogi M, Tamura K, Iwanami J, Sakata A, Fujita T, Tsukuda K, Jing F, Iwai M, Horiuchi M. Angiotensin II type 1 receptor-associated protein prevents vascular smooth muscle cell senescence via inactivation of calcineurin/nuclear factor of activated T cells pathway. J Mol Cell Cardiol 2009; 47:798-809. [DOI: 10.1016/j.yjmcc.2009.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/28/2009] [Accepted: 09/09/2009] [Indexed: 11/29/2022]
|
19
|
Martin RP, Rodrigues ES, Pacheco NAS, Corrêa SAA, Oliveira SM, Oliveira L, Nakaie CR, Shimuta SI. Distinct binding mode of 125I-AngII to AT1 receptor without the Cys18-Cys274 disulfide bridge. ACTA ACUST UNITED AC 2009; 158:14-8. [PMID: 19651161 DOI: 10.1016/j.regpep.2009.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 07/13/2009] [Accepted: 07/23/2009] [Indexed: 10/20/2022]
Abstract
Previous studies on angiotensin II (AngII) AT(1) receptor function have revealed that the N-terminal residues of AngII may modulate receptor activation by binding at the receptor extracellular site. A remarkable feature of this site is an insertion of 8 amino acids in the middle of the EC-3 loop including the Cys(274) residue that supposedly makes a disulfide bond with N-terminal Cys(18). As demonstrated by assays with Del(267-275)AT(1), the role of the Cys(18)-Cys(274) disulfide bridge is to keep a conformation of the inserted residues that allows a normal binding of the AngII N-terminal residues. C18S AT(1) receptor mutant, supposedly having a dissociated disulfide bridge, but an intact residue insertion, is constitutively activated and can less efficiently bind AngII. Similar results were observed when the S-S disulfide bond was disrupted in (C18S,C274S) AT(1) receptor. The importance of the free N-terminal amino group of Asp(1) and of the Arg(2) guanidino group for the binding of AngII to C18S mutant with EC-3 loop insertion was investigated by means of assays using AngII peptide analogues bearing a single mutation of Asp(1) for Sar(1) or Arg(2) for Lys(2), as ligands. This study showed that like AngII, [Sar(1)]-AngII can bind the C18S mutant receptor with low affinity whereas [Lys(2)]-AngII binding is still more reduced. Interestingly, when (125)I-AngII instead of (3)H-AngII was used, no significant binding of this mutant was observed although wild type AT(1) receptor was shown to bind all AngII analogues.
Collapse
Affiliation(s)
- Renan P Martin
- Department of Biophysics, Federal University of São Paulo, 04023-062 São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
20
|
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]
|
21
|
Bivol LM, Hultström M, Gudbrandsen OA, Berge RK, Iversen BM. Tetradecylthioacetic acid downregulates cyclooxygenase 2 in the renal cortex of two-kidney, one-clip hypertensive rats. Am J Physiol Regul Integr Comp Physiol 2008; 295:R1866-73. [PMID: 18843091 DOI: 10.1152/ajpregu.00850.2007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The effect of tetradecylthioacetic acid (TTA) on the cyclooxygenase (COX) system was investigated in two-kidney, one-clip (2K1C) hypertensive rats. The systolic blood pressure (BP) was increased 6 wk after clipping to 183 +/- 4 vs.127 +/- 3 mmHg in TTA-treated 2K1C rats. The COX1 protein expression was not affected either by the 2K1C procedure or by TTA treatment. COX2 expression was upregulated in both kidneys, but to a greater extent in the clipped kidney. COX2 activity was 16 +/- 3% in control and 38 +/- 2% (P < 0.001) in the clipped kidney, and COX2 protein expression was 1.3 +/- 0.04 in control and 1.6 +/- 0.12 in the clipped kidney (P = 0.006). TTA reduced COX2 activity to control levels. Subcutaneously infusion of a COX2 inhibitor did not reduce BP. Peroxisome proliferator-activated receptors (PPARs) were detected in both kidneys, and PPARdelta was upregulated in the nonclipped kidney after TTA treatment. PGE2 in renal cortex was increased in 2K1C (31 +/- 0.3 in the clipped and 28 +/- 0.2 pg/ml nonclipped kidney, P < 0.001 compared with control). TTA lowered the PGE2 to control levels. Renal blood flow (RBF) response to exogenous ANG II injected in the control and nonclipped kidney was exaggerated after indomethacin treatment but unchanged in the nonclipped kidney of the K1C TTA group. Overall, these results indicate that, after 6 wk of treatment, TTA downregulated the COX2 activity, which have potentially important effects on the regulation of renal hemodynamics but does not explain TTAs ability to lower BP.
Collapse
Affiliation(s)
- Liliana Monica Bivol
- Renal Research Group, Institute of Medicine, University of Bergen, Bergen, Norway
| | | | | | | | | |
Collapse
|
22
|
Zhang X, Wang F, Chen X, Chen Y, Ma L. Post-endocytic fates of -opioid receptor are regulated by GRK2-mediated receptor phosphorylation and distinct -arrestin isoforms. J Neurochem 2008; 106:781-92. [DOI: 10.1111/j.1471-4159.2008.05431.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Agonist-specific down regulation of mu-opioid receptors: Different cellular pathways are activated by different opioid agonists. Life Sci 2008; 82:831-9. [PMID: 18358497 DOI: 10.1016/j.lfs.2008.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Revised: 01/17/2008] [Accepted: 01/23/2008] [Indexed: 11/21/2022]
Abstract
Opioid agonists are known to induce down regulation of opioid receptors through the classical pathway that involves phosphorylation, clathrin-dependent endocytosis and lysosomal/endosomal degradation of the internalized receptors. As expected, exposure of mu-opioid receptor (MOR)-transfected HEK-293 cells to either DAMGO (a specific mu-opioid agonist) or etorphine (a wide spectrum opioid agonist) resulted in down regulation of the receptors that was blocked by the kinase inhibitor staurosporine, by hypertonic sucrose and by the lysosomal and proteasomal inhibitors chloroquine and lactacystin. High concentration of etorphine, but not of DAMGO, induced an additional process of down regulation that was resistant to staurosporine, to hypertonic sucrose and to chloroquine-lactacystin. Etorphine, but not DAMGO, also induced down regulation of mu-opioid receptors in isolated membranes of HEK cells. This membrane-delimited down regulation was blocked by selective inhibitors of protease enzymes, suggesting the involvement of membranous serine- and amino-peptidases. This membranous down regulation of opioid receptors was dependent on the concentration of etorphine and was blocked by the opioid antagonist naloxone. Etorphine induced similar down regulation in membranes of HEK-293 cells transfected with delta-opioid receptors (DOR) as well in membranes of cells that endogenously express opioid receptors. This agonist-specific membrane-delimited regulatory process appears to be physiologically relevant and should be taken into account when studying long term effects of opioid drugs.
Collapse
|
24
|
Speth RC, Karamyan VT. Brain angiotensin receptors and binding proteins. Naunyn Schmiedebergs Arch Pharmacol 2008; 377:283-93. [PMID: 18172611 DOI: 10.1007/s00210-007-0238-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Accepted: 11/26/2007] [Indexed: 12/29/2022]
Abstract
This review addresses classical and novel aspects of the brain angiotensin system. The brain contains both the AT1 and AT2 angiotensin II (Ang II) receptor subtypes which are well-characterized guanine nucleotide binding protein (G protein)-coupled receptors (GPCRs). Like other GPCRs, novel signal transduction pathways and protein interactions are being described for Ang II receptors. For brain AT1 receptors, there is a controversy regarding the identity of the active angiotensin peptide in the brain which is addressed in this review. This review also summarizes a recent discovery of a novel, membrane-bound, non-AT1, non-AT2 binding site for angiotensin peptides that appears to be brain-specific. This binding site is unmasked by a limited concentration range of the organometallic sulfhydryl-reactive agent p-chloromercuribenzoic acid (PCMB) suggesting that functional expression of this binding site may depend on the redox state of the milieu of the brain. While this binding site has similarities to a previously described soluble angiotensin-binding protein found in liver that is unmasked by PCMB, it has many different characteristics. The possible functional significance of this novel non-AT1, non-AT2 binding site for angiotensin peptides as a mediator of non-traditional actions of Ang II in the brain, e.g., stimulation of dopamine release from the striatum, as a peptidase, or as a clearance receptor, and the importance of the state of the internal environment of the brain to its function is reviewed.
Collapse
Affiliation(s)
- Robert C Speth
- Department of Pharmacology, Research Institute of Pharmaceutical Sciences, University of Mississippi, University, Oxford, MS 38677, USA.
| | | |
Collapse
|
25
|
de Godoy MAF, Rattan S. Translocation of AT1- and AT2-Receptors by Higher Concentrations of Angiotensin II in the Smooth Muscle Cells of Rat Internal Anal Sphincter. J Pharmacol Exp Ther 2006; 319:1088-95. [PMID: 16985169 DOI: 10.1124/jpet.106.108084] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies have reported bimodal effects by angiotensin II (Ang II) in the rat internal anal sphincter (IAS), a concentration-dependent contraction (at lower concentrations) and relaxation (at higher concentrations). The experiments suggest the above-mentioned responses are the result of Ang II subtype I receptor(s) (AT(1)-R) and subtype II receptor(s) (AT(2)-R) activation, respectively. These studies determined the role and mechanism of AT(2)-R-induced relaxation of the smooth muscle cells (SMCs) from the IAS in response to Ang II. Laser confocal microscopy showed that in the basal state, the AT(1)-Rs reside in the plasma membrane, whereas AT(2)-Rs are present in the cytosol. Higher concentrations of Ang II caused movement of AT(1)-R and AT(2)-R in opposite directions to the cytosol and the membrane, respectively. Losartan (AT(1)-R antagonist) but not S-(+)-1-([4-(dimethylamino)-3-methylphenyl]methyl)-5-(diphenylacetyl)-4,5,6,7-tetrahydro-1H-imidazo(4,5-c)pyridine-6-carboxylic acid (PD123319; AT(2)-R antagonist) selectively inhibited these movements. These results are based on biotinylation assays, confocal images, and Western blot analyses of the densities of AT(1)-Rs and AT(2)-Rs in the plasma membrane versus cytosolic fractions of the IAS SMCs. Ang II in higher concentrations did not change the total contents of Ang II receptors. These data combined with the functional data using measurements of IAS SMC lengths suggest that internalization of AT(1)-R and externalization of AT(2)-R may be responsible for the activation of the AT(2)-R, which leads to the relaxation of the IAS with higher concentrations of Ang II.
Collapse
MESH Headings
- Anal Canal/cytology
- Anal Canal/drug effects
- Anal Canal/metabolism
- Angiotensin II/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Biotin/metabolism
- Blotting, Western
- Cytosol/drug effects
- Cytosol/metabolism
- Fluorescent Antibody Technique
- Imidazoles/pharmacology
- In Vitro Techniques
- Losartan/pharmacology
- Male
- Microscopy, Confocal
- Muscle, Smooth/cytology
- Muscle, Smooth/drug effects
- Muscle, Smooth/metabolism
- Pyridines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Translocation, Genetic/drug effects
Collapse
Affiliation(s)
- Márcio A F de Godoy
- Jefferson Medical College of Thomas Jefferson University, Department of Medicine, Philadelphia, PA 19107, USA
| | | |
Collapse
|
26
|
Zaika O, Lara LS, Gamper N, Hilgemann DW, Jaffe DB, Shapiro MS. Angiotensin II regulates neuronal excitability via phosphatidylinositol 4,5-bisphosphate-dependent modulation of Kv7 (M-type) K+ channels. J Physiol 2006; 575:49-67. [PMID: 16777936 PMCID: PMC1819424 DOI: 10.1113/jphysiol.2006.114074] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Voltage-gated Kv7 (KCNQ) channels underlie important K+ currents in many different types of cells, including the neuronal M current, which is thought to be modulated by muscarinic stimulation via depletion of membrane phosphatidylinositol 4,5-bisphosphate (PIP2). We studied the role of modulation by angiotensin II (angioII) of M current in controlling discharge properties of superior cervical ganglion (SCG) sympathetic neurons and the mechanism of action of angioII on cloned Kv7 channels in a heterologous expression system. In SCG neurons, which endogenously express angioII AT1 receptors, application of angioII for 2 min produced an increase in neuronal excitability and a decrease in spike-frequency adaptation that partially returned to control values after 10 min of angioII exposure. The increase in excitability could be simulated in a computational model by varying only the amount of M current. Using Chinese hamster ovary (CHO) cells expressing cloned Kv7.2 + 7.3 heteromultimers and AT1 receptors studied under perforated patch clamp, angioII induced a strong suppression of the Kv7.2/7.3 current that returned to near baseline within 10 min of stimulation. The suppression was blocked by the phospholipase C inhibitor edelfosine. Under whole-cell clamp, angioII moderately suppressed the Kv7.2/7.3 current whether or not intracellular Ca2+ was clamped or Ca2+ stores depleted. Co-expression of PI(4)5-kinase in these cells sharply reduced angioII inhibition, but did not augment current amplitudes, whereas co-expression of a PIP2 5'-phosphatase sharply reduced current amplitudes, and also blunted the inhibition. The rebound of the current seen in perforated-patch recordings was blocked by the PI4-kinase inhibitor, wortmannin (50 microM), suggesting that PIP2 re-synthesis is required for current recovery. High-performance liquid chromatographic analysis of anionic phospholipids in CHO cells stably expressing AT1 receptors revealed that PIP2 and phosphatidylinositol 4-phosphate levels are to be strongly depleted after 2 min of stimulation with angioII, with a partial rebound after 10 min. The results of this study establish how angioII modulates M channels, which in turn affects the integrative properties of SCG neurons.
Collapse
Affiliation(s)
- Oleg Zaika
- Department of Physiology, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | | | | | | | | | |
Collapse
|
27
|
Wang GD, Wang XY, Hu HZ, Fang XC, Liu S, Gao N, Xia Y, Wood JD. Angiotensin receptors and actions in guinea pig enteric nervous system. Am J Physiol Gastrointest Liver Physiol 2005; 289:G614-26. [PMID: 16093423 DOI: 10.1152/ajpgi.00119.2005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Actions of ANG II on electrical and synaptic behavior of enteric neurons in the guinea pig small intestine were studied. Exposure to ANG II depolarized the membrane potential and elevated neuronal excitability. The number of responding neurons was small, with responses to ANG II in 32% of submucosal neurons and 25% of myenteric neurons. Hyperpolarizing responses were evoked by ANG II in 45% of the neurons. The hyperpolarizing responses were suppressed by alpha2-noradrenergic receptor antagonists, which suggested that the hyperpolarizing responses reflected stimulation of norepinephrine release from sympathetic neurons. Exposure to ANG II enhanced the amplitude and prolonged the duration of noradrenergic inhibitory postsynaptic potentials and suppressed the amplitude of both fast and slow excitatory postsynaptic potentials. The selective ANG II(1) receptor (AT1R) antagonists, ZD-7115 and losartan, but not a selective AT2R antagonist (PD-123319), suppressed the actions of ANG II. Western blot analysis and RT-PCR confirmed expression of AT1R protein and the mRNA transcript for the AT1R in the enteric nervous system. No expression of AT2R protein or mRNA was found. Immunoreactivity for AT1R was expressed by the majority of neurons in the gastric antrum and small and large intestine. AT1R immunoreactivity was coexpressed with calbindin, choline acetyltransferase, calretinin, neuropeptide Y, and nitric oxide synthase in subpopulations of neurons. The results suggest that formation of ANG II might have paracrine-like actions in the enteric nervous system, which include alterations in neuronal excitability and facilitated release of norepinephrine from sympathetic postganglionic axons. The enhanced presence of norepinephrine is expected to suppress fast and slow excitatory neurotransmission in the enteric microcircuits and to suppress neurogenic mucosal secretion.
Collapse
MESH Headings
- Angiotensin II/physiology
- Animals
- Blotting, Western
- Electrophysiology
- Female
- Guinea Pigs
- Humans
- Inflammation
- Intestine, Small/innervation
- Intestine, Small/physiology
- Irritable Bowel Syndrome/physiopathology
- Male
- Membrane Potentials
- Myenteric Plexus/physiology
- Norepinephrine/physiology
- RNA, Messenger/biosynthesis
- Receptor, Angiotensin, Type 1/biosynthesis
- Receptor, Angiotensin, Type 1/physiology
- Receptor, Angiotensin, Type 2/biosynthesis
- Receptor, Angiotensin, Type 2/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Synaptic Transmission/physiology
Collapse
Affiliation(s)
- Guo-Du Wang
- Dept. of Physiology and Cell Biology, The Ohio State University, College of Medicine and Public Health, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Zeng C, Wang Z, Asico LD, Hopfer U, Eisner GM, Felder RA, Jose PA. Aberrant ETB receptor regulation of AT1 receptors in immortalized renal proximal tubule cells of spontaneously hypertensive rats. Kidney Int 2005; 68:623-31. [PMID: 16014039 DOI: 10.1111/j.1523-1755.2005.00440.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The renin-angiotensin and endothelin systems interact to regulate blood pressure, in part, by affecting sodium transport in the kidney. Because angiotensin II type 1 (AT(1)) receptor activation increases ETB receptor expression in renal proximal tubule cells from Wistar-Kyoto (WKY) rat, we hypothesize that ETB receptor activation may also regulate AT(1) receptor expression. Furthermore, ETB receptor regulation of the AT(1) receptor may be different in the WKY and spontaneously hypertensive rat (SHR). METHOD AT(1) and ETB receptors were studied in immortalized renal proximal tubule cells from WKY and SHRs, using immunoblotting, confocal microscopic colocalization, and immunoprecipitation. RESULTS In WKY renal proximal tubule cells, an ETB receptor agonist, BQ3020, decreased AT(1) receptor protein in a time- and concentration-dependent manner [median effective concentration (EC(50)) = 3.2 x 10(-10) mol/L, t(1/2)= 15 hours]. The inhibitory effect of BQ3020 (10(-8) mol/L/24 hours) on AT(1) receptor protein was blocked by an ETB receptor antagonist (BQ788). However, BQ3020 (10(-8) mol/L/24 hours) increased ETB receptor protein in WKY renal proximal tubule cells. In contrast, in SHR renal proximal tubule cells, BQ3020 (10(-8) mol/L/24 hours) no longer affected AT(1) or ETB receptor protein. AT(1)/ETB receptors colocalized and coimmunoprecipitated in WKY and SHRs. BQ3020 (10(-8) mol/L/15 minutes) treatment had no effect on AT(1)/ETB coimmunoprecipitation in WKY but decreased it in SHRs. BQ3020 (10(-8) mol/L/15 minutes) treatment increased AT(1) receptor phosphorylation in WKY, but decreased it in SHRs. CONCLUSION ETB receptors regulate AT(1) receptors by direct physical receptor interaction and receptor expression. An impaired ETB receptor regulation of the AT(1) receptor may participate in the pathogenesis of high blood pressure in the SHR.
Collapse
MESH Headings
- Animals
- Cell Line, Transformed
- Endothelins/pharmacology
- Hypertension, Renal/metabolism
- Immunoprecipitation
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Peptide Fragments/pharmacology
- Phosphorylation/drug effects
- Rats
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Endothelin B/metabolism
- Renin-Angiotensin System/physiology
Collapse
Affiliation(s)
- Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
29
|
Pulakat L, Rahman S, Gray A, Knowle D, Gavini N. Roles of the intracellular regions of angiotensin II receptor AT2 in mediating reduction of intracellular cGMP levels. Cell Signal 2005; 17:395-404. [PMID: 15567070 DOI: 10.1016/j.cellsig.2004.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2004] [Revised: 08/21/2004] [Accepted: 08/23/2004] [Indexed: 02/04/2023]
Abstract
We have shown previously that the angiotensin II (Ang II) receptor AT2 reduces the intracellular levels of cGMP in Xenopus oocytes when activated by ligand binding, and the C-terminal cytoplasmic tail of the AT2 acts as a negative regulator of this function. Here we report the effects of mutations in the 2nd and 3rd intracellular loops of AT2 on AT2-mediated cGMP reduction. Mutating the highly conserved DRY motif (D141G-R142G-Y143A) of the 2nd ICL implicated in activating G(alpha) subunit of trimeric G-proteins did not affect AT2-mediated cGMP reduction. Moreover, anti-Gialpha antibody or phosphodiesterase inhibitor IBMX did not inhibit AT2-mediated cGMP reduction, suggesting that Gialpha activation and subsequent phosphodiesterase activation are not involved in this function. In contrast, mutations T250R-R251N and L255F-K256R located in the C-terminus of the 3rd ICL of AT2 retained ligand-binding properties of the wild-type AT2, and its ability to interact with the ErbB3 in yeast two-hybrid assay, but abolished AT2-mediated cGMP reduction. Similarities in the roles of ICLs of AT2 in AT2-mediated cGMP reduction in oocytes, and AT2-mediated SHP1 activation in COS-7 cells, (need of 3rd ICL for both functions and lack of involvement of DRY motif), suggest that the cascade of events in these two signaling mechanisms could be similar, and that an oocyte-specific SHP1-like protein may be involved in AT2-mediated cGMP reduction in these cells.
Collapse
Affiliation(s)
- Lakshmi Pulakat
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA
| | | | | | | | | |
Collapse
|
30
|
Shah BH, Neithardt A, Chu DB, Shah FB, Catt KJ. Role of EGF receptor transactivation in phosphoinositide 3-kinase-dependent activation of MAP kinase by GPCRs. J Cell Physiol 2005; 206:47-57. [PMID: 15920762 DOI: 10.1002/jcp.20423] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many G protein coupled receptors (GPCRs) cause phosphorylation of MAP kinases through transactivation of the epidermal growth factor receptor (EGF-R), leading to increased cell survival and growth, motility, and migration. Phosphoinositide 3-kinase (PI3K) is one of the important cell survival signaling molecules activated by EGF-R stimulation. However, the extent to which EGF-R transactivation is essential for GPCR agonist-stimulated PI3K activation is not known. Here we examined the mechanism of PI3K activation that elicits GPCR-mediated ERK1/2 activation by pathways dependent and/or independent of EGF-R transactivation in specific cell types. Immortalized hypothalamic neurons (GT1-7 cells) express endogenous gonadotropin-releasing hormone receptors (GnRH-R) and their stimulation causes marked phosphorylation of ERK1/2 and Akt (Ser 473) through transactivation of the EGF-R and recruitment of PI3K. In C9 hepatocytes, agonist activation of AT1 angiotensin II (AT1-R), lysophosphatidic acid (LPA), and EGF receptors caused phosphorylation of Akt through activation of the EGF-R in a PI3K-dependent manner. However, ERK1/2 activation by these agonists in these cells was independent of PI3K activation. In contrast, agonist stimulation of HEK 293 cells stably expressing AT1-R caused ERK1/2 phosphorylation that was independent of EGF-R transactivation but required PI3K activation. LPA signaling in these cells showed partial and complete dependence on EGF-R and PI3K, respectively. These data indicate that GPCR-induced ERK1/2 phosphorylation is dependent or independent of PI3K in specific cell types, and that the involvement of PI3K during ERK1/2 activation is not dependent solely on agonist-induced transactivation of the EGF-R.
Collapse
Affiliation(s)
- Bukhtiar H Shah
- Endocrinology and Reproduction Research Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-4510, USA.
| | | | | | | | | |
Collapse
|
31
|
Olivares-Reyes JA, Shah BH, Hernández-Aranda J, García-Caballero A, Farshori MP, García-Sáinz JA, Catt KJ. Agonist-Induced Interactions between Angiotensin AT1and Epidermal Growth Factor Receptors. Mol Pharmacol 2005; 68:356-64. [PMID: 15905421 DOI: 10.1124/mol.104.010637] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In rat hepatic C9 cells, angiotensin II (Ang II)-induced activation of angiotensin type 1 (AT(1)) receptors (AT(1)-Rs) stimulates extracellular signal-regulated kinase (ERK) 1/2 phosphorylation via transactivation of the endogenous epidermal growth factor (EGF) receptor (EGF-R) by a protein kinase C (PKC) delta/Src/Pyk2-dependent pathway. This leads to phosphorylation of the EGF-R as well as its subsequent internalization. On the other hand, EGF-induced activation of the EGF-R in C9 cells was found to cause phosphorylation of the AT(1)-R. This was prevented by selective inhibition of the intrinsic tyrosine kinase activity of the EGF-R by AG1478 [4-(3'-chloroanilino)-6,7-dimethoxy-quinazoline] and was reduced by inhibition of PKC and phosphoinositide 3-kinase. EGF-induced AT(1)-R phosphorylation was associated with a decrease in membrane-associated AT(1)-Rs and a reduced inositol phosphate response to Ang II. Agonist activation of endogenous AT(1)-Rs and EGF-Rs induced the formation of a multireceptor complex containing both the AT(1)-R and the transactivated EGF-R. The dependence of these responses on caveolin was indicated by the finding that cholesterol depletion of C9 cells abolished Ang II-induced inositol phosphate production, activation of Akt/PKB and ERK1/2, and AT(1)-R internalization. Confocal microscopy demonstrated that caveolin-1 was endogenously phosphorylated and was distributed on the plasma membrane in patches that undergo redistribution during Ang II stimulation. Agonist-induced phosphorylation and association of caveolin 1 with the AT(1)-R was observed, consistent with a scaffolding role of caveolin during transactivation of the EGF-R by Ang II. The EGF-induced AT(1)-R/caveolin association was abolished by AG1478, suggesting that activation of the EGF-R promotes the association of caveolin and the AT(1)-R.
Collapse
Affiliation(s)
- J Alberto Olivares-Reyes
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, A. P. 14-740 México, 07000 D. F., México.
| | | | | | | | | | | | | |
Collapse
|
32
|
Dhami GK, Babwah AV, Sterne-Marr R, Ferguson SSG. Phosphorylation-independent regulation of metabotropic glutamate receptor 1 signaling requires g protein-coupled receptor kinase 2 binding to the second intracellular loop. J Biol Chem 2005; 280:24420-7. [PMID: 15870073 DOI: 10.1074/jbc.m501650200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Metabotropic glutamate receptors (mGluRs) are members of a unique class of G protein-coupled receptors (class III) that include the calcium-sensing and gamma-aminobutyric acid type B receptors. The activity of mGluRs is regulated by second messenger-dependent protein kinases and G protein-coupled receptor kinases (GRKs). The attenuation of both mGluR1a and mGluR1b signaling by GRK2 is phosphorylation- and beta-arrestin-independent and requires the concomitant association of GRK2 with both the receptor and Galpha(q/11). G protein interactions are mediated, in part, by the mGluR1 intracellular second loop, but the domains required for GRK2 binding are unknown. In the present study, we showed that GRK2 binds to the second intracellular loop of mGluR1a and mGluR1b and also to the mGluR1a carboxyl-terminal tail. Alanine scanning mutagenesis revealed a discrete domain within loop 2 that contributes to GRK2 binding, and the mutation of either lysine 691 or 692 to an alanine within this domain resulted in a loss of GRK2 binding to both mGluR1a and mGluR1b. Mutation of either Lys(691) or Lys(692) prevented GRK2-mediated attenuation of mGluR1b signaling, whereas the mutation of only Lys(692) prevented GRK2-mediated inhibition of mGluR1a signaling. Thus, the mGluR1a carboxyl-terminal tail may also be involved in regulating the signaling of the mGluR1a splice variant. Taken together, our findings indicated that kinase binding to an mGluR1 domain involved in G protein-coupling is essential for the phosphorylation-independent attenuation of signaling by GRK2.
Collapse
Affiliation(s)
- Gurpreet K Dhami
- Cell Biology Research Group, Robarts Research Laboratory and Department of Physiology and Pharmacology, The University of Western Ontario, 100 Perth Drive, London, Ontario N6A 5K8, Canada
| | | | | | | |
Collapse
|
33
|
Bivol LM, Vågnes OB, Iversen BM. The renal vascular response to ANG II injection is reduced in the nonclipped kidney of two-kidney, one-clip hypertension. Am J Physiol Renal Physiol 2005; 289:F393-400. [PMID: 15784843 DOI: 10.1152/ajprenal.00319.2004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ANG II receptor 1 (AT(1)R) level in the nonclipped kidney of two-kidney, one-clip hypertension (2K1C) has shown to be unchanged despite a high circulating angiotensin (ANG) II level. To examine the vasoreactive response to ANG II in this kidney, injections of ANG II into renal artery were performed 6 wk after clipping of the kidney and compared with normotensive controls. The renal blood flow (RBF) response to 2.5 ng ANG II was measured by a Transonic transit-time flowmeter, before and after indomethacin and candesartan treatment, and analyzed by a computer program. The RBF response to 5 ng arginine-vasopressin (AVP) was examined for comparison with ANG II. The mRNA for AT(1A) and AT(1B) as well as Western blotting for AT(1)R in renal resistance vessels were determined, and plasma renin activity (PRA) was measured. Systolic blood pressure was 183 +/- 4 mmHg in 2K1C rats compared with 113 +/- 1 mmHg in controls (P < 0.001). PRA was significantly increased in 2K1C animals (P < 0.05). Injection of ANG II reduced RBF with 10 +/- 2% in the nonclipped kidney and 24 +/- 3% in controls (P < 0.001). After indomethacin, the RBF response increased from 10 +/- 2 to 20 +/- 3% (P < 0.02) in 2K1C rats and from 24 +/- 3 to 34 +/- 6% in controls (P < 0.01). The doses of candesartan needed to completely inhibit RBF response to ANG II were 30 microg/kg in the nonclipped kidney and 100 microg/kg in controls (P < 0.001). Western blotting and mRNA for AT(1A) and AT(1B) in the nonclipped kidney were similar to the controls. The results indicate that despite no difference in total AT(1)R levels, functional AT(1)R is downregulated in the nonclipped kidney of 2K1C rats.
Collapse
|
34
|
Dautzenberg FM, Gutknecht E, Van der Linden I, Olivares-Reyes JA, Dürrenberger F, Hauger RL. Cell-type specific calcium signaling by corticotropin-releasing factor type 1 (CRF1) and 2a (CRF2(a)) receptors: phospholipase C-mediated responses in human embryonic kidney 293 but not SK-N-MC neuroblastoma cells. Biochem Pharmacol 2004; 68:1833-44. [PMID: 15450949 DOI: 10.1016/j.bcp.2004.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2004] [Accepted: 07/12/2004] [Indexed: 11/25/2022]
Abstract
The human corticotropin-releasing factor (hCRF) receptors CRF1 and CRF2(a) couple to the Gs protein. It has been postulated that CRF receptors may also signal through phospholipase C (PLC). To test this hypothesis, binding and signaling properties were determined for both receptor subtypes stably expressed in human embryonic kidney 293 (HEK293) and human SK-N-MC neuroblastoma cells. CRF receptors were highly expressed and strongly coupled to Gs in HEK293 and SK-N-MC cells. However, when the calcium mobilization pathway was investigated, marked differences were observed. In SK-N-MC cells, neither CRF receptor stimulated calcium mobilization in the fluorometric imaging plate reader (FLIPR) assay, whereas activation of orexin type 1 and 2 receptors stably expressed in SK-N-MC cells revealed robust calcium responses. In contrast, intracellular calcium was strongly mobilized by agonist stimulation of hCRF1 and hCRF2(a) receptors in HEK293 cells. In HEK293 cells, potency rank orders for calcium and cAMP responses were identical for both receptors, despite a rightward shift of the dose-response curves. Complete inhibition of calcium signaling of both hCRF1 and hCRF2(a) receptors was observed in the presence of the PLC inhibitor U-73,122 whereas ryanodine, an inhibitor of calcium release channels and the protein kinase A inhibitor Rp-cAMPS were ineffective. Finally, CRF agonists produced a small but significant stimulation of inositol 1,4,5-triphosphate (IP3) accumulation in hCRF1-and hCRF2(a)-transfected HEK293 cells. These data clearly show that phospholipase C-mediated signaling of CRF receptors is dependent upon the cellular background and that in HEK293 cells human CRF receptors robustly respond in the FLIPR format.
Collapse
Affiliation(s)
- Frank M Dautzenberg
- Johnson and Johnson Research and Development, CNS Research, Turnhoutseweg 30, Beerse, Belgium.
| | | | | | | | | | | |
Collapse
|
35
|
Pulakat L, Mandavia CH, Gavini N. Role of Phe308 in the seventh transmembrane domain of the AT2 receptor in ligand binding and signaling. Biochem Biophys Res Commun 2004; 319:1138-43. [PMID: 15194486 DOI: 10.1016/j.bbrc.2004.05.092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2004] [Indexed: 11/28/2022]
Abstract
Studies on Angiotensin II (Ang II) receptor type AT1 have suggested that interaction between the two highly conserved residues, Tyr292 in the 7th transmembrane domain (TMD) and the Asp74 in the 2nd TMD, is critical for linking the Ang II binding and AT1 receptor-Gq protein coupling. In the Ang II receptor type AT2, the Asp is conserved (Asp90 in 2nd TMD), however, there is no Tyr residue in the 7th TMD and Phe308 occupies the analogous position to Tyr292 of the AT1. Replacing this Phe308 with Ala reduced receptor affinity to peptidic ligands (125)I-Ang II (K(d) = 0.37 nM) and (125)I-CGP42112A (K(d) = 0.56 nM), but retained the ability of the AT2 to reduce cGMP levels in Xenopus oocytes. Thus, the Phe308 of the AT2 does not mimic the role of Tyr292 of the AT1 in the receptor activation upon Ang II binding. We have also shown that the M8 mutant of the AT2 with the 7th TMD similar to that of wild type AT2 can couple to PLC like the AT1 and bind the AT2-specific ligands with high affinity. Since the Ang II is shown to bind to both the AT1 and the AT2 in an identical manner, we propose that the absence of Tyr in the 7th TMD of the AT2 does not prevent the receptor from coupling to Gq-protein, rather may contribute to the freedom of the AT2 to couple to trimeric G-proteins in both G- betagamma dependent and independent manners upon Ang II binding.
Collapse
Affiliation(s)
- Lakshmi Pulakat
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA.
| | | | | |
Collapse
|
36
|
Zemkova H, Balik A, Kretschmannova K, Mazna P, Stojilkovic SS. Recovery of Ins(1,4,5)-trisphosphate-dependent calcium signaling in neonatal gonadotrophs. Cell Calcium 2004; 36:89-97. [PMID: 15193857 DOI: 10.1016/j.ceca.2003.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 12/16/2003] [Accepted: 12/17/2003] [Indexed: 11/21/2022]
Abstract
Pituitary gonadotrophs express non-desensitizing gonadotropin-releasing hormone (GnRH) receptors and their activations leads to inositol 1,4,5-trisphosphate (InsP3)-dependent Ca2+ mobilization. When added in physiological concentration range GnRH induces baseline Ca2+ oscillations, whereas in higher concentrations it induces a prolonged spike response accompanied with non-oscillatory or oscillatory plateau response. Here, we studied the recovery of calcium signaling during repetitive stimulation with short (10-30 s) GnRH pulses and variable interpulse intervals in neonatal gonadotrophs perfused with Ca2+/Na+ -containing, Ca2+ -deficient/Na+ -containing, and Ca2+ -containing/Na+ -deficient media. In Ca2+/Na+ -containing medium, baseline Ca2+ oscillations recovered without refractory period and with a time constant of approximately 20 s, whereas the recovery of spike response occurred after 25-35 s refractory period and with a time constant of approximately 30 s. During repetitive GnRH stimulation, removal of Ca2+ had only a minor effect on baseline oscillations but abolished spike response, whereas removal of Na+ slightly extended duration of baseline oscillations and considerably prolonged spike response. These results indicate that two calcium handling mechanisms are operative in gonadotrophs: redistribution of calcium within InsP3-sensitive and -insensitive pools and a sodium-dependent calcium efflux followed by calcium influx. Redistribution of Ca2+ within the cell leads to rapid recovery of InsP3-dependent pool, whereas the Na+ -dependent Ca2+ efflux pathway is activated by spike response and limits the time of exposure to elevated cytosolic Ca2+ concentrations.
Collapse
Affiliation(s)
- Hana Zemkova
- Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4.
| | | | | | | | | |
Collapse
|
37
|
Santos EL, Pesquero JB, Oliveira L, Paiva ACM, Costa-Neto CM. Mutagenesis of the AT1 receptor reveals different binding modes of angiotensin II and [Sar1]-angiotensin II. ACTA ACUST UNITED AC 2004; 119:183-8. [PMID: 15120479 DOI: 10.1016/j.regpep.2004.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2003] [Revised: 01/28/2004] [Accepted: 02/04/2004] [Indexed: 11/16/2022]
Abstract
Homology modeling of the structure of the AT1 receptor, based on the high resolution rhodopsin crystal structure, indicated that it is unlikely that the binding of AngII to AT1 involves simultaneously all the receptor's residues reported in the literature to participate in this process. Site-directed mutagenesis using Ala substitution of charged residues Lys20, Arg23, Glu91 and Arg93 was performed to evaluate the participation of their side-chains in ligand binding and in triggering the cell's response. A comparative analysis by competition binding and functional assays using angiotensin II and the analog [Sar1]-angiotensin II suggests an important role for Arg23 of AT1 receptor in binding of the natural agonist. It is discussed whether some receptor's residues participate directly in the binding with AngII or whether they are part of a regulatory site.
Collapse
Affiliation(s)
- Edson L Santos
- Department of Biophysics, Escola Paulista de Medicina, UNIFESP, São Paulo 04023-062, Brazil
| | | | | | | | | |
Collapse
|
38
|
Hansen JL, Theilade J, Haunsø S, Sheikh SP. Oligomerization of Wild Type and Nonfunctional Mutant Angiotensin II Type I Receptors Inhibits Gαq Protein Signaling but Not ERK Activation. J Biol Chem 2004; 279:24108-15. [PMID: 15056658 DOI: 10.1074/jbc.m400092200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The 7-transmembrane or G protein-coupled receptors relay signals from hormones and sensory stimuli to multiple signaling systems at the intracellular face of the plasma membrane including heterotrimeric G proteins, ERK1/2, and arrestins. It is an emerging concept that 7-transmembrane receptors form oligomers; however, it is not well understood which roles oligomerization plays in receptor activation of different signaling systems. To begin to address this question, we used the angiotensin II type 1 (AT(1)) receptor, a key regulator of blood pressure and fluid homeostasis that in specific context has been described to activate ERKs without activating G proteins. By using bioluminescence resonance energy transfer, we demonstrate that AT(1) receptors exist as oligomers in transfected COS-7 cells. AT(1) oligomerization was both constitutive and receptor-specific as neither agonist, antagonist, nor co-expression with three other receptors affected the bioluminescence resonance energy transfer 2 signal. Furthermore, the oligomerization occurs early in biosynthesis before surface expression, because we could control AT(1) receptor export from the endoplasmic reticulum or Golgi by using regulated secretion/aggregation technology (RPD trade mark ). Co-expression studies of wild type AT(1) and AT(1) receptor mutants, defective in either ligand binding or G protein and ERK activation, yielded an interesting result. The mutant receptors specifically exerted a dominant negative effect on Galpha(q) activation, whereas ERK activation was preserved. These data suggest that distinctly active conformations of AT(1) oligomers can couple to each of these signaling systems and imply that oligomerization plays an active role in supporting these distinctly active conformations of AT(1) receptors.
Collapse
Affiliation(s)
- Jakob Lerche Hansen
- Laboratory of Molecular Cardiology, the Heart Centre and Copenhagen Heart Arrhythmia Research Centre, Copenhagen University Hospital Section 9312 and the Faculty of Health, University of Copenhagen, 20 Juliane Mariesvej, Copenhagen DK-2100, Denmark
| | | | | | | |
Collapse
|
39
|
Spät A, Hunyady L. Control of aldosterone secretion: a model for convergence in cellular signaling pathways. Physiol Rev 2004; 84:489-539. [PMID: 15044681 DOI: 10.1152/physrev.00030.2003] [Citation(s) in RCA: 333] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aldosterone secretion by glomerulosa cells is stimulated by angiotensin II (ANG II), extracellular K(+), corticotrophin, and several paracrine factors. Electrophysiological, fluorimetric, and molecular biological techniques have significantly clarified the molecular action of these stimuli. The steroidogenic effect of corticotrophin is mediated by adenylyl cyclase, whereas potassium activates voltage-operated Ca(2+) channels. ANG II, bound to AT(1) receptors, acts through the inositol 1,4,5-trisphosphate (IP(3))-Ca(2+)/calmodulin system. All three types of IP(3) receptors are coexpressed, rendering a complex control of Ca(2+) release possible. Ca(2+) release is followed by both capacitative and voltage-activated Ca(2+) influx. ANG II inhibits the background K(+) channel TASK and Na(+)-K(+)-ATPase, and the ensuing depolarization activates T-type (Ca(v)3.2) Ca(2+) channels. Activation of protein kinase C by diacylglycerol (DAG) inhibits aldosterone production, whereas the arachidonate released from DAG in ANG II-stimulated cells is converted by lipoxygenase to 12-hydroxyeicosatetraenoic acid, which may also induce Ca(2+) signaling. Feedback effects and cross-talk of signal-transducing pathways sensitize glomerulosa cells to low-intensity stimuli, such as physiological elevations of [K(+)] (< or =1 mM), ANG II, and ACTH. Ca(2+) signaling is also modified by cell swelling, as well as receptor desensitization, resensitization, and downregulation. Long-term regulation of glomerulosa cells involves cell growth and proliferation and induction of steroidogenic enzymes. Ca(2+), receptor, and nonreceptor tyrosine kinases and mitogen-activated kinases participate in these processes. Ca(2+)- and cAMP-dependent phosphorylation induce the transfer of the steroid precursor cholesterol from the cytoplasm to the inner mitochondrial membrane. Ca(2+) signaling, transferred into the mitochondria, stimulates the reduction of pyridine nucleotides.
Collapse
Affiliation(s)
- András Spät
- Dept. of Physiology, Semmelweis University, Faculty of Medicine, PO Box 259, H-1444 Budapest, Hungary.
| | | |
Collapse
|
40
|
Yu J, Liu B, Eramian D, Mierke D, Taylor L, Polgar P. K317, R319, and E320 within the proximal C-terminus of the bradykinin B2 receptor form a motif important for phospholipase C and phospholipase A2 but not connective tissue growth factor related signaling. J Cell Biochem 2004; 92:547-59. [PMID: 15156566 DOI: 10.1002/jcb.20075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We showed previously that large domain exchanges between the bradykinin B2 (BKB2) and angiotensin II type 1a (AT1a) receptors can result in functional hybrids. However, when we proceeded to exchange the entire bradykinin B2 receptor (BKB2R) C-terminal tail with the AT1aR C-terminus, the hybrid, while continuing to bind BK and be endocytosed as wild type (WT) BKB2R, lost much of its ability to activate phosphatidylinositol (PI) turnover or the release of arachidonic acid (ARA). In this study, we constructed chimeric receptors within the proximal C-terminus between the BKB2R and AT1aR or bradykinin B1 receptor (BKB1R). The mutant and WT receptor cDNAs were stably transfected into Rat-1 cells. Also, point mutations were generated to evaluate the role of the individual residues within this region. These chimeric studies revealed that the proximal portion of the BKB2R C-tail is crucial for G protein-linked BKB2R functions. This region could not be swapped with the AT1aR to obtain a BK activated PI turnover or ARA release. Further studies demonstrated that the distal portion (325-330) of this region is exchangeable; however, the middle portion (317-324) is not. Small motif exchanges within this section identified the KSR and EVY motifs as crucial for G(alphaq), G(alphai) related signaling of the BKB2R. Point mutations then showed that the charged amino acids K317, R319, and E320 are the residues critical for linking to PI turnover and ARA release. However, these proximal chimeras showed normal receptor uptake. Interestingly, while apparently not activating G protein-linked signaling, the proximal tail AT1aR exchange mutant and the entire C-terminus exchange hybrid continued to cause a substantial bradykinin effected increase in connective tissue growth factor (CTGF) mRNA level, as WT BKB2R.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- Arachidonic Acid/metabolism
- Arginine/genetics
- Arginine/metabolism
- Connective Tissue Growth Factor
- Endocytosis
- Glutamic Acid/genetics
- Glutamic Acid/metabolism
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Lysine/genetics
- Lysine/metabolism
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis
- Phosphatidylinositols/metabolism
- Phospholipases A/metabolism
- Phospholipases A2
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Receptor, Angiotensin, Type 1/chemistry
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Bradykinin B1/chemistry
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptor, Bradykinin B2/chemistry
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Signal Transduction
- Structure-Activity Relationship
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Jun Yu
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | |
Collapse
|
41
|
Hasan Z, Shah BH, Mahmood A, Young DB, Hussain R. The effect of mycobacterial virulence and viability on MAP kinase signalling and TNF alpha production by human monocytes. Tuberculosis (Edinb) 2003; 83:299-309. [PMID: 12972343 DOI: 10.1016/s1472-9792(03)00003-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
SETTING The success of Mycobacterium tuberculosis as a human pathogen depends on its ability to tolerate and perhaps manipulate host defense mechanisms. OBJECTIVE To determine the induction of tumour necrosis factor-alpha (TNF alpha), a central mediator of immunity, by human monocytes infected with virulent M. tuberculosis, M. leprae and attenuated M. bovis BCG. DESIGN Mycobacteria-induced cellular activation pathways of TNF alpha production was investigated using an inhibitor of protein tyrosine kinase (PTKs) and an inhibitor of mitogen-activated protein (MAP) kinases. RESULTS TNF alpha production was significantly lower during infection with virulent M. tuberculosis than with BCG and this differential response was independent of mycobacterial viability. TNF alpha production involved the PTK and MAP kinase pathways. Reduced TNF alpha induction by M. tuberculosis was associated with a reduction in the extent and duration of phosphorylation of extracellular-signal regulated kinases (ERK 1/2). Infection with M. leprae triggered low and transient ERK 1/2 activation as well as low TNF alpha production. CONCLUSION Maintenance of the differential response in both live and heat-killed preparations suggests that the reduced TNF alpha response associated with virulent mycobacteria is due to differences in the presence of components capable of triggering host pattern recognition receptors, rather than events associated with phagosome trafficking or the active release of intracellular modulators.
Collapse
Affiliation(s)
- Z Hasan
- Department of Microbiology, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Pakistan.
| | | | | | | | | |
Collapse
|
42
|
Qiu Y, Law PY, Loh HH. Mu-opioid receptor desensitization: role of receptor phosphorylation, internalization, and representation. J Biol Chem 2003; 278:36733-9. [PMID: 12860981 DOI: 10.1074/jbc.m305857200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
It is generally accepted that the internalization and desensitization of mu-opioid receptor (MOR) involves receptor phosphorylation and beta-arrestin recruitment. However, a mutant MOR, which is truncated after the amino acid residue Ser363 (MOR363D), was found to undergo phosphorylation-independent internalization and desensitization. As expected, MOR363D, missing the putative agonist-induced phosphorylation sites, did not exhibit detectable agonist-induced phosphorylation. MOR363D underwent slower internalization as reflected in the attenuation of membrane translocation of beta-arrestin 2 when compared with wild type MOR, but the level of receptor being internalized was similar to that of wild type MOR after 4 h of etorphine treatment. Furthermore, MOR363D was observed to desensitize faster than that of wild type MOR upon agonist activation. Surface biotinylation assay demonstrated that the wild type receptors recycled back to membrane after agonist-induced internalization, which contributed to the receptor resensitization and thus partially reversed the receptor desensitization. On the contrary, MOR363D did not recycle after internalization. Hence, MOR desensitization is controlled by the receptor internalization and the recycling of internalized receptor to cell surface in an active state. Taken together, our data indicated that receptor phosphorylation is not absolutely required in the internalization, but receptor phosphorylation and subsequent beta-arrestin recruitment play important roles in the resensitization of internalized receptors.
Collapse
Affiliation(s)
- Yu Qiu
- Department of Pharmacology, Medical School, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | |
Collapse
|
43
|
Hauger RL, Olivares-Reyes JA, Braun S, Catt KJ, Dautzenberg FM. Mediation of corticotropin releasing factor type 1 receptor phosphorylation and desensitization by protein kinase C: a possible role in stress adaptation. J Pharmacol Exp Ther 2003; 306:794-803. [PMID: 12734388 DOI: 10.1124/jpet.103.050088] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Protein kinase C (PKC)-mediated desensitization of the corticotropin releasing factor type 1 (CRF1) receptor was investigated in human retinoblastoma Y79 and transfected COS-7 cells. Because stimulation of Y79 cells with CRF resulted in large ( approximately 30-fold) increases in intracellular cAMP accumulation without changing inositol phosphate levels, the CRF1 receptor expressed in retinoblastoma cells couples to Gs, but not to Gq, and predominantly signals via the protein kinase A cascade. Direct activation of PKC by treatment with the phorbol ester phorbol 12-myristate 13-acetate (PMA) or 1,2-dioctanoyl-sn-glycerol (DOG) desensitized CRF1 receptors in Y79 cells, reducing the maximum for CRF- (but not forskolin)-stimulated cAMP accumulation by 56.3 +/- 1.2% and 40.4 +/- 2.1%, respectively (p < 0.001). Pretreating Y79 cells with the PKC inhibitor bisindolylmaleimide I (BIM) markedly inhibited PMA's desensitizing action on CRF-stimulated cAMP accumulation, but did not affect homologous CRF1 receptor desensitization. Retinoblastoma cells were found to express PKCalpha, betaI, betaII, delta, lambda, and RACK1. When alpha and beta isoforms of PKC were down-regulated 80 to 90% by a 48-h PMA exposure, PMA-induced CRF1 receptor desensitization was abolished. In transfected COS-7 cells the magnitude of CRF1 receptor phosphorylation after a 5-min exposure to PMA was 2.32 +/- 0.21-fold greater compared with the basal level. Pretreating COS-7 cells with BIM abolished PMA-induced CRF1 receptor phosphorylation. These studies demonstrate that protein kinase C (possibly alpha and beta isoforms) has an important role in the phosphorylation and heterologous desensitization of the CRF1 receptor.
Collapse
Affiliation(s)
- Richard L Hauger
- Department of Veternas Affairs Healthcare System and Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093-0603, USA.
| | | | | | | | | |
Collapse
|
44
|
Abstract
ATP is a key extracellular messenger that mediates the propagation of Ca 2+ waves in astrocyte networks in various regions of the CNS. ATP-mediated Ca 2+ signals play critical roles in astrocyte proliferation and differentiation and in modulating neuronal activity. The actions of ATP on astrocytes are via two distinct subtypes of P2Y purinoceptors, P2Y1 and P2Y2 receptors (P2Y1Rs and P2Y2Rs), G-protein coupled receptors that stimulate mobilization of intracellular Ca 2+ ([Ca 2+]i) via the phospholipase Cbeta-IP3 pathway. We report here that P2Y1R-mediated and P2Y2R-mediated Ca 2+ responses differentially show two forms of activity-dependent negative feedback. First, Ca 2+ responses mediated by either receptor exhibit slow depression that is independent of stimulation frequency. Second, responses mediated by P2Y1Rs, but not those mediated by P2Y2Rs, show rapid oscillations after high-frequency stimulation. We demonstrate that the oscillations are mediated by recruiting negative feedback by protein kinase C, and we map the site responsible for the effect of protein kinase C to Thr339 in the C terminus of P2Y1R. We propose that frequency-dependent changes in ATP-mediated Ca 2+ signaling pathways may modulate astrocyte function and astrocyte-neuron signaling in the CNS.
Collapse
|
45
|
Seta K, Sadoshima J. Phosphorylation of tyrosine 319 of the angiotensin II type 1 receptor mediates angiotensin II-induced trans-activation of the epidermal growth factor receptor. J Biol Chem 2003; 278:9019-26. [PMID: 12522132 DOI: 10.1074/jbc.m208017200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although tyrosine kinases are critically involved in the angiotensin II (Ang II) type 1 (AT1) receptor signaling, how AT1 receptors activate tyrosine kinases is not fully understood. We examined the structural requirements of the AT1 receptor for transactivation of the epidermal growth factor (EGF) receptor (EGFR). Studies using carboxyl terminal-truncated AT1 receptors indicated that the amino acid sequence between 312 and 337 is required for activation of EGFR. The role of the conserved YIPP motif in this sequence in transactivation of EGFR was investigated by mutating tyrosine 319. Ang II failed to activate EGFR in cells expressing AT1-Y319F, whereas EGFR was activated even without Ang II in cells expressing AT1-Y319E, which mimics the AT1 receptor phosphorylated at Tyr-319. Immunoblot analyses using anti-phospho Tyr-319-specific antibody showed that Ang II increased phosphorylation of Tyr-319. EGFR interacted with the AT1 receptor but not with AT1-Y319F in response to Ang II stimulation, whereas the EGFR-AT1 receptor interaction was inhibited in the presence of dominant negative SHP-2. The requirement of Tyr-319 seems specific for EGFR because Ang II-induced activation of other tyrosine kinases, including Src and JAK2, was preserved in cells expressing AT1-Y319F. Extracellular signal-regulated kinase activation was also maintained in AT1-Y319F through activation of Src. Overexpression of wild type AT1 receptor in cardiac fibroblasts enhanced Ang II-induced proliferation. By contrast, overexpression of AT1-Y319F failed to enhance cell proliferation. In summary, Tyr-319 of the AT1 receptor is phosphorylated in response to Ang II and plays a key role in mediating Ang II-induced transactivation of EGFR and cell proliferation, possibly through its interaction with SHP-2 and EGFR.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Animals
- COS Cells
- Calcium/metabolism
- Cell Division
- Cells, Cultured
- Conserved Sequence
- DNA, Complementary/metabolism
- ErbB Receptors/metabolism
- Fibroblasts/metabolism
- Genes, Dominant
- Humans
- Immunoblotting
- Intracellular Signaling Peptides and Proteins
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Myocardium/cytology
- Phosphorylation
- Plasmids/metabolism
- Precipitin Tests
- Protein Binding
- Protein Structure, Tertiary
- Protein Tyrosine Phosphatase, Non-Receptor Type 1
- Protein Tyrosine Phosphatase, Non-Receptor Type 11
- Protein Tyrosine Phosphatases/metabolism
- Protein-Tyrosine Kinases/metabolism
- Rats
- Rats, Wistar
- Receptor, Angiotensin, Type 1
- Receptors, Angiotensin/chemistry
- Receptors, Angiotensin/metabolism
- Sequence Homology, Amino Acid
- Time Factors
- Transcriptional Activation
- Transfection
- Tyrosine/chemistry
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Koichi Seta
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | |
Collapse
|
46
|
Kumar V, Knowle D, Gavini N, Pulakat L. Identification of the region of AT2 receptor needed for inhibition of the AT1 receptor-mediated inositol 1,4,5-triphosphate generation. FEBS Lett 2002; 532:379-86. [PMID: 12482596 DOI: 10.1016/s0014-5793(02)03713-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Increase in the intracellular inositol triphosphate (IP3) levels in Xenopus oocytes in response to expression and activation of rat angiotensin II (Ang II) receptor AT1 was inhibited by co-expression of rat AT2 receptor. To identify which region of the AT2 was involved in this inhibition, ability of three AT2 mutants to abolish this inhibition was analyzed. Deletion of the C-terminus of the AT2 did not abolish this inhibition. Replacing Ile249 in the third intracellular loop (3rd ICL) of the AT2 with proline, corresponding amino acid in the AT1, in the mutant M6, resulted in slightly reduced affinity to [125I]Ang II (K(d)=0.259 nM), however, did not abolish the inhibition. In contrast, replacing eight more amino acids in the 3rd ICL of the AT2 (at positions 241-244, 250-251 and 255-256) with that of the AT1 in the mutant M8, not only increased the affinity of the AT2 receptor to [125I]Ang II (K(d)=0.038 nM) but also abolished AT2-mediated inhibition. Interestingly, activation of the M8 by Ang II binding also resulted in increase in the intracellular IP(3) levels in oocytes. These results imply that the region of the 3rd ICL of AT2 spanning amino acids 241-256 is sufficient for the AT2-mediated inhibition of AT1-stimulated IP3 generation. Moreover, these nine mutations are also sufficient to render the AT2 with the ability to activate phospholipase C.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cytoplasm/metabolism
- DNA Mutational Analysis
- Enzyme Activation
- Inositol 1,4,5-Trisphosphate/metabolism
- Isoleucine/chemistry
- Kinetics
- Ligands
- Molecular Sequence Data
- Mutation
- Oocytes/metabolism
- Protein Binding
- Protein Structure, Tertiary
- RNA, Complementary/metabolism
- Rats
- Receptor, Angiotensin, Type 1
- Receptor, Angiotensin, Type 2
- Receptors, Angiotensin/chemistry
- Receptors, Angiotensin/metabolism
- Sequence Homology, Amino Acid
- Transcription, Genetic
- Type C Phospholipases/metabolism
- Xenopus
Collapse
Affiliation(s)
- Vikas Kumar
- Department of Biological Sciences, Bowling Green State University, Bowling Green, OH 43403, USA
| | | | | | | |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The concept that angiotensin II plays a central role in early atherogenesis, progression to atherosclerotic plaque, and the most serious clinical sequelae of coronary artery disease is the subject of considerable current interest. Results from recent large clinical trials confirm that blunting of the renin-angiotensin system through either angiotensin converting enzyme inhibition or angiotensin II type 1 receptor blockade incurs significant beneficial outcomes in patients with coronary artery disease. The exact mechanisms for these effects are not yet clear, but are suggested by studies demonstrating that suppression of the renin-angiotensin system is associated with muted vascular oxidative stress. RECENT FINDINGS As most of the biological effects of the renin-angiotensin system occur through stimulation of the angiotensin II type 1 receptor, the focus of this review is on changes in the vascular wall mediated by this receptor and primarily related to endothelial and vascular smooth muscle cells, monocyte/macrophages and platelets. The interactions between angiotensin II and nitric oxide exert particular demands on the vascular capacity to adapt to dyslipidemia, hypertension, estrogen deficiency and diabetes mellitus that appear to exacerbate atherogenesis. Associated with each of these conditions is angiotensin II-mediated stimulation of macrophages, platelet aggregation, plasminogen activator inhibitor 1, endothelial dysfunction, vascular smooth muscle cell proliferation and migration, apoptosis, leukocyte recruitment, fibrogenesis and thrombosis. SUMMARY Inhibition of the actions of angiotensin II serves a dual purpose: indirectly through reduction of mechanical stress on the vascular wall, and directly by diminished stimulation for vascular restructuring and remodeling. Collectively, data from studies published over the last year confirm and extend the notion that angiotensin II is a true cytokine prevalent at all stages of atherogenesis.
Collapse
Affiliation(s)
- William B Strawn
- Hypertension and Vascular Disease Center, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
48
|
Dhami GK, Anborgh PH, Dale LB, Sterne-Marr R, Ferguson SSG. Phosphorylation-independent regulation of metabotropic glutamate receptor signaling by G protein-coupled receptor kinase 2. J Biol Chem 2002; 277:25266-72. [PMID: 12101219 DOI: 10.1074/jbc.m203593200] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The accepted paradigm for G protein-coupled receptor kinase (GRK)-mediated desensitization of G protein-coupled receptors involves GRK-mediated receptor phosphorylation followed by the binding of arrestin proteins. Although GRKs contribute to metabotropic glutamate receptor 1 (mGluR1) inactivation, beta-arrestins do not appear to be required for mGluR1 G protein uncoupling. Therefore, we investigated whether the phosphorylation of serine and threonine residues localized within the C terminus of mGluR1a is sufficient to allow GRK2-mediated attenuation of mGluR1a signaling. We find that the truncation of the mGluR1a C-terminal tail prevents mGluR1a phosphorylation and that GRK2 does not contribute to the phosphorylation of an mGluR1 splice variant (mGluR1b). However, mGluR1a-866Delta- and mGluR1b-stimulated inositol phosphate formation is attenuated following GRK2 expression. The expression of the GRK2 C-terminal domain to block membrane translocation of endogenous GRK2 increases mGluR1a-866Delta- and mGluR1b-stimulated inositol phosphate formation, presumably by blocking membrane translocation of GRK2. In contrast, expression of the kinase-deficient GRK2-K220R mutant inhibits inositol phosphate formation by these unphosphorylated receptors. Expression of the GRK2 N-terminal domain (residues 45-185) also attenuates both constitutive and agonist-stimulated mGluR1a, mGluR1a-866Delta, and mGluR1b signaling, and the GRK2 N terminus co-precipitates with mGluR1a. Taken together, our observations indicate that attenuation of mGluR1 signaling by GRK2 is phosphorylation-independent and that the interaction of the N-terminal domain of GRK2 with mGluR1 contributes to the regulation of mGluR1 G protein coupling.
Collapse
Affiliation(s)
- Gurpreet Kaur Dhami
- Cell Biology Research Group, the John P. Robarts Research Institute, Department of Pharmacology and Toxicology, University of Western Ontario, Canada
| | | | | | | | | |
Collapse
|
49
|
Mason JN, Kozell LB, Neve KA. Regulation of dopamine D(1) receptor trafficking by protein kinase A-dependent phosphorylation. Mol Pharmacol 2002; 61:806-16. [PMID: 11901220 DOI: 10.1124/mol.61.4.806] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to use pharmacological inhibition of protein kinase A and mutation of potential protein kinase A phosphorylation sites to determine the role of protein kinase A-catalyzed phosphorylation of the dopamine D(1) receptor in agonist-stimulated desensitization and internalization of the receptor. To facilitate purification and imaging of the D(1) receptor, we attached a polyhistidine tag to the amino terminus and enhanced green fluorescent protein to the carboxyl terminus of the receptor (D(1)-EGFP). D(1)-EGFP was similar to the untagged D(1) receptor in terms of affinity for agonist and antagonist ligands, coupling to G proteins, and stimulation of cyclic AMP accumulation. D(1)-EGFP and two mutants in which either Thr268 or Ser380 was replaced with Ala were stably expressed in NS20Y neuroblastoma cells. Pretreatment with the protein kinase A inhibitor H-89 (N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide) or substitution of Ala for Thr268 reduced agonist-stimulated phosphorylation of the receptor and resulted in diminished trafficking of the receptor to the perinuclear region of the cell. Substitution of Ala for Thr268 had no effect, however, on agonist-induced receptor sequestration or desensitization of cyclic AMP accumulation. Substitution of Ala for Ser380 had no effect on D(1) receptor phosphorylation, sequestration, desensitization, or trafficking to the perinuclear region. We conclude that protein kinase A-dependent phosphorylation of the D(1) receptor on Thr268 regulates a late step in the sorting of the receptor to the perinuclear region of the cell, but that phosphorylation of Thr268 is not required for receptor sequestration or maximal desensitization of cyclic AMP accumulation.
Collapse
Affiliation(s)
- John N Mason
- Medical Research Service, Veterans Affairs Medical Center, Oregon Health and Science University, Portland, Oregon 97201, USA
| | | | | |
Collapse
|