Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
BMB Rep ; 54(7): 380-385, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34154701

ABSTRACT

Proper targeting of the ßPAK-interacting exchange factor (ßPIX)/G protein-coupled receptor kinase-interacting target protein (GIT) complex into distinct cellular compartments is essential for its diverse functions including neurite extension and synaptogenesis. However, the mechanism for translocation of this complex is still unknown. In the present study, we reported that the conventional kinesin, called kinesin-1, can transport the ßPIX/GIT complex. Additionally, ßPIX bind to KIF5A, a neuronal isoform of kinesin-1 heavy chain, but not KIF1 and KIF3. Mapping analysis revealed that the tail of KIF5s and LZ domain of ßPIX were the respective binding domains. Silencing KIF5A or the expression of a variety of mutant forms of KIF5A inhibited ßPIX targeting the neurite tips in PC12 cells. Furthermore, truncated mutants of ßPIX without LZ domain did not interact with KIF5A, and were unable to target the neurite tips in PC12 cells. These results defined kinesin-1 as a motor protein of ßPIX, and may provide new insights into ßPIX/GIT complex-dependent neuronal pathophysiology. [BMB Reports 2021; 54(7): 380-385].


Subject(s)
Cell Cycle Proteins/metabolism , Kinesins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Cell Cycle Proteins/physiology , Neurons/metabolism , PC12 Cells , Protein Isoforms/metabolism , Rats , Rho Guanine Nucleotide Exchange Factors/physiology
2.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33990469

ABSTRACT

G protein-coupled receptors (GPCRs) are gatekeepers of cellular homeostasis and the targets of a large proportion of drugs. In addition to their signaling activity at the plasma membrane, it has been proposed that their actions may result from translocation and activation of G proteins at endomembranes-namely endosomes. This could have a significant impact on our understanding of how signals from GPCR-targeting drugs are propagated within the cell. However, little is known about the mechanisms that drive G protein movement and activation in subcellular compartments. Using bioluminescence resonance energy transfer (BRET)-based effector membrane translocation assays, we dissected the mechanisms underlying endosomal Gq trafficking and activity following activation of Gq-coupled receptors, including the angiotensin II type 1, bradykinin B2, oxytocin, thromboxane A2 alpha isoform, and muscarinic acetylcholine M3 receptors. Our data reveal that GPCR-promoted activation of Gq at the plasma membrane induces its translocation to endosomes independently of ß-arrestin engagement and receptor endocytosis. In contrast, Gq activity at endosomes was found to rely on both receptor endocytosis-dependent and -independent mechanisms. In addition to shedding light on the molecular processes controlling subcellular Gq signaling, our study provides a set of tools that will be generally applicable to the study of G protein translocation and activation at endosomes and other subcellular organelles, as well as the contribution of signal propagation to drug action.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , Endocytosis/physiology , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, G-Protein-Coupled/physiology , HEK293 Cells , Humans , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , beta-Arrestins/physiology
3.
Nat Commun ; 12(1): 2815, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990566

ABSTRACT

Defining the principles of T cell migration in structurally and mechanically complex tumor microenvironments is critical to understanding escape from antitumor immunity and optimizing T cell-related therapeutic strategies. Here, we engineered nanotextured elastic platforms to study and enhance T cell migration through complex microenvironments and define how the balance between contractility localization-dependent T cell phenotypes influences migration in response to tumor-mimetic structural and mechanical cues. Using these platforms, we characterize a mechanical optimum for migration that can be perturbed by manipulating an axis between microtubule stability and force generation. In 3D environments and live tumors, we demonstrate that microtubule instability, leading to increased Rho pathway-dependent cortical contractility, promotes migration whereas clinically used microtubule-stabilizing chemotherapies profoundly decrease effective migration. We show that rational manipulation of the microtubule-contractility axis, either pharmacologically or through genome engineering, results in engineered T cells that more effectively move through and interrogate 3D matrix and tumor volumes. Thus, engineering cells to better navigate through 3D microenvironments could be part of an effective strategy to enhance efficacy of immune therapeutics.


Subject(s)
Cell Movement/physiology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Tumor Microenvironment/immunology , Tumor Microenvironment/physiology , Animals , Biomechanical Phenomena , Cells, Cultured , Extracellular Matrix/immunology , Extracellular Matrix/physiology , Gene Knockout Techniques , Genetic Engineering , Humans , Mice , Mice, Transgenic , Microtubules/physiology , Models, Biological , Nanostructures , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/physiology , Tumor Escape/immunology , Tumor Escape/physiology
4.
Cell Rep ; 34(1): 108594, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406419

ABSTRACT

Skeletal muscle regeneration after injury is essential for maintaining muscle function throughout aging. ARHGEF3, a RhoA/B-specific GEF, negatively regulates myoblast differentiation through Akt signaling independently of its GEF activity in vitro. Here, we report ARHGEF3's role in skeletal muscle regeneration revealed by ARHGEF3-KO mice. These mice exhibit indiscernible phenotype under basal conditions. Upon acute injury, however, ARHGEF3 deficiency enhances the mass/fiber size and function of regenerating muscles in both young and regeneration-defective middle-aged mice. Surprisingly, these effects occur independently of Akt but via the GEF activity of ARHGEF3. Consistently, overexpression of ARHGEF3 inhibits muscle regeneration in a Rho-associated kinase-dependent manner. We further show that ARHGEF3 KO promotes muscle regeneration through activation of autophagy, a process that is also critical for maintaining muscle strength. Accordingly, ARHGEF3 depletion in old mice prevents muscle weakness by restoring autophagy. Taken together, our findings identify a link between ARHGEF3 and autophagy-related muscle pathophysiology.


Subject(s)
Autophagy , Muscle Strength , Muscle, Skeletal/metabolism , Regeneration , Rho Guanine Nucleotide Exchange Factors/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Aging/metabolism , Animals , Cell Differentiation , Female , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myoblasts/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
5.
J Am Soc Nephrol ; 31(5): 996-1008, 2020 05.
Article in English | MEDLINE | ID: mdl-32188698

ABSTRACT

BACKGROUND: Previous studies showed that Cdc42, a member of the prototypical Rho family of small GTPases and a regulator of the actin cytoskeleton, is critical for the normal development and health of podocytes. However, upstream regulatory mechanisms for Cdc42 activity in podocytes are largely unknown. METHODS: We used a proximity-based ligation assay, BioID, to identify guanine nucleotide exchange factors that activate Cdc42 in immortalized human podocytes. We generated podocyte-specific ARHGEF7 (commonly known as ß-PIX) knockout mice by crossing ß-PIX floxed mice with Podocin-Cre mice. Using shRNA, we established cultured mouse podocytes with ß-PIX knockdown and their controls. RESULTS: We identified ß-PIX as a predominant guanine nucleotide exchange factor that interacts with Cdc42 in human podocytes. Podocyte-specific ß-PIX knockout mice developed progressive proteinuria and kidney failure with global or segmental glomerulosclerosis in adulthood. Glomerular podocyte density gradually decreased in podocyte-specific ß-PIX knockout mice, indicating podocyte loss. Compared with controls, glomeruli from podocyte-specific ß-PIX knockout mice and cultured mouse podocytes with ß-PIX knockdown exhibited significant reduction in Cdc42 activity. Loss of ß-PIX promoted podocyte apoptosis, which was mediated by the reduced activity of the prosurvival transcriptional regulator Yes-associated protein. CONCLUSIONS: These findings indicate that ß-PIX is required for the maintenance of podocyte architecture and glomerular function via Cdc42 and its downstream Yes-associated protein activities. This appears to be the first evidence that a Rho-guanine nucleotide exchange factor plays a critical role in podocytes.


Subject(s)
Podocytes/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Crosses, Genetic , Enzyme Activation , Female , Gene Knockdown Techniques , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Lipopolysaccharides/toxicity , Mice , Mice, 129 Strain , Mice, Inbred ICR , Podocytes/physiology , Podocytes/ultrastructure , Proteinuria/etiology , Proteinuria/metabolism , Proteinuria/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Rho Guanine Nucleotide Exchange Factors/deficiency , Signal Transduction , YAP-Signaling Proteins , cdc42 GTP-Binding Protein/metabolism
6.
PLoS Pathog ; 15(2): e1007573, 2019 02.
Article in English | MEDLINE | ID: mdl-30779788

ABSTRACT

Motility and phagocytosis are key processes that are involved in invasive amoebiasis disease caused by intestinal parasite Entamoeba histolytica. Previous studies have reported unconventional myosins to play significant role in membrane based motility as well as endocytic processes. EhMyosin IB is the only unconventional myosin present in E. histolytica, is thought to be involved in both of these processes. Here, we report an interaction between the SH3 domain of EhMyosin IB and c-terminal domain of EhFP10, a Rho guanine nucleotide exchange factor. EhFP10 was found to be confined to Entamoeba species only, and to contain a c-terminal domain that binds and bundles actin filaments. EhFP10 was observed to localize in the membrane ruffles, phagocytic and macropinocytic cups of E. histolytica trophozoites. It was also found in early pinosomes but not early phagosomes. A crystal structure of the c-terminal SH3 domain of EhMyosin IB (EhMySH3) in complex with an EhFP10 peptide and co-localization studies established the interaction of EhMySH3 with EhFP10. This interaction was shown to lead to inhibition of actin bundling activity and to thereby regulate actin dynamics during endocytosis. We hypothesize that unique domain architecture of EhFP10 might be compensating the absence of Wasp and related proteins in Entamoeba, which are known partners of myosin SH3 domains in other eukaryotes. Our findings also highlights the role of actin bundling during endocytosis.


Subject(s)
Entamoeba histolytica/metabolism , Myosin Type I/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Actins , Cell Movement , Cytoskeleton/immunology , Endocytosis/physiology , Entamoeba histolytica/pathogenicity , Entamoebiasis/immunology , Entamoebiasis/metabolism , Myosins , Phagocytosis , Phagosomes , Protein Binding , Protein Domains , Rho Guanine Nucleotide Exchange Factors/physiology
7.
Biochem Biophys Res Commun ; 501(1): 165-171, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29709479

ABSTRACT

The synaptic protein Neuroligin 2, similarly to its isoform Neuroligin 1, is produced by endothelial cells, but its activity in the vascular context remains unknown. This study aimed at verifying the hypothesis that Neuroligin 2, in parallel with its extraneuronal involvement in pancreatic beta cells exocytosis, modulated cytokine release from endothelial cells and consequently angiogenesis. We used in vitro approaches to modulate Neuroligin 2 expression and Neuroligin 2 null mice to test our hypotheses. In vitro, upon VEGF stimulation, Neuroligin 2 silencing strongly reduces Angiopoietin 2 release in the medium and increases the endothelial cell retention of Weibel Palade Bodies, the specialized organelles that store Angiopoietin 2 and various other cytokines. On the contrary, Neuroligin 2 overexpression almost depletes cells of Weibel Palade Bodies, independent of VEGF. In vivo, both the retina and tumor xenografts grown in NLGN2- null mice display an immature vasculature, with lower pericyte coverage and lower Tie2 phosphorylation. At the molecular level NLGN2 colocalizes with its neuronal partner collibystin, a CDC42 guanine nucleotide exchange factor, which is also expressed by endothelial cells and in turn modulates Angiopoietin 2 release. Neuroligin 2, an inhibitory synaptic protein, modulates a peculiar aspect of vascular function and could represent a novel target of therapy in various fields, from tumor angiogenesis to vascular diseases.


Subject(s)
Angiopoietin-2/metabolism , Cell Adhesion Molecules, Neuronal/physiology , Neovascularization, Physiologic , Nerve Tissue Proteins/physiology , Animals , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Retinal Vessels/cytology , Retinal Vessels/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Vascular Endothelial Growth Factor A/physiology , Weibel-Palade Bodies/physiology , von Willebrand Factor/metabolism
8.
Eur J Clin Invest ; 48 Suppl 2: e12939, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29682742

ABSTRACT

Rac-GTPases and their Rac-GEF activators play important roles in the recruitment and host defence functions of neutrophils. These proteins control the activation of adhesion molecules and the cytoskeletal dynamics that enable the adhesion, migration and tissue recruitment of neutrophils. They also regulate the effector functions that allow neutrophils to kill bacterial and fungal pathogens, and to clear debris. This review focuses on the roles of Rac-GTPases and Rac-GEFs in neutrophil adhesion, migration and recruitment.


Subject(s)
Neutrophils/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , rac1 GTP-Binding Protein/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Guanine Nucleotide Exchange Factors/physiology , Humans , Neutrophil Infiltration/physiology , Neutrophils/enzymology , Proto-Oncogene Proteins c-vav/physiology , Signal Transduction/physiology , T-Lymphoma Invasion and Metastasis-inducing Protein 1/physiology , rac GTP-Binding Proteins/physiology
9.
J Mol Med (Berl) ; 96(5): 427-444, 2018 05.
Article in English | MEDLINE | ID: mdl-29523914

ABSTRACT

We report the role of miR-1187 in regulation of osteoblast functions. Over-expression of miR-1187 inhibited osteoblast differentiation. Target prediction analysis tools and experimental validation by luciferase 3' UTR reporter assay identified BMPR-II and ArhGEF-9 as direct targets of miR-1187. ArhGEF-9 activates Cdc42 which has a major role in actin reorganization. BMP-2 also induces actin polymerization. Role of miR-1187 in actin reorganization was determined by western blotting, immunofluorescence, and in vivo gene silencing studies. Reduced protein levels of BMPR-II, activated Cdc42, and downstream signaling molecules were observed in miR-1187-transfected osteoblasts. miR-1187 over-expression resulted in decreased actin polymerization. Additionally, P-cofilin, which does not bind F-actin, was decreased in miR-1187-transfected cells. These results were corroborated by administration of BMPR-II exogenously in miR-1187-transfected osteoblasts. Silencing of miR-1187 in neonatal mice mitigated all the inhibitory effects of miR-1187 on actin cytoskeletal rearrangement. Importantly, in vivo treatment of miR-1187 inhibitor to ovariectomized BALB/c mice led to significant improvement in trabecular bone microarchitecture. Overall, miR-1187 functions as a negative regulator of osteogenesis by repressing BMPR-II and ArhGEF-9 expression thus suppressing non-Smad BMP2/Cdc42 signaling pathway and inhibiting actin reorganization. miR-1187 functions as a negative regulator of osteogenesis by repressing BMPR-II expression, which in turn, suppresses non-Smad BMP2/Cdc42 signaling pathway, thus inhibiting actin cytoskeletal rearrangement. Silencing of miR-1187 significantly improves trabecular bone microarchitecture. As miR-1187 exerts a negative regulatory role in osteoblasts function, hence, we propose that therapeutic approaches targeting miR-1187 could be useful in enhancing the bone formation and treatment of pathological conditions of bone loss.


Subject(s)
Actin Cytoskeleton/physiology , MicroRNAs/physiology , Osteoblasts/physiology , Actins/physiology , Animals , Bone Morphogenetic Protein Receptors, Type II/physiology , Cell Differentiation , Female , Mice, Inbred BALB C , Rho Guanine Nucleotide Exchange Factors/physiology
10.
Lab Invest ; 98(5): 670-681, 2018 05.
Article in English | MEDLINE | ID: mdl-29382922

ABSTRACT

Rho guanine nucleotide exchange factor 39 (ARHGEF39), also called C9orf100, is a new member of the Dbl-family of guanine nucleotide exchange factors. Although ARHGEF39 has been proven to regulate tumor progression in hepatocellular carcinoma, the downstream signaling pathway of ARHGEF39 and its clinical associations in non-small cell lung cancer (NSCLC) are currently unknown. In the present study, using MTT, colony formation, flow cytometry, mice xenografts, wound healing, and transwell assays, we showed that ARHGEF39 promoted tumor proliferation, migration, and invasion. Furthermore, ARHGEF39 promoted the expression of Cyclin A2, Cyclin D1, and MMP2 by activating Rac1, leading to increased phosphorylation of P38 and ATF2. Treatment with a P38 inhibitor counteracted the effect of ARHGEF39 overexpression on the increase in Cyclin A2, Cyclin D1, and MMP2 expression. Moreover, the elevated levels of p-P38 and p-ATF2 caused by ARHGEF39 overexpression could be inhibited by expression of a dominant negative Rac1 mutant (T17N). In addition, the inhibition of the expression of p-P38 and p-ATF2 by ARHGEF39 RNAi could be restored by the expression of a constitutively active Rac1 mutant (Q61L). A similar impact on cell growth and invasion was observed after ARHGEF39 overexpression combined with the P38 inhibitor, Rac1 T17N, or Rac1 Q61L. Using immunohistochemistry, ARHGEF39 expression was observed to correlate positively with larger tumor size in clinical samples from 109 cases of NSCLC (P = 0.008). The Kaplan-Meier test revealed that ARHGEF39 expression significantly affected the overall survival of patients with NSCLC (52.55 ± 6.40 months vs. 64.30 ± 5.40 months, P = 0.017). In conclusion, we identified that ARHGEF39 promotes tumor growth and invasion by activating the Rac1-P38-ATF2 signaling pathway, as well as increasing the expression of Cyclin A2, Cyclin D1, and MMP2 in NSCLC cells. ARHGEF39 may be a useful marker to predict poor prognosis of patients with NSCLC.


Subject(s)
Activating Transcription Factor 2/physiology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology , rac1 GTP-Binding Protein/physiology , Adult , Aged , Animals , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lung Neoplasms/mortality , Mice , Mice, Inbred BALB C , Middle Aged , Phosphorylation , Prognosis
11.
Lancet Respir Med ; 5(11): 869-880, 2017 11.
Article in English | MEDLINE | ID: mdl-29066090

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high mortality, uncertain cause, and few treatment options. Studies have identified a significant genetic risk associated with the development of IPF; however, mechanisms by which genetic risk factors promote IPF remain unclear. We aimed to identify genetic variants associated with IPF susceptibility and provide mechanistic insight using gene and protein expression analyses. METHODS: We used a two-stage approach: a genome-wide association study in patients with IPF of European ancestry recruited from nine different centres in the UK and controls selected from UK Biobank (stage 1) matched for age, sex, and smoking status; and a follow-up of associated genetic variants in independent datasets of patients with IPF and controls from two independent US samples from the Chicago consortium and the Colorado consortium (stage 2). We investigated the effect of novel signals on gene expression in large transcriptomic and genomic data resources, and examined expression using lung tissue samples from patients with IPF and controls. FINDINGS: 602 patients with IPF and 3366 controls were selected for stage 1. For stage 2, 2158 patients with IPF and 5195 controls were selected. We identified a novel genome-wide significant signal of association with IPF susceptibility near A-kinase anchoring protein 13 (AKAP13; rs62025270, odds ratio [OR] 1·27 [95% CI 1·18-1·37], p=1·32 × 10-9) and confirmed previously reported signals, including in mucin 5B (MUC5B; rs35705950, OR 2·89 [2·56-3·26], p=1·12 × 10-66) and desmoplakin (DSP; rs2076295, OR 1·44 [1·35-1·54], p=7·81 × 10-28). For rs62025270, the allele A associated with increased susceptibility to IPF was also associated with increased expression of AKAP13 mRNA in lung tissue from patients who had lung resection procedures (n=1111). We showed that AKAP13 is expressed in the alveolar epithelium and lymphoid follicles from patients with IPF, and AKAP13 mRNA expression was 1·42-times higher in lung tissue from patients with IPF (n=46) than that in lung tissue from controls (n=51). INTERPRETATION: AKAP13 is a Rho guanine nucleotide exchange factor regulating activation of RhoA, which is known to be involved in profibrotic signalling pathways. The identification of AKAP13 as a susceptibility gene for IPF increases the prospect of successfully targeting RhoA pathway inhibitors in patients with IPF. FUNDING: UK Medical Research Council, National Heart, Lung, and Blood Institute of the US National Institutes of Health, Agencia Canaria de Investigación, Innovación y Sociedad de la Información, Spain, UK National Institute for Health Research, and the British Lung Foundation.


Subject(s)
A Kinase Anchor Proteins/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Idiopathic Pulmonary Fibrosis/genetics , Minor Histocompatibility Antigens/genetics , Proto-Oncogene Proteins/genetics , White People/genetics , Aged , Alveolar Epithelial Cells/metabolism , Case-Control Studies , Europe , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , Signal Transduction/genetics , Tertiary Lymphoid Structures/genetics , rhoA GTP-Binding Protein/physiology
12.
Compr Physiol ; 7(2): 675-692, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28333386

ABSTRACT

Striated cardiac and skeletal muscles play very different roles in the body, but they are similar at the molecular level. In particular, contraction, regardless of the type of muscle, is a precise and complex process involving the integral protein myofilaments and their associated regulatory components. The smallest functional unit of muscle contraction is the sarcomere. Within the sarcomere can be found a sophisticated ensemble of proteins associated with the thick filaments (myosin, myosin binding protein-C, titin, and obscurin) and thin myofilaments (actin, troponin, tropomyosin, nebulin, and nebulette). These parallel thick and thin filaments slide across one another, pulling the two ends of the sarcomere together to regulate contraction. More specifically, the regulation of both timing and force of contraction is accomplished through an intricate network of intra- and interfilament interactions belonging to each myofilament. This review introduces the sarcomere proteins involved in striated muscle contraction and places greater emphasis on the more recently identified and less well-characterized myofilaments: cardiac myosin binding protein-C, titin, nebulin, and obscurin. © 2017 American Physiological Society. Compr Physiol 7:675-692, 2017.


Subject(s)
Myofibrils/physiology , Sarcomeres/physiology , Connectin/physiology , Humans , Muscle Contraction/physiology , Muscle Proteins/physiology , Muscle, Skeletal/physiology , Protein Serine-Threonine Kinases , Rho Guanine Nucleotide Exchange Factors/physiology , Troponin/physiology
13.
Cell Syst ; 4(1): 84-96.e6, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28065575

ABSTRACT

Mechanical signals from the extracellular matrix (ECM) and cellular geometry regulate the nuclear translocation of transcriptional regulators such as Yes-associated protein (YAP). Elucidating how physical signals control the activity of mechanosensitive proteins poses a technical challenge, because perturbations that affect cell shape may also affect protein localization indirectly. Here, we present an approach that mitigates confounding effects of cell-shape changes, allowing us to identify direct regulators of YAP localization. This method uses single-cell image analysis and statistical models that exploit the naturally occurring heterogeneity of cellular populations. Through systematic depletion of all human kinases, Rho family GTPases, GEFs, and GTPase activating proteins (GAPs), together with targeted chemical perturbations, we found that ß-PIX, a Rac1/Ccd42 GEF, and PAK2, a Rac1/Cdc42 effector, drive both YAP activation and cell-ECM adhesion turnover during cell spreading. Our observations suggest that coupling YAP to adhesion dynamics acts as a mechano-timer, allowing cells to rapidly tune gene expression in response to physical signals.


Subject(s)
Cell Shape/physiology , Nuclear Proteins/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Transcription Factors/physiology , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Adhesion/physiology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Female , GTPase-Activating Proteins/genetics , Humans , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Single-Cell Analysis/methods , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/genetics
14.
PLoS One ; 11(4): e0153387, 2016.
Article in English | MEDLINE | ID: mdl-27128976

ABSTRACT

Excessive mechanical ventilation exerts pathologic mechanical strain on lung vascular endothelium and promotes endothelial cell (EC) inflammatory activation; however, the specific mechanisms underlying EC inflammatory response caused by mechanical ventilation related cyclic stretch (CS) remain unclear. This study investigated the effects of chronic exposure to CS at physiologic (5%) and pathologic (18%) magnitude on pulmonary EC inflammatory status in control conditions and bacterial lipopolysacharide (LPS)-stimulated conditions. EC exposure to high or low CS magnitudes for 28-72 hrs had distinct effects on EC inflammatory activation. 18% CS increased surface expression of endothelial adhesion molecule ICAM1 and release of its soluble form (sICAM1) and inflammatory cytokine IL-8 by CS-stimulated pulmonary endothelial cells (EC). EC inflammatory activation was not observed in EC exposed to 5% CS. Chronic exposure to 18% CS, but not to 5% CS, augmented ICAM1 and IL-8 production and EC monolayer barrier disruption induced by LPS. 18% CS, but not 5% CS, stimulated expression of RhoA GTPase-specific guanine nucleotide exchange factor GEF-H1. GEF-H1 knockdown using gene-specific siRNA abolished 18% CS-induced ICAM1 expression and sICAM1 and IL-8 release by EC. GEF-H1 knockdown also prevented disruption of EC monolayer integrity and attenuated sICAM1 and IL-8 release in the two-hit model of EC barrier dysfunction caused by combined stimulation with 18% CS and LPS. These data demonstrate that exacerbation of inflammatory response by pulmonary endothelium exposed to excessive mechanical stretch is mediated by CS-induced induction of Rho activating protein GEF-H1.


Subject(s)
Inflammation/etiology , Inflammation/physiopathology , Pulmonary Artery/physiopathology , Capillary Permeability/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/physiology , Gene Knockdown Techniques , Humans , Inflammation/genetics , Inflammation Mediators/physiology , Lipopolysaccharides/toxicity , Pulmonary Artery/drug effects , Respiration, Artificial/adverse effects , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/physiology , Stress, Mechanical
15.
Cell Death Differ ; 23(3): 442-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26292756

ABSTRACT

While a great deal of progress has been made in understanding the molecular mechanisms that regulate retino-tectal mapping, the determinants that target retinal projections to specific layers of the optic tectum remain elusive. Here we show that two independent RGMa-peptides, C- and N-RGMa, activate two distinct intracellular pathways to regulate axonal growth. C-RGMa utilizes a Leukemia-associated RhoGEF (LARG)/Rho/Rock pathway to inhibit axonal growth. N-RGMa on the other hand relies on ϒ-secretase cleavage of the intracellular portion of Neogenin to generate an intracellular domain (NeICD) that uses LIM-only protein 4 (LMO4) to block growth. In the developing tectum (E18), overexpression of C-RGMa and dominant-negative LARG (LARG-PDZ) induced overshoots in the superficial tectal layer but not in deeper tectal layers. In younger embryos (E12), C-RGMa and LARG-PDZ prevented ectopic projections toward deeper tectal layers, indicating that C-RGMa may act as a barrier to descending axons. In contrast both N-RGMa and NeICD overexpression resulted in aberrant axonal-paths, all of which suggests that it is a repulsive guidance molecule. Thus, two RGMa fragments activate distinct pathways resulting in different axonal responses. These data reveal how retinal projections are targeted to the appropriate layer in their target tissue.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Nerve Tissue Proteins/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Animals , Cell Enlargement , Chick Embryo , Organ Specificity , Retinal Ganglion Cells/physiology , Superior Colliculi/cytology , Superior Colliculi/enzymology , Tissue Culture Techniques
17.
J Thromb Haemost ; 13(11): 2102-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334261

ABSTRACT

BACKGROUND: RhoA is an important regulator of platelet responses downstream of Gα13 , yet we still know little about its regulation in platelets. Leukemia-associated Rho guanine-nucleotide exchange factor (GEF [LARG]), a RhoA GEF, is highly expressed in platelets and may constitute a major upstream activator of RhoA. To this end, it is important to determine the role of LARG in platelet function and thrombosis. METHODS AND RESULTS: Using a platelet-specific gene knockout, we show that the absence of LARG results in a marked reduction in aggregation and dense-granule secretion in response to the thromboxane mimetic U46619 and proteinase-activated receptor 4-activating peptide, AYPGKF, but not to adenosine diphosphate. In a ferric chloride thrombosis model in vivo, this translated into a defect, under mild injury conditions. Importantly, agonist-induced RhoA activation was not affected by the absence of LARG, although basal activity was reduced, suggesting that LARG may play a housekeeper role in regulating constitutive RhoA activity. CONCLUSIONS: LARG plays an important role in platelet function and thrombosis in vivo. However, although LARG may have a role in regulating the resting activation state of RhoA, its role in regulating platelet function may principally be through RhoA-independent pathways, possibly through other Rho family members.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/physiology , Rho Guanine Nucleotide Exchange Factors/physiology , Thrombosis/blood , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Cell Degranulation/drug effects , Chlorides/toxicity , Crosses, Genetic , Ferric Compounds/toxicity , Gene Knockout Techniques , Mice , Mice, Knockout , Oligopeptides/pharmacology , Organ Specificity , Platelet Aggregation , Rho Guanine Nucleotide Exchange Factors/blood , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombosis/chemically induced
18.
PLoS One ; 10(7): e0132737, 2015.
Article in English | MEDLINE | ID: mdl-26177020

ABSTRACT

Endosomal sorting is an essential control mechanism for signaling through the epidermal growth factor receptor (EGFR). We report here that the guanine nucleotide exchange factor αPIX, which modulates the activity of Rho-GTPases, is a potent bimodal regulator of EGFR trafficking. αPIX interacts with the E3 ubiquitin ligase c-Cbl, an enzyme that attaches ubiquitin to EGFR, thereby labelling this tyrosine kinase receptor for lysosomal degradation. We show that EGF stimulation induces αPIX::c-Cbl complex formation. Simultaneously, αPIX and c-Cbl protein levels decrease, which depends on both αPIX binding to c-Cbl and c-Cbl ubiquitin ligase activity. Through interaction αPIX sequesters c-Cbl from EGFR and this results in reduced EGFR ubiquitination and decreased EGFR degradation upon EGF treatment. However, quantitatively more decisive for cellular EGFR distribution than impaired EGFR degradation is a strong stimulating effect of αPIX on EGFR recycling to the cell surface. This function depends on the GIT binding domain of αPIX but not on interaction with c-Cbl or αPIX exchange activity. In summary, our data demonstrate a previously unappreciated function of αPIX as a strong promoter of EGFR recycling. We suggest that the novel recycling regulator αPIX and the degradation factor c-Cbl closely cooperate in the regulation of EGFR trafficking: uncomplexed αPIX and c-Cbl mediate a positive and a negative feedback on EGFR signaling, respectively; αPIX::c-Cbl complex formation, however, results in mutual inhibition, which may reflect a stable condition in the homeostasis of EGF-induced signal flow.


Subject(s)
ErbB Receptors/metabolism , Animals , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Epidermal Growth Factor/physiology , Humans , Protein Binding , Protein Structure, Tertiary , Protein Transport , Proteolysis , Proto-Oncogene Proteins c-cbl/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , Ubiquitination
19.
J Biol Chem ; 290(14): 9171-82, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25694429

ABSTRACT

Recently, we have reported that the active form of Rac 1 GTPase binds to the glycogen phosphorylase muscle isoform (PYGM) and modulates its enzymatic activity leading to T cell proliferation. In the lymphoid system, Rac 1 and in general other small GTPases of the Rho family participate in the signaling cascades that are activated after engagement of the T cell antigen receptor. However, little is known about the IL-2-dependent Rac 1 activator molecules. For the first time, a signaling pathway leading to the activation of Rac 1/PYGM in response to IL-2-stimulated T cell proliferation is described. More specifically, αPIX, a known guanine nucleotide exchange factor for the small GTPases of the Rho family, preferentially Rac 1, mediates PYGM activation in Kit 225 T cells stimulated with IL-2. Using directed mutagenesis, phosphorylation of αPIX Rho-GEF serines 225 and 488 is required for activation of the Rac 1/PYGM pathway. IL-2-stimulated serine phosphorylation was corroborated in Kit 225 T cells cultures. A parallel pharmacological and genetic approach identified PKCθ as the serine/threonine kinase responsible for αPIX serine phosphorylation. The phosphorylated state of αPIX was required to activate first Rac 1 and subsequently PYGM. These results demonstrate that the IL-2 receptor activation, among other early events, leads to activation of PKCθ. To activate Rac 1 and consequently PYGM, PKCθ phosphorylates αPIX in T cells. The biological significance of this PKCθ/αPIX/Rac 1 GTPase/PYGM signaling pathway seems to be the control of different cellular responses such as migration and proliferation.


Subject(s)
Glycogen Phosphorylase/metabolism , Interleukin-12/pharmacology , T-Lymphocytes/drug effects , rac1 GTP-Binding Protein/metabolism , Base Sequence , Cell Line , Cell Proliferation/drug effects , Chemotaxis, Leukocyte/drug effects , DNA Primers , Enzyme Activation , Humans , Polymerase Chain Reaction , Rho Guanine Nucleotide Exchange Factors/physiology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
20.
Nat Cell Biol ; 16(9): 909-17, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25150978

ABSTRACT

Rho-family GTPases govern distinct types of cell migration on different extracellular matrix proteins in tissue culture or three-dimensional (3D) matrices. We searched for mechanisms selectively regulating 3D cell migration in different matrix environments and discovered a form of Cdc42-RhoA crosstalk governing cell migration through a specific pair of GTPase activator and inhibitor molecules. We first identified ßPix, a guanine nucleotide exchange factor (GEF), as a specific regulator of migration in 3D collagen using an affinity-precipitation-based GEF screen. Knockdown of ßPix specifically blocks cell migration in fibrillar collagen microenvironments, leading to hyperactive cellular protrusion accompanied by increased collagen matrix contraction. Live FRET imaging and RNAi knockdown linked this ßPix knockdown phenotype to loss of polarized Cdc42 but not Rac1 activity, accompanied by enhanced, de-localized RhoA activity. Mechanistically, collagen phospho-regulates ßPix, leading to its association with srGAP1, a GTPase-activating protein (GAP), needed to suppress RhoA activity. Our results reveal a matrix-specific pathway controlling migration involving a GEF-GAP interaction of ßPix with srGAP1 that is critical for maintaining suppressive crosstalk between Cdc42 and RhoA during 3D collagen migration.


Subject(s)
Cell Movement , Collagen/metabolism , GTPase-Activating Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/physiology , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Cell Shape , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha2beta1/metabolism , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Protein Transport , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL