1
|
Yang G, Khan A, Liang W, Xiong Z, Stegbauer J. Aortic aneurysm: pathophysiology and therapeutic options. MedComm (Beijing) 2024; 5:e703. [PMID: 39247619 PMCID: PMC11380051 DOI: 10.1002/mco2.703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Aortic aneurysm (AA) is an aortic disease with a high mortality rate, and other than surgery no effective preventive or therapeutic treatment have been developed. The renin-angiotensin system (RAS) is an important endocrine system that regulates vascular health. The ACE2/Ang-(1-7)/MasR axis can antagonize the adverse effects of the activation of the ACE/Ang II/AT1R axis on vascular dysfunction, atherosclerosis, and the development of aneurysms, thus providing an important therapeutic target for the prevention and treatment of AA. However, products targeting the Ang-(1-7)/MasR pathway still lack clinical validation. This review will outline the epidemiology of AA, including thoracic, abdominal, and thoracoabdominal AA, as well as current diagnostic and treatment strategies. Due to the highest incidence and most extensive research on abdominal AA (AAA), we will focus on AAA to explain the role of the RAS in its development, the protective function of Ang-(1-7)/MasR, and the mechanisms involved. We will also describe the roles of agonists and antagonists, suggest improvements in engineering and drug delivery, and provide evidence for Ang-(1-7)/MasR's clinical potential, discussing risks and solutions for clinical use. This study will enhance our understanding of AA and offer new possibilities and promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Guang Yang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Institute of Translational Medicine Shenzhen Second People's Hospital The First Affiliated Hospital of Shenzhen University Shenzhen China
- Department of Life Sciences Yuncheng University Yuncheng China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Abbas Khan
- Department of Nutrition and Health Promotion University of Home Economics Lahore Pakistan Lahore Pakistan
| | - Wei Liang
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Zibo Xiong
- Division of Renal Medicine Peking University Shenzhen Hospital Shenzhen China
- Shenzhen Clinical Research Center for Urology and Nephrology Shenzhen China
| | - Johannes Stegbauer
- Department of Nephrology Medical Faculty University Hospital Düsseldorf Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
2
|
West J. ACE2 trials suggest remodeling, not dilation, as primary therapeutic effect. Pulm Circ 2022; 12:e12022. [PMID: 35506063 PMCID: PMC9053010 DOI: 10.1002/pul2.12022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- James West
- Vanderbilt University Nashville Tennessee USA
| |
Collapse
|
3
|
Vasam G, S SJ, Miyat SY, Adam H, Jarajapu YP. Early onset of aging phenotype in vascular repair by Mas receptor deficiency. GeroScience 2021; 44:311-327. [PMID: 34661816 DOI: 10.1007/s11357-021-00473-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/08/2021] [Indexed: 10/20/2022] Open
Abstract
Aging is associated with impaired vascular repair following ischemic insult, largely due to reparative dysfunctions of progenitor cells. Activation of Mas receptor (MasR) was shown to reverse aging-associated vasoreparative dysfunction. This study tested the impact of MasR-deficiency on mobilization and vasoreparative functions with aging. Wild type (WT) or MasR-deficient mice (MasR-/- or MasR+/-) at 12-14 weeks (young) or middle age (11-12 months) (MA) were used in the study. Mobilization of lineage-negative, Sca-1-positive cKit-positive (LSK) cells in response to G-CSF or plerixafor was determined. Hindlimb ischemia (HLI) was induced by femoral artery ligation. Mobilization and blood flow recovery were monitored post-HLI. Radiation chimeras were made by lethal irradiation of WT or MasR-/- mice followed by administration of bone marrow cells from MasR-/- or WT mice, respectively. Nitric oxide (NO) generation by stromal-derived factor-1α (SDF) and mitochondrial reactive oxygen species (mitoROS) levels were determined by flow cytometry. Effect of A779 treatment on mobilization, blood flow recovery, and NO and ROS levels were determined in young WT and MasR+/- mice. Circulating LSK cells in basal or in response to plerixafor or G-CSF or in response to ischemic injury were lower in MasR-/- mice compared to the WT. Responses in MasR+/- mice were similar to the WT at young age but at the middle age, impairments were observed. Impaired mobilization to ischemia or G-CSF was rescued in WT → MasR-/- chimeras. NO levels were lower and mitoROS were higher in MasR-/- LSK cells compared to WT cells. A779 precipitated dysfunctions in young-MasR+/- mice similar to that observed in MA-MasR+/-, and this accompanied decreased NO generation by SDF and enhanced mitoROS levels. This study shows that mice at MA do not exhibit vasoreparative dysfunction. Either partial or total loss of MasR precipitates advanced-aging phenotype likely due to lack of NO and oxidative stress.
Collapse
Affiliation(s)
- Goutham Vasam
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58108, USA
| | - Shrinidh Joshi S
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58108, USA
| | - Su Yamin Miyat
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58108, USA
| | - Hashim Adam
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58108, USA
| | - Yagna P Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, 58108, USA.
| |
Collapse
|
4
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
5
|
Simões E Silva AC, Lanza K, Palmeira VA, Costa LB, Flynn JT. 2020 update on the renin-angiotensin-aldosterone system in pediatric kidney disease and its interactions with coronavirus. Pediatr Nephrol 2021; 36:1407-1426. [PMID: 32995920 PMCID: PMC7524035 DOI: 10.1007/s00467-020-04759-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022]
Abstract
The last decade was crucial for our understanding of the renin-angiotensin-aldosterone system (RAAS) as a two-axis, counter-regulatory system, divided into the classical axis, formed by angiotensin-converting enzyme (ACE), angiotensin II (Ang II), and the angiotensin type 1 receptor (AT1R), and the alternative axis comprising angiotensin-converting enzyme 2 (ACE2), angiotensin-(1-7) (Ang-(1-7)), and the Mas receptor. Breakthrough discoveries also took place, with other RAAS endopeptides being described, including alamandine and angiotensin A. In this review, we characterize the two RAAS axes and the role of their components in pediatric kidney diseases, including childhood hypertension (HTN), pediatric glomerular diseases, congenital abnormalities of the kidney and urinary tract (CAKUT), and chronic kidney disease (CKD). We also present recent findings on potential interactions between the novel coronavirus, SARS-CoV-2, and components of the RAAS, as well as potential implications of coronavirus disease 2019 (COVID-19) for pediatric kidney diseases.
Collapse
Affiliation(s)
- Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil.
- Pediatric Nephrology Unit, Department of Pediatrics, Faculty of Medicine, UFMG, Belo Horizonte, Brazil.
| | - Katharina Lanza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Vitória Andrade Palmeira
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Larissa Braga Costa
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Avenida Alfredo Balena, 190, 2nd floor, Room # 281, Belo Horizonte, MG, 30130-100, Brazil
| | - Joseph T Flynn
- Pediatric Nephrology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, 98105, USA
| |
Collapse
|
6
|
Anti-cancer potential of persimmon (Diospyros kaki) leaves via the PDGFR-Rac-JNK pathway. Sci Rep 2020; 10:18119. [PMID: 33093618 PMCID: PMC7581826 DOI: 10.1038/s41598-020-75140-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Persimmon leaves are known to have some beneficial effects, including ROS elimination, lipid circulation, and neuronal protection. However, their anti-cancer properties and the underlying mechanisms remain unclear. Herein, we show that treatment with the ethanol extract of persimmon, Diospyros kaki, leaves (EEDK) induces cancer cell death and inhibits cell proliferation. Using fluorescence resonance energy transfer (FRET) technology with genetically-encoded biosensors, we first found that EEDK stimulates a PDGFR-Rac signaling cascade in live cells. Moreover, we found that downstream of the PDGFR-Rac pathway, JNKs are activated by EEDK. In contrast, JNK-downstream inhibitors, such as CoCl2, T-5224, and pepstatin A, attenuated EEDK-induced cell death. Thus, we illustrate that the PDGFR-Rac-JNK signaling axis is triggered by EEDK, leading to cancer cell death, suggesting the extract of persimmon leaves may be a promising anti-cancer agent.
Collapse
|
7
|
Karnik SS, Singh KD, Tirupula K, Unal H. Significance of angiotensin 1-7 coupling with MAS1 receptor and other GPCRs to the renin-angiotensin system: IUPHAR Review 22. Br J Pharmacol 2017; 174:737-753. [PMID: 28194766 PMCID: PMC5387002 DOI: 10.1111/bph.13742] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/31/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
Angiotensins are a group of hormonal peptides and include angiotensin II and angiotensin 1-7 produced by the renin angiotensin system. The biology, pharmacology and biochemistry of the receptors for angiotensins were extensively reviewed recently. In the review, the receptor nomenclature committee was not emphatic on designating MAS1 as the angiotensin 1-7 receptor on the basis of lack of classical G protein signalling and desensitization in response to angiotensin 1-7, as well as a lack of consensus on confirmatory ligand pharmacological analyses. A review of recent publications (2013-2016) on the rapidly progressing research on angiotensin 1-7 revealed that MAS1 and two additional receptors can function as 'angiotensin 1-7 receptors', and this deserves further consideration. In this review we have summarized the information on angiotensin 1-7 receptors and their crosstalk with classical angiotensin II receptors in the context of the functions of the renin angiotensin system. It was concluded that the receptors for angiotensin II and angiotensin 1-7 make up a sophisticated cross-regulated signalling network that modulates the endogenous protective and pathogenic facets of the renin angiotensin system.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research InstituteCleveland Clinic FoundationClevelandOhioUSA
| | | | - Kalyan Tirupula
- Department of Molecular Cardiology, Lerner Research InstituteCleveland Clinic FoundationClevelandOhioUSA
- Biological E Limited, ShamirpetHyderabadIndia
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research InstituteCleveland Clinic FoundationClevelandOhioUSA
- Department of Basic Sciences, Faculty of Pharmacy and Betul Ziya Eren Genome and Stem Cell CenterErciyes UniversityKayseriTurkey
| |
Collapse
|
8
|
Tirupula KC, Zhang D, Osbourne A, Chatterjee A, Desnoyer R, Willard B, Karnik SS. MAS C-Terminal Tail Interacting Proteins Identified by Mass Spectrometry- Based Proteomic Approach. PLoS One 2015; 10:e0140872. [PMID: 26484771 PMCID: PMC4618059 DOI: 10.1371/journal.pone.0140872] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 10/01/2015] [Indexed: 11/19/2022] Open
Abstract
Propagation of signals from G protein-coupled receptors (GPCRs) in cells is primarily mediated by protein-protein interactions. MAS is a GPCR that was initially discovered as an oncogene and is now known to play an important role in cardiovascular physiology. Current literature suggests that MAS interacts with common heterotrimeric G-proteins, but MAS interaction with proteins which might mediate G protein-independent or atypical signaling is unknown. In this study we hypothesized that MAS C-terminal tail (Ct) is a major determinant of receptor-scaffold protein interactions mediating MAS signaling. Mass-spectrometry based proteomic analysis was used to comprehensively identify the proteins that interact with MAS Ct comprising the PDZ-binding motif (PDZ-BM). We identified both PDZ and non-PDZ proteins from human embryonic kidney cell line, mouse atrial cardiomyocyte cell line and human heart tissue to interact specifically with MAS Ct. For the first time our study provides a panel of PDZ and other proteins that potentially interact with MAS with high significance. A ‘cardiac-specific finger print’ of MAS interacting PDZ proteins was identified which includes DLG1, MAGI1 and SNTA. Cell based experiments with wild-type and mutant MAS lacking the PDZ-BM validated MAS interaction with PDZ proteins DLG1 and TJP2. Bioinformatics analysis suggested well-known multi-protein scaffold complexes involved in nitric oxide signaling (NOS), cell-cell signaling of neuromuscular junctions, synapses and epithelial cells. Majority of these protein hits were predicted to be part of disease categories comprising cancers and malignant tumors. We propose a ‘MAS-signalosome’ model to stimulate further research in understanding the molecular mechanism of MAS function. Identifying hierarchy of interactions of ‘signalosome’ components with MAS will be a necessary step in future to fully understand the physiological and pathological functions of this enigmatic receptor.
Collapse
Affiliation(s)
- Kalyan C. Tirupula
- Department of Molecular Cardiology, Cleveland Clinic, Ohio, United States of America
| | - Dongmei Zhang
- Proteomics Laboratory, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Appledene Osbourne
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, Ohio, United States of America
| | - Arunachal Chatterjee
- Department of Molecular Cardiology, Cleveland Clinic, Ohio, United States of America
| | - Russ Desnoyer
- Department of Molecular Cardiology, Cleveland Clinic, Ohio, United States of America
| | - Belinda Willard
- Proteomics Laboratory, Lerner Research Institute, Cleveland Clinic, Ohio, United States of America
| | - Sadashiva S. Karnik
- Department of Molecular Cardiology, Cleveland Clinic, Ohio, United States of America
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, Ohio, United States of America
- * E-mail:
| |
Collapse
|
9
|
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: 225] [Impact Index Per Article: 22.5] [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
|
10
|
Endotoxin-induced skeletal muscle wasting is prevented by angiotensin-(1-7) through a p38 MAPK-dependent mechanism. Clin Sci (Lond) 2015; 129:461-76. [PMID: 25989282 DOI: 10.1042/cs20140840] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/19/2015] [Indexed: 12/19/2022]
Abstract
Skeletal muscle atrophy induced during sepsis syndrome produced by endotoxin in the form of LPS (lipopolysaccharide), is a pathological condition characterized by the loss of strength and muscle mass, an increase in MHC (myosin heavy chain) degradation, and an increase in the expression of atrogin-1 and MuRF-1 (muscle-specific RING-finger protein 1), two ubiquitin E3 ligases belonging to the ubiquitin-proteasome system. Ang-(1-7) [Angiotensin-(1-7)], through its Mas receptor, has beneficial effects in skeletal muscle. We evaluated in vivo the role of Ang-(1-7) and Mas receptor on the muscle wasting induced by LPS injection into C57BL/10J mice. In vitro studies were performed in murine C2C12 myotubes and isolated myofibres from EDL (extensor digitorum longus) muscle. In addition, the participation of p38 MAPK (mitogen-activated protein kinase) in the Ang-(1-7) effect on the LPS-induced muscle atrophy was evaluated. Our results show that Ang-(1-7) prevents the decrease in the diameter of myofibres and myotubes, the decrease in muscle strength, the diminution in MHC levels and the induction of atrogin-1 and MuRF-1 expression, all of which are induced by LPS. These effects were reversed by using A779, a Mas antagonist. Ang-(1-7) exerts these anti-atrophic effects at least in part by inhibiting the LPS-dependent activation of p38 MAPK both in vitro and in vivo. We have demonstrated for the first time that Ang-(1-7) counteracts the skeletal muscle atrophy induced by endotoxin through a mechanism dependent on the Mas receptor that involves a decrease in p38 MAPK phosphorylation. The present study indicates that Ang-(1-7) is a novel molecule with a potential therapeutic use to improve muscle wasting during endotoxin-induced sepsis syndrome.
Collapse
|
11
|
Cabello-Verrugio C, Morales MG, Rivera JC, Cabrera D, Simon F. Renin-angiotensin system: an old player with novel functions in skeletal muscle. Med Res Rev 2015; 35:437-63. [PMID: 25764065 DOI: 10.1002/med.21343] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Skeletal muscle is a tissue that shows the most plasticity in the body; it can change in response to physiological and pathological stimuli. Among the diseases that affect skeletal muscle are myopathy-associated fibrosis, insulin resistance, and muscle atrophy. A common factor in these pathologies is the participation of the renin-angiotensin system (RAS). This system can be functionally separated into the classical and nonclassical RAS axis. The main components of the classical RAS pathway are angiotensin-converting enzyme (ACE), angiotensin II (Ang-II), and Ang-II receptors (AT receptors), whereas the nonclassical axis is composed of ACE2, angiotensin 1-7 [Ang (1-7)], and the Mas receptor. Hyperactivity of the classical axis in skeletal muscle has been associated with insulin resistance, atrophy, and fibrosis. In contrast, current evidence supports the action of the nonclassical RAS as a counter-regulator axis of the classical RAS pathway in skeletal muscle. In this review, we describe the mechanisms involved in the pathological effects of the classical RAS, advances in the use of pharmacological molecules to inhibit this axis, and the beneficial effects of stimulation of the nonclassical RAS pathway on insulin resistance, atrophy, and fibrosis in skeletal muscle.
Collapse
Affiliation(s)
- Claudio Cabello-Verrugio
- Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | | | | | | | | |
Collapse
|
12
|
Bader M, Alenina N, Andrade-Navarro MA, Santos RA. MAS and its related G protein-coupled receptors, Mrgprs. Pharmacol Rev 2014; 66:1080-105. [PMID: 25244929 DOI: 10.1124/pr.113.008136] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The Mas-related G protein-coupled receptors (Mrgprs or Mas-related genes) comprise a subfamily of receptors named after the first discovered member, Mas. For most Mrgprs, pruriception seems to be the major function based on the following observations: 1) they are relatively promiscuous in their ligand specificity with best affinities for itch-inducing substances; 2) they are expressed in sensory neurons and mast cells in the skin, the main cellular components of pruriception; and 3) they appear in evolution first in tetrapods, which have arms and legs necessary for scratching to remove parasites or other noxious substances from the skin before they create harm. Because parasites coevolved with hosts, each species faced different parasitic challenges, which may explain another striking observation, the multiple independent duplication and expansion events of Mrgpr genes in different species as a consequence of parallel adaptive evolution. Their predominant expression in dorsal root ganglia anticipates additional functions of Mrgprs in nociception. Some Mrgprs have endogenous ligands, such as β-alanine, alamandine, adenine, RF-amide peptides, or salusin-β. However, because the functions of these agonists are still elusive, the physiologic role of the respective Mrgprs needs to be clarified. The best studied Mrgpr is Mas itself. It was shown to be a receptor for angiotensin-1-7 and to exert mainly protective actions in cardiovascular and metabolic diseases. This review summarizes the current knowledge about Mrgprs, their evolution, their ligands, their possible physiologic functions, and their therapeutic potential.
Collapse
Affiliation(s)
- Michael Bader
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany (M.B., N.A., M.A.A.-N.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Lübeck, Germany (M.B.); and Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil (M.B., N.A., R.A.S.)
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany (M.B., N.A., M.A.A.-N.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Lübeck, Germany (M.B.); and Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil (M.B., N.A., R.A.S.)
| | - Miguel A Andrade-Navarro
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany (M.B., N.A., M.A.A.-N.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Lübeck, Germany (M.B.); and Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil (M.B., N.A., R.A.S.)
| | - Robson A Santos
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany (M.B., N.A., M.A.A.-N.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Lübeck, Germany (M.B.); and Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil (M.B., N.A., R.A.S.)
| |
Collapse
|
13
|
Orgaz JL, Herraiz C, Sanz-Moreno V. Rho GTPases modulate malignant transformation of tumor cells. Small GTPases 2014; 5:e29019. [PMID: 25036871 DOI: 10.4161/sgtp.29019] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Rho GTPases are involved in the acquisition of all the hallmarks of cancer, which comprise 6 biological capabilities acquired during the development of human tumors. The hallmarks include proliferative signaling, evading growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and activating invasion and metastasis programs, as defined by Hanahan and Weinberg. (1) Controlling these hallmarks are genome instability and inflammation. Emerging hallmarks are reprogramming of energy metabolism and evading immune destruction. To give a different view to the readers, we will not be focusing on invasion, metastasis, or cytoskeletal remodeling, but we will review here how Rho GTPases contribute to other hallmarks of cancer with a special emphasis on malignant transformation.
Collapse
Affiliation(s)
- Jose L Orgaz
- Randall Division of Cell and Molecular Biophysics; New Hunt's House; Guy's Campus; King's College London; London, UK
| | - Cecilia Herraiz
- Randall Division of Cell and Molecular Biophysics; New Hunt's House; Guy's Campus; King's College London; London, UK
| | - Victoria Sanz-Moreno
- Randall Division of Cell and Molecular Biophysics; New Hunt's House; Guy's Campus; King's College London; London, UK
| |
Collapse
|
14
|
Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angiotensin 1-7: novel therapeutic targets. Nat Rev Cardiol 2014; 11:413-26. [PMID: 24776703 PMCID: PMC7097196 DOI: 10.1038/nrcardio.2014.59] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Angiotensin-converting enzyme (ACE) 2 and its product angiotensin 1–7 are thought to have effects that counteract the adverse actions of other, better-known renin–angiotensin system (RAS) components Numerous experimental studies have suggested that ACE2 and angiotensin 1–7 have notable protective effects in the heart and blood vessels ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the functional importance and signalling mechanisms of angiotensin-1–7-induced actions remain unclear New pharmacological interventions targeting ACE2 are expected to be useful in clinical treatment of cardiovascular disease, especially those associated with overactivation of the conventional RAS More studies, especially randomized controlled clinical trials, are needed to clearly delineate the benefits of therapies targeting angiotensin 1–7 actions
Angiotensin-converting enzyme 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of the better-known members of the renin–angiotensin system and might, therefore, be useful therapeutic targets in patients with cardiovascular disease. Professor Jiang and colleagues review the evidence for the potential roles of these proteins in various cardiovascular conditions, including hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes. The renin–angiotensin system (RAS) has pivotal roles in the regulation of normal physiology and the pathogenesis of cardiovascular disease. Angiotensin-converting enzyme (ACE) 2, and its product angiotensin 1–7, are thought to have counteracting effects against the adverse actions of other, better known and understood, members of the RAS. The physiological and pathological importance of ACE2 and angiotensin 1–7 in the cardiovascular system are not completely understood, but numerous experimental studies have indicated that these components have protective effects in the heart and blood vessels. Here, we provide an overview on the basic properties of ACE2 and angiotensin 1–7 and a summary of the evidence from experimental and clinical studies of various pathological conditions, such as hypertension, atherosclerosis, myocardial remodelling, heart failure, ischaemic stroke, and diabetes mellitus. ACE2-mediated catabolism of angiotensin II is likely to have a major role in cardiovascular protection, whereas the relevant functions and signalling mechanisms of actions induced by angiotensin 1–7 have not been conclusively determined. The ACE2–angiotensin 1–7 pathway, however, might provide a useful therapeutic target for the treatment of cardiovascular disease, especially in patients with overactive RAS.
Collapse
Affiliation(s)
- Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Jianmin Yang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Yongtao Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Mei Dong
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Shuangxi Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Qunye Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Fang Fang Liu
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Kai Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodelling and Function Research, Qilu Hospital, Shandong University, 107 Wen Hua Xi Road, Jinan 250012, Shandong Province, China
| |
Collapse
|
15
|
Fujimoto D, Hirono Y, Goi T, Katayama K, Matsukawa S, Yamaguchi A. The activation of proteinase-activated receptor-1 (PAR1) promotes gastric cancer cell alteration of cellular morphology related to cell motility and invasion. Int J Oncol 2012; 42:565-73. [PMID: 23242308 DOI: 10.3892/ijo.2012.1738] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/23/2012] [Indexed: 12/11/2022] Open
Abstract
Cell motility proceeds by cycles of edge protrusion, adhesion and retraction. Whether these functions are coordinated by biochemical or biomechanical processes is unknown. Tumor invasion and metastasis is directly related to cell motility. We showed that stimulation of proteinase-activated receptor-1 (PAR1) can trigger an array of responses that would promote tumor cell growth and invasion. Thus, we examined aspects of PAR1 activation related to cell morphological change that might contribute to cell motility. We established a PAR1 stably transfected MKN45 gastric cancer cell line (MKN45/PAR1). We examined morphological changes, Rho family activation and overexpression of cytoskeletal protein in cells exposed to PAR1 agonists (α-thrombin and TFLLR-NH2). MKN45/PAR1 grows with an elongated and polarized morphology, extending pseudopodia at the leading edge. However, in the presence of PAR1 antagonist, MKN45/PAR1 did not show any changes in cell shape upon addition of either α-thrombin or TFLLR-NH2. Activated PAR1 induced RhoA and Rac1 phosphorylation, and subsequent overexpression of myosin IIA and filamin B which are stress fiber components that were identified by PMF analysis of peptide mass data obtained by MALDI-TOF/MS measurement. Upon stimulation of MKN45/PAR1 for 24 h with either α-thrombin or TFLLR-NH2, the distribution of both myosin IIA and filamin B proteins shifted to being distributed throughout the cytoplasm to the membrane, with more intense luminescence signals than in the absence of stimulation. These results demonstrate that PAR1 activation induces cell morphological change associated with cell motility via Rho family activation and cytoskeletal protein overexpression, and has a critical role in gastric cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Daisuke Fujimoto
- First Department of Surgery, Faculty of Medicine and Division of Bioresearch Laboratories, University of Fukui, Fukui 910-1193, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Liu C, Lv XH, Li HX, Cao X, Zhang F, Wang L, Yu M, Yang JK. Angiotensin-(1-7) suppresses oxidative stress and improves glucose uptake via Mas receptor in adipocytes. Acta Diabetol 2012; 49:291-9. [PMID: 22042130 DOI: 10.1007/s00592-011-0348-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 10/17/2011] [Indexed: 01/01/2023]
Abstract
Although reactive oxygen species (ROS) contribute to glucose intolerance induced by the renin-angiotensin system (RAS) is well documented, the role of the newly discovered pathway of RAS, angiotensin (Ang)-(1-7)/Mas axis, in this process remains unknown. Here, we examined the effect of Ang-(1-7) on oxidative stress and glucose uptake in adipocytes. We used primary cultured epididymal adipocytes from C57 mice to study Ang-(1-7) effects on glucose uptake. We also treated fully differentiated 3T3-L1 adipocytes with exogenous Ang-(1-7) or overexpression of angiotensin-converting enzyme 2 (ACE2) to induce endogenous generation of Ang-(1-7) to clarify its effects on ROS production. Intracellular ROS was measured by flow cytometry, dihydroethidium (DHE), and nitroblue tetrazolium assay. Levels of NADPH oxidase and adiponectin mRNA were measured by real-time PCR. Ang-(1-7) improved glucose uptake both in basal and insulin-stimulated states. ROS production was slightly but significantly decreased in adipocytes treated with Ang-(1-7). Additionally, Mas receptor antagonist D-Ala7-Ang-(1-7) (A779) reversed the effect of Ang-(1-7) on glucose uptake and oxidative stress. Furthermore, treatment of adipocytes with Ang-(1-7) decreased NADPH oxidase mRNA levels. We also found that oxidative stress induced by glucose oxidase-suppressed expression of adiponectin, an insulin-sensitive protein. However, the suppression of oxidative stress by Ang-(1-7) restored adiponectin expression, while A779 agonists these changes induced by Ang-(1-7). In conclusion, Ang-(1-7) can protect against oxidative stress and improve glucose metabolism in adipocytes. These results show that Ang-(1-7) is a novel target for the improvement of glucose metabolism by preventing oxidative stress.
Collapse
Affiliation(s)
- Chang Liu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Johnson JA, Hemnes AR, Perrien DS, Schuster M, Robinson LJ, Gladson S, Loibner H, Bai S, Blackwell TR, Tada Y, Harral JW, Talati M, Lane KB, Fagan KA, West J. Cytoskeletal defects in Bmpr2-associated pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2011; 302:L474-84. [PMID: 22180660 DOI: 10.1152/ajplung.00202.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The heritable form of pulmonary arterial hypertension (PAH) is typically caused by a mutation in bone morphogenic protein receptor type 2 (BMPR2), and mice expressing Bmpr2 mutations develop PAH with features similar to human disease. BMPR2 is known to interact with the cytoskeleton, and human array studies in PAH patients confirm alterations in cytoskeletal pathways. The goal of this study was to evaluate cytoskeletal defects in BMPR2-associated PAH. Expression arrays on our Bmpr2 mutant mouse lungs revealed cytoskeletal defects as a prominent molecular consequence of universal expression of a Bmpr2 mutation (Rosa26-Bmpr2(R899X)). Pulmonary microvascular endothelial cells cultured from these mice have histological and functional cytoskeletal defects. Stable transfection of different BMPR2 mutations into pulmonary microvascular endothelial cells revealed that cytoskeletal defects are common to multiple BMPR2 mutations and are associated with activation of the Rho GTPase, Rac1. Rac1 defects are corrected in cell culture and in vivo through administration of exogenous recombinant human angiotensin-converting enzyme 2 (rhACE2). rhACE2 reverses 77% of gene expression changes in Rosa26-Bmpr2(R899X) transgenic mice, in particular, correcting defects in cytoskeletal function. Administration of rhACE2 to Rosa26-Bmpr2(R899X) mice with established PAH normalizes pulmonary pressures. Together, these findings suggest that cytoskeletal function is central to the development of BMPR2-associated PAH and that intervention against cytoskeletal defects may reverse established disease.
Collapse
Affiliation(s)
- Jennifer A Johnson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2650, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhang T, Li Z, Dang H, Chen R, Liaw C, Tran TA, Boatman PD, Connolly DT, Adams JW. Inhibition of Mas G-protein signaling improves coronary blood flow, reduces myocardial infarct size, and provides long-term cardioprotection. Am J Physiol Heart Circ Physiol 2011; 302:H299-311. [PMID: 22003054 DOI: 10.1152/ajpheart.00723.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Mas receptor is a class I G-protein-coupled receptor that is expressed in brain, testis, heart, and kidney. The intracellular signaling pathways activated downstream of Mas are still largely unknown. In the present study, we examined the expression pattern and signaling of Mas in the heart and assessed the participation of Mas in cardiac ischemia-reperfusion injury. Mas mRNA and protein were present in all chambers of human hearts, with cardiomyocytes and coronary arteries being sites of enriched expression. Expression of Mas in either HEK293 cells or cardiac myocytes resulted in constitutive coupling to the G(q) protein, which in turn activated phospholipase C and caused inositol phosphate accumulation. To generate chemical tools for use in probing the function of Mas, we performed a library screen and chemistry optimization program to identify potent and selective nonpeptide agonists and inverse agonists. Mas agonists activated G(q) signaling in a dose-dependent manner and reduced coronary blood flow in isolated mouse and rat hearts. Conversely, treatment of isolated rat hearts with Mas inverse agonists improved coronary flow, reduced arrhythmias, and provided cardioprotection from ischemia-reperfusion injury, an effect that was due, at least in part, to decreased cardiomyocyte apoptosis. Participation of Mas in ischemia-reperfusion injury was confirmed in Mas knockout mice, which had reduced infarct size relative to mice with normal Mas expression. These results suggest that activation of Mas during myocardial infarction contributes to ischemia-reperfusion injury and further suggest that inhibition of Mas-G(q) signaling may provide a new therapeutic strategy directed at cardioprotection.
Collapse
Affiliation(s)
- Tong Zhang
- Arena Pharmaceuticals Incorporated, San Diego, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Iwata M, Cowling RT, Yeo SJ, Greenberg B. Targeting the ACE2-Ang-(1-7) pathway in cardiac fibroblasts to treat cardiac remodeling and heart failure. J Mol Cell Cardiol 2010; 51:542-7. [PMID: 21147120 DOI: 10.1016/j.yjmcc.2010.12.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 11/30/2010] [Accepted: 12/06/2010] [Indexed: 01/11/2023]
Abstract
Fibroblasts play a pivotal role in cardiac remodeling and the development of heart failure through the deposition of extra-cellular matrix (ECM) proteins and also by affecting cardiomyocyte growth and function. The renin-angiotensin system (RAS) is a key regulator of the cardiovascular system in health and disease and many of its effects involve cardiac fibroblasts. Levels of angiotensin II (Ang II), the main effector molecule of the RAS, are elevated in the failing heart and there is a substantial body of evidence indicating that this peptide contributes to changes in cardiac structure and function which ultimately lead to progressive worsening in heart failure. A pathway involving angiotensin converting enzyme 2 (ACE2) has the capacity to break down Ang II while generating angiotensin-(1-7) (Ang-(1-7)), a heptapeptide, which in contrast to Ang II, has cardioprotective and anti-remodeling effects. Many Ang-(1-7) actions involve cardiac fibroblasts and there is information indicating that it reduces collagen production and also may protect against cardiac hypertrophy. This report describes the effects of ACE2 and Ang-(1-7) that appear to be relevant in cardiac remodeling and heart failure and explores potential therapeutic strategies designed to increase ACE2 activity and Ang-(1-7) levels to treat these conditions. This article is part of a special issue entitled ''Key Signaling Molecules in Hypertrophy and Heart Failure.''
Collapse
Affiliation(s)
- Michikado Iwata
- Department of Medicine/Cardiology Division, University of California, San Diego, San Diego, CA, USA
| | | | | | | |
Collapse
|
20
|
da Silveira KD, Coelho FM, Vieira AT, Sachs D, Barroso LC, Costa VV, Bretas TLB, Bader M, de Sousa LP, da Silva TA, dos Santos RAS, Simões e Silva AC, Teixeira MM. Anti-inflammatory effects of the activation of the angiotensin-(1-7) receptor, MAS, in experimental models of arthritis. THE JOURNAL OF IMMUNOLOGY 2010; 185:5569-76. [PMID: 20935211 DOI: 10.4049/jimmunol.1000314] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activation of the renin-angiotensin (Ang) system induces inflammation via interaction between Ang II and type 1 receptor on leukocytes. The relevance of the new arm of the renin-Ang system, namely Ang-converting enzyme-2/Ang-(1-7)/Mas receptor, for inflammatory responses is not known and was investigated in this study. For this purpose, two experimental models were used: Ag-induced arthritis (AIA) in mice and adjuvant-induced arthritis (AdIA) in rats. Male C57BL/6 wild-type or Mas(-/-) mice were subjected to AIA and treated with Ang-(1-7), the Mas agonist AVE 0991, or vehicle. AdIA was performed in female rats that were given AVE 0991 or vehicle. In wild-type mice, Mas protein is expressed in arthritic joints. Administration of AVE 0991 or Ang-(1-7) decreased AIA-induced neutrophil accumulation, hypernociception, and production of TNF-α, IL-1β, and CXCL1. Histopathological analysis showed significant reduction of inflammation. Mechanistically, AVE 0991 reduced leukocyte rolling and adhesion, even when given after Ag challenge. Mas(-/-) mice subjected to AIA developed slightly more pronounced inflammation, as observed by greater neutrophil accumulation and cytokine release. Administration of AVE 0991 was without effect in Mas(-/-) mice subjected to AIA. In rats, administration of AVE 0991 decreased edema, neutrophil accumulation, histopathological score, and production of IL-1β and CXCL1 induced by AdIA. Therefore, activation of Mas receptors decreases neutrophil influx and cytokine production and causes significant amelioration of arthritis in experimental models of arthritis in rats and mice. This approach might represent a novel therapeutic opportunity for arthritis.
Collapse
Affiliation(s)
- Kátia Daniela da Silveira
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
He JG, Chen SL, Huang YY, Chen YL, Dong YG, Ma H. The nonpeptide AVE0991 attenuates myocardial hypertrophy as induced by angiotensin II through downregulation of transforming growth factor-beta1/Smad2 expression. Heart Vessels 2010; 25:438-43. [PMID: 20676968 DOI: 10.1007/s00380-009-1213-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Accepted: 10/08/2009] [Indexed: 11/28/2022]
Abstract
The nonpeptide AVE0991 is expected to be a putative new drug for cardiovascular diseases. However, the mechanisms for the cardioprotective actions of AVE0991 are still not fully understood. We planned to determine whether AVE0991 attenuates the angiotensin II (AngII)-induced myocardial hypertrophy and whether these AVE0991 effects involved transforming growth factor beta1 (TGF-beta1) and Smad2. A rat model of neonatal myocardial hypertrophy was induced by AngII. The AngII group significantly increased in protein content, surface area, and [(3)H]leucine incorporation efficiency by cardiomyocytes, compared to those of the control group (P < 0.01). The AngII group also had elevated TGF-beta1 and Smad2 expression (P < 0.01). These AngII-induced changes were significantly attenuated by AVE0991 in a dose-dependent manner. In our study, these actions of AngII (10(-6) mol/l) were significantly inhibited by both concentrations of AVE0991 (10(-5) mol/l and 10(-7) mol/l). Moreover, the high AVE0991 group had significantly better inhibition of myocardial hypertrophy than the low AVE0991 group. Meanwhile, the beneficial effects of AVE0991 were completely abolished when the cardiomyocytes were pretreated with Ang-(1-7) receptor antagonist A-779 (10(-6) mol/l). These results suggested that AVE0991 prevented AngII-inducing myocardial hypertrophy in a dose-dependent fashion, a process that may be associated with the inhibition of TGF-beta1/Smad2 signaling.
Collapse
Affiliation(s)
- Jian-Gui He
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | | | | | | | | | | |
Collapse
|
22
|
Singh A, Boyer JL, Der CJ, Zohn IE. Transformation by a nucleotide-activated P2Y receptor is mediated by activation of Galphai, Galphaq and Rho-dependent signaling pathways. J Mol Signal 2010; 5:11. [PMID: 20653955 PMCID: PMC2917412 DOI: 10.1186/1750-2187-5-11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 07/23/2010] [Indexed: 11/14/2022] Open
Abstract
Background Nucleotide-actived P2Y receptors play critical roles in the growth of tumor cells by regulating cellular proliferation, differentiation and survival. Results Here we demonstrate that an avian P2Y purinoceptor (tP2YR) with unique pharmacological and signal transduction properties induces morphologic and growth transformation of rodent fibroblasts. tP2YR induced a transformed phenotype similar to the mas oncogene, a G protein-coupled receptor which causes transformation by activation of Rac-dependent pathways. tP2YR-transformed cells exhibited increased steady-state activation of Rac1 and RhoA. Like activated Rho GTPases, tP2YR cooperated with activated Raf and caused synergistic transformation of NIH3T3 cells. Our data indicate that the ability of tP2YR to cause transformation is due to its unique ability among purinergic receptors to simultaneously activate Gαq and Gαi. Co-expression of constitutively activated mutants of these two Gα subunits caused the same transformed phenotype as tP2YR and Mas. Furthermore, transformation by both tP2YR and Mas was blocked by pharmacological inhibition of GαI by pertussis toxin (PTX) indicating an essential role for Gαi in transformation by these G-protein coupled receptors. Conclusions Our data suggest that coordinated activation of Gαq and Gαi may link the tP2YR and possibility the Mas oncogene with signaling pathways resulting in activation of Rho family proteins to promote cellular transformation.
Collapse
Affiliation(s)
- Anurag Singh
- Linebergher Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27599, USA.
| | | | | | | |
Collapse
|
23
|
Lin WZ, Li ZF, Tsang SY, Lung LKW, Wang DK, Chan WY, Zhu YK, Lee SST, Cheung WT. Identification and characterization of a novel CXC chemokine in xenograft tumor induced by mas-overexpressing cells. Int J Cancer 2009; 125:1316-27. [PMID: 19408311 DOI: 10.1002/ijc.24440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Overexpressions of G protein-coupled receptor (GPCR) with elevated downstream signaling events have been reported in various tumors. However, the cellular mechanism that GPCR overexpression leads to tumor formation is largely unknown. The orphan GPCR mas was originally isolated from a human epidermoid carcinoma. In vivo studies of mas-overexpressing cells suggested that xenograft tumor formation was positively correlated with the levels of mas expression. Histochemical analysis indicated that xenograft tumor consisted of mas-transfected and stromal cells. Biochemical analyses revealed that cells overexpressing mas exhibited significantly increased anchorage-independent growth, whereas there was no significant difference in cell proliferation in comparison with empty vector-transfected control cells. Expression profiling using mRNA differential display and Northern analysis indicated an elevated expression of GRO and a novel CXC chemokines, tumor-induced factor (TIF), in mas-transfected cells and xenograft tumor. Bacterially expressed recombinant TIF was found to act as a neutrophil chemoattractant in a chemotactic assay. These results suggest that mas overexpression enables anchorage-independent growth of transformed cells, and interplays of secreted chemokines with stromal cells modulate xenograft tumor formation. Importantly, a novel CXC chemokine, TIF, was identified in the xenograft tumor tissues.
Collapse
Affiliation(s)
- Wen-Zhen Lin
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gava E, Samad-Zadeh A, Zimpelmann J, Bahramifarid N, Kitten GT, Santos RA, Touyz RM, Burns KD. Angiotensin-(1-7) activates a tyrosine phosphatase and inhibits glucose-induced signalling in proximal tubular cells. Nephrol Dial Transplant 2009; 24:1766-73. [PMID: 19144997 PMCID: PMC2684752 DOI: 10.1093/ndt/gfn736] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background. In the diabetic kidney, stimulation of mitogen-activated protein kinases (MAPKs) leads to extracellular matrix protein synthesis. In the proximal tubule, angiotensin-(1–7) [Ang-(1–7)] blocks activation of MAPKs by angiotensin II. We studied the effect of Ang-(1–7) on signalling responses in LLC-PK1 cells in normal (5 mM) or high (25 mM) glucose. Methods. The p38 MAPK was assayed by immunoblot, Src homology 2-containing protein-tyrosine phosphatase-1 (SHP-1) activity was measured after immunoprecipitation, cell protein synthesis was determined by [3H]-leucine incorporation and transforming growth factor-β1 (TGF-β1), fibronectin and collagen IV were assayed by immunoblots and/or ELISA. Results. High glucose stimulated p38 MAPK. This response was inhibited by Ang-(1–7) in a concentration-dependent fashion, an effect reversed by the receptor Mas antagonist A-779. Ang-(1–7) increased SHP-1 activity, via the receptor Mas. An inhibitor of tyrosine phosphatase, phenylarsine oxide, reversed the inhibitory effect of Ang-(1–7) on high glucose-stimulated p38 MAPK. Ang-(1–7) inhibited high glucose-stimulated protein synthesis, and blocked the stimulatory effect of glucose on TGF-β1. Conversely, Ang-(1–7) had no effect on glucose-stimulated synthesis of fibronectin or collagen IV. Conclusions. These data indicate that in proximal tubular cells, binding of Ang-(1–7) to the receptor Mas stimulates SHP-1, associated with the inhibition of glucose-stimulated p38 MAPK. Ang-(1–7) selectively inhibits glucose-stimulated protein synthesis and TGF-β1. In diabetic nephropathy, Ang-(1–7) may partly counteract the profibrotic effects of high glucose.
Collapse
Affiliation(s)
- Elisandra Gava
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Health Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Alenina N, Xu P, Rentzsch B, Patkin EL, Bader M. Genetically altered animal models for Mas and angiotensin-(1-7). Exp Physiol 2007; 93:528-37. [PMID: 18156169 DOI: 10.1113/expphysiol.2007.040345] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mas is the receptor for angiotensin-(1-7) and is involved in cardiovascular and neuronal regulation, in which the heptapeptide also plays a major role. Mas-deficient mice have been generated by us, and their characterization has shown that Mas has important functions in behaviour and cardiovascular regulation. These mice exhibit increased anxiety but, despite an enhanced long-term potentiation in the hippocampus, do not perform better in learning experiments. When Mas-deficient mice are backcrossed to the FVB/N genetic background, a cardiovascular phenotype is uncovered, in that the backcrossed animals become hypertensive. Concordant with our detection by fluorescent in situ hybridization of Mas mRNA in mouse endothelium, this phenotype is caused by endothelial dysfunction based on a dysbalance between nitric oxide and reactive oxygen species in the vessel wall. In agreement with these data, transgenic spontaneously hypertensive stroke-prone rats overexpressing ACE2 in the vessel wall exhibit reduced blood pressure as a result of improved endothelial function. Moreover, angiotensin-(1-7) overexpression in transgenic rats has cardioprotective and haemodynamic effects. In conclusion, the angiotensin-(1-7)-Mas axis has important functional implications for vascular regulation and blood pressure control, particularly in pathophysiological situations.
Collapse
Affiliation(s)
- Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Strasse 10, D-13092 Berlin-Buch, Germany
| | | | | | | | | |
Collapse
|
26
|
Shutes A, Onesto C, Picard V, Leblond B, Schweighoffer F, Der CJ. Specificity and Mechanism of Action of EHT 1864, a Novel Small Molecule Inhibitor of Rac Family Small GTPases. J Biol Chem 2007; 282:35666-78. [DOI: 10.1074/jbc.m703571200] [Citation(s) in RCA: 241] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
27
|
Santos EL, Reis RI, Silva RG, Shimuta SI, Pecher C, Bascands JL, Schanstra JP, Oliveira L, Bader M, Paiva ACM, Costa-Neto CM, Pesquero JB. Functional rescue of a defective angiotensin II AT1 receptor mutant by the Mas protooncogene. ACTA ACUST UNITED AC 2007; 141:159-67. [PMID: 17320985 DOI: 10.1016/j.regpep.2006.12.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2006] [Revised: 12/23/2006] [Accepted: 12/23/2006] [Indexed: 11/19/2022]
Abstract
Earlier studies with Mas protooncogene, a member of the G-protein-coupled receptor family, have proposed this gene to code for a functional AngII receptor, however further results did not confirm this assumption. In this work we investigated the hypothesis that a heterodimeration AT(1)/Mas could result in a functional interaction between both receptors. For this purpose, CHO or COS-7 cells were transfected with the wild-type AT(1) receptor, a non-functional AT(1) receptor double mutant (C18F-K20A) and Mas or with WT/Mas and C18F-K20A/Mas. Cells single-expressing Mas or C18F/K20A did not show any binding for AngII. The co-expression of the wild-type AT(1) receptor and Mas showed a binding profile similar to that observed for the wild-type AT(1) expressed alone. Surprisingly, the co-expression of the double mutant C18F/K20A and Mas evoked a total recovery of the binding affinity for AngII to a level similar to that obtained for the wild-type AT(1). Functional measurements using inositol phosphate and extracellular acidification rate assays also showed a clear recovery of activity for AngII on cells co-expressing the mutant C18F/K20A and Mas. In addition, immunofluorescence analysis localized the AT(1) receptor mainly at the plasma membrane and the mutant C18F-K20A exclusively inside the cells. However, the co-expression of C18F-K20A mutant with the Mas changed the distribution pattern of the mutant, with intense signals at the plasma membrane, comparable to those observed in cells expressing the wild-type AT(1) receptor. These results support the hypothesis that Mas is able to rescue binding and functionality of the defective C18F-K20A mutant by dimerization.
Collapse
MESH Headings
- Amino Acid Sequence
- Angiotensin II/metabolism
- Animals
- CHO Cells
- COS Cells
- Cell Membrane/metabolism
- Chlorocebus aethiops
- Cricetinae
- Cricetulus
- Fluoresceins
- Fluorescent Antibody Technique, Direct
- Fluorescent Dyes
- Indoles
- Inhibitory Concentration 50
- Inositol Phosphates/analysis
- Inositol Phosphates/metabolism
- Models, Chemical
- Molecular Sequence Data
- Mutation
- Polymerase Chain Reaction
- Proto-Oncogenes/genetics
- Receptor, Angiotensin, Type 1/chemistry
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Transfection
Collapse
Affiliation(s)
- Edson L Santos
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, 04023-062 São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Darbro BW, Lee KM, Nguyen NK, Domann FE, Klingelhutz AJ. Methylation of the p16(INK4a) promoter region in telomerase immortalized human keratinocytes co-cultured with feeder cells. Oncogene 2006; 25:7421-33. [PMID: 16767161 PMCID: PMC1894570 DOI: 10.1038/sj.onc.1209729] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Human keratinocytes grown in co-culture with fibroblast feeder cells have an extended in vitro lifespan and delayed accumulation of the tumor suppressor protein p16(INK4a) when compared to the same cells grown on tissue culture plastic alone. Previous studies have indicated that human keratinocytes can be immortalized by telomerase activity alone when grown in co-culture with feeder cells, suggesting that loss of the p16(INK4a)/Rb pathway is not required for immortalization. Using two independent human keratinocyte cell strains, we found that exogenous telomerase expression and co-culture with feeder cells results in efficient extension of lifespan without an apparent crisis. However, when these cells were transferred from the co-culture environment to plastic alone they experienced only a brief period of slowed growth before continuing to proliferate indefinitely. Examination of immortal cell lines demonstrated p16(INK4a) promoter methylation had occurred in both the absence and presence of feeder cells. Reintroduction of p16(INK4a) into immortal cell lines resulted in rapid growth arrest. Our results suggest that p16(INK4a)/Rb-induced telomere-independent senescence, although delayed in the presence of feeders, still provides a proliferation barrier to human keratinocytes in this culture system and that extended culture of telomerase-transduced keratinocytes on feeders can lead to the methylation of p16(INK4a).
Collapse
Affiliation(s)
- BW Darbro
- Interdisciplinary Program in Molecular Biology and Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - KM Lee
- Department of Microbiology and Holden Cancer Center, University of Iowa, Iowa City, IA, USA
| | - NK Nguyen
- Department of Microbiology and Holden Cancer Center, University of Iowa, Iowa City, IA, USA
| | - FE Domann
- Department of Radiation Oncology and the Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA, USA
| | - AJ Klingelhutz
- Department of Microbiology and Holden Cancer Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
29
|
Hellner K, Walther T, Schubert M, Albrecht D. Angiotensin-(1–7) enhances LTP in the hippocampus through the G-protein-coupled receptor Mas. Mol Cell Neurosci 2005; 29:427-35. [PMID: 15950155 DOI: 10.1016/j.mcn.2005.03.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2004] [Revised: 03/03/2005] [Accepted: 03/28/2005] [Indexed: 10/25/2022] Open
Abstract
The renin-angiotensin system not only plays a critical role in blood pressure control but is also involved in learning and memory mechanisms. In addition to angiotensin (Ang) II, Ang-(1-7) may also have important biological activities in the brain. Here, we show for the first time that Ang-(1-7) enhances long-term potentiation (LTP) in the CA1 region of the hippocampus. Our studies with AT1 receptor antagonists and selective Ang-(1-7) receptor antagonists demonstrate the existence of a distinct Ang-(1-7) receptor in the brain, the G-protein-coupled receptor Mas, encoded by the Mas protooncogene. We also show that the genetic deletion of this receptor abolishes the Ang-(1-7)-induced enhancement of LTP. Thus, we firstly demonstrate that Ang-(1-7) influences the induction of LTP in limbic structures implicating its distinct function in learning and memory mechanisms; secondly, we have identified Mas as a functional receptor for Ang-(1-7) in the brain.
Collapse
Affiliation(s)
- K Hellner
- Institute of Neurophysiology (Charité, CCM), Humboldt University Berlin, Tucholskystr. 2, D-10117 Berlin, Germany
| | | | | | | |
Collapse
|
30
|
Palmby TR, Abe K, Karnoub AE, Der CJ. Vav Transformation Requires Activation of Multiple GTPases and Regulation of Gene Expression. Mol Cancer Res 2004. [DOI: 10.1158/1541-7786.702.2.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Although Vav can act as a guanine nucleotide exchange factor for RhoA, Rac1, and Cdc42, its transforming activity has been ascribed primarily to its ability to activate Rac1. However, because activated Vav, but not Rac-specific guanine nucleotide exchange factors, exhibits very potent focus-forming transforming activity when assayed in NIH 3T3 cells, Vav transforming activity must also involve activation of Rac-independent pathways. In this study, we determined the involvement of other Rho family proteins and their signaling pathways in Vav transformation. We found that RhoA, Rac1, and Cdc42 functions are all required for Vav transforming activity. Furthermore, we determined that Vav activation of nuclear factor-κB and the Jun NH2-terminal kinase mitogen-activated protein kinase (MAPK) is necessary for full transformation by Vav, whereas p38 MAPK does not seem to play an important role. We also determined that Vav is a weak activator of Elk-1 via a Ras- and MAPK/extracellular signal-regulated kinase kinase–dependent pathway, and this activity was essential for Vav transformation. Thus, we conclude that full Vav transforming activation is mediated by the activation of multiple small GTPases and their subsequent activation of signaling pathways that regulate changes in gene expression. Because Vav is activated by the epidermal growth factor receptor and other tyrosine kinases involved in cancer development, defining the role of aberrant Vav signaling may identify activities of receptor tyrosine kinases important for human oncogenesis.
Collapse
Affiliation(s)
- Todd R. Palmby
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Karon Abe
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Antoine E. Karnoub
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Channing J. Der
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Gandhi PN, Gibson RM, Tong X, Miyoshi J, Takai Y, Konieczkowski M, Sedor JR, Wilson-Delfosse AL. An activating mutant of Rac1 that fails to interact with Rho GDP-dissociation inhibitor stimulates membrane ruffling in mammalian cells. Biochem J 2004; 378:409-19. [PMID: 14629200 PMCID: PMC1223982 DOI: 10.1042/bj20030979] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2003] [Revised: 11/11/2003] [Accepted: 11/21/2003] [Indexed: 11/17/2022]
Abstract
Rac1, a member of the Rho family of small GTP-binding proteins, is involved in the regulation of the actin cytoskeleton via activation of lamellipodia and membrane ruffle formation. RhoGDI (Rho-family-specific GDP-dissociation inhibitor) forms a complex with Rho proteins in the cytosol of mammalian cells. It not only regulates guanine nucleotide binding to Rho proteins, but may also function as a molecular shuttle to carry Rho proteins from an inactive cytosolic pool to the membrane for activation. These studies tested if RhoGDI is necessary for the translocation of Rac1 from the cytosol to the plasma membrane for the formation of membrane ruffles. We describe a novel mutant of Rac1, R66E (Arg66-->Glu), that fails to bind RhoGDI. This RhoGDI-binding-defective mutation is combined with a Rac1-activating mutation G12V, resulting in a double-mutant [Rac1(G12V/R66E)] that fails to interact with RhoGDI in COS-7 cells, but remains constitutively activated. This double mutant stimulates membrane ruffling to a similar extent as that observed after epidermal growth factor treatment of non-transfected cells. To confirm that Rac1 can signal ruffle formation in the absence of interaction with RhoGDI, Rac1(G12V) was overexpressed in cultured mesangial cells derived from a RhoGDI knockout mouse. Rac1-mediated membrane ruffling was indistinguishable between the RhoGDI(-/-) and RhoGDI(+/+) cell lines. In both the COS-7 and cultured mesangial cells, Rac1(G12V) and Rac1(G12V/R66E) co-localize with membrane ruffles. These findings suggest that interaction with RhoGDI is not essential in the mechanism by which Rac1 translocates to the plasma membrane to stimulate ruffle formation.
Collapse
Affiliation(s)
- Payal N Gandhi
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Fiegen D, Haeusler LC, Blumenstein L, Herbrand U, Dvorsky R, Vetter IR, Ahmadian MR. Alternative Splicing of Rac1 Generates Rac1b, a Self-activating GTPase. J Biol Chem 2004; 279:4743-9. [PMID: 14625275 DOI: 10.1074/jbc.m310281200] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Rac1b was recently identified in malignant colorectal tumors as an alternative splice variant of Rac1 containing a 19-amino acid insertion next to the switch II region. The structures of Rac1b in the GDP- and the GppNHp-bound forms, determined at a resolution of 1.75 A, reveal that the insertion induces an open switch I conformation and a highly mobile switch II. As a consequence, Rac1b has an accelerated GEF-independent GDP/GTP exchange and an impaired GTP hydrolysis, which is restored partially by GTPase-activating proteins. Interestingly, Rac1b is able to bind the GTPase-binding domain of PAK but not full-length PAK in a GTP-dependent manner, suggesting that the insertion does not completely abolish effector interaction. The presented study provides insights into the structural and biochemical mechanism of a self-activating GTPase.
Collapse
Affiliation(s)
- Dennis Fiegen
- Max-Planck-Institut für molekulare Physiologie, Abteilung Strukturelle Biologie, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Santos RAS, e Silva ACS, Maric C, Silva DMR, Machado RP, de Buhr I, Heringer-Walther S, Pinheiro SVB, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss HP, Speth R, Walther T. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci U S A 2003; 100:8258-63. [PMID: 12829792 PMCID: PMC166216 DOI: 10.1073/pnas.1432869100] [Citation(s) in RCA: 1361] [Impact Index Per Article: 61.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The renin-angiotensin system plays a critical role in blood pressure control and body fluid and electrolyte homeostasis. Besides angiotensin (Ang) II, other Ang peptides, such as Ang III [Ang-(2-8)], Ang IV [Ang-(3-8)], and Ang-(1-7) may also have important biological activities. Ang-(1-7) has become an angiotensin of interest in the past few years, because its cardiovascular and baroreflex actions counteract those of Ang II. Unique angiotensin-binding sites specific for this heptapeptide and studies with a selective Ang-(1-7) antagonist indicated the existence of a distinct Ang-(1-7) receptor. We demonstrate that genetic deletion of the G protein-coupled receptor encoded by the Mas protooncogene abolishes the binding of Ang-(1-7) to mouse kidneys. Accordingly, Mas-deficient mice completely lack the antidiuretic action of Ang-(1-7) after an acute water load. Ang-(1-7) binds to Mas-transfected cells and elicits arachidonic acid release. Furthermore, Mas-deficient aortas lose their Ang-(1-7)-induced relaxation response. Collectively, these findings identify Mas as a functional receptor for Ang-(1-7) and provide a clear molecular basis for the physiological actions of this biologically active peptide.
Collapse
Affiliation(s)
- Robson A. S. Santos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Ana C. Simoes e Silva
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Christine Maric
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Denise M. R. Silva
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Raquel Pillar Machado
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Insa de Buhr
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Silvia Heringer-Walther
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Sergio Veloso B. Pinheiro
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Myriam Teresa Lopes
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Michael Bader
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Elizabeth P. Mendes
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Virgina Soares Lemos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Heinz-Peter Schultheiss
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Robert Speth
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
| | - Thomas Walther
- Department of Physiology and Biophysics, Federal
University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil;
Department of Medicine, Georgetown University,
Washington, DC 20057; Department of Cardiology
and Pneumology, University Hospital Benjamin Franklin, Free University, 12200
Berlin, Germany; Max Delbrück Center, 13125
Berlin, Germany; and Department of Veterinary
and Comparative Anatomy, Pharmacology, and Physiology, Washington State
University, Pullman, WA 99164-6520
- To whom correspondence should be addressed at: Benjamin Franklin Medical
Center, Department of Cardiology and Pneumology, Free University of Berlin,
Hindenburgdamm 30, 12200 Berlin, Germany. E-mail:
| |
Collapse
|
34
|
Pratt RL, Kinch MS. Activation of the EphA2 tyrosine kinase stimulates the MAP/ERK kinase signaling cascade. Oncogene 2002; 21:7690-9. [PMID: 12400011 DOI: 10.1038/sj.onc.1205758] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2002] [Revised: 06/06/2002] [Accepted: 07/06/2002] [Indexed: 01/11/2023]
Abstract
Intracellular signaling by receptor tyrosine kinases regulates many different aspects of cell behavior. Recent studies in our laboratory and others have demonstrated that the EphA2 receptor tyrosine kinase critically regulates tumor cell growth, migration and invasiveness. Although the cellular consequences of EphA2 signaling have been the focus of recent attention, the biochemical changes that are triggered by ligand-mediated activation of EphA2 remain largely unknown. Herein, we demonstrate that ligand stimulation of EphA2 promotes the nucleus translocation and phosphorylation of ERK kinases, followed by an increase in nuclear induction of the Elk-1 transcription factor. Ligand-mediated activation allows EphA2 to form a molecular complex with the SHC and GRB2 adaptor proteins. Specifically, we demonstrate that tyrosine phosphorylated EphA2 interacts with the PTB and SH2 domains of SHC. We also show that the interaction of EphA2 with GRB2 is indirect and mediated by SHC and that this complex is necessary for EphA2-mediated activation of ERK kinases. These studies provide a novel mechanism to demonstrate how EphA2 can convey information from the cell exterior to the nucleus.
Collapse
Affiliation(s)
- Rebecca L Pratt
- Department of Basic Medical Sciences, Purdue University Cancer Center, West Lafayette, Indiana, IN 47907-1246, USA
| | | |
Collapse
|
35
|
Cheng L, Rossman KL, Mahon GM, Worthylake DK, Korus M, Sondek J, Whitehead IP. RhoGEF specificity mutants implicate RhoA as a target for Dbs transforming activity. Mol Cell Biol 2002; 22:6895-905. [PMID: 12215546 PMCID: PMC134030 DOI: 10.1128/mcb.22.19.6895-6905.2002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dbs is a Rho-specific guanine nucleotide exchange factor (RhoGEF) that exhibits transforming activity when overexpressed in NIH 3T3 mouse fibroblasts. Like many RhoGEFs, the in vitro catalytic activity of Dbs is not limited to a single substrate. It can catalyze the exchange of GDP for GTP on RhoA and Cdc42, both of which are expressed in most cell types. This lack of substrate specificity, which is relatively common among members of the RhoGEF family, complicates efforts to determine the molecular basis of their transforming activity. We have recently determined crystal structures of several RhoGEFs bound to their cognate GTPases and have used these complexes to predict structural determinants dictating the specificities of coupling between RhoGEFs and GTPases. Guided by this information, we mutated Dbs to alter significantly its relative exchange activity for RhoA versus Cdc42 and show that the transformation potential of Dbs correlates with exchange on RhoA but not Cdc42. Supporting this conclusion, oncogenic Dbs activates endogenous RhoA but not endogenous Cdc42 in NIH 3T3 cells. Similarly, a competitive inhibitor that blocks RhoA activation also blocks Dbs-mediated transformation. In conclusion, this study highlights the usefulness of specificity mutants of RhoGEFs as tools to genetically dissect the multiple signaling pathways potentially activated by overexpressed or oncogenic RhoGEFs. These ideas are exemplified for Dbs, which is strongly implicated in the transformation of NIH 3T3 cells via RhoA and not Cdc42.
Collapse
Affiliation(s)
- Li Cheng
- Department of Microbiology and Molecular Genetics, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark 07103-2714, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Pruitt K, Pruitt WM, Bilter GK, Westwick JK, Der CJ. Raf-independent deregulation of p38 and JNK mitogen-activated protein kinases are critical for Ras transformation. J Biol Chem 2002; 277:31808-17. [PMID: 12082106 DOI: 10.1074/jbc.m203964200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activated Ras, but not Raf, causes transformation of RIE-1 epithelial cells, supporting the importance of Raf-independent pathways in mediating Ras transformation. The p38 and JNK mitogen-activated protein kinase cascades are activated by Ras via Raf-independent effector function. Therefore, we determined whether p38 and JNK activation are involved in Ras transformation of RIE-1 epithelial cells. Rather surprisingly, we found that pharmacologic inhibition of p38, together with Raf activation of ERK, was sufficient to mimic the morphologic and growth transformation caused by oncogenic Ras. p38 inhibition together with ERK activation also caused the same alterations in cyclin D1 and p21(CIP1) expression caused by Ras and induced an autocrine growth factor loop important for transformation. Finally, in contrast to p38, we found that JNK activation promoted Ras transformation, and that Ras deregulation of p38 and JNK was not mediated by activation of the Rac small GTPase. We conclude that a key action of Raf-independent effector pathways important for Ras transformation may involve inhibition of p38 and activation of JNK.
Collapse
Affiliation(s)
- Kevin Pruitt
- University of North Carolina, Lineberger Comprehensive Cancer Center, Department of Pharmacology, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
During the development and progression of human cancer, cells undergo numerous changes in morphology, proliferation, and transcriptional profile. Over the past couple of decades there have been intense efforts to understand the molecular mechanisms involved, and members of the Ras superfamily of small GTPases have emerged as important players. Mutated versions of the Ras genes were first identified in human cancers some 20 years ago, but more recently, the Rho branch of the family has been receiving increased attention. In addition to the experimental evidence implicating Rho GTPase signaling in promoting malignant transformation, genetic analysis of human cancers has now revealed a few examples of direct alterations in the genes encoding regulators of Rho GTPases. In this review, we discuss the evidence implicating Rho GTPases in transformation and metastasis, as well as the progress made toward identifying their biochemical mechanism of action.
Collapse
Affiliation(s)
- Aron B Jaffe
- CRC Oncogene and Signal Transduction Group, University College London, UK
| | | |
Collapse
|
38
|
Booden MA, Siderovski DP, Der CJ. Leukemia-associated Rho guanine nucleotide exchange factor promotes G alpha q-coupled activation of RhoA. Mol Cell Biol 2002; 22:4053-61. [PMID: 12024019 PMCID: PMC133844 DOI: 10.1128/mcb.22.12.4053-4061.2002] [Citation(s) in RCA: 147] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leukemia-associated Rho guanine-nucleotide exchange factor (LARG) belongs to the subfamily of Dbl homology RhoGEF proteins (including p115 RhoGEF and PDZ-RhoGEF) that possess amino-terminal regulator of G protein signaling (RGS) boxes also found within GTPase-accelerating proteins (GAPs) for heterotrimeric G protein alpha subunits. p115 RhoGEF stimulates the intrinsic GTP hydrolysis activity of G alpha 12/13 subunits and acts as an effector for G13-coupled receptors by linking receptor activation to RhoA activation. The presence of RGS box and Dbl homology domains within LARG suggests this protein may also function as a GAP toward specific G alpha subunits and couple G alpha activation to RhoA-mediating signaling pathways. Unlike the RGS box of p115 RhoGEF, the RGS box of LARG interacts not only with G alpha 12 and G alpha 13 but also with G alpha q. In cellular coimmunoprecipitation studies, the LARG RGS box formed stable complexes with the transition state mimetic forms of G alpha q, G alpha 12, and G alpha 13. Expression of the LARG RGS box diminished the transforming activity of oncogenic G protein-coupled receptors (Mas, G2A, and m1-muscarinic cholinergic) coupled to G alpha q and G alpha 13. Activated G alpha q, as well as G alpha 12 and G alpha 13, cooperated with LARG and caused synergistic activation of RhoA, suggesting that all three G alpha subunits stimulate LARG-mediated activation of RhoA. Our findings suggest that the RhoA exchange factor LARG, unlike the related p115 RhoGEF and PDZ-RhoGEF proteins, can serve as an effector for Gq-coupled receptors, mediating their functional linkage to RhoA-dependent signaling pathways.
Collapse
Affiliation(s)
- Michelle A Booden
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 27599, USA.
| | | | | |
Collapse
|
39
|
Lambert JM, Karnoub AE, Graves LM, Campbell SL, Der CJ. Role of MLK3-mediated activation of p70 S6 kinase in Rac1 transformation. J Biol Chem 2002; 277:4770-7. [PMID: 11713255 DOI: 10.1074/jbc.m109379200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The signaling pathways that mediate the transforming activity of the Rac1 GTPase remain to be determined. In the present study, we used effector domain mutants of the constitutively activated Rac(61L) mutant that display differential transforming activities and differential activation of downstream effector pathways to investigate the contribution of p70 S6 kinase (p70(S6K)) to Rac1 transformation and to decipher the signaling pathways leading from Rac1 to p70(S6K). First, we found that Rac1 transforming activity could be dissociated from Rac1 activation of p70(S6K). A weakly transforming Rac1 mutant retained the ability to activate p70(S6K), whereas some potently transforming effector mutants were impaired in their ability to activate p70(S6K). These data suggest that p70(S6K) is not necessary to promote full Rac1 transforming activity. We also found a strong correlation between the ability of the Rac(61L) effector mutants to activate p70(S6K) and their ability to activate the JNK mitogen-activated protein kinase. We found that the MLK3 serine/threonine kinase activated JNK and p70(S6K), whereas activation of p70(S6K) by Rac(61L) was significantly inhibited by dominant-negative MLK3. Additionally, the ability of the Rac(61L) effector mutants to activate MLK3 correlated well with their ability to activate p70(S6K) and JNK. Taken together, these results provide evidence that Rac1 coordinately activates p70(S6K) and JNK via MLK3 activation. Finally, we found that co-expression of wild type, but not kinase-dead, MLK3 significantly inhibited Rac1 transforming activity. These results suggest that MLK3 may be a negative regulator of the growth-promoting and transforming properties of Rac1.
Collapse
Affiliation(s)
- John M Lambert
- Lineberger Comprehensive Cancer Center, Department of Pharmacology and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA
| | | | | | | | | |
Collapse
|
40
|
Recio JA, Merlino G. Hepatocyte growth factor/scatter factor activates proliferation in melanoma cells through p38 MAPK, ATF-2 and cyclin D1. Oncogene 2002; 21:1000-8. [PMID: 11850817 DOI: 10.1038/sj.onc.1205150] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2001] [Revised: 10/21/2001] [Accepted: 10/30/2001] [Indexed: 11/09/2022]
Abstract
Members of the mitogen-activated protein kinase (MAPK) superfamily, including p38 kinase and SAPK/JNK, play a central role in mediating cellular response to environmental stress, growth factors and cytokines. Hepatocyte growth factor/scatter factor (HGF/SF) is a multifunctional cytokine capable of eliciting mitogenic, motogenic and morphogenetic activities in responsive cells, and has been implicated in tumor development and metastasis. Binding of HGF/SF to its tyrosine kinase receptor c-Met stimulates multiple signal transduction pathways, leading to the activation of numerous transcription factors. We here report that HGF/SF can induce cyclin D1 expression in mouse melanoma cells, and that this up-regulation is mediated in part by the activating transcription factor-2 (ATF-2). HGF/SF-mediated phosphorylation of ATF-2 was reduced in the presence of either the p38 kinase-specific inhibitor SB203580, a dominant negative p38 mutant, the SAPK/JNK inhibitor JNK-interacting protein-1 (JIP-1), or the phosphatidylinositol 3-kinase (PI3K)-specific inhibitor LY294002. Activation of p38 kinase by HGF/SF was partially blocked by the PI3K-specific inhibitor as well. The upstream kinases for p38, MKK3/6, did not become activated following HGF/SF exposure, and ATF-2 activation was undiminished by transient transfection of a dominant negative MKK6 mutant. However, transcriptional up-regulation of cyclin D1 by HGF/SF was partially inhibited by the p38 kinase-specific inhibitor, and cyclin D1 protein induction was partially blocked by a dominant negative ATF-2 mutant. Notably, the p38 kinase-specific inhibitor was able to block melanoma cell proliferation but not motility. We conclude that the ATF-2 transcription factor becomes activated by HGF/SF through p38 MAPK and SAPK/JNK. Moreover, the p38-ATF-2 pathway can help mediate proliferation signals in tumor cells through transcriptional activation of key cell cycle regulators.
Collapse
Affiliation(s)
- Juan A Recio
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264, USA
| | | |
Collapse
|
41
|
Martin CB, Mahon GM, Klinger MB, Kay RJ, Symons M, Der CJ, Whitehead IP. The thrombin receptor, PAR-1, causes transformation by activation of Rho-mediated signaling pathways. Oncogene 2001; 20:1953-63. [PMID: 11360179 DOI: 10.1038/sj.onc.1204281] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2000] [Revised: 01/12/2001] [Accepted: 01/18/2001] [Indexed: 11/09/2022]
Abstract
We utilized a cDNA expression library derived from the B6SutA(1) mouse myeloid progenitor cell line to search for novel oncogenes that promote growth transformation of NIH3T3 cells. A 2.2 kb transforming cDNA was recovered that encodes the wild type thrombin-stimulated G protein-coupled receptor PAR-1. In addition to its potent focus forming activity, constitutive overexpression of PAR-1 in NIH3T3 cells promoted the loss of anchorage- and serum-dependent growth. Although inhibitors of thrombin failed to block PAR-1 transforming activity, a PAR-1 mutant that cannot be cleaved by thrombin was nontransforming. Since the foci of transformed cells induced by PAR-1 bear a striking resemblance to those induced by activated RhoA, we determined if PAR-1 transformation was due to the aberrant activation of a specific Rho family member. Like RhoA, PAR-1 cooperated with activated Raf-1 and caused synergistic enhancement of transforming activity, induced stress fibers when microinjected into porcine aortic endothelial cells, stimulated the activity of the serum response factor and NF-kappaB transcription factors, and PAR-1 transformation was blocked by co-expression of dominant negative RhoA. Finally, PAR-1 transforming activity was blocked by pertussis toxin and by co-expression of the RGS domain of Lsc, implicating Galpha(i) and Galpha(12)/Galpha(13) subunits, respectively, as mediators of PAR-1 transformation. Taken together, these observations suggest that PAR-1 growth transformation is mediated, in part, by activation of RhoA.
Collapse
Affiliation(s)
- C B Martin
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina NC 27599-7295, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Bauerfeld CP, Hershenson MB, Page K. Cdc42, but not RhoA, regulates cyclin D1 expression in bovine tracheal myocytes. Am J Physiol Lung Cell Mol Physiol 2001; 280:L974-82. [PMID: 11290522 DOI: 10.1152/ajplung.2001.280.5.l974] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously demonstrated that Rac1 increased cyclin D1 promoter activity in an extracellular signal-regulated kinase (ERK)-independent, antioxidant-sensitive manner. Here, we examined the regulation of cyclin D1 expression by Cdc42 and RhoA. Overexpression of active Cdc42, but not of RhoA, induced transcription from the cyclin D1 promoter. Furthermore, dominant negative Cdc42, but not RhoA, attenuated platelet-derived growth factor-mediated activation of the cyclin D1 promoter. Overexpression of active Cdc42 increased cyclin D1 protein abundance in COS cells. Cdc42-induced cyclin D1 promoter activation was independent of ERK as evidenced by insensitivity to PD-98059, an inhibitor of mitogen-activated protein kinase/ERK kinase (MEK). Furthermore, Cdc42 was neither sufficient nor required for activation of ERK. Similar to Rac1-induced cyclin D1 expression, pretreatment with the antioxidants catalase and ebselen inhibited Cdc42-mediated transcription from the cyclin D1 promoter. Finally, like Rac1, active Cdc42 induced transactivation of the cyclin D1 promoter cAMP response element binding protein/activating transcription factor-2 binding site. Together, these data suggest that in airway smooth muscle cells, Cdc42 and Rac1 share a common signaling pathway to cyclin D1 promoter activation.
Collapse
Affiliation(s)
- C P Bauerfeld
- Department of Pediatrics, University of Chicago, Chicago, Illinois 60637-1470, USA
| | | | | |
Collapse
|
43
|
Abstract
Jun : Fos and Jun : ATF complexes represent two classes of AP-1 dimers that (1) preferentially bind to either heptameric or octameric AP-1 binding sites, and (2) are differently regulated by cellular signaling pathways and oncogene products. To discriminate between the functions of Jun : Fos, Jun : ATF and Jun : Jun, mutants were developed that restrict the ability of Jun to dimerize either to itself, or to Fos(-like) or ATF(-like) partners. Introduction of these mutants in chicken embryo fibroblasts shows that Jun : Fra2 and Jun : ATF2 dimers play distinct, complementary roles in in vitro oncogenesis by inducing either anchorage independence or growth factor independence, respectively. v-Jun : ATF2 rather than v-Jun : Fra2 triggers the development of primary fibrosarcomas in the chicken wing. Genes encoding extracellular matrix components seem to constitute an important subset of v-Jun : ATF2-target genes. Repression of the matrix component SPARC by Jun is essential for the induction of fibrosarcomas. Avian primary cells transformed by either Jun : Fra2 or Jun : ATF2 thus provide powerful tools for the investigation of the downstream pathways involved in oncogenesis. Further genetic studies with Jun dimerization mutants will be required to be precise and extend the specific roles of the Jun : Fos and Jun : ATF dimers during cancer progression in avian and mammalian systems.
Collapse
Affiliation(s)
- H van Dam
- Department of Molecular Cell Biology, Leiden University Medical Center, Sylvius Laboratories, PO Box 9503, 2300 RA Leiden, The Netherlands
| | | |
Collapse
|
44
|
Abstract
G protein coupled receptors (GPCRs) constitute the largest family of cell surface receptors, with more than 1000 members, and are responsible for converting a diverse array of extracellular stimuli into intracellular signaling events. Most members of the family have defined roles in intermediary metabolism and generally perform these functions in well-differentiated cells. However, there is an increasing awareness that some GPCRs can also regulate proliferative signaling pathways and that chronic stimulation or mutational activation of receptors can lead to oncogenic transformation. Activating mutations in GPCRs are associated with several types of human tumors and some receptors exhibit potent oncogenic activity due to agonist overexpression. Additionally, expression screening analyses for novel oncogenes identified GPCRs whose expression causes the oncogenic transformation of NIH3T3 mouse fibroblasts. These include Mas, G2A, and the PAR-1 thrombin receptor. In this review we summarize the signaling and transforming properties of these GPCR oncoproteins. What has emerged from these studies is the delineation of a GTPase cascade where transforming GPCRs cause aberrant growth regulation via activation of Rho family small GTPases.
Collapse
Affiliation(s)
- I P Whitehead
- Department of Microbiology and Molecular Genetics, UMDNJ-New Jersey Medical School, Newark, New Jersey, NJ 07103-2714, USA
| | | | | |
Collapse
|
45
|
Braga VM, Betson M, Li X, Lamarche-Vane N. Activation of the small GTPase Rac is sufficient to disrupt cadherin-dependent cell-cell adhesion in normal human keratinocytes. Mol Biol Cell 2000; 11:3703-21. [PMID: 11071901 PMCID: PMC15031 DOI: 10.1091/mbc.11.11.3703] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
To achieve strong adhesion to their neighbors and sustain stress and tension, epithelial cells develop many different specialized adhesive structures. Breakdown of these structures occurs during tumor progression, with the development of a fibroblastic morphology characteristic of metastatic cells. During Ras transformation, Rac-signaling pathways participate in the disruption of cadherin-dependent adhesion. We show that sustained Rac activation per se is sufficient to disassemble cadherin-mediated contacts in keratinocytes, in a concentration- and time-dependent manner. Cadherin receptors are removed from junctions before integrin receptors, suggesting that pathways activated by Rac can specifically interfere with cadherin function. We mapped an important region for disruption of junctions to the putative second effector domain of the Rac protein. Interestingly, although this region overlaps the domain necessary to induce lamellipodia, we demonstrate that the disassembly of cadherin complexes is a new Rac activity, distinct from Rac-dependent lamellipodia formation. Because Rac activity is also necessary for migration, Rac is a good candidate to coordinately regulate cell-cell and cell-substratum adhesion during tumorigenesis.
Collapse
Affiliation(s)
- V M Braga
- Medical Research Council Laboratory for Molecular Cell Biology and the Department of Biochemistry and Molecular Biology, University College London, London WC1E 6BT, United Kingdom.
| | | | | | | |
Collapse
|
46
|
Voss A, Malberg H, Schumann A, Wessel N, Walther T, Stepan H, Faber R. Baroreflex sensitivity, heart rate, and blood pressure variability in normal pregnancy. Am J Hypertens 2000; 13:1218-25. [PMID: 11078183 DOI: 10.1016/s0895-7061(00)01199-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Heart rate variability is a relevant predictor of cardiovascular risk in humans. However, to use heart and blood pressure (BP) variability or baroreflex sensitivity as markers for hypertensive pregnancy disorders, it is first necessary to describe these parameters in normal pregnancy. To accommodate the complexities of autonomic cardiovascular control we added parameter domains of nonlinear dynamics to conventional linear methods of time and frequency domains. The BP of 27 women with normal pregnancy and 14 nonpregnant women were monitored at a high resolution (200 Hz sampling frequency) using a Portapres for 30 min. The pregnant women were divided into groups of 32 or less or greater than 32 weeks of gestation. Pregnant and nonpregnant women were classified into subclasses of maternal age of less than 28 or 28 or more years. Except for two single parameter domains, we found no significant differences in heart rate and BP variability for pregnant women with different gestational age or different maternal age. Moreover, no significant differences in spontaneous baroreflex sensitivity could be found between pregnant women regardless of either their age or gestational age. In contrast, all measures of nonlinear dynamics of heart rate variability as well as all parameter domains of spontaneous baroreflex sensitivity showed significant changes between pregnant and nonpregnant women, whereas BP variability did not differ between those groups. This complex assessment of autonomic cardiovascular regulation has shown that the parameters tested are stable in the second half of normal pregnancy, and might have the potential to be excellent indicators of pathophysiologic conditions.
Collapse
Affiliation(s)
- A Voss
- University of Applied Sciences, Jena, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Page K, Li J, Wang Y, Kartha S, Pestell RG, Hershenson MB. Regulation of cyclin D(1) expression and DNA synthesis by phosphatidylinositol 3-kinase in airway smooth muscle cells. Am J Respir Cell Mol Biol 2000; 23:436-43. [PMID: 11017907 DOI: 10.1165/ajrcmb.23.4.3953] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We have shown in bovine tracheal myocytes that extracellular signal-regulated kinase (ERK) and Rac1 function as upstream activators of transcription from the cyclin D(1) promoter. We now examine the role of phosphatidylinositol (PI) 3-kinase in this process. PI 3-kinase activity was increased by platelet-derived growth factor (PDGF) and attenuated by the PI 3-kinase inhibitors wortmannin and LY294002. These inhibitors also decreased cyclin D(1) promoter activity, protein abundance, and DNA synthesis. Overexpression of the active catalytic subunit of PI 3-kinase (p110(PI) (3-K)CAAX) was sufficient to activate the cyclin D(1) promoter. Wortmannin and LY294002 failed to attenuate PDGF-induced ERK activation, and overexpression of p110(PI) (3-K)CAAX was insufficient to activate ERK. p110(PI) (3-K)CAAX-induced cyclin D(1) promoter activity was not blocked by PD98059, an inhibitor of mitogen-activated protein kinase/ERK kinase. We next examined whether PI 3-kinase and the 21-kD guanidine triphosphatase Rac1 regulate cyclin D(1) promoter activity by similar mechanisms. p110(PI) (3-K)CAAX-induced cyclin D(1) promoter activity was decreased by two inhibitors of Rac1-mediated signaling, catalase and diphenylene iodonium. Further, PDGF, PI 3-kinase, and Rac1 each activated the cyclin D(1) promoter at the cyclic adenosine monophosphate response element binding protein (CREB)/activating transcription factor (ATF)-2 binding site, as evidenced by expression of a CREB/ATF-2 reporter plasmid. Finally, PI 3-kinase and Rac1-induced CREB/ATF-2 transactivation were each inhibited by catalase. Together, these data suggest that in airway smooth muscle (ASM) cells, PI 3-kinase regulates transcription from the cyclin D(1) promoter and DNA synthesis in an ERK-independent manner. Further, PI 3-kinase and Rac1 regulate ASM cell cycle traversal via a common cis-regulatory element in the cyclin D(1) promoter.
Collapse
Affiliation(s)
- K Page
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
48
|
Camoretti-Mercado B, Liu HW, Halayko AJ, Forsythe SM, Kyle JW, Li B, Fu Y, McConville J, Kogut P, Vieira JE, Patel NM, Hershenson MB, Fuchs E, Sinha S, Miano JM, Parmacek MS, Burkhardt JK, Solway J. Physiological control of smooth muscle-specific gene expression through regulated nuclear translocation of serum response factor. J Biol Chem 2000; 275:30387-93. [PMID: 10866994 DOI: 10.1074/jbc.m000840200] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Prolonged serum deprivation induces a structurally and functionally contractile phenotype in about 1/6 of cultured airway myocytes, which exhibit morphological elongation and accumulate abundant contractile apparatus-associated proteins. We tested the hypothesis that transcriptional activation of genes encoding these proteins accounts for their accumulation during this phenotypic transition by measuring the transcriptional activities of the murine SM22 and human smooth muscle myosin heavy chain promoters during transient transfection in subconfluent, serum fed or 7 day serum-deprived cultured canine tracheal smooth muscle cells. Contrary to our expectation, SM22 and smooth muscle myosin heavy chain promoter activities (but not viral murine sarcoma virus-long terminal repeat promoter activity) were decreased in long term serum-deprived myocytes by at least 8-fold. Because serum response factor (SRF) is a required transcriptional activator of these and other smooth muscle-specific promoters, we evaluated the expression and function of SRF in subconfluent and long term serum-deprived cells. Whole cell SRF mRNA and protein were maintained at high levels in serum-deprived myocytes, but SRF transcription-promoting activity, nuclear SRF binding to consensus CArG sequences, and nuclear SRF protein were reduced. Furthermore, immunocytochemistry revealed extranuclear redistribution of SRF in serum-deprived myocytes; nuclear localization of SRF was restored after serum refeeding. These results uncover a novel mechanism for physiological control of smooth muscle-specific gene expression through extranuclear redistribution of SRF and consequent down-regulation of its transcription-promoting activity.
Collapse
Affiliation(s)
- B Camoretti-Mercado
- Department of Medicine, University of Chicago, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zohn IE, Klinger M, Karp X, Kirk H, Symons M, Chrzanowska-Wodnicka M, Der CJ, Kay RJ. G2A is an oncogenic G protein-coupled receptor. Oncogene 2000; 19:3866-77. [PMID: 10951580 DOI: 10.1038/sj.onc.1203731] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
G2A is a heptahelical cell surface protein that has recently been described as a potential tumor suppressor, based on its ability to counteract transformation of pre-B cells and fibroblasts by Bcr-Abl, an oncogenic tyrosine kinase. We have isolated cDNAs encoding G2A in the course of screening libraries for clones that cause oncogenic transformation of NIH3T3 fibroblasts. When expressed at high levels in NIH3T3 cells by retroviral transduction, G2A induced a full range of phenotypes characteristic of oncogenic transformation, including loss of contact inhibition, anchorage-independent survival and proliferation, reduced dependence on serum, and tumorigenicity in mice. When expressed by transfection, G2A greatly enhanced the ability of a weakly oncogenic form of Raf-1 to transform NIH3T3 cells. These results demonstrate that G2A is potently oncogenic both on its own and in cooperation with another oncogene. Expression of G2A in fibroblasts and endothelial cells resulted in changes in cell morphology and cytoskeleton structure that were equivalent to those induced by the G protein subunit Galpha13. Transformation of NIH3T3 cells via G2A expression was completely suppressed by co-expression of LscRGS, a GTPase activating protein that suppresses signaling by Galpha12 and Galpha13. Hyperactivity of Galpha12 or Galpha13 has previously been shown to result in activation of Rho GTPases. G2A expression resulted in activation of Rho, and transformation via G2A was suppressed by a dominant negative form of RhoA. These results indicate that G2A may be directly coupled to Galpha13, and that it is the activation of this Rho-activating Galpha protein which is responsible for the ability of G2A to transform fibroblasts.
Collapse
MESH Headings
- 3T3 Cells/pathology
- Amino Acid Sequence
- Animals
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Transformation, Neoplastic
- Cytoskeleton/genetics
- Cytoskeleton/ultrastructure
- DNA, Complementary
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Endothelium, Vascular/pathology
- GTP-Binding Protein alpha Subunit, Gi2
- GTP-Binding Protein alpha Subunits, G12-G13
- GTP-Binding Protein alpha Subunits, Gi-Go
- GTP-Binding Protein alpha Subunits, Gq-G11
- GTP-Binding Proteins/genetics
- GTP-Binding Proteins/metabolism
- Genes, ras
- Hematopoietic Stem Cells/physiology
- Heterotrimeric GTP-Binding Proteins/metabolism
- Hybridomas
- Mice
- Molecular Sequence Data
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-raf/genetics
- Proto-Oncogene Proteins c-raf/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled
- Serum Response Factor
- Signal Transduction
- Transcription, Genetic
- rac1 GTP-Binding Protein/genetics
- rac1 GTP-Binding Protein/metabolism
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- I E Zohn
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill 27599-7038, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Palmieri SJ, Nebl T, Pope RK, Seastone DJ, Lee E, Hinchcliffe EH, Sluder G, Knecht D, Cardelli J, Luna EJ. Mutant Rac1B expression in Dictyostelium: effects on morphology, growth, endocytosis, development, and the actin cytoskeleton. CELL MOTILITY AND THE CYTOSKELETON 2000; 46:285-304. [PMID: 10962483 DOI: 10.1002/1097-0169(200008)46:4<285::aid-cm6>3.0.co;2-n] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Rac1 is a small G-protein in the Ras superfamily that has been implicated in the control of cell growth, adhesion, and the actin-based cytoskeleton. To investigate the role of Rac1 during motile processes, we have established Dictyostelium cell lines that conditionally overexpress epitope-tagged Dictyostelium discoideum wild-type Rac1B (DdRac1B) or a mutant DdRac1B protein. Expression of endogenous levels of myc- or GFP-tagged wild-type DdRac1B had minimal effect on cellular morphologies and behaviors. By contrast, expression of a constitutively active mutant (G12-->V or Q61-->L) or a dominant negative mutant (T17-->N) generated amoebae with characteristic cellular defects. The morphological appearance of actin-containing structures, intracellular levels of F-actin, and cellular responses to chemoattractant closely paralleled the amount of active DdRac1B, indicating a role in upregulating actin cytoskeletal activities. Expression of any of the three mutants inhibited cell growth and cytokinesis, and delayed multicellular development, suggesting that DdRac1B plays important regulatory role(s) during these processes. No significant effects were observed on binding or internalization of latex beads in suspension or on intracellular membrane trafficking. Cells expressing DdRac1B-G12V exhibited defects in fluid-phase endocytosis and the longest developmental delays; DdRac1B-Q61L produced the strongest cytokinesis defect; and DdRac1B-T17N generated intermediate phenotypes. These conditionally expressed DdRac1B proteins should facilitate the identification and characterization of the Rac1 signaling pathway in an organism that is amenable to both biochemical and molecular genetic manipulations.
Collapse
Affiliation(s)
- S J Palmieri
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|