1
|
Kong CH, Dries E. Rad protein: An essential player in L-type Ca2+ channel localization and modulation in cardiomyocytes. J Gen Physiol 2024; 156:e202413629. [PMID: 39172109 PMCID: PMC11344166 DOI: 10.1085/jgp.202413629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Rad is an emerging key Cav1.2 modulator. In the present issue of JGP, Elmore, Ahern et al. examine how the Rad C-terminus affects its subcellular distribution and Cav1.2 regulation.
Collapse
Affiliation(s)
- Cherrie H.T. Kong
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Eef Dries
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Zhu Y, Yang X, Yang Y, Yan X, Li C, Chen S. Identification and Functional Analysis of Ras-Related Associated with Diabetes Gene ( rrad) in Edwardsiella piscicida-Resistant Individuals of Japanese Flounder ( Paralichthys olivaceus). Int J Mol Sci 2024; 25:10532. [PMID: 39408905 PMCID: PMC11476895 DOI: 10.3390/ijms251910532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/09/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Ras-related associated with diabetes (RRAD) is a member of the Ras GTPase superfamily that plays a role in several cellular functions, such as cell proliferation and differentiation. In particular, the superfamily acts as an NF-κB signaling pathway inhibitor and calcium regulator to participate in the immune response pathway. A recent transcriptome study revealed that rrad was expressed in the spleen of disease-resistant Japanese flounder (Paralichthys olivaceus) individuals compared with disease-susceptible individuals, and the results were also verified by qPCR. Thus, the present study aimed to explore how rrad regulates antimicrobial immunity via the NF-κB pathway. First, the coding sequence of P. olivaceus rrad was identified. The sequence was 1092 bp in length, encoding 364 amino acids. Based on phylogenetic and structural relationship analyses, P. olivaceus rrad appeared to be more closely related to teleosts. Next, rrad expression differences between disease-resistant and disease-susceptible individuals in immune-related tissues were evaluated, and the results revealed that rrad was expressed preferentially in the spleen of disease-resistant individuals. In response to Edwardsiella piscicida infection, rrad expression in the spleen changed. In vitro, co-culture was carried out to assess the hypo-methylated levels of the rrad promoter in the disease-resistant spleen, which was consistent with the high mRNA expression. The siRNA-mediated knockdown of rrad performed with the gill cell line of P. olivaceus affected many rrad-network-related genes, i.e., dcp1b, amagt, rus1, rapgef1, ralbp1, plce1, rasal1, nckipsd, prkab2, cytbc-1, sh3, and others, as well as some inflammation-related genes, such as bal2 and Il-1β. In addition, flow cytometry analysis showed that rrad overexpression was more likely to induce cell apoptosis, with establishing a link between rrad's function and its potential roles in regulating the NF-κB pathway. Thus,. the current study provided some clarity in terms of understanding the immune response about rrad gene differences between disease-resistant and disease-susceptible P. olivaceus individuals. This study provides a molecular basis for fish rrad gene functional analysis and may serve as a reference for in-depth of bacterial disease resistance of teleost.
Collapse
Affiliation(s)
- Ying Zhu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.Y.); (C.L.)
| | - Xinsheng Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.Y.); (C.L.)
| | - Yingming Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.Y.); (X.Y.); (S.C.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Xu Yan
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.Y.); (X.Y.); (S.C.)
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China; (X.Y.); (C.L.)
| | - Songlin Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.Y.); (X.Y.); (S.C.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
3
|
Rio-Vilariño A, del Puerto-Nevado L, García-Foncillas J, Cebrián A. Ras Family of Small GTPases in CRC: New Perspectives for Overcoming Drug Resistance. Cancers (Basel) 2021; 13:3757. [PMID: 34359657 PMCID: PMC8345156 DOI: 10.3390/cancers13153757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer remains among the cancers with the highest incidence, prevalence, and mortality worldwide. Although the development of targeted therapies against the EGFR and VEGFR membrane receptors has considerably improved survival in these patients, the appearance of resistance means that their success is still limited. Overactivation of several members of the Ras-GTPase family is one of the main actors in both tumour progression and the lack of response to cytotoxic and targeted therapies. This fact has led many resources to be devoted over the last decades to the development of targeted therapies against these proteins. However, they have not been as successful as expected in their move to the clinic so far. In this review, we will analyse the role of these Ras-GTPases in the emergence and development of colorectal cancer and their relationship with resistance to targeted therapies, as well as the status and new advances in the design of targeted therapies against these proteins and their possible clinical implications.
Collapse
Affiliation(s)
| | | | - Jesús García-Foncillas
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| | - Arancha Cebrián
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| |
Collapse
|
4
|
Application of the antitussive agents oxelaidin and butamirate as anti-glioma agents. Sci Rep 2021; 11:10145. [PMID: 33980886 PMCID: PMC8115262 DOI: 10.1038/s41598-021-89238-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 03/10/2021] [Indexed: 11/09/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor with a strong tendency of relapse and resistance to chemotherapy, but we currently lack non-toxic agents that effectively treat GBM. In this study, high-throughput screening of FDA-approved drugs was performed to identify safe and effective molecules and test their effect on GBM cell lines, LN229, U87 and T98G. Cough suppressants, oxelaidin and butamirate inhibited GBM growth. A Ras family GTPase, Ras-related associated with diabetes (RRAD), contributes to activation of STAT3, which is essential for survival and growth of many cancer types. Interestingly, oxelaidin and butamirate did not affect proliferation in RRAD negative GBM cells. Docking simulation analyses revealed selective interactions between oxelaidin and RRAD. The mechanism by which butamirate and oxelaidin inhibits GBM cell growth involves the suppression of STAT3 transcriptional activity, leading to down-regulation of cyclin D1 and survivin. In addition, components of RRAD-associated signaling cascades, including p-EGFR, p-Akt, and p-STAT3, were inhibited upon oxelaidin treatment. Intraperitoneal administration of oxelaidin or butamirate markedly suppressed tumor growth in a glioblastoma xenograft mouse model without significant adverse effects. Our collective findings indicate that oxelaidin and butamirate exert anti-tumor effects in glioblastoma, supporting its utility as a novel therapeutic candidate for glioblastoma.
Collapse
|
5
|
Belbachir N, Portero V, Al Sayed ZR, Gourraud JB, Dilasser F, Jesel L, Guo H, Wu H, Gaborit N, Guilluy C, Girardeau A, Bonnaud S, Simonet F, Karakachoff M, Pattier S, Scott C, Burel S, Marionneau C, Chariau C, Gaignerie A, David L, Genin E, Deleuze JF, Dina C, Sauzeau V, Loirand G, Baró I, Schott JJ, Probst V, Wu JC, Redon R, Charpentier F, Le Scouarnec S. RRAD mutation causes electrical and cytoskeletal defects in cardiomyocytes derived from a familial case of Brugada syndrome. Eur Heart J 2020; 40:3081-3094. [PMID: 31114854 DOI: 10.1093/eurheartj/ehz308] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/13/2018] [Accepted: 05/02/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS The Brugada syndrome (BrS) is an inherited cardiac disorder predisposing to ventricular arrhythmias. Despite considerable efforts, its genetic basis and cellular mechanisms remain largely unknown. The objective of this study was to identify a new susceptibility gene for BrS through familial investigation. METHODS AND RESULTS Whole-exome sequencing performed in a three-generation pedigree with five affected members allowed the identification of one rare non-synonymous substitution (p.R211H) in RRAD, the gene encoding the RAD GTPase, carried by all affected members of the family. Three additional rare missense variants were found in 3/186 unrelated index cases. We detected higher levels of RRAD transcripts in subepicardium than in subendocardium in human heart, and in the right ventricle outflow tract compared to the other cardiac compartments in mice. The p.R211H variant was then subjected to electrophysiological and structural investigations in human cardiomyocytes derived from induced pluripotent stem cells (iPSC-CMs). Cardiomyocytes derived from induced pluripotent stem cells from two affected family members exhibited reduced action potential upstroke velocity, prolonged action potentials and increased incidence of early afterdepolarizations, with decreased Na+ peak current amplitude and increased Na+ persistent current amplitude, as well as abnormal distribution of actin and less focal adhesions, compared with intra-familial control iPSC-CMs Insertion of p.R211H-RRAD variant in control iPSCs by genome editing confirmed these results. In addition, iPSC-CMs from affected patients exhibited a decreased L-type Ca2+ current amplitude. CONCLUSION This study identified a potential new BrS-susceptibility gene, RRAD. Cardiomyocytes derived from induced pluripotent stem cells expressing RRAD variant recapitulated single-cell electrophysiological features of BrS, including altered Na+ current, as well as cytoskeleton disturbances.
Collapse
Affiliation(s)
- Nadjet Belbachir
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent Portero
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Zeina R Al Sayed
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Jean-Baptiste Gourraud
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Florian Dilasser
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Laurence Jesel
- CHU Strasbourg, Service de Cardiologie, Strasbourg, France
| | - Hongchao Guo
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Haodi Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathalie Gaborit
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | | | - Aurore Girardeau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Stephanie Bonnaud
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Floriane Simonet
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Matilde Karakachoff
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | | | - Carol Scott
- The Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Sophie Burel
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Céline Marionneau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Caroline Chariau
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France
| | - Anne Gaignerie
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France
| | - Laurent David
- INSERM, CNRS, UNIV Nantes, CHU Nantes, SFR François Bonamy, iPSC core facility, Nantes, France.,Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, UNIV Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | | | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Institut de Génomique, CEA, Evry, France
| | - Christian Dina
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Vincent Sauzeau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Gervaise Loirand
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Isabelle Baró
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| | - Jean-Jacques Schott
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Vincent Probst
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Joseph C Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard Redon
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Flavien Charpentier
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France.,l'institut du thorax, CHU Nantes, Nantes, France
| | - Solena Le Scouarnec
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, 8 quai Moncousu, 44007 Nantes cedex 1, France
| |
Collapse
|
6
|
Withers CN, Brown DM, Byiringiro I, Allen MR, Condon KW, Satin J, Andres DA. Rad GTPase is essential for the regulation of bone density and bone marrow adipose tissue in mice. Bone 2017; 103:270-280. [PMID: 28732776 PMCID: PMC6886723 DOI: 10.1016/j.bone.2017.07.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/12/2017] [Accepted: 07/16/2017] [Indexed: 01/03/2023]
Abstract
The small GTP-binding protein Rad (RRAD, Ras associated with diabetes) is the founding member of the RGK (Rad, Rem, Rem2, and Gem/Kir) family that regulates cardiac voltage-gated Ca2+ channel function. However, its cellular and physiological functions outside of the heart remain to be elucidated. Here we report that Rad GTPase function is required for normal bone homeostasis in mice, as Rad deletion results in significantly lower bone mass and higher bone marrow adipose tissue (BMAT) levels. Dynamic histomorphometry in vivo and primary calvarial osteoblast assays in vitro demonstrate that bone formation and osteoblast mineralization rates are depressed, while in vitro osteoclast differentiation is increased, in the absence of Rad. Microarray analysis revealed that canonical osteogenic gene expression (Runx2, osterix, etc.) is not altered in Rad-/- calvarial osteoblasts; instead robust up-regulation of matrix Gla protein (MGP, +11-fold), an inhibitor of extracellular matrix mineralization and a protein secreted during adipocyte differentiation, was observed. Strikingly, Rad deficiency also resulted in significantly higher marrow adipose tissue levels in vivo and promoted spontaneous in vitro adipogenesis of primary calvarial osteoblasts. Adipogenic differentiation of wildtype calvarial osteoblasts resulted in the loss of endogenous Rad protein, further supporting a role for Rad in the control of BMAT levels. These findings reveal a novel in vivo function for Rad and establish a role for Rad signaling in the complex physiological control of skeletal homeostasis and bone marrow adiposity.
Collapse
Affiliation(s)
- Catherine N Withers
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, BBSRB, 741 S Limestone Street, Lexington, KY 40536-0509, USA.
| | - Drew M Brown
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Innocent Byiringiro
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Keith W Condon
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | - Jonathan Satin
- Department of Physiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298, USA.
| | - Douglas A Andres
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, BBSRB, 741 S Limestone Street, Lexington, KY 40536-0509, USA.
| |
Collapse
|
7
|
Yeom SY, Nam DH, Park C. RRAD promotes EGFR-mediated STAT3 activation and induces temozolomide resistance of malignant glioblastoma. Mol Cancer Ther 2014; 13:3049-61. [PMID: 25313011 DOI: 10.1158/1535-7163.mct-14-0244] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastoma multiforme (GBM) is an extremely aggressive brain cancer with a median survival of less than 2 years. GBM is characterized by abnormal activation of receptor tyrosine kinase and constitutively activated STAT3. Although EGFR phosphorylation and STAT3 activation are essential for the maintenance of GBM cancer stem cells, the molecular mechanism underlying endosome-mediated STAT3 activation is not fully understood. In the current study, we showed that GTP-binding protein RRAD (RAS associated with diabetes, RAD) physically associates with EGFR, and EEA1, enhancing the stability and endosome-associated nuclear translocation of EGFR. Functionally, RRAD contributes to the activation of STAT3 and expression of the stem cell factors OCT4, NANOG, and SOX2, thereby enhancing self-renewing ability, tumor sphere formation, EMT, and in vivo tumorigenesis. Most importantly, RRAD contributes to poor survival in patients with GBM. RRAD expression is correlated with temozolomide resistance, and, conversely, depletion of RRAD leads to sensitization of highly temozolomide-resistant GBM cells. Our data collectively support a novel function of RRAD in STAT3 activation and provide evidence that RRAD acts as a positive regulator in the EGFR signaling pathway. These results demonstrate a critical role for RRAD in GBM tumorigenesis and provide a rationale for the development of pharmacologic inhibitors of RRAD in GBM.
Collapse
Affiliation(s)
- Seon-Yong Yeom
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chaehwa Park
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
8
|
Sari AN, Kacan M, Unsal D, Sahan Firat S, Kemal Buharalioglu C, Vezir O, Korkmaz B, Cuez T, Canacankatan N, Sucu N, Ayaz L, Tamer Gumus L, Gorur A, Tunctan B. Contribution of RhoA/Rho-kinase/MEK1/ERK1/2/iNOS pathway to ischemia/reperfusion-induced oxidative/nitrosative stress and inflammation leading to distant and target organ injury in rats. Eur J Pharmacol 2013; 723:234-45. [PMID: 24296316 DOI: 10.1016/j.ejphar.2013.11.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/20/2013] [Accepted: 11/22/2013] [Indexed: 12/26/2022]
Abstract
The small G protein RhoA and its downstream effector Rho-kinase play an important role in various physiopathological processes including ischemia/reperfusion (I/R) injury. Reactive oxygen and nitrogen species produced by iNOS and NADPH oxidase are important mediators of inflammation and organ injury following an initial localized I/R event. The aim of this study was to determine whether RhoA/Rho-kinase signaling pathway increases the expression and activity of MEK1, ERK1/2, iNOS, gp91(phox), and p47(phox), and peroxynitrite formation which result in oxidative/nitrosative stress and inflammation leading to hindlimb I/R-induced injury in kidney as a distant organ and gastrocnemius muscle as a target organ. I/R-induced distant and target organ injury was performed by using the rat hindlimb tourniquet model. I/R caused an increase in the expression and/or activity of RhoA, MEK1, ERK1/2, iNOS, gp91(phox), p47(phox), and 3-nitrotyrosine and nitrotyrosine levels in the tissues. Although Rho-kinase activity was increased by I/R in the kidney, its activity was decreased in the muscle. Serum and tissue MDA levels and MPO activity were increased following I/R. I/R also caused an increase in SOD and catalase activities associated with decreased GSH levels in the tissues. Y-27632, a selective Rho-kinase inhibitor, (100µg/kg, i.p.; 1h before reperfusion) prevented the I/R-induced changes except Rho-kinase activity in the muscle. These results suggest that activation of RhoA/Rho-kinase/MEK1/ERK1/2/iNOS pathway associated with oxidative/nitrosative stress and inflammation contributes to hindlimb I/R-induced distant organ injury in rats. It also seems that hindlimb I/R induces target organ injury via upregulation of RhoA/MEK1/ERK1/2/iNOS pathway associated with decreased Rho-kinase activity.
Collapse
Affiliation(s)
- A Nihal Sari
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Meltem Kacan
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Demet Unsal
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Seyhan Sahan Firat
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - C Kemal Buharalioglu
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Ozden Vezir
- Department of Cardiovascular Surgery, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Belma Korkmaz
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Tuba Cuez
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey
| | - Necmiye Canacankatan
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Nehir Sucu
- Department of Cardiovascular Surgery, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Lokman Ayaz
- Department of Medicinal Biochemistry, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Lulufer Tamer Gumus
- Department of Medicinal Biochemistry, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Aysegul Gorur
- Department of Biochemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Bahar Tunctan
- Department of Pharmacology, Faculty of Pharmacy, Yenisehir Campus, Mersin University, 33169 Mersin, Turkey.
| |
Collapse
|
9
|
Role of Rho-kinase and its inhibitors in pulmonary hypertension. Pharmacol Ther 2013; 137:352-64. [DOI: 10.1016/j.pharmthera.2012.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 10/27/2012] [Indexed: 11/20/2022]
|
10
|
Activity-dependent subcellular cotrafficking of the small GTPase Rem2 and Ca2+/CaM-dependent protein kinase IIα. PLoS One 2012; 7:e41185. [PMID: 22815963 PMCID: PMC3399833 DOI: 10.1371/journal.pone.0041185] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/18/2012] [Indexed: 11/19/2022] Open
Abstract
Background Rem2 is a small monomeric GTP-binding protein of the RGK family, whose known functions are modulation of calcium channel currents and alterations of cytoskeletal architecture. Rem2 is the only RGK protein found predominantly in the brain, where it has been linked to synaptic development. We wished to determine the effect of neuronal activity on the subcellular distribution of Rem2 and its interacting partners. Results We show that Rem2 undergoes activity-and N-Methyl-D-Aspartate Receptor (NMDAR)-dependent translocation in rat hippocampal neurons. This redistribution of Rem2, from a diffuse pattern to one that is highly punctate, is dependent on Ca2+ influx, on binding to calmodulin (CaM), and also involves an auto-inhibitory domain within the Rem2 distal C-terminus region. We found that Rem2 can bind to Ca2+/CaM-dependent protein kinase IIα (CaMKII) a in Ca2+/CaM-dependent manner. Furthermore, our data reveal a spatial and temporal correlation between NMDAR-dependent clustering of Rem2 and CaMKII in neurons, indicating co-assembly and co-trafficking in neurons. Finally, we show that inhibiting CaMKII aggregation in neurons and HEK cells reduces Rem2 clustering, and that Rem2 affects the baseline distribution of CaMKII in HEK cells. Conclusions Our data suggest a novel function for Rem2 in co-trafficking with CaMKII, and thus potentially expose a role in neuronal plasticity.
Collapse
|
11
|
Luo Y, Zhang M, Zhang J, Zhang J, Chen C, Chen YE, Xiong JW, Zhu X. Platelet-derived growth factor induces Rad expression through Egr-1 in vascular smooth muscle cells. PLoS One 2011; 6:e19408. [PMID: 21559360 PMCID: PMC3084842 DOI: 10.1371/journal.pone.0019408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 04/05/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Ras associated with diabetes (Rad) inhibits vascular lesion formation by reducing the attachment and migration of vascular smooth muscle cells (VSMCs). However, the transcriptional regulation of Rad in VSMCs is unclear. METHODOLOGY AND PRINCIPAL FINDINGS We found that Platelet-Derived Growth Factor (PDGF)induced Rad expression in a time- and dose-dependent manner in rat aortic smooth muscle cells (RASMCs) using quantitative real-time PCR. By serial deletion analysis of the Rad promoter, we identified that two GC-rich early growth response-1 (Egr-1) binding sites are essential for PDGF-induced Rad promoter activation. Overexpression of Egr-1 in RASMCs strongly stimulated Rad expression while the Egr-1 corepressor, NGFI-A binding protein 2 (NAB2), repressed PDGF-induced Rad up-regulation in a dose-dependent manner. Direct binding of Egr-1 to the Rad promoter region was further confirmed by chromatin immunoprecipitation assays. CONCLUSIONS Our results demonstrate that Rad is regulated by PDGF through the transcriptional factor Egr-1 in RASMCs.
Collapse
Affiliation(s)
- Yan Luo
- The Institute of Molecular Medicine, Peking University, Beijing, China
| | - Meiling Zhang
- The Institute of Molecular Medicine, Peking University, Beijing, China
| | - Ji Zhang
- The Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jifeng Zhang
- The Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Chunlei Chen
- The Institute of Molecular Medicine, Peking University, Beijing, China
| | - Y. Eugene Chen
- The Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jing-Wei Xiong
- The Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiaojun Zhu
- The Institute of Molecular Medicine, Peking University, Beijing, China
- * E-mail:
| |
Collapse
|
12
|
Lee I, Yeom SY, Lee SJ, Kang WK, Park C. A novel senescence-evasion mechanism involving Grap2 and Cyclin D interacting protein inactivation by Ras associated with diabetes in cancer cells under doxorubicin treatment. Cancer Res 2010; 70:4357-65. [PMID: 20460530 DOI: 10.1158/0008-5472.can-09-3791] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ras associated with diabetes (Rad) is a Ras-related GTPase that promotes cell growth by accelerating cell cycle transitions. Rad knockdown induced cell cycle arrest and premature senescence without additional cellular stress in multiple cancer cell lines, indicating that Rad expression might be critical for the cell cycle in these cells. To investigate the precise function of Rad in this process, we used human Rad as bait in a yeast two-hybrid screening system and sought Rad-interacting proteins. We identified the Grap2 and cyclin D interacting protein (GCIP)/DIP1/CCNDBP1/HHM, a cell cycle-inhibitory molecule, as a binding partner of Rad. Further analyses revealed that Rad binds directly to GCIP in vitro and coimmunoprecipitates with GCIP from cell lysates. Rad translocates GCIP from the nucleus to the cytoplasm, thereby inhibiting the tumor suppressor activity of GCIP, which occurs in the nucleus. Furthermore, in the presence of Rad, GCIP loses its ability to reduce retinoblastoma phosphorylation and inhibit cyclin D1 activity. The function of Rad in transformation is also evidenced by increased telomerase activity and colony formation according to Rad expression level. In vivo tumorigenesis analyses revealed that tumors derived from Rad knockdown cells were significantly smaller than those from control cells (P = 0.0131) and the preestablished tumors are reduced in size after the injection of siRad (P = 0.0064). Therefore, we propose for the first time that Rad may promote carcinogenesis at least in part by inhibiting GCIP-mediated tumor suppression.
Collapse
Affiliation(s)
- Inkyoung Lee
- Biomedical Research Institute, Samsung Medical Center and Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | |
Collapse
|
13
|
Correll RN, Pang C, Niedowicz DM, Finlin BS, Andres DA. The RGK family of GTP-binding proteins: regulators of voltage-dependent calcium channels and cytoskeleton remodeling. Cell Signal 2008; 20:292-300. [PMID: 18042346 PMCID: PMC2254326 DOI: 10.1016/j.cellsig.2007.10.028] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 10/30/2007] [Indexed: 02/05/2023]
Abstract
RGK proteins constitute a novel subfamily of small Ras-related proteins that function as potent inhibitors of voltage-dependent (VDCC) Ca(2+) channels and regulators of actin cytoskeletal dynamics. Within the larger Ras superfamily, RGK proteins have distinct regulatory and structural characteristics, including nonconservative amino acid substitutions within regions known to participate in nucleotide binding and hydrolysis and a C-terminal extension that contains conserved regulatory sites which control both subcellular localization and function. RGK GTPases interact with the VDCC beta-subunit (Ca(V)beta) and inhibit Rho/Rho kinase signaling to regulate VDCC activity and the cytoskeleton respectively. Binding of both calmodulin and 14-3-3 to RGK proteins, and regulation by phosphorylation controls cellular trafficking and the downstream signaling of RGK proteins, suggesting that a complex interplay between interacting protein factors and trafficking contribute to their regulation.
Collapse
Affiliation(s)
- Robert N Correll
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | | | | | | | | |
Collapse
|
14
|
Willsky GR, Chi LH, Liang Y, Gaile DP, Hu Z, Crans DC. Diabetes-altered gene expression in rat skeletal muscle corrected by oral administration of vanadyl sulfate. Physiol Genomics 2006; 26:192-201. [PMID: 16684804 DOI: 10.1152/physiolgenomics.00196.2005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Treatment with vanadium, a representative of a class of antidiabetic compounds, alleviates diabetic hyperglycemia and hyperlipidemia. Oral administration of vanadium compounds in animal models and humans does not cause clinical symptoms of hypoglycemia, a common problem for diabetic patients with insulin treatment. Gene expression, using Affymetrix arrays, was examined in muscle from streptozotocin-induced diabetic and normal rats in the presence or absence of oral vanadyl sulfate treatment. This treatment affected normal rats differently from diabetic rats, as demonstrated by two-way ANOVA of the full array data. Diabetes altered the expression of 133 genes, and the expression of 30% of these genes dysregulated in diabetes was normalized by vanadyl sulfate treatment. For those genes, the ratio of expression in normal animals to the expression in diabetic animals showed a strong negative correlation with the ratio of expression in diabetic animals to the expression in diabetic animals treated with vanadyl sulfate ( P = −0.85). The genes identified belong to six major metabolic functional groups: lipid metabolism, oxidative stress, muscle structure, protein breakdown and biosynthesis, the complement system, and signal transduction. The identification of oxidative stress genes, coupled with the known oxidative chemistry of vanadium, implicates reactive oxygen species in the action of this class of compounds. These results imply that early transition metals or compounds formed from their chemical interactions with other metabolites may act as general transcription modulators, a role not usually associated with this class of compounds.
Collapse
Affiliation(s)
- Gail R Willsky
- Department of Biochemistry, School of Medicine and Biomedical Sciences, The State University of New York (SUNY) at Buffalo, Buffalo, New York 14214, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Ilany J, Bilan PJ, Kapur S, Caldwell JS, Patti ME, Marette A, Kahn CR. Overexpression of Rad in muscle worsens diet-induced insulin resistance and glucose intolerance and lowers plasma triglyceride level. Proc Natl Acad Sci U S A 2006; 103:4481-6. [PMID: 16537411 PMCID: PMC1450197 DOI: 10.1073/pnas.0511246103] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rad is a low molecular weight GTPase that is overexpressed in skeletal muscle of some patients with type 2 diabetes mellitus and/or obesity. Overexpression of Rad in adipocytes and muscle cells in culture results in diminished insulin-stimulated glucose uptake. To further elucidate the potential role of Rad in vivo, we have generated transgenic (tg) mice that overexpress Rad in muscle using the muscle creatine kinase (MCK) promoter-enhancer. Rad tg mice have a 6- to 12-fold increase in Rad expression in muscle as compared to wild-type littermates. Rad tg mice grow normally and have normal glucose tolerance and insulin sensitivity, but have reduced plasma triglyceride levels. On a high-fat diet, Rad tg mice develop more severe glucose intolerance than the wild-type mice; this is due to increased insulin resistance in muscle, as exemplified by a rightward shift in the dose-response curve for insulin stimulated 2-deoxyglucose uptake. There is also a unexpected further reduction of the plasma triglyceride levels that is associated with increased levels of lipoprotein lipase in the Rad tg mice. These results demonstrate a potential synergistic interaction between increased expression of Rad and high-fat diet in creation of insulin resistance and altered lipid metabolism present in type 2 diabetes.
Collapse
Affiliation(s)
- Jacob Ilany
- *Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215; and
| | - Philip J. Bilan
- *Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215; and
| | - Sonia Kapur
- Laval University, Quebec, QC, Canada G1K 7P4
| | - James S. Caldwell
- *Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215; and
| | - Mary-Elizabeth Patti
- *Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215; and
| | | | - C. Ronald Kahn
- *Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
16
|
Hawke TJ, Kanatous SB, Martin CM, Goetsch SC, Garry DJ. Rad is temporally regulated within myogenic progenitor cells during skeletal muscle regeneration. Am J Physiol Cell Physiol 2005; 290:C379-87. [PMID: 16221735 DOI: 10.1152/ajpcell.00270.2005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The successful use of myogenic progenitor cells for therapeutic applications requires an understanding of the intrinsic and extrinsic cues involved in their regulation. Herein we demonstrate the expression pattern and transcriptional regulation of Rad, a prototypical member of a family of novel Ras-related GTPases, during mammalian development and skeletal muscle regeneration. Rad was identified using microarray analysis, which revealed robust upregulation of its expression during skeletal muscle regeneration. Our current findings demonstrate negligible Rad expression with resting adult skeletal muscle; however, after muscle injury, Rad is expressed within the myogenic progenitor cell population. Rad expression is significantly increased and localized to the myogenic progenitor cell population during the early phases of regeneration and within the newly regenerated myofibers during the later phases of regeneration. Immunohistochemical analysis demonstrated that Rad and MyoD are coexpressed within the myogenic progenitor cell population of regenerating skeletal muscle. This expression profile of Rad during skeletal muscle regeneration is consistent with the proposed roles for Rad in the inhibition of L-type Ca(2+) channel activity and the inhibition of Rho/RhoA kinase activity. We also have demonstrated that known myogenic transcription factors (MEF2, MyoD, and Myf-5) can increase the transcriptional activity of the Rad promoter and that this ability is significantly enhanced by the presence of the Ca(2+)-dependent phosphatase calcineurin. Furthermore, this enhanced transcriptional activity appears to be dependent on the presence of a conserved NFAT binding motif within the Rad promoter. Taken together, these data define Rad as a novel factor within the myogenic progenitor cells of skeletal muscle and identify key regulators of its transcriptional activity.
Collapse
Affiliation(s)
- Thomas J Hawke
- School of Kinesiology and Health Science, York Univ., 4700 Keele St., Toronto, ON, Canada M3J 1P3.
| | | | | | | | | |
Collapse
|
17
|
Fu M, Zhang J, Tseng YH, Cui T, Zhu X, Xiao Y, Mou Y, De Leon H, Chang MMJ, Hamamori Y, Kahn CR, Chen YE. Rad GTPase attenuates vascular lesion formation by inhibition of vascular smooth muscle cell migration. Circulation 2005; 111:1071-7. [PMID: 15710763 DOI: 10.1161/01.cir.0000156439.55349.ad] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Rad (Ras associated with diabetes) GTPase is a prototypic member of a new subfamily of Ras-related GTPases with unique structural features, although its physiological role remains largely unknown. In the present study, we characterized the Rad function in vascular smooth muscle cells (VSMCs) and the influence of adenovirus-mediated Rad (Ad-Rad) gene delivery on vascular remodeling after experimental angioplasty. METHODS AND RESULTS We documented for the first time that neointimal formation using balloon-injured rat carotid arteries was associated with a significant increase in Rad expression as determined by immunohistochemistry and quantitative real-time reverse-transcriptase polymerase chain reaction. The levels of Rad expression in VSMCs were highly induced by platelet-derived growth factor and tumor necrosis factor-alpha. Morphometric analyses 14 days after injury revealed significantly diminished neointimal formation in the Ad-Rad-treated carotid arteries compared with Ad-GFP or PBS controls, whereas the mutated form of Rad GTPase, which can bind GDP but not GTP, increased neointimal formation. Overexpression of Rad significantly inhibited the attachment and migration of VSMCs. In addition, Rad expression dramatically reduced the formation of focal contacts and stress fibers in VSMCs by blocking the Rho/ROK signaling pathway. CONCLUSIONS Our data clearly identified Rad GTPase as a novel and critical mediator that inhibits vascular lesion formation. Manipulation of the Rad signaling pathway may provide new therapeutic approaches that will limit vascular pathological remodeling.
Collapse
Affiliation(s)
- Mingui Fu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Ga 30310, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
ROCKs, or Rho kinases, are serine/threonine kinases that are involved in many aspects of cell motility, from smooth-muscle contraction to cell migration and neurite outgrowth. Recent experiments have defined new functions of ROCKs in cells, including centrosome positioning and cell-size regulation, which might contribute to various physiological and pathological states.
Collapse
Affiliation(s)
- Kirsi Riento
- Ludwig Institute for Cancer Research, Royal Free and University College School of Medicine, 91 Riding House Street, London W1W 7BS, UK.
| | | |
Collapse
|
19
|
Kimura N, Shimada N, Ishijima Y, Fukuda M, Takagi Y, Ishikawa N. Nucleoside diphosphate kinases in mammalian signal transduction systems: recent development and perspective. J Bioenerg Biomembr 2003; 35:41-7. [PMID: 12848340 DOI: 10.1023/a:1023489722460] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The role of nucleoside diphosphate (NDP) kinase with special reference to mammalian signal transduction systems was described. The interaction between NDP kinases and G proteins was reevaluated in view of their protein structural information and its significance was extended further on the basis of recent findings obtained with small molecular weight G proteins such as Rad, menin, and Rac. Meanwhile, observations suggesting involvement of NDP kinases in the regulation of cell growth and differentiation led to the realization that NDP kinases may play a crucial role in receptor tyrosine kinase signal transduction systems. In fact, a number of experimental results, particularly obtained with PC12 cells, implicate that NDP kinases appear to regulate differentiation marker proteins and cell-cycle-associated proteins cooperatively. Consequently, we propose a hypothesis that NDP kinases might act like a molecular switch to determine the cell fate toward proliferation or differentiation in response to environmental signals.
Collapse
Affiliation(s)
- Narimichi Kimura
- Cellular Signaling Research Group, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan.
| | | | | | | | | | | |
Collapse
|
20
|
Sonna LA, Gaffin SL, Pratt RE, Cullivan ML, Angel KC, Lilly CM. Effect of acute heat shock on gene expression by human peripheral blood mononuclear cells. J Appl Physiol (1985) 2002; 92:2208-20. [PMID: 11960976 DOI: 10.1152/japplphysiol.01002.2001] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We studied the effect of heat shock on gene expression by normal human cells. Peripheral blood mononuclear cells (PBMCs) were obtained from healthy adults. Paired samples from each subject were subjected to either 20 min of heat shock (43 degrees C) or control (37 degrees C) conditions and then returned to 37 degrees C. RNA was isolated 160 min later, and five representative samples were analyzed on Affymetrix gene chip arrays containing approximately 12,600 probes. A biologically meaningful effect was defined as a statistically significant, twofold or greater difference in expression of sequences that were detected in all five experiments under control (downregulated sequences) or heat shock (upregulated sequences) conditions. Changes occurred in 395 sequences (227 increased by heat shock, 168 decreased), representing 353 Unigene numbers, in every functional category previously implicated in the heat shock response. By RT-PCR, we confirmed the findings for one upregulated sequence (Rad, a G protein) and one downregulated sequence (osteopontin, a cytokine). We conclude that heat shock causes extensive gene expression changes in PBMCs, affecting all functional categories of the heat shock response.
Collapse
Affiliation(s)
- Larry A Sonna
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, Natick, Massachusetts 01760, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Ward Y, Yap SF, Ravichandran V, Matsumura F, Ito M, Spinelli B, Kelly K. The GTP binding proteins Gem and Rad are negative regulators of the Rho-Rho kinase pathway. J Cell Biol 2002; 157:291-302. [PMID: 11956230 PMCID: PMC2199248 DOI: 10.1083/jcb.200111026] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The cytoskeletal changes that alter cellular morphogenesis and motility depend upon a complex interplay among molecules that regulate actin, myosin, and other cytoskeletal components. The Rho family of GTP binding proteins are important upstream mediators of cytoskeletal organization. Gem and Rad are members of another family of small GTP binding proteins (the Rad, Gem, and Kir family) for which biochemical functions have been mostly unknown. Here we show that Gem and Rad interface with the Rho pathway through association with the Rho effectors, Rho kinase (ROK) alpha and beta. Gem binds ROKbeta independently of RhoA in the ROKbeta coiled-coil region adjacent to the Rho binding domain. Expression of Gem inhibited ROKbeta-mediated phosphorylation of myosin light chain and myosin phosphatase, but not LIM kinase, suggesting that Gem acts by modifying the substrate specificity of ROKbeta. Gem or Rad expression led to cell flattening and neurite extension in N1E-115 neuroblastoma cells. In interference assays, Gem opposed ROKbeta- and Rad opposed ROKalpha-mediated cell rounding and neurite retraction. Gem did not oppose cell rounding initiated by ROKbeta containing a deletion of the Gem binding region, demonstrating that Gem binding to ROKbeta is required for the effects observed. In epithelial or fibroblastic cells, Gem or Rad expression resulted in stress fiber and focal adhesion disassembly. In addition, Gem reverted the anchorage-independent growth and invasiveness of Dbl-transformed fibroblasts. These results identify physiological roles for Gem and Rad in cytoskeletal regulation mediated by ROK.
Collapse
Affiliation(s)
- Yvona Ward
- Cell and Cancer Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Finlin BS, Gau CL, Murphy GA, Shao H, Kimel T, Seitz RS, Chiu YF, Botstein D, Brown PO, Der CJ, Tamanoi F, Andres DA, Perou CM. RERG is a novel ras-related, estrogen-regulated and growth-inhibitory gene in breast cancer. J Biol Chem 2001; 276:42259-67. [PMID: 11533059 DOI: 10.1074/jbc.m105888200] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Using microarray analysis, we identified a unique ras superfamily gene, termed RERG (ras-related and estrogen-regulated growth inhibitor), whose expression was decreased or lost in a significant percentage of primary human breast tumors that show a poor clinical prognosis. Importantly, high RERG expression correlated with expression of a set of genes that define a breast tumor subtype that is estrogen receptor-positive and associated with a slow rate of tumor cell proliferation and a favorable prognosis for these cancer patients. RERG mRNA expression was induced rapidly in MCF-7 cells stimulated by beta-estradiol and repressed by tamoxifen treatment. Like Ras, RERG protein exhibited intrinsic GDP/GTP binding and GTP hydrolysis activity. Unlike Ras proteins, RERG lacks a known recognition signal for COOH-terminal prenylation and was localized primarily in the cytoplasm. Expression of RERG protein in MCF-7 breast carcinoma cells resulted in a significant inhibition of both anchorage-dependent and anchorage-independent growth in vitro and inhibited tumor formation in nude mice. These features of RERG are strikingly different from most Ras superfamily GTP-binding pro-teins and suggest that the loss of RERG expression may contribute to breast tumorigenesis.
Collapse
Affiliation(s)
- B S Finlin
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Piddini E, Schmid JA, de Martin R, Dotti CG. The Ras-like GTPase Gem is involved in cell shape remodelling and interacts with the novel kinesin-like protein KIF9. EMBO J 2001; 20:4076-87. [PMID: 11483511 PMCID: PMC149163 DOI: 10.1093/emboj/20.15.4076] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Gem belongs to the Rad/Gem/Kir (RGK) subfamily of Ras-related GTPases, which also comprises Rem, Rem2 and Ges. The RGK family members Ges and Rem have been shown to produce endothelial cell sprouting and reorganization of the actin cytoskeleton upon overexpression. Here we show that high intracellular Gem levels promote profound changes in cell morphology and we investigate how this phenotype arises dynamically. We also show that this effect requires intact microtubules and microfilaments, and that Gem is associated with both cytoskeletal components. In order to investigate the mechanisms of Gem recruitment to the cytoskeleton, we performed a yeast two-hybrid screen and identified a novel kinesin-like protein, termed KIF9, as a new Gem interacting partner. We further show that Gem and KIF9 interact by co-immunoprecipitation. Furthermore, Gem and KIF9 display identical patterns of gene expression in different tissues and developmental stages. The Gem- KIF9 interaction reported here is the first molecular link between RGK family members and the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Eugenia Piddini
- EMBL, Cell Biology and Biophysics Programme, Meyerhofstrasse 1, 69117 Heidelberg, Germany, Department of Vascular Biology and Thrombosis Research, University of Vienna, Vienna International Research Cooperation Centre, A-1235 Vienna, Austria and Cavaliere Ottolenghi Scientific Institute, Università degli Studi di Torino A.O. San Luigi Gonzaga Regione Gonzole, 10, I-10043 Orbassano (TO), Italy Corresponding authors e-mail: or
E.Piddini and J.A.Schmid contributed equally to this work
| | - Johannes A. Schmid
- EMBL, Cell Biology and Biophysics Programme, Meyerhofstrasse 1, 69117 Heidelberg, Germany, Department of Vascular Biology and Thrombosis Research, University of Vienna, Vienna International Research Cooperation Centre, A-1235 Vienna, Austria and Cavaliere Ottolenghi Scientific Institute, Università degli Studi di Torino A.O. San Luigi Gonzaga Regione Gonzole, 10, I-10043 Orbassano (TO), Italy Corresponding authors e-mail: or
E.Piddini and J.A.Schmid contributed equally to this work
| | - Rainer de Martin
- EMBL, Cell Biology and Biophysics Programme, Meyerhofstrasse 1, 69117 Heidelberg, Germany, Department of Vascular Biology and Thrombosis Research, University of Vienna, Vienna International Research Cooperation Centre, A-1235 Vienna, Austria and Cavaliere Ottolenghi Scientific Institute, Università degli Studi di Torino A.O. San Luigi Gonzaga Regione Gonzole, 10, I-10043 Orbassano (TO), Italy Corresponding authors e-mail: or
E.Piddini and J.A.Schmid contributed equally to this work
| | - Carlos G. Dotti
- EMBL, Cell Biology and Biophysics Programme, Meyerhofstrasse 1, 69117 Heidelberg, Germany, Department of Vascular Biology and Thrombosis Research, University of Vienna, Vienna International Research Cooperation Centre, A-1235 Vienna, Austria and Cavaliere Ottolenghi Scientific Institute, Università degli Studi di Torino A.O. San Luigi Gonzaga Regione Gonzole, 10, I-10043 Orbassano (TO), Italy Corresponding authors e-mail: or
E.Piddini and J.A.Schmid contributed equally to this work
| |
Collapse
|
24
|
Pan JY, Fieles WE, White AM, Egerton MM, Silberstein DS. Ges, A human GTPase of the Rad/Gem/Kir family, promotes endothelial cell sprouting and cytoskeleton reorganization. J Cell Biol 2000; 149:1107-16. [PMID: 10831614 PMCID: PMC2174817 DOI: 10.1083/jcb.149.5.1107] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2000] [Accepted: 04/21/2000] [Indexed: 11/22/2022] Open
Abstract
Rad, Gem/Kir, and mRem (RGK) represent a unique GTPase family with largely unknown functions (Reynet, C., and C.R. Kahn. 1993. Science. 262:1441-1444; Cohen, L., R. Mohr, Y. Chen, M. Huang, R. Kato, D. Dorin, F. Tamanoi, A. Goga, D. Afar, N. Rosenberg, and O. Witte. Proc. Natl. Acad. Sci. USA. 1994. 91:12448-12452; Maguire, J., T. Santoro, P. Jensen, U. Siebenlist, J. Yewdell, and K. Kelly. 1994. Science. 265:241-244; Finlin, B.S., and D.A. Andres. 1997. J. Biol. Chem. 272:21982-21988). We report that Ges (GTPase regulating endothelial cell sprouting), a human RGK protein expressed in the endothelium, functions as a potent morphogenic switch in endothelial cells (ECs). Ges function is sufficient to substitute for angiogenic growth factor/extracellular matrix (ECM) signals in promoting EC sprouting, since overexpression of Ges in ECs cultured on glass leads to the development of long cytoplasmic extensions and reorganization of the actin cytoskeleton. Ges function is also necessary for Matrigel-induced EC sprouting, since this event is blocked by its dominant negative mutant, Ges(T94N), predicted to prevent the activation of endogenous Ges through sequestration of its guanine nucleotide exchange factor. Thus, Ges appears to be a key transducer linking extracellular signals to cytoskeleton/morphology changes in ECs.
Collapse
MESH Headings
- Actins/analysis
- Actins/metabolism
- Base Sequence
- Biocompatible Materials
- Blotting, Northern
- Blotting, Western
- Cells, Cultured
- Collagen
- Cytoskeleton/metabolism
- Drug Combinations
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Extracellular Matrix/metabolism
- GTP Phosphohydrolases/analysis
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Gene Expression Regulation, Enzymologic/physiology
- Growth Substances/pharmacology
- Humans
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Laminin
- Molecular Sequence Data
- Monomeric GTP-Binding Proteins/genetics
- Monomeric GTP-Binding Proteins/metabolism
- Neovascularization, Physiologic/physiology
- Proteoglycans
- RNA, Messenger/analysis
- Sequence Homology, Amino Acid
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transfection
- Umbilical Arteries/cytology
- Vinculin/analysis
- Vinculin/metabolism
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Julie Y. Pan
- Enabling Science and Technology-Biology, AstraZeneca Pharmaceuticals, Wilmington, Delaware 19850-5437
| | - William E. Fieles
- Enabling Science and Technology-Biology, AstraZeneca Pharmaceuticals, Wilmington, Delaware 19850-5437
| | - Anne M. White
- Department of Cancer and Infection, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield, Cheshire, SK10 4TG United Kingdom
| | - Mark M. Egerton
- Department of Cancer and Infection, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield, Cheshire, SK10 4TG United Kingdom
| | - David S. Silberstein
- Enabling Science and Technology-Biology, AstraZeneca Pharmaceuticals, Wilmington, Delaware 19850-5437
| |
Collapse
|