1
|
Fu L, Adu-Amankwaah J, Sang L, Tang Z, Gong Z, Zhang X, Li T, Sun H. Gender differences in GRK2 in cardiovascular diseases and its interactions with estrogen. Am J Physiol Cell Physiol 2023; 324:C505-C516. [PMID: 36622065 DOI: 10.1152/ajpcell.00407.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
G protein-coupled receptor kinase 2 (GRK2) is a multifunctional protein involved in regulating G protein-coupled receptor (GPCR) and non-GPCR signaling in the body. In the cardiovascular system, increased expression of GRK2 has been implicated in the occurrence and development of several cardiovascular diseases (CVDs). Recent studies have found gender differences in GRK2 in the cardiovascular system under physiological and pathological conditions, where GRK2's expression and activity are increased in males than in females. The incidence of CVDs in premenopausal women is lower than in men of the same age, which is related to estrogen levels. Given the shared location of GRK2 and estrogen receptors, estrogen may interact with GRK2 by modulating vital molecules such as calmodulin (CaM), caveolin, RhoA, nitrate oxide (NO), and mouse double minute 2 homolog (Mdm2), via signaling pathways mediated by estrogen's genomic (ERα and ERβ), and non-genomic (GPER) receptors, conferring cardiovascular protection in females. Highlighting the gender differences in GRK2 and understanding its interaction with estrogen in the cardiovascular system is pertinent in treating gender-related CVDs. As a result, this article explores the gender differences of GRK2 in the cardiovascular system and its relationship with estrogen during disease conditions. Estrogen's protective and therapeutic effects and its mechanism on GRK2-related cardiovascular diseases have also been discussed.
Collapse
Affiliation(s)
- Lu Fu
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Lili Sang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China.,National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Ziqing Tang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zheng Gong
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China.,School of Public Affairs & Governance, Silliman University, Dumaguete, Philippines
| | - Xiaoyan Zhang
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Tao Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| |
Collapse
|
2
|
Svec KV, Howe AK. Protein Kinase A in cellular migration—Niche signaling of a ubiquitous kinase. Front Mol Biosci 2022; 9:953093. [PMID: 35959460 PMCID: PMC9361040 DOI: 10.3389/fmolb.2022.953093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022] Open
Abstract
Cell migration requires establishment and maintenance of directional polarity, which in turn requires spatial heterogeneity in the regulation of protrusion, retraction, and adhesion. Thus, the signaling proteins that regulate these various structural processes must also be distinctly regulated in subcellular space. Protein Kinase A (PKA) is a ubiquitous serine/threonine kinase involved in innumerable cellular processes. In the context of cell migration, it has a paradoxical role in that global inhibition or activation of PKA inhibits migration. It follows, then, that the subcellular regulation of PKA is key to bringing its proper permissive and restrictive functions to the correct parts of the cell. Proper subcellular regulation of PKA controls not only when and where it is active but also specifies the targets for that activity, allowing the cell to use a single, promiscuous kinase to exert distinct functions within different subcellular niches to facilitate cell movement. In this way, understanding PKA signaling in migration is a study in context and in the elegant coordination of distinct functions of a single protein in a complex cellular process.
Collapse
Affiliation(s)
- Kathryn V. Svec
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Alan K. Howe
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, V T, United States
- University of Vermont Cancer Center, University of Vermont, Burlington, VT, United States
- *Correspondence: Alan K. Howe,
| |
Collapse
|
3
|
Schuster T, Geiger H. Septins in Stem Cells. Front Cell Dev Biol 2021; 9:801507. [PMID: 34957123 PMCID: PMC8695968 DOI: 10.3389/fcell.2021.801507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/24/2021] [Indexed: 12/01/2022] Open
Abstract
Septins were first described in yeast. Due to extensive research in non-yeast cells, Septins are now recognized across all species as important players in the regulation of the cytoskeleton, in the establishment of polarity, for migration, vesicular trafficking and scaffolding. Stem cells are primarily quiescent cells, and this actively maintained quiescent state is critical for proper stem cell function. Equally important though, stem cells undergo symmetric or asymmetric division, which is likely linked to the level of symmetry found in the mother stem cell. Due to the ability to organize barriers and be able to break symmetry in cells, Septins are thought to have a significant impact on organizing quiescence as well as the mode (symmetric vs asymmetric) of stem cell division to affect self-renewal versus differentiation. Mechanisms of regulating mammalian quiescence and symmetry breaking by Septins are though still somewhat elusive. Within this overview article, we summarize current knowledge on the role of Septins in stem cells ranging from yeast to mice especially with respect to quiescence and asymmetric division, with a special focus on hematopoietic stem cells.
Collapse
Affiliation(s)
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
4
|
Electrostatic Forces Mediate the Specificity of RHO GTPase-GDI Interactions. Int J Mol Sci 2021; 22:ijms222212493. [PMID: 34830380 PMCID: PMC8622166 DOI: 10.3390/ijms222212493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 01/13/2023] Open
Abstract
Three decades of research have documented the spatiotemporal dynamics of RHO family GTPase membrane extraction regulated by guanine nucleotide dissociation inhibitors (GDIs), but the interplay of the kinetic mechanism and structural specificity of these interactions is as yet unresolved. To address this, we reconstituted the GDI-controlled spatial segregation of geranylgeranylated RHO protein RAC1 in vitro. Various biochemical and biophysical measurements provided unprecedented mechanistic details for GDI function with respect to RHO protein dynamics. We determined that membrane extraction of RHO GTPases by GDI occurs via a 3-step mechanism: (1) GDI non-specifically associates with the switch regions of the RHO GTPases; (2) an electrostatic switch determines the interaction specificity between the C-terminal polybasic region of RHO GTPases and two distinct negatively-charged clusters of GDI1; (3) a non-specific displacement of geranylgeranyl moiety from the membrane sequesters it into a hydrophobic cleft, effectively shielding it from the aqueous milieu. This study substantially extends the model for the mechanism of GDI-regulated RHO GTPase extraction from the membrane, and could have implications for clinical studies and drug development.
Collapse
|
5
|
Ménesi D, Klement É, Ferenc G, Fehér A. The Arabidopsis Rho of Plants GTPase ROP1 Is a Potential Calcium-Dependent Protein Kinase (CDPK) Substrate. PLANTS (BASEL, SWITZERLAND) 2021; 10:2053. [PMID: 34685862 PMCID: PMC8539224 DOI: 10.3390/plants10102053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/25/2021] [Indexed: 11/16/2022]
Abstract
Plant Rho-type GTPases (ROPs) are versatile molecular switches involved in a number of signal transduction pathways. Although it is well known that they are indirectly linked to protein kinases, our knowledge about their direct functional interaction with upstream or downstream protein kinases is scarce. It is reasonable to suppose that similarly to their animal counterparts, ROPs might also be regulated by phosphorylation. There is only, however, very limited experimental evidence to support this view. Here, we present the analysis of two potential phosphorylation sites of AtROP1 and two types of potential ROP-kinases. The S74 site of AtROP1 has been previously shown to potentially regulate AtROP1 activation dependent on its phosphorylation state. However, the kinase phosphorylating this evolutionarily conserved site could not be identified: we show here that despite of the appropriate phosphorylation site consensus sequences around S74 neither the selected AGC nor CPK kinases phosphorylate S74 of AtROP1 in vitro. However, we identified several phosphorylation sites other than S74 for the CPK17 and 34 kinases in AtROP1. One of these sites, S97, was tested for biological relevance. Although the mutation of S97 to alanine (which cannot be phosphorylated) or glutamic acid (which mimics phosphorylation) somewhat altered the protein interaction strength of AtROP1 in yeast cells, the mutant proteins did not modify pollen tube growth in an in vivo test.
Collapse
Affiliation(s)
- Dalma Ménesi
- Institute of Plant Biology, Biological Research Centre of the Eötvös Lóránd Research Network, 6726 Szeged, Hungary; (D.M.); (G.F.)
| | - Éva Klement
- Laboratory of Proteomics Research, Biological Research Centre of the Eötvös Lóránd Research Network, 6726 Szeged, Hungary; or
- Single Cell Omics ACF, Hungarian Centre of Excellence for Molecular Medicine, 6726 Szeged, Hungary
| | - Györgyi Ferenc
- Institute of Plant Biology, Biological Research Centre of the Eötvös Lóránd Research Network, 6726 Szeged, Hungary; (D.M.); (G.F.)
| | - Attila Fehér
- Institute of Plant Biology, Biological Research Centre of the Eötvös Lóránd Research Network, 6726 Szeged, Hungary; (D.M.); (G.F.)
- Department of Plant Biology, University of Szeged, 6726 Szeged, Hungary
| |
Collapse
|
6
|
Capote AE, Batra A, Warren CM, Chowdhury SAK, Wolska BM, Solaro RJ, Rosas PC. B-arrestin-2 Signaling Is Important to Preserve Cardiac Function During Aging. Front Physiol 2021; 12:696852. [PMID: 34512376 PMCID: PMC8430342 DOI: 10.3389/fphys.2021.696852] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Experiments reported here tested the hypothesis that β-arrestin-2 is an important element in the preservation of cardiac function during aging. We tested this hypothesis by aging β-arrestin-2 knock-out (KO) mice, and wild-type equivalent (WT) to 12-16months. We developed the rationale for these experiments on the basis that angiotensin II (ang II) signaling at ang II receptor type 1 (AT1R), which is a G-protein coupled receptor (GPCR) promotes both G-protein signaling as well as β-arrestin-2 signaling. β-arrestin-2 participates in GPCR desensitization, internalization, but also acts as a scaffold for adaptive signal transduction that may occur independently or in parallel to G-protein signaling. We have previously reported that biased ligands acting at the AT1R promote β-arrestin-2 signaling increasing cardiac contractility and reducing maladaptations in a mouse model of dilated cardiomyopathy. Although there is evidence that ang II induces maladaptive senescence in the cardiovascular system, a role for β-arrestin-2 signaling has not been studied in aging. By echocardiography, we found that compared to controls aged KO mice exhibited enlarged left atria and left ventricular diameters as well as depressed contractility parameters with preserved ejection fraction. Aged KO also exhibited depressed relaxation parameters when compared to WT controls at the same age. Moreover, cardiac dysfunction in aged KO mice was correlated with alterations in the phosphorylation of myofilament proteins, such as cardiac myosin binding protein-C, and myosin regulatory light chain. Our evidence provides novel insights into a role for β-arrestin-2 as an important signaling mechanism that preserves cardiac function during aging.
Collapse
Affiliation(s)
- Andrielle E. Capote
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - Ashley Batra
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - Chad M. Warren
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - Shamim A. K. Chowdhury
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - Beata M. Wolska
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
- Department of Medicine, Division of Cardiology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - R. John Solaro
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| | - Paola C. Rosas
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
7
|
Mosaddeghzadeh N, Ahmadian MR. The RHO Family GTPases: Mechanisms of Regulation and Signaling. Cells 2021; 10:1831. [PMID: 34359999 PMCID: PMC8305018 DOI: 10.3390/cells10071831] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022] Open
Abstract
Much progress has been made toward deciphering RHO GTPase functions, and many studies have convincingly demonstrated that altered signal transduction through RHO GTPases is a recurring theme in the progression of human malignancies. It seems that 20 canonical RHO GTPases are likely regulated by three GDIs, 85 GEFs, and 66 GAPs, and eventually interact with >70 downstream effectors. A recurring theme is the challenge in understanding the molecular determinants of the specificity of these four classes of interacting proteins that, irrespective of their functions, bind to common sites on the surface of RHO GTPases. Identified and structurally verified hotspots as functional determinants specific to RHO GTPase regulation by GDIs, GEFs, and GAPs as well as signaling through effectors are presented, and challenges and future perspectives are discussed.
Collapse
Affiliation(s)
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich Heine University, Universitätsstrasse 1, Building 22.03.05, 40225 Düsseldorf, Germany;
| |
Collapse
|
8
|
Molecular subversion of Cdc42 signalling in cancer. Biochem Soc Trans 2021; 49:1425-1442. [PMID: 34196668 PMCID: PMC8412110 DOI: 10.1042/bst20200557] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Cdc42 is a member of the Rho family of small GTPases and a master regulator of the actin cytoskeleton, controlling cell motility, polarity and cell cycle progression. This small G protein and its regulators have been the subject of many years of fruitful investigation and the advent of functional genomics and proteomics has opened up new avenues of exploration including how it functions at specific locations in the cell. This has coincided with the introduction of new structural techniques with the ability to study small GTPases in the context of the membrane. The role of Cdc42 in cancer is well established but the molecular details of its action are still being uncovered. Here we review alterations found to Cdc42 itself and to key components of the signal transduction pathways it controls in cancer. Given the challenges encountered with targeting small G proteins directly therapeutically, it is arguably the regulators of Cdc42 and the effector signalling pathways downstream of the small G protein which will be the most tractable targets for therapeutic intervention. These will require interrogation in order to fully understand the global signalling contribution of Cdc42, unlock the potential for mapping new signalling axes and ultimately produce inhibitors of Cdc42 driven signalling.
Collapse
|
9
|
Vicente-Soler J, Soto T, Franco A, Cansado J, Madrid M. The Multiple Functions of Rho GTPases in Fission Yeasts. Cells 2021; 10:1422. [PMID: 34200466 PMCID: PMC8228308 DOI: 10.3390/cells10061422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 01/20/2023] Open
Abstract
The Rho family of GTPases represents highly conserved molecular switches involved in a plethora of physiological processes. Fission yeast Schizosaccharomyces pombe has become a fundamental model organism to study the functions of Rho GTPases over the past few decades. In recent years, another fission yeast species, Schizosaccharomyces japonicus, has come into focus offering insight into evolutionary changes within the genus. Both fission yeasts contain only six Rho-type GTPases that are spatiotemporally controlled by multiple guanine-nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs), and whose intricate regulation in response to external cues is starting to be uncovered. In the present review, we will outline and discuss the current knowledge and recent advances on how the fission yeasts Rho family GTPases regulate essential physiological processes such as morphogenesis and polarity, cellular integrity, cytokinesis and cellular differentiation.
Collapse
Affiliation(s)
| | | | | | - José Cansado
- Yeast Physiology Group, Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.V.-S.); (T.S.); (A.F.)
| | - Marisa Madrid
- Yeast Physiology Group, Departamento de Genética y Microbiología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain; (J.V.-S.); (T.S.); (A.F.)
| |
Collapse
|
10
|
Machin PA, Tsonou E, Hornigold DC, Welch HCE. Rho Family GTPases and Rho GEFs in Glucose Homeostasis. Cells 2021; 10:cells10040915. [PMID: 33923452 PMCID: PMC8074089 DOI: 10.3390/cells10040915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of glucose homeostasis leading to metabolic syndrome and type 2 diabetes is the cause of an increasing world health crisis. New intriguing roles have emerged for Rho family GTPases and their Rho guanine nucleotide exchange factor (GEF) activators in the regulation of glucose homeostasis. This review summates the current knowledge, focusing in particular on the roles of Rho GEFs in the processes of glucose-stimulated insulin secretion by pancreatic β cells and insulin-stimulated glucose uptake into skeletal muscle and adipose tissues. We discuss the ten Rho GEFs that are known so far to regulate glucose homeostasis, nine of which are in mammals, and one is in yeast. Among the mammalian Rho GEFs, P-Rex1, Vav2, Vav3, Tiam1, Kalirin and Plekhg4 were shown to mediate the insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane and/or insulin-stimulated glucose uptake in skeletal muscle or adipose tissue. The Rho GEFs P-Rex1, Vav2, Tiam1 and β-PIX were found to control the glucose-stimulated release of insulin by pancreatic β cells. In vivo studies demonstrated the involvement of the Rho GEFs P-Rex2, Vav2, Vav3 and PDZ-RhoGEF in glucose tolerance and/or insulin sensitivity, with deletion of these GEFs either contributing to the development of metabolic syndrome or protecting from it. This research is in its infancy. Considering that over 80 Rho GEFs exist, it is likely that future research will identify more roles for Rho GEFs in glucose homeostasis.
Collapse
Affiliation(s)
- Polly A. Machin
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
| | - Elpida Tsonou
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - David C. Hornigold
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - Heidi C. E. Welch
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Correspondence: ; Tel.: +44-(0)1223-496-596
| |
Collapse
|
11
|
Batra A, Warren CM, Ke Y, McCann M, Halas M, Capote AE, Liew CW, Solaro RJ, Rosas PC. Deletion of P21-activated kinase-1 induces age-dependent increased visceral adiposity and cardiac dysfunction in female mice. Mol Cell Biochem 2021; 476:1337-1349. [PMID: 33389497 PMCID: PMC7925422 DOI: 10.1007/s11010-020-03993-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
It is known that there is an age-related progression in diastolic dysfunction, especially prevalent in postmenopausal women, who develop heart failure with preserved ejection fraction (HFpEF, EF > 50%). Mechanisms and therapies are poorly understood, but there are strong correlations between obesity and HFpEF. We have tested the hypothesis that P21-activated kinase-1 (PAK1) preserves cardiac function and adipose tissue homeostasis during aging in female mice. Previous demonstrations in male mice by our lab that PAK1 activity confers cardio-protection against different stresses formed the rationale for this hypothesis. Our studies compared young (3-6 months) and middle-aged (12-15 months) female and male PAK1 knock-out mice (PAK1-/-) and wild-type (WT) equivalent. Female WT mice exhibited increased cardiac PAK1 abundance during aging. By echocardiography, compared to young WT female mice, middle-aged WT female mice showed enlargement of the left atrium as well as thickening of posterior wall and increased left ventricular mass; however, all contraction and relaxation parameters were preserved during aging. Compared to WT controls, middle-aged PAK1-/- female mice demonstrated worsening of cardiac function involving a greater enlargement of the left atrium, ventricular hypertrophy, and diastolic dysfunction. Moreover, with aging PAK1-/- female mice, unlike male PAK1-/- mice, exhibited increased adiposity with increased accumulation of visceral adipose tissue. Our data provide evidence for the significance of PAK1 signaling as an element in the preservation of cardiac function and adipose tissue homeostasis in females during aging.
Collapse
Affiliation(s)
- Ashley Batra
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Chad M Warren
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Yunbo Ke
- Department of Anesthesiology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Maximilian McCann
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Monika Halas
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrielle E Capote
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Chong Wee Liew
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - R John Solaro
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Paola C Rosas
- Department of Physiology & Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Ngo ATP, Parra-Izquierdo I, Aslan JE, McCarty OJT. Rho GTPase regulation of reactive oxygen species generation and signalling in platelet function and disease. Small GTPases 2021; 12:440-457. [PMID: 33459160 DOI: 10.1080/21541248.2021.1878001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
13
|
MMP-9 Signaling Pathways That Engage Rho GTPases in Brain Plasticity. Cells 2021; 10:cells10010166. [PMID: 33467671 PMCID: PMC7830260 DOI: 10.3390/cells10010166] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
The extracellular matrix (ECM) has been identified as a critical factor affecting synaptic function. It forms a functional scaffold that provides both the structural support and the reservoir of signaling molecules necessary for communication between cellular constituents of the central nervous system (CNS). Among numerous ECM components and modifiers that play a role in the physiological and pathological synaptic plasticity, matrix metalloproteinase 9 (MMP-9) has recently emerged as a key molecule. MMP-9 may contribute to the dynamic remodeling of structural and functional plasticity by cleaving ECM components and cell adhesion molecules. Notably, MMP-9 signaling was shown to be indispensable for long-term memory formation that requires synaptic remodeling. The core regulators of the dynamic reorganization of the actin cytoskeleton and cell adhesion are the Rho family of GTPases. These proteins have been implicated in the control of a wide range of cellular processes occurring in brain physiology and pathology. Here, we discuss the contribution of Rho GTPases to MMP-9-dependent signaling pathways in the brain. We also describe how the regulation of Rho GTPases by post-translational modifications (PTMs) can influence these processes.
Collapse
|
14
|
Agbaegbu Iweka C, Hussein RK, Yu P, Katagiri Y, Geller HM. The lipid phosphatase-like protein PLPPR1 associates with RhoGDI1 to modulate RhoA activation in response to axon growth inhibitory molecules. J Neurochem 2021; 157:494-507. [PMID: 33320336 DOI: 10.1111/jnc.15271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 11/29/2022]
Abstract
Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1) is a member of a family of lipid phosphatase related proteins, integral membrane proteins characterized by six transmembrane domains. This family of proteins is enriched in the brain and recent data indicate potential pleiotropic functions in several different contexts. An inherent ability of this family of proteins is to induce morphological changes, and we have previously reported that members of this family interact with each other and may function co-operatively. However, the function of PLPPR1 is not yet understood. Here we show that the expression of PLPPR1 reduces the inhibition of neurite outgrowth of cultured mouse hippocampal neurons by chondroitin sulfate proteoglycans and the retraction of neurites of Neuro-2a cells by lysophosphatidic acid (LPA). Further, we show that PLPPR1 reduces the activation of Ras homolog family member A (RhoA) by LPA in Neuro-2a cells, and that this is because of an association of PLPPR1with the Rho-specific guanine nucleotide dissociation inhibitor (RhoGDI1). These results establish a novel signaling pathway for the PLPPR1 protein.
Collapse
Affiliation(s)
- Chinyere Agbaegbu Iweka
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA.,Department of Neuroscience, Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
| | - Rowan K Hussein
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yasuhiro Katagiri
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, National Heart Lung and Blood Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
15
|
Barreto N, Caballero M, Bonfanti AP, de Mato FCP, Munhoz J, da Rocha-E-Silva TAA, Sutti R, Vitorino-Araujo JL, Verinaud L, Rapôso C. Spider venom components decrease glioblastoma cell migration and invasion through RhoA-ROCK and Na +/K +-ATPase β2: potential molecular entities to treat invasive brain cancer. Cancer Cell Int 2020; 20:576. [PMID: 33327966 PMCID: PMC7745393 DOI: 10.1186/s12935-020-01643-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Glioblastoma (GB) cells have the ability to migrate and infiltrate the normal parenchyma, leading to the formation of recurrent tumors often adjacent to the surgical extraction site. We recently showed that Phoneutria nigriventer spider venom (PnV) has anticancer effects mainly on the migration of human GB cell lines (NG97 and U-251). The present work aimed to investigate the effects of isolated components from the venom on migration, invasiveness, morphology and adhesion of GB cells, also evaluating RhoA-ROCK signaling and Na+/K+-ATPase β2 (AMOG) involvement. METHODS Human (NG97) GB cells were treated with twelve subfractions (SFs-obtained by HPLC from PnV). Migration and invasion were evaluated by scratch wound healing and transwell assays, respectively. Cell morphology and actin cytoskeleton were shown by GFAP and phalloidin labeling. The assay with fibronectin coated well plate was made to evaluate cell adhesion. Western blotting demonstrated ROCK and AMOG levels and a ROCK inhibitor was used to verify the involvement of this pathway. Values were analyzed by the GraphPad Prism software package and the level of significance was determinate using one-way analysis of variance (ANOVA) followed by Dunnett's multiple comparisons test. RESULTS Two (SF1 and SF11) of twelve SFs, decreased migration and invasion compared to untreated control cells. Both SFs also altered actin cytoskeleton, changed cell morphology and reduced adhesion. SF1 and SF11 increased ROCK expression and the inhibition of this protein abolished the effects of both subfractions on migration, morphology and adhesion (but not on invasion). SF11 also increased Na+/K+-ATPase β2. CONCLUSION All components of the venom were evaluated and two SFs were able to impair human glioblastoma cells. The RhoA effector, ROCK, was shown to be involved in the mechanisms of both PnV components. It is possible that AMOG mediates the effect of SF11 on the invasion. Further investigations to isolate and biochemically characterize the molecules are underway.
Collapse
Affiliation(s)
- Natália Barreto
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | - Marcus Caballero
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | - Amanda Pires Bonfanti
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | - Felipe Cezar Pinheiro de Mato
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | - Jaqueline Munhoz
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.,Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | | | - Rafael Sutti
- Faculdade de Ciências Médicas, Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - João Luiz Vitorino-Araujo
- Disciplina de Neurocirurgia, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Liana Verinaud
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, UNICAMP, São Paulo, Brazil
| | - Catarina Rapôso
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo, 13083-865, Brazil.
| |
Collapse
|
16
|
Lachowski D, Cortes E, Matellan C, Rice A, Lee DA, Thorpe SD, del Río Hernández AE. G Protein-Coupled Estrogen Receptor Regulates Actin Cytoskeleton Dynamics to Impair Cell Polarization. Front Cell Dev Biol 2020; 8:592628. [PMID: 33195261 PMCID: PMC7649801 DOI: 10.3389/fcell.2020.592628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Mechanical forces regulate cell functions through multiple pathways. G protein-coupled estrogen receptor (GPER) is a seven-transmembrane receptor that is ubiquitously expressed across tissues and mediates the acute cellular response to estrogens. Here, we demonstrate an unidentified role of GPER as a cellular mechanoregulator. G protein-coupled estrogen receptor signaling controls the assembly of stress fibers, the dynamics of the associated focal adhesions, and cell polarization via RhoA GTPase (RhoA). G protein-coupled estrogen receptor activation inhibits F-actin polymerization and subsequently triggers a negative feedback that transcriptionally suppresses the expression of monomeric G-actin. Given the broad expression of GPER and the range of cytoskeletal changes modulated by this receptor, our findings position GPER as a key player in mechanotransduction.
Collapse
Affiliation(s)
- Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| | - David A. Lee
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, London, United Kingdom
| | - Stephen D. Thorpe
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, London, United Kingdom
- UCD School of Medicine, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Dysregulation of Rho GTPases in Human Cancers. Cancers (Basel) 2020; 12:cancers12051179. [PMID: 32392742 PMCID: PMC7281333 DOI: 10.3390/cancers12051179] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 01/28/2023] Open
Abstract
Rho GTPases play central roles in numerous cellular processes, including cell motility, cell polarity, and cell cycle progression, by regulating actin cytoskeletal dynamics and cell adhesion. Dysregulation of Rho GTPase signaling is observed in a broad range of human cancers, and is associated with cancer development and malignant phenotypes, including metastasis and chemoresistance. Rho GTPase activity is precisely controlled by guanine nucleotide exchange factors, GTPase-activating proteins, and guanine nucleotide dissociation inhibitors. Recent evidence demonstrates that it is also regulated by post-translational modifications, such as phosphorylation, ubiquitination, and sumoylation. Here, we review the current knowledge on the role of Rho GTPases, and the precise mechanisms controlling their activity in the regulation of cancer progression. In addition, we discuss targeting strategies for the development of new drugs to improve cancer therapy.
Collapse
|
18
|
Rice A, Cortes E, Lachowski D, Oertle P, Matellan C, Thorpe SD, Ghose R, Wang H, Lee DA, Plodinec M, del Río Hernández AE. GPER Activation Inhibits Cancer Cell Mechanotransduction and Basement Membrane Invasion via RhoA. Cancers (Basel) 2020; 12:E289. [PMID: 31991740 PMCID: PMC7073197 DOI: 10.3390/cancers12020289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 01/11/2023] Open
Abstract
The invasive properties of cancer cells are intimately linked to their mechanical phenotype, which can be regulated by intracellular biochemical signalling. Cell contractility, induced by mechanotransduction of a stiff fibrotic matrix, and the epithelial-mesenchymal transition (EMT) promote invasion. Metastasis involves cells pushing through the basement membrane into the stroma-both of which are altered in composition with cancer progression. Agonists of the G protein-coupled oestrogen receptor (GPER), such as tamoxifen, have been largely used in the clinic, and interest in GPER, which is abundantly expressed in tissues, has greatly increased despite a lack of understanding regarding the mechanisms which promote its multiple effects. Here, we show that specific activation of GPER inhibits EMT, mechanotransduction and cell contractility in cancer cells via the GTPase Ras homolog family member A (RhoA). We further show that GPER activation inhibits invasion through an in vitro basement membrane mimic, similar in structure to the pancreatic basement membrane that we reveal as an asymmetric bilayer, which differs in composition between healthy and cancer patients.
Collapse
Affiliation(s)
- Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
| | - Ernesto Cortes
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
| | - Dariusz Lachowski
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
| | - Philipp Oertle
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland;
| | - Carlos Matellan
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
| | - Stephen D. Thorpe
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK; (S.D.T.); (D.A.L.)
| | - Ritobrata Ghose
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain;
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China;
| | - David A. Lee
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK; (S.D.T.); (D.A.L.)
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, 4056 Basel, Switzerland;
| | - Armando E. del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (A.R.); (E.C.); (D.L.); (C.M.)
| |
Collapse
|
19
|
Bober P, Alexovič M, Tomková Z, Kilík R, Sabo J. RHOA and mDia1 Promotes Apoptosis of Breast Cancer Cells Via a High Dose of Doxorubicin Treatment. Open Life Sci 2019; 14:619-627. [PMID: 33817200 PMCID: PMC7874778 DOI: 10.1515/biol-2019-0070] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 09/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background Transforming RhoA proteins (RHOA) and their downstream Diaphanous homolog 1 proteins (DIAPH1) or mDia1 participate in the regulation of actin cytoskeleton which plays critical role in cells, i.e., morphologic changes and apoptosis. Methodology To determine the cell viability the real time cell analysis (RTCA) and flow cytometry were used. To perform proteomic analysis, the label-free quantitative method and post-translation modification by the nano-HPLC and ESI-MS ion trap mass analyser were used. Results The results of the cell viability showed an increase of dead cells (around 30 %) in MCF-7/DOX-1 (i.e., 1μM of doxorubicin was added to MCF-7/WT breast cancer cell line) compared to MCF-7/WT (control) after 24 h doxorubicin (DOX) treatment. The signalling pathway of the Regulation of actin cytoskeleton (p<0.0026) was determined, where RHOA and mDia1 proteins were up-regulated. Also, post-translational modification analysis of these proteins in MCF-7/DOX-1 cells revealed dysregulation of the actin cytoskeleton, specifically the collapse of actin stress fibbers due to phosphorylation of RHOA at serine 188 and mDia1 at serine 22, resulting in their deactivation and cell apoptosis. Conclusion These results pointed to an assumed role of DOX to dysregulation of actin cytoskeleton and cell death.
Collapse
Affiliation(s)
- Peter Bober
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Zuzana Tomková
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Róbert Kilík
- 1st Department of Surgery, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P.J. Šafárik in Košice, Trieda SNP1, 04011 Košice, Slovakia
| |
Collapse
|
20
|
Lažetić V, Joseph BB, Bernazzani SM, Fay DS. Actin organization and endocytic trafficking are controlled by a network linking NIMA-related kinases to the CDC-42-SID-3/ACK1 pathway. PLoS Genet 2018; 14:e1007313. [PMID: 29608564 PMCID: PMC5897031 DOI: 10.1371/journal.pgen.1007313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 04/12/2018] [Accepted: 03/19/2018] [Indexed: 01/07/2023] Open
Abstract
Molting is an essential process in the nematode Caenorhabditis elegans during which the epidermal apical extracellular matrix, termed the cuticle, is detached and replaced at each larval stage. The conserved NIMA-related kinases NEKL-2/NEK8/NEK9 and NEKL-3/NEK6/NEK7, together with their ankyrin repeat partners, MLT-2/ANKS6, MLT-3/ANKS3, and MLT-4/INVS, are essential for normal molting. In nekl and mlt mutants, the old larval cuticle fails to be completely shed, leading to entrapment and growth arrest. To better understand the molecular and cellular functions of NEKLs during molting, we isolated genetic suppressors of nekl molting-defective mutants. Using two independent approaches, we identified CDC-42, a conserved Rho-family GTPase, and its effector protein kinase, SID-3/ACK1. Notably, CDC42 and ACK1 regulate actin dynamics in mammals, and actin reorganization within the worm epidermis has been proposed to be important for the molting process. Inhibition of NEKL-MLT activities led to strong defects in the distribution of actin and failure to form molting-specific apical actin bundles. Importantly, this phenotype was reverted following cdc-42 or sid-3 inhibition. In addition, repression of CDC-42 or SID-3 also suppressed nekl-associated defects in trafficking, a process that requires actin assembly and disassembly. Expression analyses indicated that components of the NEKL-MLT network colocalize with both actin and CDC-42 in specific regions of the epidermis. Moreover, NEKL-MLT components were required for the normal subcellular localization of CDC-42 in the epidermis as well as wild-type levels of CDC-42 activation. Taken together, our findings indicate that the NEKL-MLT network regulates actin through CDC-42 and its effector SID-3. Interestingly, we also observed that downregulation of CDC-42 in a wild-type background leads to molting defects, suggesting that there is a fine balance between NEKL-MLT and CDC-42-SID-3 activities in the epidermis.
Collapse
Affiliation(s)
- Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY
| | - Braveen B. Joseph
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY
| | - Sarina M. Bernazzani
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY
- * E-mail:
| |
Collapse
|
21
|
Lei L, Huang M, Su L, Xie D, Mamuya FA, Ham O, Tsuji K, Păunescu TG, Yang B, Lu HAJ. Manganese promotes intracellular accumulation of AQP2 via modulating F-actin polymerization and reduces urinary concentration in mice. Am J Physiol Renal Physiol 2017; 314:F306-F316. [PMID: 29046300 DOI: 10.1152/ajprenal.00391.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aquaporin-2 (AQP2) is a water channel protein expressed in principal cells (PCs) of the kidney collecting ducts (CDs) and plays a critical role in mediating water reabsorption and urine concentration. AQP2 undergoes both regulated trafficking mediated by vasopressin (VP) and constitutive recycling, which is independent of VP. For both pathways, actin cytoskeletal dynamics is a key determinant of AQP2 trafficking. We report here that manganese chloride (MnCl2) is a novel and potent regulator of AQP2 trafficking in cultured cells and in the kidney. MnCl2 treatment promoted internalization and intracellular accumulation of AQP2. The effect of MnCl2 on the intracellular accumulation of AQP2 was associated with activation of RhoA and actin polymerization without modification of AQP2 phosphorylation. Although the level of total and phosphorylated AQP2 did not change, MnCl2 treatment impeded VP-induced phosphorylation of AQP2 at its serine-256, -264, and -269 residues and dephosphorylation at serine 261. In addition, MnCl2 significantly promoted F-actin polymerization along with downregulation of RhoA activity and prevented VP-induced membrane accumulation of AQP2. Finally, MnCl2 treatment in mice resulted in significant polyuria and reduced urinary concentration, likely due to intracellular relocation of AQP2 in the PCs of kidney CDs. More importantly, the reduced urinary concentration caused by MnCl2 treatment in animals was not corrected by VP. In summary, our study identified a novel effect of MnCl2 on AQP2 trafficking through modifying RhoA activity and actin polymerization and uncovered its potent impact on water diuresis in vivo.
Collapse
Affiliation(s)
- Lei Lei
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Ming Huang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Limin Su
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China.,Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Dongping Xie
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | - Fahmy A Mamuya
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Onju Ham
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Kenji Tsuji
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Teodor G Păunescu
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University , Beijing , People's Republic of China
| | - Hua A Jenny Lu
- Program in Membrane Biology, Center for Systems Biology, and Division of Nephrology, Department of Medicine, Massachusetts General Hospital , Boston, Massachusetts.,Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
22
|
Interaction between Rho GTPases and 14-3-3 Proteins. Int J Mol Sci 2017; 18:ijms18102148. [PMID: 29036929 PMCID: PMC5666830 DOI: 10.3390/ijms18102148] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/25/2023] Open
Abstract
The Rho GTPase family accounts for as many as 20 members. Among them, the archetypes RhoA, Rac1, and Cdc42 have been the most well-characterized. Like all members of the small GTPases superfamily, Rho proteins act as molecular switches to control cellular processes by cycling between active, GTP-bound and inactive, GDP-bound states. The 14-3-3 family proteins comprise seven isoforms. They exist as dimers (homo- or hetero-dimer) in cells. They function by binding to Ser/Thr phosphorylated intracellular proteins, which alters the conformation, activity, and subcellular localization of their binding partners. Both 14-3-3 proteins and Rho GTPases regulate cell cytoskeleton remodeling and cell migration, which suggests a possible interaction between the signaling pathways regulated by these two groups of proteins. Indeed, more and more emerging evidence indicates the mutual regulation of these two signaling pathways. There have been many documented reviews of 14-3-3 protein and Rac1 separately, but there is no review regarding the interaction and mutual regulation of these two groups of proteins. Thus, in this article we thoroughly review all the reported interactions between the signaling pathways regulated by 14-3-3 proteins and Rho GTPases (mostly Rac1).
Collapse
|
23
|
Smith CA, Miner AS, Barbee RW, Ratz PH. Metabolic Stress-Induced Activation of AMPK and Inhibition of Constitutive Phosphoproteins Controlling Smooth Muscle Contraction: Evidence for Smooth Muscle Fatigue? Front Physiol 2017; 8:681. [PMID: 28943852 PMCID: PMC5596101 DOI: 10.3389/fphys.2017.00681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/24/2017] [Indexed: 11/23/2022] Open
Abstract
Metabolic stress diminishes smooth muscle contractile strength by a poorly defined mechanism. To test the hypothesis that metabolic stress activates a compensatory cell signaling program to reversibly downregulate contraction, arterial rings and bladder muscle strips in vitro were deprived of O2 and glucose for 30 and 60 min (“starvation”) to induce metabolic stress, and the phosphorylation status of proteins involved in regulation of contraction and metabolic stress were assessed in tissues under basal and stimulated conditions. A 15–30 min recovery period (O2 and glucose repletion) tested whether changes induced by starvation were reversible. Starvation decreased basal phosphorylation of myosin regulatory light chain (MLC-pS19) and of the rho kinase (ROCK) downstream substrates cofilin (cofilin-pS3) and myosin phosphatase targeting subunit MYPT1 (MYPT1-pT696 and MYPT1-pT853), and abolished the ability of contractile stimuli to cause a strong, sustained contraction. Starvation increased basal phosphorylation of AMPK (AMPK-pT172) and 3 downstream AMPK substrates, acetyl-CoA carboxylase (ACC-pS79), rhoA (rhoA-pS188), and phospholamban (PLB-pS16). Increases in rhoA-pS188 and PLB-pS16 would be expected to inhibit contraction. Recovery restored basal AMPK-pT172 and MLC-pS19 to control levels, and restored contraction. In AMPKα2 deficient mice (AMPKα2-/-), the basal level of AMPK-pT172 was reduced by 50%, and MLC-pS19 was elevated by 50%, but AMPKα2-/- did not prevent starvation-induced contraction inhibition nor enhance recovery from starvation. These results indicate that constitutive AMPK activity participates in constitutive regulation of contractile proteins, and suggest that AMPK activation is necessary, but may not be sufficient, to cause smooth muscle contraction inhibition during metabolic stress.
Collapse
Affiliation(s)
- Corey A Smith
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Amy S Miner
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Robert W Barbee
- Departments of Emergency Medicine and Physiology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Paul H Ratz
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| |
Collapse
|
24
|
Florke RR, Young GT, Hamann MJ. Unraveling a model of TCL/RhoJ allosterism using TC10 reverse chimeras. Small GTPases 2017; 11:138-145. [PMID: 28696829 DOI: 10.1080/21541248.2017.1347599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
In addition to the classical regulation of GTPase activity by effector proteins, investigating the variations in the amino acid sequence and structures of GTPases often provides insights into regulatory mechanisms that are more GTPase-specific. TCL/RhoJ is a Rho GTPase most closely related to Cdc42 and TC10; however, its nucleotide exchange activity is distinctly influenced by N-terminal amino acids 17-20 and the more distal amino acids 121-129. In this short study, we have further explored the differences between TCL and its homolog TC10 and show that its unique mode of allosteric regulation requires broader diversification of its amino acid sequence than previously appreciated.
Collapse
Affiliation(s)
| | - Grace T Young
- Biology Department, Bemidji State University, Bemidji, MN, USA
| | | |
Collapse
|
25
|
Zhao K, Wang L, Li T, Zhu M, Zhang C, Chen L, Zhao P, Zhou H, Yu S, Yang X. The role of miR-451 in the switching between proliferation and migration in malignant glioma cells: AMPK signaling, mTOR modulation and Rac1 activation required. Int J Oncol 2017; 50:1989-1999. [PMID: 28440461 PMCID: PMC5435333 DOI: 10.3892/ijo.2017.3973] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/03/2017] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM), WHO grade IV astrocytoma, is the most common primary neoplasm of the central nervous system (CNS) and has the highest malignancy and mortality rates. The invasive nature of GBM complicates surgical resection and restricts chemotherapeutic access, contributing to poor patient prognosis. The migration of tumor cells is closely related to the tumor cell proliferation. The acquisition of migratory capability, in addition to intracellular factors, is proposed to be a crucial mechanism during the progression of invasion. Using qRT-PCR analysis, we determined that the expression of miR-451 in glioma tissue was lower than in control brain tissue, especially in the central portions of the tumor. In glioma cell lines, we found that decreased miR-451 expression suppressed tumor cell proliferation but enhanced migration with a concomitant low level of CAB39/AMPK/mTOR pathway activation and high level of Rac1/cofilin pathway activation, respectively. Notably, the effect of miR-451 on cytological behavior and on the activation of mTOR and Rac1 was limited when AMPKα1 expression was knocked-down with a synthetic shRNA. We suggest that the glioma microenvironment results in heterogeneity of miR-451 expression. Our data indicated that miR-451 relays environmental signals by upregulating the activity of AMPK signaling, thereby modulating the activation of mTOR and Rac1/cofilin which, in turn, play key roles in glioma cell proliferation and migration, respectively. Our results highlight the need to consider opposing roles of a therapeutic target which, while suppressing tumor cell proliferation, could also promote cell infiltration.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Leilei Wang
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Meng Zhu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Lei Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Pengfei Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hua Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
26
|
Kim HJ, Choi HS, Park JH, Kim MJ, Lee HG, Petersen RB, Kim YS, Park JB, Choi EK. Regulation of RhoA activity by the cellular prion protein. Cell Death Dis 2017; 8:e2668. [PMID: 28300846 PMCID: PMC5386549 DOI: 10.1038/cddis.2017.37] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/18/2016] [Accepted: 01/10/2017] [Indexed: 01/28/2023]
Abstract
The cellular prion protein (PrPC) is a highly conserved glycosylphosphatidylinositol (GPI)-anchored membrane protein that is involved in the signal transduction during the initial phase of neurite outgrowth. The Ras homolog gene family member A (RhoA) is a small GTPase that is known to have an essential role in regulating the development, differentiation, survival, and death of neurons in the central nervous system. Although recent studies have shown the dysregulation of RhoA in a variety of neurodegenerative diseases, the role of RhoA in prion pathogenesis remains unclear. Here, we investigated the regulation of RhoA-mediated signaling by PrPC using both in vitro and in vivo models and found that overexpression of PrPC significantly induced RhoA inactivation and RhoA phosphorylation in hippocampal neuronal cells and in the brains of transgenic mice. Using siRNA-mediated depletion of endogenous PrPC and overexpression of disease-associated mutants of PrPC, we confirmed that PrPC induced RhoA inactivation, which accompanied RhoA phosphorylation but reduced the phosphorylation levels of LIM kinase (LIMK), leading to cofilin activation. In addition, PrPC colocalized with RhoA, and the overexpression of PrPC significantly increased neurite outgrowth in nerve growth factor-treated PC12 cells through RhoA inactivation. However, the disease-associated mutants of PrPC decreased neurite outgrowth compared with wild-type PrPC. Moreover, inhibition of Rho-associated kinase (ROCK) substantially facilitated neurite outgrowth in NGF-treated PC12 cells, similar to the effect induced by PrPC. Interestingly, we found that the induction of RhoA inactivation occurred through the interaction of PrPC with RhoA and that PrPC enhanced the interaction between RhoA and p190RhoGAP (a GTPase-activating protein). These findings suggest that the interactions of PrPC with RhoA and p190RhoGAP contribute to neurite outgrowth by controlling RhoA inactivation and RhoA-mediated signaling and that disease-associated mutations of PrPC impair RhoA inactivation, which in turn leads to prion-related neurodegeneration.
Collapse
Affiliation(s)
- Hee-Jun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea
| | - Hong-Seok Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea.,Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jeong-Ho Park
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea.,Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Mo-Jong Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea.,Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Republic of Korea
| | - Hyoung-Gon Lee
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Robert Bob Petersen
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.,Departments of Neuroscience and Neurology, Case Western Reserve University, Cleveland, OH, USA
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea.,Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Eun-Kyoung Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Republic of Korea.,Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
27
|
Girouard MP, Pool M, Alchini R, Rambaldi I, Fournier AE. RhoA Proteolysis Regulates the Actin Cytoskeleton in Response to Oxidative Stress. PLoS One 2016; 11:e0168641. [PMID: 27992599 PMCID: PMC5167403 DOI: 10.1371/journal.pone.0168641] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/05/2016] [Indexed: 11/24/2022] Open
Abstract
The small GTPase RhoA regulates the actin cytoskeleton to affect multiple cellular processes including endocytosis, migration and adhesion. RhoA activity is tightly regulated through several mechanisms including GDP/GTP cycling, phosphorylation, glycosylation and prenylation. Previous reports have also reported that cleavage of the carboxy-terminus inactivates RhoA. Here, we describe a novel mechanism of RhoA proteolysis that generates a stable amino-terminal RhoA fragment (RhoA-NTF). RhoA-NTF is detectable in healthy cells and tissues and is upregulated following cell stress. Overexpression of either RhoA-NTF or the carboxy-terminal RhoA cleavage fragment (RhoA-CTF) induces the formation of disorganized actin stress fibres. RhoA-CTF also promotes the formation of disorganized actin stress fibres and nuclear actin rods. Both fragments disrupt the organization of actin stress fibres formed by endogenous RhoA. Together, our findings describe a novel RhoA regulatory mechanism.
Collapse
Affiliation(s)
- Marie-Pier Girouard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Rue University, Montréal, Québec, Canada
| | - Madeline Pool
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Rue University, Montréal, Québec, Canada
| | - Ricardo Alchini
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Rue University, Montréal, Québec, Canada
| | - Isabel Rambaldi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Rue University, Montréal, Québec, Canada
| | - Alyson E. Fournier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Rue University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
28
|
Hodgson L, Spiering D, Sabouri-Ghomi M, Dagliyan O, DerMardirossian C, Danuser G, Hahn KM. FRET binding antenna reports spatiotemporal dynamics of GDI-Cdc42 GTPase interactions. Nat Chem Biol 2016; 12:802-809. [PMID: 27501396 PMCID: PMC5030135 DOI: 10.1038/nchembio.2145] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/16/2016] [Indexed: 12/29/2022]
Abstract
Guanine-nucleotide dissociation inhibitors (GDIs) are negative regulators of Rho family GTPases that sequester the GTPases away from the membrane. Here we ask how GDI-Cdc42 interaction regulates localized Cdc42 activation for cell motility. The sensitivity of cells to overexpression of Rho family pathway components led us to a new biosensor, GDI.Cdc42 FLARE, in which Cdc42 is modified with a fluorescence resonance energy transfer (FRET) 'binding antenna' that selectively reports Cdc42 binding to endogenous GDIs. Similar antennae could also report GDI-Rac1 and GDI-RhoA interaction. Through computational multiplexing and simultaneous imaging, we determined the spatiotemporal dynamics of GDI-Cdc42 interaction and Cdc42 activation during cell protrusion and retraction. This revealed remarkably tight coordination of GTPase release and activation on a time scale of 10 s, suggesting that GDI-Cdc42 interactions are a critical component of the spatiotemporal regulation of Cdc42 activity, and not merely a mechanism for global sequestration of an inactivated pool of signaling molecules.
Collapse
Affiliation(s)
- Louis Hodgson
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine.,Department of Pharmacology and Lineberger Cancer Center, University of North Carolina at Chapel Hill
| | - Désirée Spiering
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine
| | | | - Onur Dagliyan
- Department of Pharmacology and Lineberger Cancer Center, University of North Carolina at Chapel Hill
| | | | - Gaudenz Danuser
- Department of Cell Biology, The Scripps Research Institute.,Department of Cell Biology, Harvard Medical School
| | - Klaus M Hahn
- Department of Pharmacology and Lineberger Cancer Center, University of North Carolina at Chapel Hill
| |
Collapse
|
29
|
Abstract
Rho GTPases regulate cytoskeletal and cell adhesion dynamics and thereby coordinate a wide range of cellular processes, including cell migration, cell polarity and cell cycle progression. Most Rho GTPases cycle between a GTP-bound active conformation and a GDP-bound inactive conformation to regulate their ability to activate effector proteins and to elicit cellular responses. However, it has become apparent that Rho GTPases are regulated by post-translational modifications and the formation of specific protein complexes, in addition to GTP-GDP cycling. The canonical regulators of Rho GTPases - guanine nucleotide exchange factors, GTPase-activating proteins and guanine nucleotide dissociation inhibitors - are regulated similarly, creating a complex network of interactions to determine the precise spatiotemporal activation of Rho GTPases.
Collapse
Affiliation(s)
- Richard G Hodge
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Anne J Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
30
|
Smithers CC, Overduin M. Structural Mechanisms and Drug Discovery Prospects of Rho GTPases. Cells 2016; 5:E26. [PMID: 27304967 PMCID: PMC4931675 DOI: 10.3390/cells5020026] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/28/2016] [Accepted: 06/07/2016] [Indexed: 12/25/2022] Open
Abstract
Rho GTPases regulate cellular morphology and dynamics, and some are key drivers of cancer progression. This superfamily offers attractive potential targets for therapeutic intervention, with RhoA, Rac1 and Cdc42 being prime examples. The challenges in developing agents that act on these signaling enzymes include the lack of obvious druggable pockets and their membrane-bound activities. However, progress in targeting the similar Ras protein is illuminating new strategies for specifically inhibiting oncogenic GTPases. The structures of multiple signaling and regulatory states of Rho proteins have been determined, and the post-translational modifications including acylation and phosphorylation points have been mapped and their functional effects examined. The development of inhibitors to probe the significance of overexpression and mutational hyperactivation of these GTPases underscores their importance in cancer progression. The ability to integrate in silico, in vitro, and in vivo investigations of drug-like molecules indicates the growing tractability of GTPase systems for lead optimization. Although no Rho-targeted drug molecules have yet been clinically approved, this family is clearly showing increasing promise for the development of precision medicine and combination cancer therapies.
Collapse
Affiliation(s)
- Cameron C Smithers
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| | - Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
31
|
Yoon C, Cho SJ, Aksoy BA, Park DJ, Schultz N, Ryeom SW, Yoon SS. RETRACTED: Chemotherapy Resistance in Diffuse-Type Gastric Adenocarcinoma Is Mediated by RhoA Activation in Cancer Stem-Like Cells. Clin Cancer Res 2016; 22:971-83. [PMID: 26482039 PMCID: PMC4823002 DOI: 10.1158/1078-0432.ccr-15-1356] [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: 06/09/2015] [Accepted: 09/28/2015] [Indexed: 12/21/2022]
Abstract
PURPOSE The Lauren diffuse type of gastric adenocarcinoma (DGA), as opposed to the intestinal type (IGA), often harbors mutations in RHOA, but little is known about the role of RhoA in DGA. EXPERIMENTAL DESIGN We examined RhoA activity and RhoA pathway inhibition in DGA cell lines and in two mouse xenograft models. RhoA activity was also assessed in patient tumor samples. RESULTS RhoA activity was higher in DGA compared with IGA cell lines and was further increased when grown as spheroids to enrich for cancer stem-like cells (CSCs) or when sorted using the gastric CSC marker CD44. RhoA shRNA or the RhoA inhibitor Rhosin decreased expression of the stem cell transcription factor, Sox2, and decreased spheroid formation by 78% to 81%. DGA spheroid cells had 3- to 5-fold greater migration and invasion than monolayer cells, and this activity was Rho-dependent. Diffuse GA spheroid cells were resistant in a cytotoxicity assay to 5-fluorouracil and cisplatin chemotherapy, and this resistance could be reversed with RhoA pathway inhibition. In two xenograft models, cisplatin inhibited tumor growth by 40% to 50%, RhoA inhibition by 32% to 60%, and the combination by 77% to 83%. In 288 patient tumors, increased RhoA activity correlated with worse overall survival in DGA patients (P = 0.017) but not in IGA patients (P = 0.612). CONCLUSIONS RhoA signaling promotes CSC phenotypes in DGA cells. Increased RhoA activity is correlated with worse overall survival in DGA patients, and RhoA inhibition can reverse chemotherapy resistance in DGA CSC and in tumor xenografts. Thus, the RhoA pathway is a promising new target in DGA patients.
Collapse
Affiliation(s)
- Changhwan Yoon
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Soo-Jeong Cho
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Bülent Arman Aksoy
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York
| | - Do Joong Park
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, New York, New York
| | - Sandra W Ryeom
- Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
32
|
Tong J, Li L, Ballermann B, Wang Z. Phosphorylation and Activation of RhoA by ERK in Response to Epidermal Growth Factor Stimulation. PLoS One 2016; 11:e0147103. [PMID: 26816343 PMCID: PMC4729484 DOI: 10.1371/journal.pone.0147103] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 12/29/2015] [Indexed: 12/21/2022] Open
Abstract
The small GTPase RhoA has been implicated in various cellular activities, including the formation of stress fibers, cell motility, and cytokinesis. In addition to the canonical GTPase cycle, recent findings have suggested that phosphorylation further contributes to the tight regulation of Rho GTPases. Indeed, RhoA is phosphorylated on serine 188 (188S) by a number of protein kinases. We have recently reported that Rac1 is phosphorylated on threonine 108 (108T) by extracellular signal-regulated kinases (ERK) in response to epidermal growth factor (EGF) stimulation. Here, we provide evidence that RhoA is phosphorylated by ERK on 88S and 100T in response to EGF stimulation. We show that ERK interacts with RhoA and that this interaction is dependent on the ERK docking site (D-site) at the C-terminus of RhoA. EGF stimulation enhanced the activation of the endogenous RhoA. The phosphomimetic mutant, GFP-RhoA S88E/T100E, when transiently expressed in COS-7 cells, displayed higher GTP-binding than wild type RhoA. Moreover, the expression of GFP-RhoA S88E/T100E increased actin stress fiber formation in COS-7 cells, which is consistent with its higher activity. In contrast to Rac1, phosphorylation of RhoA by ERK does not target RhoA to the nucleus. Finally, we show that regardless of the phosphorylation status of RhoA and Rac1, substitution of the RhoA PBR with the Rac1 PBR targets RhoA to the nucleus and substitution of Rac1 PBR with RhoA PBR significantly reduces the nuclear localization of Rac1. In conclusion, ERK phosphorylates RhoA on 88S and 100T in response to EGF, which upregulates RhoA activity.
Collapse
Affiliation(s)
- Junfeng Tong
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Laiji Li
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Barbara Ballermann
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Zhixiang Wang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Signal Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- * E-mail:
| |
Collapse
|
33
|
Bruurs LJM, Donker L, Zwakenberg S, Zwartkruis FJ, Begthel H, Knisely AS, Posthuma G, van de Graaf SFJ, Paulusma CC, Bos JL. ATP8B1-mediated spatial organization of Cdc42 signaling maintains singularity during enterocyte polarization. J Cell Biol 2015; 210:1055-63. [PMID: 26416959 PMCID: PMC4586737 DOI: 10.1083/jcb.201505118] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The disease-associated phospholipid flippase ATP8B1 decreases Cdc42 mobility at the apical membrane to ensure the formation of a single apical domain and to maintain healthy lumen architecture. During yeast cell polarization localization of the small GTPase, cell division control protein 42 homologue (Cdc42) is clustered to ensure the formation of a single bud. Here we show that the disease-associated flippase ATPase class I type 8b member 1 (ATP8B1) enables Cdc42 clustering during enterocyte polarization. Loss of this regulation results in increased apical membrane size with scattered apical recycling endosomes and permits the formation of more than one apical domain, resembling the singularity defect observed in yeast. Mechanistically, we show that to become apically clustered, Cdc42 requires the interaction between its polybasic region and negatively charged membrane lipids provided by ATP8B1. Disturbing this interaction, either by ATP8B1 depletion or by introduction of a Cdc42 mutant defective in lipid binding, increases Cdc42 mobility and results in apical membrane enlargement. Re-establishing Cdc42 clustering, by tethering it to the apical membrane or lowering its diffusion, restores normal apical membrane size in ATP8B1-depleted cells. We therefore conclude that singularity regulation by Cdc42 is conserved between yeast and human and that this regulation is required to maintain healthy tissue architecture.
Collapse
Affiliation(s)
- Lucas J M Bruurs
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Lisa Donker
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Susan Zwakenberg
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Fried J Zwartkruis
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Harry Begthel
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3508 AD Utrecht, Netherlands
| | - A S Knisely
- Institute of Liver Studies, King's College Hospital, London SE5 9RS, England, UK
| | - George Posthuma
- Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Centre, 1105 AZ Amsterdam, Netherlands
| | - Coen C Paulusma
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Centre, 1105 AZ Amsterdam, Netherlands
| | - Johannes L Bos
- Molecular Cancer Research and Cancer Genomics Netherlands, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| |
Collapse
|
34
|
Amphetamine activates Rho GTPase signaling to mediate dopamine transporter internalization and acute behavioral effects of amphetamine. Proc Natl Acad Sci U S A 2015; 112:E7138-47. [PMID: 26553986 DOI: 10.1073/pnas.1511670112] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute amphetamine (AMPH) exposure elevates extracellular dopamine through a variety of mechanisms that include inhibition of dopamine reuptake, depletion of vesicular stores, and facilitation of dopamine efflux across the plasma membrane. Recent work has shown that the DAT substrate AMPH, unlike cocaine and other nontransported blockers, can also stimulate endocytosis of the plasma membrane dopamine transporter (DAT). Here, we show that when AMPH enters the cytoplasm it rapidly stimulates DAT internalization through a dynamin-dependent, clathrin-independent process. This effect, which can be observed in transfected cells, cultured dopamine neurons, and midbrain slices, is mediated by activation of the small GTPase RhoA. Inhibition of RhoA activity with C3 exotoxin or a dominant-negative RhoA blocks AMPH-induced DAT internalization. These actions depend on AMPH entry into the cell and are blocked by the DAT inhibitor cocaine. AMPH also stimulates cAMP accumulation and PKA-dependent inactivation of RhoA, thus providing a mechanism whereby PKA- and RhoA-dependent signaling pathways can interact to regulate the timing and robustness of AMPH's effects on DAT internalization. Consistent with this model, the activation of D1/D5 receptors that couple to PKA in dopamine neurons antagonizes RhoA activation, DAT internalization, and hyperlocomotion observed in mice after AMPH treatment. These observations support the existence of an unanticipated intracellular target that mediates the effects of AMPH on RhoA and cAMP signaling and suggest new pathways to target to disrupt AMPH action.
Collapse
|
35
|
Wang Y, Chen Y, Wu M, Lan T, Wu Y, Li Y, Qian H. Type II cyclic guanosine monophosphate-dependent protein kinase inhibits Rac1 activation in gastric cancer cells. Oncol Lett 2015; 10:502-508. [PMID: 26171059 DOI: 10.3892/ol.2015.3173] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 03/27/2015] [Indexed: 12/13/2022] Open
Abstract
Enhanced motility of cancer cells is a critical step in promoting tumor metastasis, which remains the major cause of gastric cancer-associated mortality. The small GTPase Rac1 is a key signaling component in the regulation of cell migration. Previous studies have demonstrated that Rac1 activity may be regulated by protein kinase G (PKG); however, the underlying mechanism is not yet clear. The current study aimed to investigate the effect of type II cyclic guanosine monophosphate (cGMP)-dependent protein kinase (PKG II) on Rac1 activity. The human gastric cancer cell line AGS was infected with adenoviral constructs encoding PKG II to increase the expression of this enzyme, and treated with a cGMP analog (8-pCPT-cGMP) to induce its activation. A Transwell assay was employed to measure cell migration, and the activity of Rac1 was assessed using a pull-down assay. Immunoprecipitation was used to isolate the Rac1 protein. Phosphorylation of phosphatidylinositol 4,5 bisphosphate 3 kinase (PI3K) and its downstream effecter protein kinase B (Akt) are associated with lysophosphatidic acid (LPA)-induced motility/migration of cancer cells. Extracellular signal regulated kinase (ERK) is the major signaling molecule of the Mitogen activated protein kinase (MAPK) mediated signaling pathway. ERK and its upstream activator MAPK kinase (MEK) are also involved in LPA-induced motility/migration of cancer cells. Phosphorylation of PI3K/Akt, MEK/ERK and enriched Rac1 were detected by western blotting. The results revealed that blocking the activation of Rac1 by ectopically expressing an inactive Rac1 mutant (T17N) impeded LPA-induced cell migration. Increased PKG II activity inhibited LPA-induced migration and LPA-induced activation of Rac1; however, it had no effect on the phosphorylation of Rac1. PKG II also inhibited the activation of PI3K/Akt and MEK/ERK mediated signaling, which is important for LPA-induced Rac1 activation. These results suggest that PKG II affects LPA-stimulated migration of AGS cells by blocking Rac1 activation, via inhibition of PI3K/Akt and MEK/ERK mediated signaling.
Collapse
Affiliation(s)
- Ying Wang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Min Wu
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ting Lan
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yan Wu
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yueying Li
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hai Qian
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
36
|
Bonfrate L, Procino G, Wang DQH, Svelto M, Portincasa P. A novel therapeutic effect of statins on nephrogenic diabetes insipidus. J Cell Mol Med 2015; 19:265-82. [PMID: 25594563 PMCID: PMC4407600 DOI: 10.1111/jcmm.12422] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 08/01/2014] [Indexed: 12/12/2022] Open
Abstract
Statins competitively inhibit hepatic 3-hydroxy-3-methylglutaryl-coenzyme A reductase, resulting in reduced plasma total and low-density lipoprotein cholesterol levels. Recently, it has been shown that statins exert additional ‘pleiotropic’ effects by increasing expression levels of the membrane water channels aquaporin 2 (AQP2). AQP2 is localized mainly in the kidney and plays a critical role in determining cellular water content. This additional effect is independent of cholesterol homoeostasis, and depends on depletion of mevalonate-derived intermediates of sterol synthetic pathways, i.e. farnesylpyrophosphate and geranylgeranylpyrophosphate. By up-regulating the expression levels of AQP2, statins increase water reabsorption by the kidney, thus opening up a new avenue in treating patients with nephrogenic diabetes insipidus (NDI), a hereditary disease that yet lacks high-powered and limited side effects therapy. Aspects related to water balance determined by AQP2 in the kidney, as well as standard and novel therapeutic strategies of NDI are discussed.
Collapse
Affiliation(s)
- Leonilde Bonfrate
- Department of Biomedical Sciences and Human Oncology, Internal Medicine, University Medical School, Bari, Italy
| | | | | | | | | |
Collapse
|
37
|
Cox AD, Der CJ. Ras history: The saga continues. Small GTPases 2014; 1:2-27. [PMID: 21686117 DOI: 10.4161/sgtp.1.1.12178] [Citation(s) in RCA: 516] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/17/2010] [Accepted: 05/24/2010] [Indexed: 12/24/2022] Open
Abstract
Although the roots of Ras sprouted from the rich history of retrovirus research, it was the discovery of mutationally activated RAS genes in human cancer in 1982 that stimulated an intensive research effort to understand Ras protein structure, biochemistry and biology. While the ultimate goal has been developing anti-Ras drugs for cancer treatment, discoveries from Ras have laid the foundation for three broad areas of science. First, they focused studies on the origins of cancer to the molecular level, with the subsequent discovery of genes mutated in cancer that now number in the thousands. Second, elucidation of the biochemical mechanisms by which Ras facilitates signal transduction established many of our fundamental concepts of how a normal cell orchestrates responses to extracellular cues. Third, Ras proteins are also founding members of a large superfamily of small GTPases that regulate all key cellular processes and established the versatile role of small GTP-binding proteins in biology. We highlight some of the key findings of the last 28 years.
Collapse
Affiliation(s)
- Adrienne D Cox
- Department of Radiation Oncology; Lineberger Comprehensive Cancer Center; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | | |
Collapse
|
38
|
Collins C, Osborne LD, Guilluy C, Chen Z, O'Brien ET, Reader JS, Burridge K, Superfine R, Tzima E. Haemodynamic and extracellular matrix cues regulate the mechanical phenotype and stiffness of aortic endothelial cells. Nat Commun 2014; 5:3984. [PMID: 24917553 PMCID: PMC4068264 DOI: 10.1038/ncomms4984] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 04/29/2014] [Indexed: 01/16/2023] Open
Abstract
Endothelial cell (ECs) lining blood vessels express many mechanosensors, including platelet endothelial cell adhesion molecule-1 (PECAM-1), that convert mechanical force to biochemical signals. While it is accepted that mechanical stresses and the mechanical properties of ECs regulate vessel health, the relationship between force and biological response remains elusive. Here we show that ECs integrate mechanical forces and extracellular matrix (ECM) cues to modulate their own mechanical properties. We demonstrate that the ECM influences EC response to tension on PECAM-1. ECs adherent on collagen display divergent stiffening and focal adhesion growth compared to ECs on fibronectin. This is due to PKA-dependent serine phosphorylation and inactivation of RhoA. PKA signaling regulates focal adhesion dynamics and EC compliance in response to shear stress in vitro and in vivo. Our study identifies a ECM-specific, mechanosensitive signaling pathway that regulates EC compliance and may serve as an atheroprotective mechanism maintains blood vessel integrity in vivo.
Collapse
Affiliation(s)
- Caitlin Collins
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Lukas D Osborne
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Christophe Guilluy
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Zhongming Chen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - E Tim O'Brien
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - John S Reader
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Keith Burridge
- 1] Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA [2] Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA [3] McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Richard Superfine
- Department of Physics and Astronomy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Ellie Tzima
- 1] Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA [2] Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA [3] McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
39
|
Hobbs GA, Zhou B, Cox AD, Campbell SL. Rho GTPases, oxidation, and cell redox control. Small GTPases 2014; 5:e28579. [PMID: 24809833 DOI: 10.4161/sgtp.28579] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
While numerous studies support regulation of Ras GTPases by reactive oxygen and nitrogen species, the Rho subfamily has received considerably less attention. Over the last few years, increasing evidence is emerging that supports the redox sensitivity of Rho GTPases. Moreover, as Rho GTPases regulate the cellular redox state by controlling enzymes that generate and convert reactive oxygen and nitrogen species, redox feedback loops likely exist. Here, we provide an overview of cellular oxidants, Rho GTPases, and their inter-dependence.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA
| | - Bingying Zhou
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA
| | - Adrienne D Cox
- Department of Pharmacology; University of North Carolina; Chapel Hill, NC USA; Department of Radiation Oncology, University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| | - Sharon L Campbell
- Department of Biochemistry and Biophysics; University of North Carolina; Chapel Hill, NC USA; Lineberger Comprehensive Cancer Center; University of North Carolina; Chapel Hill, NC USA
| |
Collapse
|
40
|
Motawea HKB, Blazek AD, Zirwas MJ, Pleister AP, Ahmed AAE, McConnell BK, Chotani MA. Delocalization of Endogenous A-kinase Antagonizes Rap1-Rho-α 2C-Adrenoceptor Signaling in Human Microvascular Smooth Muscle Cells. JOURNAL OF CYTOLOGY & MOLECULAR BIOLOGY 2014; 1:1000002. [PMID: 24701590 PMCID: PMC3970818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The second messenger cyclic AMP (cAMP) plays a vital role in the physiology of the cardiovascular system, including vasodilation of large blood vessels. This study focused on cAMP signaling in peripheral blood vessels, specifically in human vascular smooth muscle (microVSM) cells explanted from skin punch biopsy arterioles (also known as resistance vessels) of healthy volunteers. Using these human microVSM we recently demonstrated cAMP activation of exchange protein activated by cAMP (Epac), the Ras-related small GTPase Rap1A, and RhoA-ROCK-F-actin signaling in human microVSM to increase expression and cell surface translocation of functional α2C-adrenoceptors (α2C-ARs) that mediate vasoconstriction. Protein-protein association with the actin-binding protein filamin-2 and phosphorylation of filamin-2 Ser2113 by cAMP-Rap1A-Rho-ROCK signaling were necessary for receptor translocation in these cells. Although cAMP activated A-kinase in these cells, these effects were independent of A-kinase, and suggested compartmentalized A-kinase local signaling facilitated by A-kinase anchoring proteins (AKAPs). In this study we globally disrupted A-kinase-AKAP interactions by the anchoring inhibitor decoy peptide Ht31 and examined the effect on α2C-AR expression, translocation, and function in quiescent microVSM treated with the adenylyl cyclase activator and cAMP elevating agent forskolin. The results show that Ht31, but not the control peptide Ht31-P, reduced forskolin-stimulated Ser133 phosphorylation of A-kinase substrate CREB, reduced α2C-AR mRNA levels, reduced cell surface translocated receptors, and attenuated agonist-triggered receptor functional responses. Together, the results suggest that compartmentalized cAMP signaling elicits a selective cellular response in microVSM, which may have relevance to arteriole physiological function and responses.
Collapse
Affiliation(s)
- Hanaa K. B. Motawea
- Center for Cardiovascular & Pulmonary Research, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Faculty of Pharmacy, Department of Pharmacology & Toxicology, Helwan University, Helwan, Egypt
| | - Alisa D. Blazek
- Davis Heart and Lung Research Institute, Ohio State University, Columbus, Ohio, USA
| | - Matthew J. Zirwas
- Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Adam P. Pleister
- Internal Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Amany A. E. Ahmed
- Faculty of Pharmacy, Department of Pharmacology & Toxicology, Helwan University, Helwan, Egypt
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Texas Medical Center, Houston, Texas, USA
| | - Maqsood A. Chotani
- Center for Cardiovascular & Pulmonary Research, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Davis Heart and Lung Research Institute, Ohio State University, Columbus, Ohio, USA
- Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
41
|
Membrane ERα attenuates myocardial fibrosis via RhoA/ROCK-mediated actin remodeling in ovariectomized female infarcted rats. J Mol Med (Berl) 2013; 92:43-51. [DOI: 10.1007/s00109-013-1103-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 10/21/2013] [Accepted: 10/30/2013] [Indexed: 10/26/2022]
|
42
|
Takahashi M, Dillon TJ, Liu C, Kariya Y, Wang Z, Stork PJS. Protein kinase A-dependent phosphorylation of Rap1 regulates its membrane localization and cell migration. J Biol Chem 2013; 288:27712-23. [PMID: 23946483 PMCID: PMC3784689 DOI: 10.1074/jbc.m113.466904] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 08/13/2013] [Indexed: 11/06/2022] Open
Abstract
The small G protein Rap1 can mediate "inside-out signaling" by recruiting effectors to the plasma membrane that signal to pathways involved in cell adhesion and cell migration. This action relies on the membrane association of Rap1, which is dictated by post-translational prenylation as well as by a stretch of basic residues within its carboxyl terminus. One feature of this stretch of acidic residues is that it lies adjacent to a functional phosphorylation site for the cAMP-dependent protein kinase PKA. This phosphorylation has two effects on Rap1 action. One, it decreases the level of Rap1 activity as measured by GTP loading and the coupling of Rap1 to RapL, a Rap1 effector that couples Rap1 GTP loading to integrin activation. Two, it destabilizes the membrane localization of Rap1, promoting its translocation into the cytoplasm. These two actions, decreased GTP loading and decreased membrane localization, are related, as the translocation of Rap1-GTP into the cytoplasm is associated with its increased GTP hydrolysis and inactivation. The consequences of this phosphorylation in Rap1-dependent cell adhesion and cell migration were also examined. Active Rap1 mutants that lack this phosphorylation site had a minimal effect on cell adhesion but strongly reduced cell migration, when compared with an active Rap1 mutant that retained the phosphorylation site. This suggests that optimal cell migration is associated with cycles of Rap1 activation, membrane egress, and inactivation, and requires the regulated phosphorylation of Rap1 by PKA.
Collapse
Affiliation(s)
| | | | | | - Yumi Kariya
- From the Vollum Institute, and
- the Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Zhiping Wang
- From the Vollum Institute, and
- Department of Surgery, Oregon Health and Science University, Portland, Oregon 97239 and
| | | |
Collapse
|
43
|
Phosphorylation of Rac1 T108 by extracellular signal-regulated kinase in response to epidermal growth factor: a novel mechanism to regulate Rac1 function. Mol Cell Biol 2013; 33:4538-51. [PMID: 24043306 DOI: 10.1128/mcb.00822-13] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Accumulating evidence has implicated Rho GTPases, including Rac1, in many aspects of cancer development. Recent findings suggest that phosphorylation might further contribute to the tight regulation of Rho GTPases. Interestingly, sequence analysis of Rac1 shows that Rac1 T108 within the (106)PNTP(109) motif is likely an extracellular signal-regulated kinase (ERK) phosphorylation site and that Rac1 also has an ERK docking site, (183)KKRKRKCLLL(192) (D site), at the C terminus. Indeed, we show here that both transfected and endogenous Rac1 interacts with ERK and that this interaction is mediated by its D site. Green fluorescent protein (GFP)-Rac1 is threonine (T) phosphorylated in response to epidermal growth factor (EGF), and EGF-induced Rac1 threonine phosphorylation is dependent on the activation of ERK. Moreover, mutant Rac1 with the mutation of T108 to alanine (A) is not threonine phosphorylated in response to EGF. In vitro ERK kinase assay further shows that pure active ERK phosphorylates purified Rac1 but not mutant Rac1 T108A. We also show that Rac1 T108 phosphorylation decreases Rac1 activity, partially due to inhibiting its interaction with phospholipase C-γ1 (PLC-γ1). T108 phosphorylation targets Rac1 to the nucleus, which isolates Rac1 from other guanine nucleotide exchange factors (GEFs) and hinders Rac1's role in cell migration. We conclude that Rac1 T108 is phosphorylated by ERK in response to EGF, which plays an important role in regulating Rac1.
Collapse
|
44
|
Li DB, Yang GJ, Xu HW, Fu ZX, Wang SW, Hu SJ. Regulation on RhoA in Vascular Smooth Muscle Cells Under Inflammatory Stimulation Proposes a Novel Mechanism Mediating the Multiple-Beneficial Action of Acetylsalicylic Acid. Inflammation 2013; 36:1403-14. [DOI: 10.1007/s10753-013-9680-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
45
|
Sabbatini ME, Williams JA. Cholecystokinin-mediated RhoGDI phosphorylation via PKCα promotes both RhoA and Rac1 signaling. PLoS One 2013; 8:e66029. [PMID: 23776598 PMCID: PMC3679036 DOI: 10.1371/journal.pone.0066029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 05/07/2013] [Indexed: 01/18/2023] Open
Abstract
RhoA and Rac1 have been implicated in the mechanism of CCK-induced amylase secretion from pancreatic acini. In all cell types studied to date, inactive Rho GTPases are present in the cytosol bound to the guanine nucleotide dissociation inhibitor RhoGDI. Here, we identified the switch mechanism regulating RhoGDI1-Rho GTPase dissociation and RhoA translocation upon CCK stimulation in pancreatic acini. We found that both Gα13 and PKC, independently, regulate CCK-induced RhoA translocation and that the PKC isoform involved is PKCα. Both RhoGDI1 and RhoGDI3, but not RhoGDI2, are expressed in pancreatic acini. Cytosolic RhoA and Rac1 are associated with RhoGDI1, and CCK-stimulated PKCα activation releases the complex. Overexpression of RhoGDI1, by binding RhoA, inhibits its activation, and thereby, CCK-induced apical amylase secretion. RhoA translocation is also inhibited by RhoGDI1. Inactive Rac1 influences CCK-induced RhoA activation by preventing RhoGDI1 from binding RhoA. By mutational analysis we found that CCK-induced PKCα phosphorylation on RhoGDI1 at Ser96 releases RhoA and Rac1 from RhoGDI1 to facilitate Rho GTPases signaling.
Collapse
Affiliation(s)
- Maria Eugenia Sabbatini
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America.
| | | |
Collapse
|
46
|
Ntantie E, Gonyo P, Lorimer EL, Hauser AD, Schuld N, McAllister D, Kalyanaraman B, Dwinell MB, Auchampach JA, Williams CL. An adenosine-mediated signaling pathway suppresses prenylation of the GTPase Rap1B and promotes cell scattering. Sci Signal 2013; 6:ra39. [PMID: 23716716 DOI: 10.1126/scisignal.2003374] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During metastasis, cancer cells acquire the ability to dissociate from each other and migrate, which is recapitulated in vitro as cell scattering. The small guanosine triphosphatase (GTPase) Rap1 opposes cell scattering by promoting cell-cell adhesion, a function that requires its prenylation, or posttranslational modification with a carboxyl-terminal isoprenoid moiety, to enable its localization at cell membranes. Thus, signaling cascades that regulate the prenylation of Rap1 offer a mechanism to control the membrane localization of Rap1. We identified a signaling cascade initiated by adenosine A2B receptors that suppressed the prenylation of Rap1B through phosphorylation of Rap1B, which decreased its interaction with the chaperone protein SmgGDS (small GTPase guanosine diphosphate dissociation stimulator). These events promoted the cytosolic and nuclear accumulation of nonprenylated Rap1B and diminished cell-cell adhesion, resulting in cell scattering. We found that nonprenylated Rap1 was more abundant in mammary tumors than in normal mammary tissue in rats and that activation of adenosine receptors delayed Rap1B prenylation in breast, lung, and pancreatic cancer cell lines. Our findings support a model in which high concentrations of extracellular adenosine, such as those that arise in the tumor microenvironment, can chronically activate A2B receptors to suppress Rap1B prenylation and signaling at the cell membrane, resulting in reduced cell-cell contact and promoting cell scattering. Inhibiting A2B receptors may be an effective method to prevent metastasis.
Collapse
Affiliation(s)
- Elizabeth Ntantie
- 1Department of Pharmacology and Toxicology, Cancer Center, and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Riou P, Kjær S, Garg R, Purkiss A, George R, Cain R, Bineva G, Reymond N, McColl B, Thompson A, O’Reilly N, McDonald N, Parker P, Ridley A. 14-3-3 proteins interact with a hybrid prenyl-phosphorylation motif to inhibit G proteins. Cell 2013; 153:640-53. [PMID: 23622247 PMCID: PMC3690454 DOI: 10.1016/j.cell.2013.03.044] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 01/22/2013] [Accepted: 03/29/2013] [Indexed: 12/13/2022]
Abstract
Signaling through G proteins normally involves conformational switching between GTP- and GDP-bound states. Several Rho GTPases are also regulated by RhoGDI binding and sequestering in the cytosol. Rnd proteins are atypical constitutively GTP-bound Rho proteins, whose regulation remains elusive. Here, we report a high-affinity 14-3-3-binding site at the C terminus of Rnd3 consisting of both the Cys241-farnesyl moiety and a Rho-associated coiled coil containing protein kinase (ROCK)-dependent Ser240 phosphorylation site. 14-3-3 binding to Rnd3 also involves phosphorylation of Ser218 by ROCK and/or Ser210 by protein kinase C (PKC). The crystal structure of a phosphorylated, farnesylated Rnd3 peptide with 14-3-3 reveals a hydrophobic groove in 14-3-3 proteins accommodating the farnesyl moiety. Functionally, 14-3-3 inhibits Rnd3-induced cell rounding by translocating it from the plasma membrane to the cytosol. Rnd1, Rnd2, and geranylgeranylated Rap1A interact similarly with 14-3-3. In contrast to the canonical GTP/GDP switch that regulates most Ras superfamily members, our results reveal an unprecedented mechanism for G protein inhibition by 14-3-3 proteins.
Collapse
Affiliation(s)
- Philippe Riou
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Svend Kjær
- Protein Purification Facility, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Andrew Purkiss
- Structural Biology Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Roger George
- Protein Purification Facility, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Robert J. Cain
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Ganka Bineva
- Peptide Synthesis Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Nicolas Reymond
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Brad McColl
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Andrew J. Thompson
- MRC Centre for Neurodegeneration Research, De Crespigny Park, King's College London, London SE5 8AF, UK
- The Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London SW3 6JB, UK
| | - Nicola O’Reilly
- Peptide Synthesis Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Neil Q. McDonald
- Structural Biology Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Malet Street, Birkbeck College, University of London, London WC1E 7HX, UK
| | - Peter J. Parker
- Division of Cancer Studies, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Protein Phosphorylation Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields, London WC2A 3LY, UK
| | - Anne J. Ridley
- Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
| |
Collapse
|
48
|
Falkenberg CV, Loew LM. Computational analysis of Rho GTPase cycling. PLoS Comput Biol 2013; 9:e1002831. [PMID: 23326220 PMCID: PMC3542069 DOI: 10.1371/journal.pcbi.1002831] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 10/22/2012] [Indexed: 01/05/2023] Open
Abstract
The Rho family of GTPases control actin organization during diverse cellular responses (migration, cytokinesis and endocytosis). Although the primary members of this family (RhoA, Rac and Cdc42) have different downstream effects on actin remodeling, the basic mechanism involves targeting to the plasma membrane and activation by GTP binding. Our hypothesis is that the details of GTPase cycling between membrane and cytosol are key to the differential upstream regulation of these biochemical switches. Accordingly, we developed a modeling framework to analyze experimental data for these systems. This analysis can reveal details of GDI-mediated cycling and help distinguish between GDI-dependent and -independent mechanisms, including vesicle trafficking and direct association-dissociation of GTPase with membrane molecules. Analysis of experimental data for Rac membrane cycling reveals that the lower apparent affinity of GDI for RacGTP compared to RacGDP can be fully explained by the faster dissociation of the latter from the membrane. Non-dimensional steady-state solutions for membrane fraction of GTPase are presented in multidimensional charts. This methodology is then used to analyze glucose stimulated Rac cycling in pancreatic β-cells. The charts are used to illustrate the effects of GEFs/GAPs and regulated affinities between GTPases and membrane and/or GDI on the amount of membrane bound GTPase. In a similar fashion, the charts can be used as a guide in assessing how targeted modifications may compensate for altered GTPase-GDI balance in disease scenarios. Among the functions of the small GTPases Rac, RhoA and Cdc42 are the regulation of protein traffic, insulin secretion, cell shape, survival and motility. The last two are important steps for tumor growth and metastasis. The function of these proteins relies on their expression levels, proper membrane localization and activation. In addition, all three proteins compete for the same protein ‘GDI’, which modulates their cycling. These proteins are ubiquitous in mammalian cells, but also studied in simpler in vitro systems and cultured yeast. Here we show, using a series of computational analyses, that for each of these experimental systems the dominant pathway for membrane cycling of GTPases seems to differ. This means that the researcher interested in the physiological function of any of those proteins must make sure that the experimental system is appropriate. We present a methodology to identify the dominant pathways by measuring the apparent membrane dissociation rate of the protein as a function of GDI concentration. We provide charts generated from parametric scans. This analysis is then applied to the Rac-dependent insulin secretion pathway in pancreatic ß-cells, revealing that direct signaling between Rac and the membrane is an essential mechanism that emerges from the data.
Collapse
Affiliation(s)
- Cibele Vieira Falkenberg
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Leslie M. Loew
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
49
|
Lutz S, Mohl M, Rauch J, Weber P, Wieland T. RhoGEF17, a Rho-specific guanine nucleotide exchange factor activated by phosphorylation via cyclic GMP-dependent kinase Iα. Cell Signal 2012. [PMID: 23195829 DOI: 10.1016/j.cellsig.2012.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
RhoGEF17, the product of the ARHGEF17 gene, is a Rho-specific guanine nucleotide exchange factor (GEF) with an unusual structure and so far unknown function. In order to get insights in its regulation, we studied a variety of signaling pathways for activation of recombinantly expressed RhoGEF17. We found that in the presence of stable cGMP analogs RhoGEF17 associates with and is phosphorylated by co-expressed cGKIα at distinct phosphorylation sites leading to a cooperative activation of RhoA, the Rho dependent kinases (ROCK) and serum response factor-induced gene transcription. Activation of protein kinase A did not induce phosphorylation of RhoGEF17 nor altered its activity. Furthermore, we obtained evidence for a ROCK-driven positive feedback mechanism involving serine/threonine protein phosphatases, which further enhanced cGMP/cGKIα-induced RhoGEF17 activation. By using mutants of RhoA which are phosphorylation resistant to cGK or mimic phosphorylation at serine 188, we could show that RhoGEF17 is able to activate RhoA independently of its phosphorylation state. Together with the ROCK-enforced activation of RhoGEF17 by cGMP/cGKIα, this might explain why expression of RhoGEF17 switches the inhibitory effect of cGMP/cGKIα on serum-induced RhoA activation into a stimulatory one. We conclude that RhoGEF17, depending on its expression profile and level, might drastically alter the effect of cGMP/cGK involving signaling pathways on RhoA-activated downstream effectors.
Collapse
Affiliation(s)
- Susanne Lutz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, University of Heidelberg, Maybachstrasse 14, 68169 Mannheim, Germany
| | | | | | | | | |
Collapse
|
50
|
Oishi A, Makita N, Sato J, Iiri T. Regulation of RhoA signaling by the cAMP-dependent phosphorylation of RhoGDIα. J Biol Chem 2012; 287:38705-15. [PMID: 23012358 DOI: 10.1074/jbc.m112.401547] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RhoA plays a pivotal role in regulating cell shape and movement. Protein kinase A (PKA) inhibits RhoA signaling and thereby induces a characteristic morphological change, cell rounding. This has been considered to result from cAMP-induced phosphorylation of RhoA at Ser-188, which induces a stable RhoA-GTP-RhoGDIα complex and sequesters RhoA to the cytosol. However, few groups have shown RhoA phosphorylation in intact cells. Here we show that phosphorylation of RhoGDIα but not RhoA plays an essential role in the PKA-induced inhibition of RhoA signaling and in the morphological changes using cardiac fibroblasts. The knockdown of RhoGDIα by siRNA blocks cAMP-induced cell rounding, which is recovered by RhoGDIα-WT expression but not when a RhoGDIα-S174A mutant is expressed. PKA phosphorylates RhoGDIα at Ser-174 and the phosphorylation of RhoGDIα is likely to induce the formation of a active RhoA-RhoGDIα complex. Our present results thus reveal a principal molecular mechanism underlying G(s)/cAMP-induced cross-talk with G(q)/G(13)/RhoA signaling.
Collapse
Affiliation(s)
- Atsuro Oishi
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo 113-8655, Japan
| | | | | | | |
Collapse
|