151
|
Abstract
Rho-type small GTPases are involved in cytokinesis in various organisms, but their precise roles and regulation remain unclear. Rho proteins function as molecular switches by cycling between the active GTP-bound and inactive GDP-bound states; the GTP-bound proteins in turn interact with their downstream effectors to transmit the signal. Biochemical assays using Rho-binding domains of effector proteins have been used to specifically pull down GTP-bound Rho proteins from cell extracts. Here, we describe the application of such a method in combination with cell-cycle synchronization in the budding yeast Saccharomyces cerevisiae; this approach allows dissection of the activity of Rho1 at different stages of cytokinesis. We also present data showing the importance of caution in interpreting such biochemical data and of comparing to the results obtained with other approaches where possible. The principle of this protocol is also applicable to analyses of other Rho-type GTPases and cell-cycle events.
Collapse
|
152
|
Inhibition of the RhoA GTPase Activity Increases Sensitivity of Melanoma Cells to UV Radiation Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:2696952. [PMID: 26823948 PMCID: PMC4707346 DOI: 10.1155/2016/2696952] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/25/2015] [Indexed: 01/16/2023]
Abstract
Ultraviolet radiation is the main cause of DNA damage to melanocytes and development of melanoma, one of the most lethal human cancers, which leads to metastasis due to uncontrolled cell proliferation and migration. These phenotypes are mediated by RhoA, a GTPase overexpressed or overactivated in highly aggressive metastatic tumors that plays regulatory roles in cell cycle progression and cytoskeleton remodeling. This work explores whether the effects of UV on DNA damage, motility, proliferation, and survival of human metastatic melanoma cells are mediated by the RhoA pathway. Mutant cells expressing dominant-negative (MeWo-RhoA-N19) or constitutively active RhoA (MeWo-RhoA-V14) were generated and subjected to UV radiation. A slight reduction in migration and invasion was observed in MeWo and MeWo-RhoA-V14 cells but not in MeWo-RhoA-N19 cells, which presented inefficient motility and invasiveness associated with stress fibers fragmentation. Proliferation and survival of RhoA-deficient cells were drastically reduced by UV compared to cells displaying normal or high RhoA activity, suggesting increased sensitivity to UV. Loss of RhoA activity also caused less efficient DNA repair, with elevated levels of DNA lesions such as strand breaks and cyclobutane pyrimidine dimers (CPDs). Thus, RhoA mediates genomic stability and represents a potential target for sensitizing metastatic tumors to genotoxic agents.
Collapse
|
153
|
González Wusener AE, González Á, Nakamura F, Arregui CO. PTP1B triggers integrin-mediated repression of myosin activity and modulates cell contractility. Biol Open 2015; 5:32-44. [PMID: 26700725 PMCID: PMC4728310 DOI: 10.1242/bio.015883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell contractility and migration by integrins depends on precise regulation of protein tyrosine kinase and Rho-family GTPase activities in specific spatiotemporal patterns. Here we show that protein tyrosine phosphatase PTP1B cooperates with β3 integrin to activate the Src/FAK signalling pathway which represses RhoA-myosin-dependent contractility. Using PTP1B null (KO) cells and PTP1B reconstituted (WT) cells, we determined that some early steps following cell adhesion to fibronectin and vitronectin occurred robustly in WT cells, including aggregation of β3 integrins and adaptor proteins, and activation of Src/FAK-dependent signalling at small puncta in a lamellipodium. However, these events were significantly impaired in KO cells. We established that cytoskeletal strain and cell contractility was highly enhanced at the periphery of KO cells compared to WT cells. Inhibition of the Src/FAK signalling pathway or expression of constitutive active RhoA in WT cells induced a KO cell phenotype. Conversely, expression of constitutive active Src or myosin inhibition in KO cells restored the WT phenotype. We propose that this novel function of PTP1B stimulates permissive conditions for adhesion and lamellipodium assembly at the protruding edge during cell spreading and migration. Summary: Here we show that protein tyrosine phosphatase PTP1B cooperates with β3 integrin to transiently repress RhoA-myosin-dependent contractility, stimulating adhesion and lamellipodium assembly during cell spreading and migration.
Collapse
Affiliation(s)
- Ana E González Wusener
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| | - Ángela González
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| | - Fumihiko Nakamura
- Hematology Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02445, USA
| | - Carlos O Arregui
- IIB-INTECH, Universidad Nacional de San Martín, 1650 San Martín, Buenos Aires, Argentina
| |
Collapse
|
154
|
Salinas RP, Ortiz Flores RM, Distel JS, Aguilera MO, Colombo MI, Berón W. Coxiella burnetii Phagocytosis Is Regulated by GTPases of the Rho Family and the RhoA Effectors mDia1 and ROCK. PLoS One 2015; 10:e0145211. [PMID: 26674774 PMCID: PMC4682630 DOI: 10.1371/journal.pone.0145211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 12/01/2015] [Indexed: 01/09/2023] Open
Abstract
The GTPases belonging to the Rho family control the actin cytoskeleton rearrangements needed for particle internalization during phagocytosis. ROCK and mDia1 are downstream effectors of RhoA, a GTPase involved in that process. Coxiella burnetii, the etiologic agent of Q fever, is internalized by the host´s cells in an actin-dependent manner. Nevertheless, the molecular mechanism involved in this process has been poorly characterized. This work analyzes the role of different GTPases of the Rho family and some downstream effectors in the internalization of C. burnetii by phagocytic and non-phagocytic cells. The internalization of C. burnetii into HeLa and RAW cells was significantly inhibited when the cells were treated with Clostridium difficile Toxin B which irreversibly inactivates members of the Rho family. In addition, the internalization was reduced in HeLa cells that overexpressed the dominant negative mutants of RhoA, Rac1 or Cdc42 or that were knocked down for the Rho GTPases. The pharmacological inhibition or the knocking down of ROCK diminished bacterium internalization. Moreover, C. burnetii was less efficiently internalized in HeLa cells overexpressing mDia1-N1, a dominant negative mutant of mDia1, while the overexpression of the constitutively active mutant mDia1-ΔN3 increased bacteria uptake. Interestingly, when HeLa and RAW cells were infected, RhoA, Rac1 and mDia1 were recruited to membrane cell fractions. Our results suggest that the GTPases of the Rho family play an important role in C. burnetii phagocytosis in both HeLa and RAW cells. Additionally, we present evidence that ROCK and mDia1, which are downstream effectors of RhoA, are involved in that process.
Collapse
Affiliation(s)
- Romina P. Salinas
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Rodolfo M. Ortiz Flores
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Jesús S. Distel
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Milton O. Aguilera
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - María I. Colombo
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
| | - Walter Berón
- Instituto de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo—CONICET, Mendoza, 5500, Argentina
- * E-mail:
| |
Collapse
|
155
|
Yang Y, Zhao B, Ji Z, Zhang G, Zhang J, Li S, Guo G, Lin H. CRMPs colocalize and interact with cytoskeleton in hippocampal neurons. Int J Clin Exp Med 2015; 8:22337-22344. [PMID: 26885211 PMCID: PMC4729997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/05/2015] [Indexed: 06/05/2023]
Abstract
CRMP family proteins (CRMPs) are widely expressed in the developing neurons, mediating a variety of fundamental functions such as growth cone guidance, neuronal polarity and axon elongation. However, whether all the CRMP proteins interact with cytoskeleton remains unknown. In this study, we found that in cultured hippocampal neurons, CRMPs mainly colocalized with tubulin and actin network in neurites. In growth cones, CRMPs colocalized with tubulinmainly in the central (C-) domain and transition zone (T-zone), less in the peripheral (P-) domain and colocalized with actin in all the C-domain, T-zone and P-domain. The correlation efficiency of CRMPs between actin was significantly higher than that between tubulin, especially in growth cones. We successfully constructed GST-CRMPs plasmids, expressed and purified the GST-CRMP proteins. By GST-pulldown assay, all the CRMP family proteins were found to beinteracted with cytoskeleton proteins. Taken together, we revealed that CRMPs were colocalized with cytoskeleton in hippocampal neurons, especially in growth cones. CRMPs can interact with both tubulin and actin, thus mediating neuronal development.
Collapse
Affiliation(s)
- Yuhao Yang
- Department of Orthopedics, The First Affiliated Hospital of Jinan UniversityGuangzhou 510630, China
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Bo Zhao
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan UniversityGuangzhou 510630, China
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan UniversityGuangzhou 510630, China
| | - Jifeng Zhang
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Sumei Li
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Guoqing Guo
- Department of Anatomy, Medical College of Jinan UniversityGuangzhou 510630, China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan UniversityGuangzhou 510630, China
| |
Collapse
|
156
|
Otsu K, Harada H. Rho GTPases in ameloblast differentiation. JAPANESE DENTAL SCIENCE REVIEW 2015; 52:32-40. [PMID: 28408954 PMCID: PMC5382790 DOI: 10.1016/j.jdsr.2015.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/04/2015] [Accepted: 09/22/2015] [Indexed: 01/12/2023] Open
Abstract
During tooth development, ameloblasts differentiate from inner enamel epithelial cells to enamel-forming cells by modulating the signal pathways mediating epithelial–mesenchymal interaction and a cell-autonomous gene network. The differentiation process of epithelial cells is characterized by marked changes in their morphology and polarity, accompanied by dynamic cytoskeletal reorganization and changes in cell–cell and cell–matrix adhesion over time. Functional ameloblasts are tall, columnar, polarized cells that synthesize and secrete enamel-specific proteins. After deposition of the full thickness of enamel matrix, ameloblasts become smaller and regulate enamel maturation. Recent significant advances in the fields of molecular biology and genetics have improved our understanding of the regulatory mechanism of the ameloblast cell life cycle, mediated by the Rho family of small GTPases. They act as intracellular molecular switch that transduce signals from extracellular stimuli to the actin cytoskeleton and the nucleus. In our review, we summarize studies that provide current evidence for Rho GTPases and their involvement in ameloblast differentiation. In addition to the Rho GTPases themselves, their downstream effectors and upstream regulators have also been implicated in ameloblast differentiation.
Collapse
Affiliation(s)
- Keishi Otsu
- Corresponding author. Tel.: +81 19 651 5111x5881; fax: +81 19 908 8017.
| | | |
Collapse
|
157
|
Gong X, Tan M, Gao Y, Chen K, Guo G. CRMP‑5 interacts with actin to regulate neurite outgrowth. Mol Med Rep 2015; 13:1179-85. [PMID: 26677106 PMCID: PMC4732841 DOI: 10.3892/mmr.2015.4662] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 10/29/2015] [Indexed: 11/17/2022] Open
Abstract
CRMP family proteins (CRMPs) are abundantly expressed in the developing nervous system mediating growth cone guidance, neuronal polarity and axon elongation. CRMP-5 has been indicated to serve a critical role in neurite outgrowth. However, the detailed mechanisms of how CRMP-5 regulates neurite outgrowth remain unclear. In the current study, co-immunoprecipitation was used to identify the fact that CRMP-5 interacted with the actin and tubulin cytoskeleton networks in the growth cones of developing hippocampal neurons. CRMP-5 exhibited increased affinity towards actin when compared with microtubules. Immunocytochemistry was used to identify the fact that CRMP-5 colocalized with actin predominantly in the C-domain and T-zone in growth cones. In addition, genetic inhibition of CRMP-5 by siRNA suppressed the expression of actin, growth cone development and neurite outgrowth. Overexpression of CRMP-5 promoted the interaction with actin, growth cone development and hippocampal neurite outgrowth. Taken together, these data suggest that CRMP-5 is able to interact with the actin cytoskeleton network in the growth cone and affect growth cone development and neurite outgrowth via this interaction in developing hippocampal neurons.
Collapse
Affiliation(s)
- Xiaobing Gong
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Minghui Tan
- Department of Anatomy, Medical College of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Yuan Gao
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Keen Chen
- Department of Anatomy, Medical College of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Guoqing Guo
- Department of Anatomy, Medical College of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
158
|
Modulation of RhoA GTPase Activity Sensitizes Human Cervix Carcinoma Cells to γ-Radiation by Attenuating DNA Repair Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6012642. [PMID: 26649141 PMCID: PMC4662998 DOI: 10.1155/2016/6012642] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/21/2015] [Indexed: 11/28/2022]
Abstract
Radiotherapy with γ-radiation is widely used in cancer treatment to induce DNA damage reducing cell proliferation and to kill tumor cells. Although RhoA GTPase overexpression/hyperactivation is observed in many malignancies, the effect of RhoA activity modulation on cancer radiosensitivity has not been previously investigated. Here, we generated stable HeLa cell clones expressing either the dominant negative RhoA-N19 or the constitutively active RhoA-V14 and compared the responses of these cell lines with those of parental HeLa cells, after treatment with low doses of γ-radiation. HeLa-RhoA-N19 and HeLa-RhoA-V14 clones displayed reduced proliferation and survival compared to parental cells after radiation and became arrested at cell cycle stages correlated with increased cellular senescence and apoptosis. Also, Chk1/Chk2 and histone H2A phosphorylation data, as well as comet assays, suggest that the levels of DNA damage and DNA repair activation and efficiency in HeLa cell lines are correlated with active RhoA. In agreement with these results, RhoA inhibition by C3 toxin expression drastically affected homologous recombination (HR) and nonhomologous end joining (NHEJ). These data suggest that modulation of RhoA GTPase activity impairs DNA damage repair, increasing HeLa cell radiosensitivity.
Collapse
|
159
|
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: 68] [Impact Index Per Article: 6.8] [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
|
160
|
Policastro GM, Becker ML. Osteogenic growth peptide and its use as a bio-conjugate in regenerative medicine applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:449-64. [DOI: 10.1002/wnan.1376] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 07/18/2015] [Accepted: 08/12/2015] [Indexed: 12/13/2022]
Affiliation(s)
| | - Matthew L. Becker
- Departments of Polymer Science and Biomedical Engineering; University of Akron; Akron OH USA
| |
Collapse
|
161
|
Leyme A, Marivin A, Perez-Gutierrez L, Nguyen LT, Garcia-Marcos M. Integrins activate trimeric G proteins via the nonreceptor protein GIV/Girdin. J Cell Biol 2015; 210:1165-84. [PMID: 26391662 PMCID: PMC4586755 DOI: 10.1083/jcb.201506041] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/20/2015] [Indexed: 01/04/2023] Open
Abstract
Signal transduction via integrins and G protein-coupled receptors is critical to control cell behavior. These two receptor classes have been traditionally believed to trigger distinct and independent signaling cascades in response to extracellular cues. Here, we report a novel mechanism of integrin signaling that requires activation of the trimeric G protein Gαi by the nonreceptor guanine nucleotide exchange factor (GEF) GIV (also known as Girdin), a metastasis-associated protein. We demonstrate that GIV enhances integrin-dependent cell responses upon extracellular matrix stimulation and makes tumor cells more invasive. These responses include remodeling of the actin cytoskeleton and PI3K-dependent signaling, resulting in enhanced haptotaxis and invasion. We show that both GIV and its substrate Gαi3 are recruited to active integrin complexes and that tumor cells engineered to express GEF-deficient GIV fail to transduce integrin signals into proinvasive responses via a Gβγ-PI3K axis. Our discoveries delineate a novel mechanism by which integrin signaling is rewired during metastasis to result in increased tumor invasiveness.
Collapse
Affiliation(s)
- Anthony Leyme
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | - Arthur Marivin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | | | - Lien T Nguyen
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
162
|
Shen B, Estevez B, Xu Z, Kreutz B, Karginov A, Bai Y, Qian F, Norifumi U, Mosher D, Du X. The interaction of Gα13 with integrin β1 mediates cell migration by dynamic regulation of RhoA. Mol Biol Cell 2015; 26:3658-70. [PMID: 26310447 PMCID: PMC4603935 DOI: 10.1091/mbc.e15-05-0274] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/11/2015] [Indexed: 12/13/2022] Open
Abstract
Heterotrimeric G protein Gα13 is known to transmit G protein-coupled receptor (GPCR) signals leading to activation of RhoA and plays a role in cell migration. The mechanism underlying the role of Gα13 in cell migration, however, remains unclear. Recently we found that Gα13 interacts with the cytoplasmic domain of integrin β3 subunits in platelets via a conserved ExE motif. Here we show that a similar direct interaction between Gα13 and the cytoplasmic domain of the integrin β1 subunit plays a critical role in β1-dependent cell migration. Point mutation of either glutamic acid in the Gα13-binding (767)EKE motif in β1 or treatment with a peptide derived from the Gα13-binding sequence of β1 abolished Gα13-β1 interaction and inhibited β1 integrin-dependent cell spreading and migration. We further show that the Gα13-β1 interaction mediates β1 integrin-dependent Src activation and transient RhoA inhibition during initial cell adhesion, which is in contrast to the role of Gα13 in mediating GPCR-dependent RhoA activation. These data indicate that Gα13 plays dynamic roles in both stimulating RhoA via a GPCR pathway and inhibiting RhoA via an integrin signaling pathway. This dynamic regulation of RhoA activity is critical for cell migration on β1 integrin ligands.
Collapse
Affiliation(s)
- Bo Shen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Brian Estevez
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Zheng Xu
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Barry Kreutz
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Andrei Karginov
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Yanyan Bai
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Feng Qian
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612 Department of Internal Medicine, Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Ohio State University, Columbus, OH 43210
| | - Urao Norifumi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612 Center for Wound Healing and Tissue Regeneration, Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Deane Mosher
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin-Madison, Madison, WI 53792
| | - Xiaoping Du
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
163
|
Burke SL, Hammell M, Ambros V. Robust Distal Tip Cell Pathfinding in the Face of Temperature Stress Is Ensured by Two Conserved microRNAS in Caenorhabditis elegans. Genetics 2015; 200:1201-18. [PMID: 26078280 PMCID: PMC4574240 DOI: 10.1534/genetics.115.179184] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/10/2015] [Indexed: 12/26/2022] Open
Abstract
Biological robustness, the ability of an organism to maintain a steady-state output as genetic or environmental inputs change, is critical for proper development. MicroRNAs have been implicated in biological robustness mechanisms through their post-transcriptional regulation of genes and gene networks. Previous research has illustrated examples of microRNAs promoting robustness as part of feedback loops and genetic switches and by buffering noisy gene expression resulting from environmental and/or internal changes. Here we show that the evolutionarily conserved microRNAs mir-34 and mir-83 (homolog of mammalian mir-29) contribute to the robust migration pattern of the distal tip cells in Caenorhabditis elegans by specifically protecting against stress from temperature changes. Furthermore, our results indicate that mir-34 and mir-83 may modulate the integrin signaling involved in distal tip cell migration by potentially targeting the GTPase cdc-42 and the beta-integrin pat-3. Our findings suggest a role for mir-34 and mir-83 in integrin-controlled cell migrations that may be conserved through higher organisms. They also provide yet another example of microRNA-based developmental robustness in response to a specific environmental stress, rapid temperature fluctuations.
Collapse
Affiliation(s)
- Samantha L Burke
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Molly Hammell
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | - Victor Ambros
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
164
|
Kumar G, Ho CC, Co CC. Cell-Substrate Interactions Feedback to Direct Cell Migration along or against Morphological Polarization. PLoS One 2015; 10:e0133117. [PMID: 26186588 PMCID: PMC4506050 DOI: 10.1371/journal.pone.0133117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/24/2015] [Indexed: 12/19/2022] Open
Abstract
In response to external stimuli, cells polarize morphologically into teardrop shapes prior to moving in the direction of their blunt leading edge through lamellipodia extension and retraction of the rear tip. This textbook description of cell migration implies that the initial polarization sets the direction of cell migration. Using microfabrication techniques to control cell morphologies and the direction of migration without gradients, we demonstrate that after polarization, lamelipodia extension and attachment can feedback to change and even reverse the initial morphological polarization. Cells do indeed migrate faster in the direction of their morphologically polarization. However, feedback from subsequent lamellipodia extension and attachment can be so powerful as to induce cells to reverse and migrate against their initial polarization, albeit at a slower speed. Constitutively active mutants of RhoA show that RhoA stimulates cell motility when cells are guided either along or against their initial polarization. Cdc42 activation and inhibition, which results in loss of directional motility during chemotaxis, only reduces the speed of migration without altering the directionality of migration on the micropatterns. These results reveal significant differences between substrate directed cell migration and that induced by chemotactic gradients.
Collapse
Affiliation(s)
- Girish Kumar
- Biomedical, Chemical, and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221–0012, United States of America
| | - Chia-Chi Ho
- Biomedical, Chemical, and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221–0012, United States of America
| | - Carlos C. Co
- Biomedical, Chemical, and Environmental Engineering, University of Cincinnati, Cincinnati, OH, 45221–0012, United States of America
- * E-mail:
| |
Collapse
|
165
|
Gralka M, Kroy K. Inelastic mechanics: A unifying principle in biomechanics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3025-37. [PMID: 26151340 DOI: 10.1016/j.bbamcr.2015.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/13/2015] [Accepted: 06/26/2015] [Indexed: 01/16/2023]
Abstract
Many soft materials are classified as viscoelastic. They behave mechanically neither quite fluid-like nor quite solid-like - rather a bit of both. Biomaterials are often said to fall into this class. Here, we argue that this misses a crucial aspect, and that biomechanics is essentially damage mechanics, at heart. When deforming an animal cell or tissue, one can hardly avoid inducing the unfolding of protein domains, the unbinding of cytoskeletal crosslinkers, the breaking of weak sacrificial bonds, and the disruption of transient adhesions. We classify these activated structural changes as inelastic. They are often to a large degree reversible and are therefore not plastic in the proper sense, but they dissipate substantial amounts of elastic energy by structural damping. We review recent experiments involving biological materials on all scales, from single biopolymers over cells to model tissues, to illustrate the unifying power of this paradigm. A deliberately minimalistic yet phenomenologically very rich mathematical modeling framework for inelastic biomechanics is proposed. It transcends the conventional viscoelastic paradigm and suggests itself as a promising candidate for a unified description and interpretation of a wide range of experimental data. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Matti Gralka
- Institute for Theoretical Physics, University of Leipzig, Bruederstr. 16, 04103 Leipzig, Germany.
| | - Klaus Kroy
- Institute for Theoretical Physics, University of Leipzig, Bruederstr. 16, 04103 Leipzig, Germany.
| |
Collapse
|
166
|
Yu OM, Brown JH. G Protein-Coupled Receptor and RhoA-Stimulated Transcriptional Responses: Links to Inflammation, Differentiation, and Cell Proliferation. Mol Pharmacol 2015; 88:171-80. [PMID: 25904553 PMCID: PMC4468647 DOI: 10.1124/mol.115.097857] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/22/2015] [Indexed: 01/06/2023] Open
Abstract
The low molecular weight G protein RhoA (rat sarcoma virus homolog family member A) serves as a node for transducing signals through G protein-coupled receptors (GPCRs). Activation of RhoA occurs through coupling of G proteins, most prominently, G12/13, to Rho guanine nucleotide exchange factors. The GPCR ligands that are most efficacious for RhoA activation include thrombin, lysophosphatidic acid, sphingosine-1-phosphate, and thromboxane A2. These ligands also stimulate proliferation, differentiation, and inflammation in a variety of cell and tissues types. The molecular events underlying these responses are the activation of transcription factors, transcriptional coactivators, and downstream gene programs. This review describes the pathways leading from GPCRs and RhoA to the regulation of activator protein-1, NFκB (nuclear factor κ-light-chain-enhancer of activated B cells), myocardin-related transcription factor A, and Yes-associated protein. We also focus on the importance of two prominent downstream transcriptional gene targets, the inflammatory mediator cyclooxygenase 2, and the matricellular protein cysteine-rich angiogenic inducer 61 (CCN1). Finally, we describe the importance of GPCR-induced activation of these pathways in the pathophysiology of cancer, fibrosis, and cardiovascular disease.
Collapse
Affiliation(s)
- Olivia M Yu
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| | - Joan Heller Brown
- Department of Pharmacology (O.Y., J.H.B.) and Biomedical Sciences Graduate Program, University of California at San Diego, La Jolla, California (O.Y.)
| |
Collapse
|
167
|
Apolipoprotein E3 Inhibits Rho to Regulate the Mechanosensitive Expression of Cox2. PLoS One 2015; 10:e0128974. [PMID: 26068461 PMCID: PMC4465925 DOI: 10.1371/journal.pone.0128974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 05/01/2015] [Indexed: 11/30/2022] Open
Abstract
Apolipoprotein E3 (apoE3) is thought to protect against atherosclerosis by enhancing reverse cholesterol transport. However, apoE3 also has cholesterol-independent effects that contribute to its anti-atherogenic properties. These include altering extracellular matrix protein synthesis and inhibiting vascular smooth muscle cell proliferation. Both of these cholesterol-independent effects result from an apoE3-mediated induction of cyclooxygenase-2 (Cox2). Nevertheless, how apoE3 regulates Cox2 remains unknown. Here, we show that apoE3 inhibits the activation of Rho, which reduces the formation of actin stress fibers and focal adhesions and results in cellular softening. Inhibition of Rho-Rho kinase signaling or direct cellular softening recapitulates the effect of apoE3 on Cox2 expression while a constitutively active Rho mutant overrides the apoE3 effect on both intracellular stiffness and Cox2. Thus, our results describe a previously unidentified mechanism by which an atheroprotective apolipoprotein uses Rho to control cellular mechanics and Cox2.
Collapse
|
168
|
WEHI-3 cells inhibit adipocyte differentiation in 3T3-L1 cells. Biochem Biophys Res Commun 2015; 462:105-11. [DOI: 10.1016/j.bbrc.2015.04.064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 04/10/2015] [Indexed: 11/19/2022]
|
169
|
CRMP4 and CRMP2 Interact to Coordinate Cytoskeleton Dynamics, Regulating Growth Cone Development and Axon Elongation. Neural Plast 2015; 2015:947423. [PMID: 26064693 PMCID: PMC4442009 DOI: 10.1155/2015/947423] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/13/2015] [Accepted: 04/15/2015] [Indexed: 11/18/2022] Open
Abstract
Cytoskeleton dynamics are critical phenomena that underpin many fundamental cellular processes. Collapsin response mediator proteins (CRMPs) are highly expressed in the developing nervous system, mediating growth cone guidance, neuronal polarity, and axonal elongation. However, whether and how CRMPs associate with microtubules and actin coordinated cytoskeletal dynamics remain unknown. In this study, we demonstrated that CRMP2 and CRMP4 interacted with tubulin and actin in vitro and colocalized with the cytoskeleton in the transition-zone in developing growth cones. CRMP2 and CRMP4 also interacted with one another coordinately to promote growth cone development and axonal elongation. Genetic silencing of CRMP2 enhanced, whereas overexpression of CRMP2 suppressed, the inhibitory effects of CRMP4 knockdown on axonal development. In addition, knockdown of CRMP2 or overexpression of truncated CRMP2 reversed the promoting effect of CRMP4. With the overexpression of truncated CRMP2 or CRMP4 lacking the cytoskeleton interaction domain, the promoting effect of CRMP was suppressed. These data suggest a model in which CRMP2 and CRMP4 form complexes to bridge microtubules and actin and thus work cooperatively to regulate growth cone development and axonal elongation.
Collapse
|
170
|
Activation of mechanosensitive ion channel TRPV4 normalizes tumor vasculature and improves cancer therapy. Oncogene 2015; 35:314-22. [PMID: 25867067 PMCID: PMC4948740 DOI: 10.1038/onc.2015.83] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 12/31/2014] [Accepted: 02/16/2015] [Indexed: 12/19/2022]
Abstract
Tumor vessels are characterized by abnormal morphology and hyper-permeability that together cause inefficient delivery of chemotherapeutic agents. Although VEGF has been established as a critical regulator of tumor angiogenesis, the role of mechanical signaling in the regulation of tumor vasculature or tumor endothelial cell (TEC) function is not known. Here, we show that the mechanosensitive ion channel TRPV4 regulates tumor angiogenesis and tumor vessel maturation via modulation of TEC mechanosensitivity. We found that TEC exhibit reduced TRPV4 expression and function, which is correlated with aberrant mechanosensitivity towards ECM stiffness, increased migration and abnormal angiogenesis by TEC. Further, syngeneic tumor experiments revealed that the absence of TRPV4 induced increased vascular density, vessel diameter and reduced pericyte coverage resulting in enhanced tumor growth in TRPV4 KO mice. Importantly, overexpression or pharmacological activation of TRPV4 restored aberrant TEC mechanosensitivity, migration and normalized abnormal angiogenesis in vitro by modulating Rho activity. Finally, a small molecule activator of TRPV4, GSK1016790A, in combination with anti-cancer drug Cisplatin, significantly reduced tumor growth in WT mice by inducing vessel maturation. Our findings demonstrate TRPV4 channels to be critical regulators of tumor angiogenesis and represent a novel target for anti-angiogenic and vascular normalization therapies.
Collapse
|
171
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
172
|
Rac1 GTPase-deficient HeLa cells present reduced DNA repair, proliferation, and survival under UV or gamma irradiation. Mol Cell Biochem 2015; 404:281-97. [DOI: 10.1007/s11010-015-2388-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
|
173
|
Shankar J, Nabi IR. Actin cytoskeleton regulation of epithelial mesenchymal transition in metastatic cancer cells. PLoS One 2015; 10:e0119954. [PMID: 25756282 PMCID: PMC4355409 DOI: 10.1371/journal.pone.0119954] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/26/2015] [Indexed: 11/17/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is associated with loss of the cell-cell adhesion molecule E-cadherin and disruption of cell-cell junctions as well as with acquisition of migratory properties including reorganization of the actin cytoskeleton and activation of the RhoA GTPase. Here we show that depolymerization of the actin cytoskeleton of various metastatic cancer cell lines with Cytochalasin D (Cyt D) reduces cell size and F-actin levels and induces E-cadherin expression at both the protein and mRNA level. Induction of E-cadherin was dose dependent and paralleled loss of the mesenchymal markers N-cadherin and vimentin. E-cadherin levels increased 2 hours after addition of Cyt D in cells showing an E-cadherin mRNA response but only after 10-12 hours in HT-1080 fibrosarcoma and MDA-MB-231 cells in which E-cadherin mRNA level were only minimally affected by Cyt D. Cyt D treatment induced the nuclear-cytoplasmic translocation of EMT-associated SNAI 1 and SMAD1/2/3 transcription factors. In non-metastatic MCF-7 breast cancer cells, that express E-cadherin and represent a cancer cell model for EMT, actin depolymerization with Cyt D induced elevated E-cadherin while actin stabilization with Jasplakinolide reduced E-cadherin levels. Elevated E-cadherin levels due to Cyt D were associated with reduced activation of Rho A. Expression of dominant-negative Rho A mutant increased and dominant-active Rho A mutant decreased E-cadherin levels and also prevented Cyt D induction of E-cadherin. Reduced Rho A activation downstream of actin remodelling therefore induces E-cadherin and reverses EMT in cancer cells. Cyt D treatment inhibited migration and, at higher concentrations, induced cytotoxicity of both HT-1080 fibrosarcoma cells and normal Hs27 fibroblasts, but only induced mesenchymal-epithelial transition in HT-1080 cancer cells. Our studies suggest that actin remodelling is an upstream regulator of EMT in metastatic cancer cells.
Collapse
Affiliation(s)
- Jay Shankar
- Departments of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Ivan R Nabi
- Departments of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
174
|
Ura B, Scrimin F, Zanconati F, Arrigoni G, Monasta L, Romano A, Banco R, Zweyer M, Milani D, Ricci G. Two-dimensional gel electrophoresis analysis of the leiomyoma interstitial fluid reveals altered protein expression with a possible involvement in pathogenesis. Oncol Rep 2015; 33:2219-26. [PMID: 25738828 DOI: 10.3892/or.2015.3827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/30/2014] [Indexed: 11/05/2022] Open
Abstract
Uterine leiomyoma is the most common smooth benign neoplasm. In the present study, we analyzed the global interstitial fluid (IF) profile of leiomyoma vs. normal myometrium to identify protein dysregulation involved in leiomyoma pathogenesis. Two-dimensional gel electrophoresis and mass spectrometry were used to generate and compare the global interstitial fluid profiles of the leiomyoma and of the normal tissue. Two proteins were validated by immunohistochemistry. By comparing the interstitial fluid profile of the leiomyoma with that of the normal myometrium, the levels of seven proteins were found to be significantly different: four structural organization proteins (desmin, prelamin-A/C, transgelin and α-actinin-1), an inflammatory response (α1-antitrypsin), a response to oxidative stress (peroxiredoxin-2), and a folding protein (heat shock 70 kDa protein 1A/1B). Desmin, α1-antitrypsin and peroxiredoxin-2 were upregulated in the leiomyoma, whereas heat shock 70 kDa protein 1A/1B, α-actinin-1, prelamin-A/C and transgelin were downregulated. Desmin and α1-antitrypsin were further validated by immunohistochemistry. By identifying proteins with altered expression levels compared to the myometrium from several pathways of the leiomyoma pathogenesis, we found the leiomyoma interstitial fluid to have a characteristic proteomic profile. A better appreciation of the pathophysiology of the disease can be useful in the development of conservative treatments that serve as viable alternatives to hysterectomy.
Collapse
Affiliation(s)
- Blendi Ura
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, Italy
| | - Federica Scrimin
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, Italy
| | - Fabrizio Zanconati
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Lorenzo Monasta
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, Italy
| | - Andrea Romano
- UCO Pathological Anatomy and Histology, Azienda Ospedaliera-Universitaria Ospedali Riuniti, Trieste, Italy
| | - Rubina Banco
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, Italy
| | - Marina Zweyer
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health - IRCCS 'Burlo Garofolo', Trieste, Italy
| |
Collapse
|
175
|
Son K, Smith TC, Luna EJ. Supervillin binds the Rac/Rho-GEF Trio and increases Trio-mediated Rac1 activation. Cytoskeleton (Hoboken) 2015; 72:47-64. [PMID: 25655724 DOI: 10.1002/cm.21210] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/21/2015] [Indexed: 01/06/2023]
Abstract
We investigated cross-talk between the membrane-associated, myosin II-regulatory protein supervillin and the actin-regulatory small GTPases Rac1, RhoA, and Cdc42. Supervillin knockdown reduced Rac1-GTP loading, but not the GTP loading of RhoA or Cdc42, in HeLa cells with normal levels of the Rac1-activating protein Trio. No reduction in Rac1-GTP loading was observed when supervillin levels were reduced in Trio-depleted cells. Conversely, overexpression of supervillin isoform 1 (SV1) or, especially, isoform 4 (SV4) increased Rac1 activation. Inhibition of the Trio-mediated Rac1 guanine nucleotide exchange activity with ITX3 partially blocked the SV4-mediated increase in Rac1-GTP. Both SV4 and SV1 co-localized with Trio at or near the plasma membrane in ruffles and cell surface projections. Two sequences within supervillin bound directly to Trio spectrin repeats 4-7: SV1-171, which contains N-terminal residues found in both SV1 and SV4 and the SV4-specific differentially spliced coding exons 3, 4, and 5 within SV4 (SV4-E345; SV4 amino acids 276-669). In addition, SV4-E345 interacted with the homologous sequence in rat kalirin (repeats 4-7, amino acids 531-1101). Overexpressed SV1-174 and SV4-E345 affected Rac1-GTP loading, but only in cells with endogenous levels of Trio. Trio residues 771-1057, which contain both supervillin-interaction sites, exerted a dominant-negative effect on cell spreading. Supervillin and Trio knockdowns, separately or together, inhibited cell spreading, suggesting that supervillin regulates the Rac1 guanine nucleotide exchange activity of Trio, and potentially also kalirin, during cell spreading and lamellipodia extension.
Collapse
Affiliation(s)
- Kyonghee Son
- Department of Cell and Developmental Biology, Program in Cell & Developmental Dynamics, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | | |
Collapse
|
176
|
Prolyl hydroxylase regulates axonal rewiring and motor recovery after traumatic brain injury. Cell Death Dis 2015; 6:e1638. [PMID: 25675298 PMCID: PMC4669805 DOI: 10.1038/cddis.2015.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 12/23/2014] [Accepted: 12/29/2014] [Indexed: 12/12/2022]
Abstract
Prolyl 4-hydroxylases (PHDs; PHD1, PHD2, and PHD3) are a component of cellular oxygen sensors that regulate the adaptive response depending on the oxygen concentration stabilized by hypoxia/stress-regulated genes transcription. In normoxic condition, PHD2 is required to stabilize hypoxia inducible factors. Silencing of PHD2 leads to the activation of intracellular signaling including RhoA and Rho-associated protein kinase (ROCK), which are key regulators of neurite growth. In this study, we determined that genetic or pharmacological inhibition of PHD2 in cultured cortical neurons prevents neurite elongation through a ROCK-dependent mechanism. We then explored the role of PHDs in axonal reorganization following a traumatic brain injury in adult mice. Unilateral destruction of motor cortex resulted in behavioral deficits due to disruption of the corticospinal tract (CST), a part of the descending motor pathway. In the spinal cord, sprouting of fibers from the intact side of the CST into the denervated side is thought to contribute to the recovery process following an injury. Intracortical infusion of PHD inhibitors into the intact side of the motor cortex abrogated spontaneous formation of CST collaterals and functional recovery after damage to the sensorimotor cortex. These findings suggest PHDs have an important role in the formation of compensatory axonal networks following an injury and may represent a new molecular target for the central nervous system disorders.
Collapse
|
177
|
Bernusso VA, Machado-Neto JA, Pericole FV, Vieira KP, Duarte AS, Traina F, Hansen MD, Olalla Saad ST, Barcellos KS. Imatinib restores VASP activity and its interaction with Zyxin in BCR–ABL leukemic cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:388-95. [PMID: 25450971 DOI: 10.1016/j.bbamcr.2014.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 12/24/2022]
|
178
|
Chen C, Xie J, Rajappa R, Deng L, Fredberg J, Yang L. Interleukin-1β and tumor necrosis factor-α increase stiffness and impair contractile function of articular chondrocytes. Acta Biochim Biophys Sin (Shanghai) 2015; 47:121-9. [PMID: 25520178 DOI: 10.1093/abbs/gmu116] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) are major proinflammatory cytokines involved in osteoarthritis (OA). These cytokines disturb chondrocyte metabolism by suppressing the synthesis of extracellular matrix proteins and stimulating the release of catabolic proteases, but little is known about their role in chondrocyte mechanics. Thus, the aim of this study was to measure the effects of IL-1β and TNF-α on the mechanical properties of the chondrocytes. Chondrocytes from goat knee joints were cultured in 96-well plates. The cellular stiffness and contractile function were probed using optical magnetic twisting cytometry, the cytoskeleton and the expression of extracellular matrix proteins were visualized using immunofluorescent staining, and chondrocyte phenotypical expression was measured by western blot analysis. Results showed that chondrocyte stiffness was dramatically decreased by disruption of F-actin but was unaffected by disruption of the intermediate filament vimentin. Treatment with 10 ng/ml IL-1β or 40 ng/ml TNF-α for 24 h substantially increased the expression level of F-actin and cellular stiffness, and impaired cell stiffening in response to the contractile agonist histamine, but these effects were blocked by the Rho-associated protein kinase inhibitor Y27632. In conclusion, IL-1β and TNF-α substantially change the mechanical properties of the chondrocytes in vitro. While changes of chondrocyte mechanics in vivo during OA progression remain unclear, this finding reveals a prominent role of these cytokines in cellular mechanics and provides insight for anti-cytokine therapies of OA.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | - Jing Xie
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, China
| | - Ravikumar Rajappa
- Tissue Engineering Laboratories, Veterans Affairs Boston Healthcare System, Boston, MA 02115, USA Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China
| | - Jeffrey Fredberg
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
179
|
Zhang CC, Li R, Jiang H, Lin S, Rogalski JC, Liu K, Kast J. Development and application of a quantitative multiplexed small GTPase activity assay using targeted proteomics. J Proteome Res 2015; 14:967-76. [PMID: 25569337 DOI: 10.1021/pr501010v] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Small GTPases are a family of key signaling molecules that are ubiquitously expressed in various types of cells. Their activity is often analyzed by western blot, which is limited by its multiplexing capability, the quality of isoform-specific antibodies, and the accuracy of quantification. To overcome these issues, a quantitative multiplexed small GTPase activity assay has been developed. Using four different binding domains, this assay allows the binding of up to 12 active small GTPase isoforms simultaneously in a single experiment. To accurately quantify the closely related small GTPase isoforms, a targeted proteomic approach, i.e., selected/multiple reaction monitoring, was developed, and its functionality and reproducibility were validated. This assay was successfully applied to human platelets and revealed time-resolved coactivation of multiple small GTPase isoforms in response to agonists and differential activation of these isoforms in response to inhibitor treatment. This widely applicable approach can be used for signaling pathway studies and inhibitor screening in many cellular systems.
Collapse
Affiliation(s)
- Cheng-Cheng Zhang
- The Biomedical Research Centre, ∥The Centre for Blood Research, University of British Columbia , Vancouver, BC V6T 1Z3, Canada
| | | | | | | | | | | | | |
Collapse
|
180
|
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: 2.6] [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
|
181
|
Meighan CM, Kelly VE, Krahe EC, Gaeta AJ. α integrin cytoplasmic tails can rescue the loss of Rho-family GTPase signaling in the C. elegans somatic gonad. Mech Dev 2015; 136:111-22. [PMID: 25576691 DOI: 10.1016/j.mod.2014.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/02/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
Integrin signaling relies on multiple, distinct pathways to impact a diverse set of cell behaviors. The Rho family of GTPases are well-established downstream signaling partners of integrins that regulate cell shape, polarity, and migration. The nematode C. elegans provides a simple in vivo system for studying both integrins and the Rho family. Our previous work showed that the C. elegans α integrin cytoplasmic tails have tissue-specific functions during development. Here, we use chimeric α integrins to show that the cytoplasmic tails can rescue the loss of the Rho family of GTPases in three cell types in the somatic gonad. Knockdown of rho-1 by RNAi causes defects in sheath cell actin organization, ovulation, and vulva morphology. Chimeric α integrin ina-1 with the pat-2 cytoplasmic tail can rescue both actin organization and ovulation after rho-1 RNAi, yet cannot restore vulva morphology. Knockdown of cdc-42 by RNAi causes defects in sheath cell actin organization, ovulation, vulva morphology, and distal tip cell migration. Chimeric α integrin pat-2 with the ina-1 cytoplasmic tail can rescue vulva morphology defects and distal tip cell migration after cdc-42 RNAi, yet cannot restore sheath cell actin organization or ovulation. Disruption of Rac yields the same phenotype in distal tip cells regardless of α integrin cytoplasmic tail composition. Taken together, the cytoplasmic tails of α integrins can bypass signaling from members of the Rho family of GTPases during development.
Collapse
Affiliation(s)
| | - Victoria E Kelly
- Christopher Newport University, Newport News, VA 23606, United States
| | - Elena C Krahe
- Christopher Newport University, Newport News, VA 23606, United States
| | - Adriel J Gaeta
- Christopher Newport University, Newport News, VA 23606, United States
| |
Collapse
|
182
|
Bodman JAR, Yang Y, Logan MR, Eitzen G. Yeast translation elongation factor-1A binds vacuole-localized Rho1p to facilitate membrane integrity through F-actin remodeling. J Biol Chem 2015; 290:4705-4716. [PMID: 25561732 DOI: 10.1074/jbc.m114.630764] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rho GTPases are molecular switches that modulate a variety of cellular processes, most notably those involving actin dynamics. We have previously shown that yeast vacuolar membrane fusion requires re-organization of actin filaments mediated by two Rho GTPases, Rho1p and Cdc42p. Cdc42p initiates actin polymerization to facilitate membrane tethering; Rho1p has a role in the late stages of vacuolar fusion, but its mode of action is unknown. Here, we identified eEF1A as a vacuolar Rho1p-interacting protein. eEF1A (encoded by the TEF1 and TEF2 genes in yeast) is an aminoacyl-tRNA transferase needed during protein translation. eEF1A also has a second function that is independent of translation; it binds and organizes actin filaments into ordered cable structures. Here, we report that eEF1A interacts with Rho1p via a C-terminal subdomain. This interaction occurs predominantly when both proteins are in the GDP-bound state. Therefore, eEF1A is an atypical downstream effector of Rho1p. eEF1A does not promote vacuolar fusion; however, overexpression of the Rho1p-interacting subdomain affects vacuolar morphology. Vacuoles were destabilized and prone to leakage when treated with the eEF1A inhibitor narciclasine. We propose a model whereby eEF1A binds to Rho1p-GDP on the vacuolar membrane; it is released upon Rho1p activation and then bundles actin filaments to stabilize fused vacuoles. Therefore, the Rho1p-eEF1A complex acts to spatially localize a pool of eEF1A to vacuoles where it can readily organize F-actin.
Collapse
Affiliation(s)
- James A R Bodman
- From the Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Yang Yang
- From the Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Michael R Logan
- From the Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Gary Eitzen
- From the Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
183
|
Microtubule-dependent modulation of adhesion complex composition. PLoS One 2014; 9:e115213. [PMID: 25526367 PMCID: PMC4272306 DOI: 10.1371/journal.pone.0115213] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/19/2014] [Indexed: 12/24/2022] Open
Abstract
The microtubule network regulates the turnover of integrin-containing adhesion complexes to stimulate cell migration. Disruption of the microtubule network results in an enlargement of adhesion complex size due to increased RhoA-stimulated actomyosin contractility, and inhibition of adhesion complex turnover; however, the microtubule-dependent changes in adhesion complex composition have not been studied in a global, unbiased manner. Here we used label-free quantitative mass spectrometry-based proteomics to determine adhesion complex changes that occur upon microtubule disruption with nocodazole. Nocodazole-treated cells displayed an increased abundance of the majority of known adhesion complex components, but no change in the levels of the fibronectin-binding α5β1 integrin. Immunofluorescence analyses confirmed these findings, but revealed a change in localisation of adhesion complex components. Specifically, in untreated cells, α5-integrin co-localised with vinculin at peripherally located focal adhesions and with tensin at centrally located fibrillar adhesions. In nocodazole-treated cells, however, α5-integrin was found in both peripherally located and centrally located adhesion complexes that contained both vinculin and tensin, suggesting a switch in the maturation state of adhesion complexes to favour focal adhesions. Moreover, the switch to focal adhesions was confirmed to be force-dependent as inhibition of cell contractility with the Rho-associated protein kinase inhibitor, Y-27632, prevented the nocodazole-induced conversion. These results highlight a complex interplay between the microtubule cytoskeleton, adhesion complex maturation state and intracellular contractile force, and provide a resource for future adhesion signaling studies. The proteomics data have been deposited in the ProteomeXchange with identifier PXD001183.
Collapse
|
184
|
Wang N, Wang M, Zhu YH, Grosel TW, Sun D, Kudryashov DS, Wu JQ. The Rho-GEF Gef3 interacts with the septin complex and activates the GTPase Rho4 during fission yeast cytokinesis. Mol Biol Cell 2014; 26:238-55. [PMID: 25411334 PMCID: PMC4294672 DOI: 10.1091/mbc.e14-07-1196] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Rho GTPases, activated by Rho guanine nucleotide exchange factors (GEFs), are conserved molecular switches for signal transductions that regulate diverse cellular processes, including cell polarization and cytokinesis. The fission yeast Schizosaccharomyces pombe has six Rho GTPases (Cdc42 and Rho1-Rho5) and seven Rho GEFs (Scd1, Rgf1-Rgf3, and Gef1-Gef3). The GEFs for Rho2-Rho5 have not been unequivocally assigned. In particular, Gef3, the smallest Rho GEF, was barely studied. Here we show that Gef3 colocalizes with septins at the cell equator. Gef3 physically interacts with septins and anillin Mid2 and depends on them to localize. Gef3 coprecipitates with GDP-bound Rho4 in vitro and accelerates nucleotide exchange of Rho4, suggesting that Gef3 is a GEF for Rho4. Consistently, Gef3 and Rho4 are in the same genetic pathways to regulate septum formation and/or cell separation. In gef3∆ cells, the localizations of two potential Rho4 effectors--glucanases Eng1 and Agn1--are abnormal, and active Rho4 level is reduced, indicating that Gef3 is involved in Rho4 activation in vivo. Moreover, overexpression of active Rho4 or Eng1 rescues the septation defects of mutants containing gef3∆. Together our data support that Gef3 interacts with the septin complex and activates Rho4 GTPase as a Rho GEF for septation in fission yeast.
Collapse
Affiliation(s)
| | - Mo Wang
- Department of Molecular Genetics
| | | | | | | | | | - Jian-Qiu Wu
- Department of Molecular Genetics Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
185
|
Miller NLG, Kleinschmidt EG, Schlaepfer DD. RhoGEFs in cell motility: novel links between Rgnef and focal adhesion kinase. Curr Mol Med 2014; 14:221-34. [PMID: 24467206 DOI: 10.2174/1566524014666140128110339] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 07/08/2013] [Accepted: 12/02/2013] [Indexed: 11/22/2022]
Abstract
Rho guanine exchange factors (GEFs) are a large, diverse family of proteins defined by their ability to catalyze the exchange of GDP for GTP on small GTPase proteins such as Rho family members. GEFs act as integrators from varied intra- and extracellular sources to promote spatiotemporal activity of Rho GTPases that control signaling pathways regulating cell proliferation and movement. Here we review recent studies elucidating roles of RhoGEF proteins in cell motility. Emphasis is placed on Dbl-family GEFs and connections to development, integrin signaling to Rho GTPases regulating cell adhesion and movement, and how these signals may enhance tumor progression. Moreover, RhoGEFs have additional domains that confer distinctive functions or specificity. We will focus on a unique interaction between Rgnef (also termed Arhgef28 or p190RhoGEF) and focal adhesion kinase (FAK), a non-receptor tyrosine kinase that controls migration properties of normal and tumor cells. This Rgnef-FAK interaction activates canonical GEF-dependent RhoA GTPase activity to govern contractility and also functions as a scaffold in a GEF-independent manner to enhance FAK activation. Recent studies have also brought to light the importance of specific regions within the Rgnef pleckstrin homology (PH) domain for targeting the membrane. As revealed by ongoing Rgnef-FAK investigations, exploring GEF roles in cancer will yield fundamental new information on the molecular mechanisms promoting tumor spread and metastasis.
Collapse
Affiliation(s)
| | | | - D D Schlaepfer
- University of California San Diego, Moores Cancer Center, Department of Reproductive Medicine, MC 0803, 3855 Health Sciences Dr., La Jolla, CA 92093 USA.
| |
Collapse
|
186
|
Marjoram RJ, Lessey EC, Burridge K. Regulation of RhoA activity by adhesion molecules and mechanotransduction. Curr Mol Med 2014; 14:199-208. [PMID: 24467208 PMCID: PMC3929014 DOI: 10.2174/1566524014666140128104541] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 07/05/2013] [Accepted: 12/02/2013] [Indexed: 12/26/2022]
Abstract
The low molecular weight GTP-binding protein RhoA regulates many cellular events, including cell migration, organization of the cytoskeleton, cell adhesion, progress through the cell cycle and gene expression. Physical forces influence these cellular processes in part by regulating RhoA activity through mechanotransduction of cell adhesion molecules (e.g. integrins, cadherins, Ig superfamily molecules). RhoA activity is regulated by guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) that are themselves regulated by many different signaling pathways. Significantly, the engagement of many cell adhesion molecules can affect RhoA activity in both positive and negative ways. In this brief review, we consider how RhoA activity is regulated downstream from cell adhesion molecules and mechanical force. Finally, we highlight the importance of mechanotransduction signaling to RhoA in normal cell biology as well as in certain pathological states.
Collapse
Affiliation(s)
| | | | - K Burridge
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
187
|
Goicoechea SM, Awadia S, Garcia-Mata R. I'm coming to GEF you: Regulation of RhoGEFs during cell migration. Cell Adh Migr 2014; 8:535-49. [PMID: 25482524 PMCID: PMC4594598 DOI: 10.4161/cam.28721] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cell migration is a highly regulated multistep process that requires the coordinated regulation of cell adhesion, protrusion, and contraction. These processes require numerous protein–protein interactions and the activation of specific signaling pathways. The Rho family of GTPases plays a key role in virtually every aspect of the cell migration cycle. The activation of Rho GTPases is mediated by a large and diverse family of proteins; the guanine nucleotide exchange factors (RhoGEFs). GEFs work immediately upstream of Rho proteins to provide a direct link between Rho activation and cell–surface receptors for various cytokines, growth factors, adhesion molecules, and G protein-coupled receptors. The regulated targeting and activation of RhoGEFs is essential to coordinate the migratory process. In this review, we summarize the recent advances in our understanding of the role of RhoGEFs in the regulation of cell migration.
Collapse
Key Words
- DH, Dbl-homology
- DHR, DOCK homology region
- DOCK, dedicator of cytokinesis
- ECM, extracellular matrix
- EGF, epidermal growth factor
- FA, focal adhesion
- FN, fibronectin
- GAP, GTPase activating protein
- GDI, guanine nucleotide dissociation inhibitor
- GEF, guanine nucleotide exchange factor
- GPCR, G protein-coupled receptor
- HGF, hepatocyte growth factor
- LPA, lysophosphatidic acid
- MII, myosin II
- PA, phosphatidic acid
- PDGF, platelet-derived growth factor
- PH, pleckstrin-homology
- PIP2, phosphatidylinositol 4, 5-bisphosphate
- PIP3, phosphatidylinositol (3, 4, 5)-trisphosphate.
- Rho GEFs
- Rho GTPases
- bFGF, basic fibroblast growth factor
- cell migration
- cell polarization
- focal adhesions
- guanine nucleotide exchange factors
Collapse
Affiliation(s)
- Silvia M Goicoechea
- a Department of Biological Sciences ; University of Toledo ; Toledo , OH USA
| | | | | |
Collapse
|
188
|
RHGF-1/PDZ-RhoGEF and retrograde DLK-1 signaling drive neuronal remodeling on microtubule disassembly. Proc Natl Acad Sci U S A 2014; 111:16568-73. [PMID: 25359212 DOI: 10.1073/pnas.1410263111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neurons remodel their connectivity in response to various insults, including microtubule disruption. How neurons sense microtubule disassembly and mount remodeling responses by altering genetic programs in the soma are not well defined. Here we show that in response to microtubule disassembly, the Caenorhabditis elegans PLM neuron remodels by retracting its synaptic branch and overextending the primary neurite. This remodeling required RHGF-1, a PDZ-Rho guanine nucleotide exchange factor (PDZ-RhoGEF) that was associated with and inhibited by microtubules. Independent of the myosin light chain activation, RHGF-1 acted through Rho-dependent kinase LET-502/ROCK and activated a conserved, retrograde DLK-1 MAPK (DLK-1/dual leucine zipper kinase) pathway, which triggered synaptic branch retraction and overgrowth of the PLM neurite in a dose-dependent manner. Our data represent a neuronal remodeling paradigm during development that reshapes the neural circuit by the coordinated removal of the dysfunctional synaptic branch compartment and compensatory extension of the primary neurite.
Collapse
|
189
|
Activin B antagonizes RhoA signaling to stimulate mesenchymal morphology and invasiveness of clear cell renal cell carcinomas. PLoS One 2014; 9:e111276. [PMID: 25343250 PMCID: PMC4208853 DOI: 10.1371/journal.pone.0111276] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022] Open
Abstract
Activin B belongs to the TGFβ family of growth factors and is upregulated in clear cell renal cell carcinoma cells by hypoxia inducible factors. Expression of Activin B is required for tumor growth in vivo and tumor cell invasion in vitro. Here we show that activation of RhoA signaling counteracts Activin B mediated disassembly of actin stress fibers, mesenchymal cell morphology and invasiveness, whereas inhibition of RhoA rescues these effects in Activin B knockdown cells. Conversely, Activin B inhibits RhoA signaling suggesting that there is an antagonistic connection between both pathways. In addition we found that Rac1 plays an opposite role to RhoA, i.e. activation of Rac1 initiates loss of actin stress fibers, promotes a mesenchymal cell morphology and induces invasion in Activin B knockown cells, whereas inhibition of Rac1 abolishes these Activin B effects. Collectively, our data provide evidence that reduction of RhoA signaling by Activin B together with persistent Rac1 activity is a prerequisite for inducing an invasive phenotype in clear cell renal cell carcinoma.
Collapse
|
190
|
Abstract
The detection of Ras superfamily GTPase activity in innate immune cells is important when studying signaling events elicited by various ligands and cellular processes. The development of high-affinity probes detecting the activated, GTP-bound form of small GTPases has significantly enhanced our understanding of initiation and termination of GTPase-regulated signaling pathways. These probes are created by fusing a high-affinity GTPase-binding domain derived from a specific downstream effector protein to glutathione S-transferase (GST). Such domains bind preferentially to the GTP-bound form of the upstream Rho or Ras GTPase. Coupling these probes to beads enables extraction of the complex and subsequent quantification of the active GTP-binding protein by immunoblotting. Although effector domains that discriminate efficiently between GDP- and GTP-bound states and highly specific antibodies are not yet available for every small GTPase, analysis of certain members of the Rho and Ras GTPase family is now routinely performed. Here, we describe affinity-based pulldown assays for detection of Rho GTPase (Rac1/2, Cdc42, RhoA/B) and Rap1/2 activity in stimulated neutrophils or macrophages.
Collapse
|
191
|
Wan YJ, Yang Y, Leng QL, Lan B, Jia HY, Liu YH, Zhang CZ, Cao Y. Vav1 increases Bcl-2 expression by selective activation of Rac2-Akt in leukemia T cells. Cell Signal 2014; 26:2202-9. [PMID: 24880064 DOI: 10.1016/j.cellsig.2014.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 01/06/2023]
Abstract
Vav proteins are guanine nucleotide exchange factors (GEFs) that activate a group of small G proteins (GTPases). Vav1 is predominantly expressed in hematopoietic cells, whereas Vav2 and Vav3 are ubiquitously distributed in almost all human tissues. All three Vav proteins contain conserved structural motifs and associate with a variety of cellular activities including proliferation, migration, and survival. Previous observation with Jurkat leukemia T cells showed that Vav1 possessed anti-apoptotic activity by enhancing Bcl-2 transcription. However the mechanism has not been unveiled. Here, we explored the effectors of Vav1 in promoting Bcl-2 expression in Jurkat cells and revealed that Rac2-Akt was specifically evoked by the expression of Vav1, but not Vav2 or Vav3. Although all three Vav isoforms existed in Jurkat cells, Rac2 was distinguishably activated by Vav1 and that led to enhanced Bcl-2 expression and cell survival. Akt was modulated downstream of Vav1-Rac2, and the activation of Akt was indispensable in the enhanced transcription of Bcl-2. Intriguingly, neither Vav2 nor Vav3 was able to activate Rac2-Akt pathway as determined by gene silencing approach. Our data illustrated a unique role of Vav1 in T leukemia survival by selectively triggering Rac2-Akt axis and elevating the expression of anti-apoptotic Bcl-2.
Collapse
Affiliation(s)
- Ya-Juan Wan
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Yin Yang
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Qian-Li Leng
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Bei Lan
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Hui-Yan Jia
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Yao-Hui Liu
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Cui-Zhu Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China
| | - Youjia Cao
- Key Laboratory of Microbial Functional Genomics of Ministry of Education, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China; State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, PR China.
| |
Collapse
|
192
|
Huff LP, DeCristo MJ, Cox AD. Effector recruitment method to study spatially regulated activation of Ras and Rho GTPases. Methods Mol Biol 2014; 1120:263-83. [PMID: 24470032 DOI: 10.1007/978-1-62703-791-4_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Ras and Rho family GTPases control a wide variety of cellular processes, and the signaling downstream of these GTPases is influenced by their subcellular localization when activated. Since only a minority of total cellular GTPases is active, observation of the total subcellular distribution of GTPases does not reveal where active GTPases are localized. In this chapter, we describe the use of effector recruitment assays to monitor the subcellular localization of active Ras and Rho family GTPases. The recruitment assay relies on preferential binding of downstream effectors to active GTPases versus inactive GTPases. Tagging the GTPase-binding-domain (GBD) of a downstream effector with a fluorescent protein produces a probe that is recruited to compartments where GTPases are active. We describe an example of a recruitment assay using the GBD of PAK1 to monitor Rac1 activity and explain how the assay can be expanded to determine the subcellular localization of activation of other GTPases.
Collapse
Affiliation(s)
- Lauren P Huff
- Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | |
Collapse
|
193
|
Bellas E, Chen CS. Forms, forces, and stem cell fate. Curr Opin Cell Biol 2014; 31:92-7. [PMID: 25269668 DOI: 10.1016/j.ceb.2014.09.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/15/2014] [Indexed: 01/26/2023]
Abstract
Cells change their shape and mechanics dramatically during development and tissue healing in response to morphogens, cell-cell contact, adhesion to extracellular matrix, and more. Several regulatory links have been described between cell shape, cytoskeletal tension, matrix adhesiveness and stiffness, and recent studies have begun to uncover how these mechanotransduction pathways can impact transcriptional signaling and cell fate decision. The integrated mechanisms linking cell forces, form and fate are likely critical for driving normal morphogenesis, tissue development, and healing. Dysregulation of these mechanisms may also tip the scale from normal to diseased states. Here, we highlight mechanisms that alter cell shape and mechanics, and the pathways affected by these changes.
Collapse
Affiliation(s)
- Evangelia Bellas
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States; The Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, United States.
| |
Collapse
|
194
|
Yamamoto T, Ugawa Y, Yamashiro K, Shimoe M, Tomikawa K, Hongo S, Kochi S, Ideguchi H, Maeda H, Takashiba S. Osteogenic differentiation regulated by Rho-kinase in periodontal ligament cells. Differentiation 2014; 88:33-41. [PMID: 25278479 DOI: 10.1016/j.diff.2014.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 08/26/2014] [Accepted: 09/03/2014] [Indexed: 12/23/2022]
Abstract
The periodontal ligament is a multifunctional soft connective tissue, which functions not only as a cushion supporting the teeth against occlusal force, but is also a source of osteogenic cells that can regenerate neighboring hard tissues. Periodontal ligament cells (PDL cells) contain heterogeneous cell populations, including osteogenic cell progenitors. However, the precise mechanism underlying the differentiation process remains elusive. Cell differentiation is regulated by the local biochemical and mechanical microenvironment that can modulate gene expression and cell morphology by altering actin cytoskeletal organization mediated by Rho-associated, coiled-coil containing protein kinase (ROCK). To determine its role in PDL cell differentiation, we examined the effects of ROCK on cytoskeletal changes and kinetics of gene expression during osteogenic differentiation. PDL cells were isolated from human periodontal ligament on extracted teeth and cultured in osteogenic medium for 14 days. Y-27632 was used for ROCK inhibition assay. Osteogenic phenotype was determined by monitoring alkaline phosphatase (ALP) activity and calcium deposition by Alizarin Red staining. ROCK-induced cytoskeletal changes were examined by immunofluorescence analysis of F-actin and myosin light chain 2 (MLC2) expression. Real-time PCR was performed to examine the kinetics of osteogenic gene expression. F-actin and phospho-MLC2 were markedly induced during osteogenic differentiation, which coincided with upregulation of ALP activity and mineralization. Subsequent inhibition assay indicated that Y-27632 significantly inhibited F-actin and phospho-MLC2 expression in a dose-dependent manner with concomitant partial reversal of the PDL cell osteogenic phenotype. PCR array analysis of osteogenic gene expression indicated that extracellular matrix genes, such as fibronectin 1, collagen type I and III, and biglycan, were significantly downregulated by Y27632. These findings indicated crucial effects of ROCK in cytoskeletal reorganization and differentiation of PDL cells toward osteogenic cells. ROCK contributes to induction of osteogenic differentiation by synergistic increases in extracellular matrix gene expression in PDL cells.
Collapse
Affiliation(s)
- Tadashi Yamamoto
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Yuki Ugawa
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Keisuke Yamashiro
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Masayuki Shimoe
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Kazuya Tomikawa
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Shoichi Hongo
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Shinsuke Kochi
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Hidetaka Ideguchi
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Hiroshi Maeda
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Shogo Takashiba
- Department of Pathophysiology-Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan.
| |
Collapse
|
195
|
Abstract
The Ras superfamily of small GTPases is composed of more than 150 members, which share a conserved structure and biochemical properties, acting as binary molecular switches turned on by binding GTP and off by hydrolyzing GTP to GDP. However, despite considerable structural and biochemical similarities, these proteins play multiple and divergent roles, being versatile and key regulators of virtually all fundamental cellular processes. Conversely, their dysfunction plays a crucial role in the pathogenesis of serious human diseases, including cancer and developmental syndromes. Fuelled by the original identification in 1982 of mutationally activated and transforming human Ras genes in human cancer cell lines, a variety of powerful experimental techniques have been intensively focused on discovering and studying structure, biochemistry, and biology of Ras and Ras-related small GTPases, leading to fundamental research breakthroughs into identification and structural and functional characterization of a huge number of Ras superfamily members, as well as of their multiple regulators and effectors. In this review we provide a general overview of the major milestones that eventually allowed to unlock the secret treasure chest of this large and important superfamily of proteins.
Collapse
|
196
|
Chang YW, Huang YS. Arsenite-activated JNK signaling enhances CPEB4-Vinexin interaction to facilitate stress granule assembly and cell survival. PLoS One 2014; 9:e107961. [PMID: 25237887 PMCID: PMC4169592 DOI: 10.1371/journal.pone.0107961] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
Stress granules (SGs) are compartmentalized messenger ribonucleoprotein particles (mRNPs) where translationally repressed mRNAs are stored when cells encounter environmental stress. Cytoplasmic polyadenylation element-binding protein (CPEB)4 is a sequence-specific RNA-binding protein and translational regulator. In keeping with the results obtained from the study of other RNA-binding proteins, we found CPEB4 localized in SGs in various arsenite-treated cells. In this study, we identified that Vinexin, a CPEB4-interacting protein, is a novel component of SGs. Vinexin is a SH3-domain-containing adaptor protein and affects cell migration through its association with Vinculin to localize at focal adhesions (FAs). Unexpectedly, Vinexin is translocated from FAs to SGs under arsenite-induced stress. The recruitment of Vinexin to SGs depends on its interaction with CPEB4 and influences SG formation and cell survival. Arsenite-activated c-Jun N-terminal kinase (JNK) signaling enhances the association between CPEB4 and Vinexin, which consequently facilitates SG localization of Vinexin. Taken together, this study uncovers a novel interaction between a translational regulator and an adaptor protein to influence SG assembly and cell survival.
Collapse
Affiliation(s)
- Yu-Wei Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
197
|
Tetraspanin CD9 regulates cell contraction and actin arrangement via RhoA in human vascular smooth muscle cells. PLoS One 2014; 9:e106999. [PMID: 25184334 PMCID: PMC4153684 DOI: 10.1371/journal.pone.0106999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/07/2014] [Indexed: 02/07/2023] Open
Abstract
The most prevalent cardiovascular diseases arise from alterations in vascular smooth muscle cell (VSMC) morphology and function. Tetraspanin CD9 has been previously implicated in regulating vascular pathologies; however, insight into how CD9 may regulate adverse VSMC phenotypes has not been provided. We utilized a human model of aortic smooth muscle cells to understand the consequences of CD9 deficiency on VSMC phenotypes. Upon knocking down CD9, the cells developed an abnormally small and rounded morphology. We determined that this morphological change was due to a lack of typical parallel actin arrangement. We also found similar total RhoA but decreased GTP-bound (active) RhoA levels in CD9 deficient cells. As a result, cells lacking a full complement of CD9 were less contractile than their control treated counterparts. Upon restoration of RhoA activity in the CD9 deficient cells, the phenotype was reversed and cell contraction was restored. Conversely, inhibition of RhoA activity in the control cells mimicked the CD9-deficient cell phenotype. Thus, alteration in CD9 expression was sufficient to profoundly disrupt cellular actin arrangement and endogenous cell contraction by interfering with RhoA signaling. This study provides insight into how CD9 may regulate previously described vascular smooth muscle cell pathophysiology.
Collapse
|
198
|
Circulating tumor cells exhibit a biologically aggressive cancer phenotype accompanied by selective resistance to chemotherapy. Cancer Lett 2014; 325:99-107. [PMID: 25016063 DOI: 10.1016/j.canlet.2012.06.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 05/31/2012] [Accepted: 06/24/2012] [Indexed: 12/14/2022]
Abstract
With prostate cancer (PCa), circulating tumor cells (CTCs) and disseminated tumor cells (DTCs) portend a poor clinical prognosis. Their unknown biology precludes rational therapeutic design. We demonstrate that CTC and DTC cell lines, established from mice bearing human PCa orthotopic implants, exhibit increased cellular invasion in vitro, increased metastasis in mice, and express increased epithelial to mesenchymal transition biomarkers. Further, they are selectively resistant to growth inhibition by mitoxantrone-like agents. These findings demonstrate that CTC formation is accompanied by phenotypic progression without obligate reversion. Their increased metastatic potential, selective therapeutic resistance, and differential expression of potential therapeutic targets provide a rational basis to test further interventions.
Collapse
|
199
|
Chircop M. Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells. Small GTPases 2014; 5:29770. [PMID: 24988197 DOI: 10.4161/sgtp.29770] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Rho GTPases regulate a diverse range of cellular functions primarily through their ability to modulate microtubule dynamics and the actin-myosin cytoskeleton. Both of these cytoskeletal structures are crucial for a mitotic cell division. Specifically, their assembly and disassembly is tightly regulated in a temporal manner to ensure that each mitotic stage occurs in the correct sequential order and not prematurely until the previous stage is completed. Thus, it is not surprising that the Rho GTPases, RhoA, and Cdc42, have reported roles in several stages of mitosis: cell cortex stiffening during cell rounding, mitotic spindle formation, and bi-orient attachment of the spindle microtubules to the kinetochore and during cytokinesis play multiple roles in establishing the division plane, assembly, and activation of the contractile ring, membrane ingression, and abscission. Here, I review the molecular mechanisms regulating the spatial and temporal activation of RhoA and Cdc42 during mitosis, and how this is critical for mitotic progression and completion.
Collapse
Affiliation(s)
- Megan Chircop
- Children's Medical Research Institute; The University of Sydney; Westmead, Australia
| |
Collapse
|
200
|
Muñoz S, Manjón E, Sánchez Y. The putative exchange factor Gef3p interacts with Rho3p GTPase and the septin ring during cytokinesis in fission yeast. J Biol Chem 2014; 289:21995-2007. [PMID: 24947517 DOI: 10.1074/jbc.m114.548792] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The small GTP-binding proteins of the Rho family and its regulatory proteins play a central role in cytokinetic actomyosin ring assembly and cytokinesis. Here we show that the fission yeast guanine nucleotide exchange factor Gef3p interacts with Rho3p at the division site. Gef3p contains a putative DH homology domain and a BAR/IMD-like domain. The protein localized to the division site late in mitosis, where it formed a ring that did not constrict with actomyosin ring (cytokinetic actomyosin ring) invagination; instead, it split into a double ring that resembled the septin ring. Gef3p co-localized with septins and Mid2p and required septins and Mid2p for its localization. Gef3p interacts physically with the GTP-bound form of Rho3p. Although Gef3p is not essential for cell separation, the simultaneous disruption of gef3(+) and Rho3p-interacting proteins, such as Sec8p, an exocyst component, Apm1p, a subunit of the clathrin adaptor complex or For3p, an actin-polymerizing protein, yielded cells with strong defects in septation and polarity respectively. Our results suggest that interactions between septins and Rho-GEFs provide a new targeting mechanism for GTPases in cytokinesis, in this case probably contributing to Rho3p function in vesicle tethering and vesicle trafficking in the later steps of cell separation.
Collapse
Affiliation(s)
- Sofía Muñoz
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| | - Elvira Manjón
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| | - Yolanda Sánchez
- From the Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas and Departamento de Microbiología y Genética, Universidad de Salamanca, C/ Zacarías González, s/n. 37007 Salamanca, Spain
| |
Collapse
|