Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 516
Filter
1.
Cells ; 12(23)2023 11 21.
Article in English | MEDLINE | ID: mdl-38067102

ABSTRACT

Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here we show that RhoA in SCs is indispensable to have correct muscle regeneration and hypertrophy. In particular, the absence of RhoA in SCs prevents a correct SC fusion both to other RhoA-deleted SCs (regeneration context) and to growing control myofibers (hypertrophy context). We demonstrated that RhoA is dispensable for SCs proliferation and differentiation; however, RhoA-deleted SCs have an inefficient movement even if their cytoskeleton assembly is not altered. Proliferative myoblast and differentiated myotubes without RhoA display a decreased expression of Chordin, suggesting a crosstalk between these genes for myoblast fusion regulation. These findings demonstrate the importance of RhoA in SC fusion regulation and its requirement to achieve an efficient skeletal muscle homeostasis restoration.


Subject(s)
Cell Fusion , Muscle Fibers, Skeletal , Satellite Cells, Skeletal Muscle , rhoA GTP-Binding Protein , Humans , Cell Communication , Hypertrophy/metabolism , Satellite Cells, Skeletal Muscle/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/physiology
2.
Curr Biol ; 31(6): 1326-1336.e5, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33581074

ABSTRACT

Epithelia must eliminate apoptotic cells to preserve tissue barriers and prevent inflammation.1 Several different mechanisms exist for apoptotic clearance, including efferocytosis2,3 and apical extrusion.4,5 We found that extrusion was the first-line response to apoptosis in cultured monolayers and in zebrafish epidermis. During extrusion, the apoptotic cell elicited active lamellipodial protrusions and assembly of a contractile extrusion ring in its neighbors. Depleting E-cadherin compromised both the contractile ring and extrusion, implying that a cadherin-dependent pathway allows apoptotic cells to engage their neighbors for extrusion. We identify RhoA as the cadherin-dependent signal in the neighbor cells and show that it is activated in response to contractile tension from the apoptotic cell. This mechanical stimulus is conveyed by a myosin-VI-dependent mechanotransduction pathway that is necessary both for extrusion and to preserve the epithelial barrier when apoptosis was stimulated. Earlier studies suggested that release of sphingosine-1-phosphate (S1P) from apoptotic cells might define where RhoA was activated. However, we found that, although S1P is necessary for extrusion, its contribution does not require a localized source of S1P in the epithelium. We therefore propose a unified view of how RhoA is stimulated to engage neighbor cells for apoptotic extrusion. Here, tension-sensitive mechanotransduction is the proximate mechanism that activates RhoA specifically in the immediate neighbors of apoptotic cells, but this also must be primed by S1P in the tissue environment. Together, these elements provide a coincidence detection system that confers robustness on the extrusion response.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Mechanotransduction, Cellular , Zebrafish , rhoA GTP-Binding Protein/physiology , Animals , Cadherins/genetics , Lysophospholipids , Sphingosine/analogs & derivatives
3.
BMC Urol ; 21(1): 5, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407350

ABSTRACT

BACKGROUND: Emerging evidence has noted the important participation of microRNAs (miRNAs) in several human diseases including cancer. This research was launched to probe the function of miR-381 in bladder cancer (BCa) progression. METHODS: Twenty-eight patients with primary BCa were included in this study. Cancer tissues and the adjacent normal tissues were obtained. Aberrantly expressed miRNAs in BCa tissues were analyzed using miRNA microarrays. miR-381 expression in the bladder and paired tumor tissues, and in BCa and normal cell lines was determined. The target relationship between miR-381 and BMI1 was predicted online and validated through a luciferase assay. Gain-of-functions of miR-381 and BMI1 were performed to identify their functions on BCa cell behaviors as well as tumor growth in vivo. The involvement of the Rho/ROCK signaling was identified. RESULTS: miR-381 was poor regulated in BCa tissues and cells (all p < 0.05). A higher miR-381 level indicated a better prognosis of patients with BCa. Artificial up-regulation of miR-381 inhibited proliferation, invasion, migration, resistance to apoptosis, and tumor formation ability of BCa T24 and RT4 cells (all p < 0.05). miR-381 was found to directly bind to BMI1 and was negatively correlated with BMI1 expression. Overexpression of BMI1 partially blocked the tumor suppressing roles of miR-381 in cell malignancy and tumor growth (all p < 0.05). In addition, miR-381 led to decreased RhoA phosphorylation and ROCK2 activation, which were also reversed by BMI1 (all p < 0.05). Artificial inhibition of the Rho/ROCK signaling blocked the functions of BMI1 in cell growth and metastasis (all p < 0.05). CONCLUSION: The study evidenced that miR-381 may act as a beneficiary biomarker in BCa patients. Up-regulation of miR-381 suppresses BCa development both in vivo and in vitro through BMI1 down-regulation and the Rho/ROCK inactivation.


Subject(s)
MicroRNAs/physiology , Polycomb Repressive Complex 1/physiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , Cell Proliferation , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Signal Transduction
4.
Eur J Pharmacol ; 894: 173815, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33345850

ABSTRACT

Parkinson's disease (PD) is a complex and widespread neurodegenerative disease characterized by depletion of midbrain dopaminergic (DA) neurons. Key issues are the development of therapies that can stop or reverse the disease progression, identification of dependable biomarkers, and better understanding of the pathophysiological mechanisms of PD. RhoA-ROCK signals appear to have an important role in PD symptoms, making it a possible approach for PD treatment strategies. Activation of RhoA-ROCK (Rho-associated coiled-coil containing protein kinase) appears to stimulate various PD risk factors including aggregation of alpha-synuclein (αSyn), dysregulation of autophagy, and activation of apoptosis. This manuscript reviews current updates about the biology and function of the RhoA-ROCK pathway and discusses the possible role of this signaling pathway in causing the pathogenesis of PD. We conclude that inhibition of the RhoA-ROCK signaling pathway may have high translational potential and could be a promising therapeutic target in PD.


Subject(s)
Parkinson Disease/drug therapy , Parkinson Disease/etiology , Signal Transduction , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , Animals , Axons/metabolism , Humans , Microglia/metabolism , Signal Transduction/drug effects , alpha-Synuclein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/chemistry , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors
5.
Science ; 370(6522)2020 12 11.
Article in English | MEDLINE | ID: mdl-33303584

ABSTRACT

Embryo polarization is critical for mouse development; however, neither the regulatory clock nor the molecular trigger that it activates is known. Here, we show that the embryo polarization clock reflects the onset of zygotic genome activation, and we identify three factors required to trigger polarization. Advancing the timing of transcription factor AP-2 gamma (Tfap2c) and TEA domain transcription factor 4 (Tead4) expression in the presence of activated Ras homolog family member A (RhoA) induces precocious polarization as well as subsequent cell fate specification and morphogenesis. Tfap2c and Tead4 induce expression of actin regulators that control the recruitment of apical proteins on the membrane, whereas RhoA regulates their lateral mobility, allowing the emergence of the apical domain. Thus, Tfap2c, Tead4, and RhoA are regulators for the onset of polarization and cell fate segregation in the mouse.


Subject(s)
Biological Clocks/physiology , Blastocyst/physiology , Cell Polarity/physiology , DNA-Binding Proteins/physiology , Muscle Proteins/physiology , Transcription Factor AP-2/physiology , Transcription Factors/physiology , rhoA GTP-Binding Protein/physiology , Actins/metabolism , Animals , Biological Clocks/genetics , Blastocyst/cytology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Polarity/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Female , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , RNA Interference , TEA Domain Transcription Factors , Transcription Factor AP-2/genetics , Transcription Factors/genetics , rhoA GTP-Binding Protein/genetics
6.
J Cell Mol Med ; 24(18): 10693-10704, 2020 09.
Article in English | MEDLINE | ID: mdl-32725958

ABSTRACT

Endometriosis is a benign gynaecological disease appearing with pelvic pain, rising dysmenorrhoea and infertility seriously impacting on 10% of reproductive-age females. This research attempts to demonstrate the function and molecular mechanism of RhoA/ROCK pathway on epithelial-mesenchymal transition (EMT) and proliferation in endometriosis. The expression of Rho family was abnormally changed in endometriotic lesions; in particular, RhoA and ROCK1/2 were significantly elevated. Overexpression of RhoA in human eutopic endometrial epithelial cells (eutopic EECs) enhanced the cell mobility, epithelial-mesenchymal transition (EMT) and proliferation, and RhoA knockdown exhibited the opposite function. Oestrogen up-regulated the RhoA activity and expression of RhoA and ROCK1/2. RhoA overexpression reinforced the effect of oestrogen on promoting EMT and proliferation, and RhoA knockdown impaired the effect of oestrogen. oestrogen receptor α (ERα) was involved with the regulation of oestrogen on EMT and proliferation and up-regulated RhoA activity and expression of RhoA and ROCK1/2. The function of ERα was modulated by the change in RhoA expression. Furthermore, phosphorylated ERK that was enhanced by oestrogen and ERα promoted the protein expression of RhoA/ROCK pathway. Endometriosis mouse model revealed that oestrogen enhanced the size and weight of endometriotic lesions. The expression of RhoA and phosphorylated ERK in mouse endometriotic lesions was significantly elevated by oestrogen. We conclude that abnormal activated RhoA/ROCK pathway in endometriosis is responsible for the function of oestrogen/ERα/ERK signalling, which promoted EMT and proliferation and resulted in the development of endometriosis.


Subject(s)
Endometriosis/pathology , Endometrium/pathology , Epithelial-Mesenchymal Transition/physiology , Estrogens/physiology , Signal Transduction/physiology , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , Adult , Animals , Cells, Cultured , Disease Models, Animal , Endometriosis/surgery , Endometrium/drug effects , Endometrium/transplantation , Epithelial-Mesenchymal Transition/drug effects , Estradiol/pharmacology , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Ovarian Cysts/etiology , Ovarian Cysts/surgery , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , rho-Associated Kinases/biosynthesis , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/biosynthesis , rhoA GTP-Binding Protein/genetics
7.
J Am Soc Nephrol ; 31(8): 1762-1780, 2020 08.
Article in English | MEDLINE | ID: mdl-32709711

ABSTRACT

BACKGROUND: Diabetic nephropathy (dNP), now the leading cause of ESKD, lacks efficient therapies. Coagulation protease-dependent signaling modulates dNP, in part via the G protein-coupled, protease-activated receptors (PARs). Specifically, the cytoprotective protease-activated protein C (aPC) protects from dNP, but the mechanisms are not clear. METHODS: A combination of in vitro approaches and mouse models evaluated the role of aPC-integrin interaction and related signaling in dNP. RESULTS: The zymogen protein C and aPC bind to podocyte integrin-ß3, a subunit of integrin-αvß3. Deficiency of this integrin impairs thrombin-mediated generation of aPC on podocytes. The interaction of aPC with integrin-αvß3 induces transient binding of integrin-ß3 with G α13 and controls PAR-dependent RhoA signaling in podocytes. Binding of aPC to integrin-ß3via its RGD sequence is required for the temporal restriction of RhoA signaling in podocytes. In podocytes lacking integrin-ß3, aPC induces sustained RhoA activation, mimicking the effect of thrombin. In vivo, overexpression of wild-type aPC suppresses pathologic renal RhoA activation and protects against dNP. Disrupting the aPC-integrin-ß3 interaction by specifically deleting podocyte integrin-ß3 or by abolishing aPC's integrin-binding RGD sequence enhances RhoA signaling in mice with high aPC levels and abolishes aPC's nephroprotective effect. Pharmacologic inhibition of PAR1, the pivotal thrombin receptor, restricts RhoA activation and nephroprotects RGE-aPChigh and wild-type mice.Conclusions aPC-integrin-αvß3 acts as a rheostat, controlling PAR1-dependent RhoA activation in podocytes in diabetic nephropathy. These results identify integrin-αvß3 as an essential coreceptor for aPC that is required for nephroprotective aPC-PAR signaling in dNP.


Subject(s)
Diabetic Nephropathies/prevention & control , Integrin beta3/physiology , Podocytes/physiology , Protein C/physiology , rhoA GTP-Binding Protein/physiology , Animals , Cytoprotection , Endothelial Protein C Receptor/physiology , GTP-Binding Protein alpha Subunits, G12-G13/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptor, PAR-1/physiology
8.
Front Immunol ; 11: 219, 2020.
Article in English | MEDLINE | ID: mdl-32133004

ABSTRACT

Inhalation of crystalline silica (SiO2) is a risk factor of systemic autoimmune diseases such as systemic sclerosis (SSc) and fibrotic pulmonary disorders such as silicosis. A defect of apoptotic cell clearance (i.e., efferocytosis, a key process in the resolution of inflammation) is reported in macrophages from patients with fibrotic or autoimmune diseases. However, the precise links between SiO2 exposure and efferocytosis impairment remain to be determined. Answering to this question may help to better link innate immunity and fibrosis. In this study, we first aim to determine whether SiO2 might alter efferocytosis capacities of human and mouse macrophages. We secondly explore possible mechanisms explaining efferocytosis impairment, with a specific focus on macrophage polarization and on the RhoA/ROCK pathway, a key regulator of cytoskeleton remodeling and phagocytosis. Human monocyte-derived macrophages (MDM) and C57BL/6J mice exposed to SiO2 and to CFSE-positive apoptotic Jurkat cells were analyzed by flow cytometry to determine their efferocytosis index (EI). The effects of ROCK inhibitors (Y27632 and Fasudil) on EI of SiO2-exposed MDM and MDM from SSc patients were evaluated in vitro. Our results demonstrated that SiO2 significantly decreased EI of human MDM in vitro and mouse alveolar macrophages in vivo. In human MDM, this SiO2-associated impairment of efferocytosis, required the expression of the membrane receptor SR-B1 and was associated with a decreased expression of M2 polarization markers (CD206, CD204, and CD163). F-actin staining, RhoA activation and impairment of efferocytosis, all induced by SiO2, were reversed by ROCK inhibitors. Moreover, the EI of MDM from SSc patients was similar to the EI of in vitro- SiO2-exposed MDM and Y27632 significantly increased SSc MDM efferocytosis capacities, suggesting a likewise activation of the RhoA/ROCK pathway in SSc. Altogether, our results demonstrate that SiO2 exposure may contribute to the impairment of efferocytosis capacities of mouse and human macrophages but also of MDM in SiO2-associated autoimmune diseases and fibrotic disorders such as SSc; in this context, the silica/RhoA/ROCK pathway may constitute a relevant therapeutic target.


Subject(s)
Apoptosis/drug effects , Macrophages/drug effects , Phagocytosis/drug effects , Scleroderma, Systemic/chemically induced , Silicon Dioxide/toxicity , Animals , Female , Humans , Jurkat Cells , Macrophages/physiology , Mice , Mice, Inbred C57BL , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology
9.
J Pharmacol Exp Ther ; 373(2): 214-219, 2020 05.
Article in English | MEDLINE | ID: mdl-32144123

ABSTRACT

Sickle cell disease (SCD) is associated with overactive bladder (OAB). Detrusor overactivity, a component of OAB, is present in an SCD mouse, but the molecular mechanisms for this condition are not well-defined. We hypothesize that nitric oxide (NO)/ ras homolog gene family (Rho) A/Rho-associated kinase (ROCK) dysregulation is a mechanism for detrusor overactivity and that NO-releasing nanoparticles (NO-nps), a novel NO delivery system, may serve to treat this condition. Male adult SCD transgenic, combined endothelial NO synthases (eNOSs) and neuronal NOS (nNOS) gene-deficient (dNOS-/-), and wild-type (WT) mice were used. Empty nanoparticle or NO-np was injected into the bladder, followed by cystometric studies. The expression levels of phosphorylated eNOS (Ser-1177), protein kinase B (Akt) (Ser-473), nNOS (Ser-1412), and myosin phosphatase target subunit 1 (MYPT1) (Thr-696) were assessed in the bladder. SCD and dNOS-/- mice had a greater (P < 0.05) number of voiding and nonvoiding contractions compared with WT mice, and they were normalized by NO-np treatment. eNOS (Ser-1177) and AKT (Ser-473) phosphorylation were decreased (P < 0.05) in the bladder of SCD compared with WT mice and reversed by NO-np. Phosphorylated MYPT1, a marker of the RhoA/ROCK pathway, was increased (P < 0.05) in the bladder of SCD mice compared with WT and reversed by NO-np. nNOS phosphorylation on positive (Ser-1412) regulatory site was decreased (P < 0.05) in the bladder of SCD mice compared with WT and was not affected by NO-np. NO-nps did not affect any of the measured parameters in WT mice. In conclusion, dysregulation of NO and RhoA/ROCK pathways is associated with detrusor overactivity in SCD mice; NO-np reverses these molecular derangements in the bladder and decreases detrusor overactivity. SIGNIFICANCE STATEMENT: Voiding abnormalities commonly affect patients with sickle cell disease (SCD) but are problematic to treat. Clarification of the science for this condition in an animal model of SCD may lead to improved interventions for it. Our findings suggest that novel topical delivery of a vasorelaxant agent nitric oxide into the bladder of these mice corrects overactive bladder by improving deranged bladder physiology regulatory signaling.


Subject(s)
Nanoparticles/therapeutic use , Nitric Oxide/physiology , Urinary Bladder, Overactive/drug therapy , rho-Associated Kinases/physiology , Anemia, Sickle Cell/complications , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase/physiology , Phosphorylation , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology
10.
Commun Biol ; 3(1): 76, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060357

ABSTRACT

Activity-dependent remodeling of excitatory connections underpins memory formation in the brain. Serotonin receptors are known to contribute to such remodeling, yet the underlying molecular machinery remains poorly understood. Here, we employ high-resolution time-lapse FRET imaging in neuroblastoma cells and neuronal dendrites to establish that activation of serotonin receptor 5-HT4 (5-HT4R) rapidly triggers spatially-restricted RhoA activity and G13-mediated phosphorylation of cofilin, thus locally boosting the filamentous actin fraction. In neuroblastoma cells, this leads to cell rounding and neurite retraction. In hippocampal neurons in situ, 5-HT4R-mediated RhoA activation triggers maturation of dendritic spines. This is paralleled by RhoA-dependent, transient alterations in cell excitability, as reflected by increased spontaneous synaptic activity, apparent shunting of evoked synaptic responses, and enhanced long-term potentiation of excitatory transmission. The 5-HT4R/G13/RhoA signaling thus emerges as a previously unrecognized molecular pathway underpinning use-dependent functional remodeling of excitatory synaptic connections.


Subject(s)
Actins/metabolism , Dendritic Spines/physiology , Receptors, Serotonin, 5-HT4/physiology , Synapses/physiology , rhoA GTP-Binding Protein/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/physiology , Animals , Animals, Newborn , Cells, Cultured , Female , Long-Term Potentiation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT4/genetics , Signal Transduction/genetics , Synaptic Transmission/physiology
11.
Mol Biol Cell ; 31(8): 741-752, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32049581

ABSTRACT

Collective cell migration plays crucial roles in tissue remodeling, wound healing, and cancer cell invasion. However, its underlying mechanism remains unknown. Previously, we showed that the RhoA-targeting guanine nucleotide exchange factor Solo (ARHGEF40) is required for tensile force-induced RhoA activation and proper organization of keratin-8/keratin-18 (K8/K18) networks. Here, we demonstrate that Solo knockdown significantly increases the rate at which Madin-Darby canine kidney cells collectively migrate on collagen gels. However, it has no apparent effect on the migratory speed of solitary cultured cells. Therefore, Solo decelerates collective cell migration. Moreover, Solo localized to the anteroposterior regions of cell-cell contact sites in collectively migrating cells and was required for the local accumulation of K8/K18 filaments in the forward areas of the cells. Partial Rho-associated protein kinase (ROCK) inhibition or K18 or plakoglobin knockdown also increased collective cell migration velocity. These results suggest that Solo acts as a brake for collective cell migration by generating pullback force at cell-cell contact sites via the RhoA-ROCK pathway. It may also promote the formation of desmosomal cell-cell junctions related to K8/K18 filaments and plakoglobin.


Subject(s)
Cell Movement/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/physiology , Amides/pharmacology , Animals , Cell Polarity , Collagen , Cytoskeleton/physiology , Desmosomes/physiology , Dogs , Gels , Gene Knockdown Techniques , Keratin-18/antagonists & inhibitors , Keratin-18/genetics , Keratin-18/physiology , Keratin-8/antagonists & inhibitors , Keratin-8/genetics , Keratin-8/physiology , Madin Darby Canine Kidney Cells , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Stress, Mechanical , Time-Lapse Imaging , gamma Catenin/antagonists & inhibitors , gamma Catenin/genetics , gamma Catenin/physiology , rac1 GTP-Binding Protein/physiology , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/physiology
12.
Curr Biol ; 29(22): 3874-3886.e9, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31679934

ABSTRACT

The specification of an axon and its subsequent outgrowth are key steps during neuronal polarization, a prerequisite to wire the brain. The Rho-guanosine triphosphatase (GTPase) RhoA is believed to be a central player in these processes. However, its physiological role has remained undefined. Here, genetic loss- and gain-of-function experiments combined with time-lapse microscopy, cell culture, and in vivo analysis show that RhoA is not involved in axon specification but confines the initiation of neuronal polarization and axon outgrowth during development. Biochemical analysis and super-resolution microscopy together with molecular and pharmacological manipulations reveal that RhoA restrains axon growth by activating myosin-II-mediated actin arc formation in the growth cone to prevent microtubules from protruding toward the leading edge. Through this mechanism, RhoA regulates the duration of axon growth and pause phases, thus controlling the tightly timed extension of developing axons. Thereby, this work unravels physiologically relevant players coordinating actin-microtubule interactions during axon growth.


Subject(s)
Axons/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Axons/physiology , Brain/embryology , Brain/metabolism , Cell Polarity/physiology , Female , Gain of Function Mutation/genetics , Growth Cones/metabolism , Loss of Function Mutation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/metabolism , Myosin Type II/metabolism , Neurogenesis/physiology , Neurons/metabolism , rhoA GTP-Binding Protein/physiology
13.
Mol Nutr Food Res ; 63(22): e1900489, 2019 11.
Article in English | MEDLINE | ID: mdl-31483951

ABSTRACT

SCOPE: Podocytes are a component of glomerular filtration barrier with interdigitating foot processes. The podocyte function depends on the dynamics of actin cytoskeletal and focal adhesion crucial for foot process structure. This study investigates the renoprotective effects of eucalyptol on the F-actin cytoskeleton formation and focal adhesion assembly in glucose-loaded podocytes and diabetic kidneys. METHODS AND RESULTS: Eucalyptol at 1-20 µm reverses the reduction of cellular level of F-actin, ezrin, cortactin, and Arp2/3 in 33 mm glucose-loaded mouse podocytes, and oral administration of 10 mg kg-1 eucalyptol elevates tissue levels of actin cytoskeletal proteins reduced in db/db mouse kidneys. Eucalyptol inhibits podocyte morphological changes, showing F-actin cytoskeleton formation in cortical regions and agminated F-actin along the cell periphery. Eucalyptol induces focal adhesion proteins of paxillin, vinculin, talin1, FAK, and Src in glucose-exposed podocytes and diabetic kidneys. Additionally, GTP-binding Rac1, Cdc42, Rho A, and ROCK are upregulated in glucose-stimulated podocytes and diabetic kidneys, which is attenuated by supplying eucalyptol. Rho A gene depletion partially diminishes GSK3ß induction of podocytes by glucose. CONCLUSION: Eucalyptol ameliorates F-actin cytoskeleton formation and focal adhesion assembly through blockade of the Rho signaling pathway, entailing partial involvement of GSK3ß, which may inhibit barrier dysfunction of podocytes and resultant proteinuria.


Subject(s)
Actin Cytoskeleton/drug effects , Diabetic Nephropathies/drug therapy , Eucalyptol/pharmacology , Focal Adhesions/drug effects , Glucose/toxicity , Podocytes/drug effects , Actin Cytoskeleton/physiology , Animals , Cells, Cultured , Glomerular Basement Membrane/physiology , Glycogen Synthase Kinase 3 beta/physiology , Male , Mice , Mice, Inbred C57BL , rhoA GTP-Binding Protein/physiology
14.
Philos Trans R Soc Lond B Biol Sci ; 374(1779): 20180229, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31431179

ABSTRACT

Cells respond and adapt to their physical environments and to the mechanical forces that they experience. The translation of physical forces into biochemical signalling pathways is known as mechanotransduction. In this review, we focus on two aspects of mechanotransduction. First, we consider how forces exerted on cell adhesion molecules at the cell surface regulate the RhoA signalling pathway by controlling the activities of guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). In the second part of the review, we discuss how the nucleus contributes to mechanotransduction as a physical structure connected to the cytoskeleton. We focus on recent studies that have either severed the connections between the nucleus and the cytoskeleton, or that have entirely removed the nucleus from cells. These actions reduce the levels of active RhoA, thereby altering the mechanical properties of cells and decreasing their ability to generate tension and respond to external mechanical forces. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.


Subject(s)
Cell Membrane/physiology , Cell Nucleus/physiology , Mechanotransduction, Cellular/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology , Humans
15.
Sci Rep ; 9(1): 8302, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31165762

ABSTRACT

Muscle differentiation is controlled by adhesion and growth factor-dependent signalling through common effectors that regulate muscle-specific transcriptional programs. Here we report that mDiaphanous1, an effector of adhesion-dependent RhoA-signalling, negatively regulates myogenesis at the level of Myogenin expression. In myotubes, over-expression of mDia1ΔN3, a RhoA-independent mutant, suppresses Myogenin promoter activity and expression. We investigated mDia1-interacting proteins that may counteract mDia1 to permit Myogenin expression and timely differentiation. Using yeast two-hybrid and mass-spectrometric analysis, we report that mDia1 has a stage-specific interactome, including Prohibitin2, MyoD, Akt2, and ß-Catenin, along with a number of proteosomal and mitochondrial components. Of these interacting partners, Prohibitin2 colocalises with mDia1 in cytoplasmic punctae in myotubes. We mapped the interacting domains of mDia1 and Phb2, and used interacting (mDia1ΔN3/Phb2 FL or mDia1ΔN3/Phb2-Carboxy) and non-interacting pairs (mDia1H + P/Phb2 FL or mDia1ΔN3/Phb2-Amino) to dissect the functional consequences of this partnership on Myogenin promoter activity. Co-expression of full-length as well as mDia1-interacting domains of Prohibitin2 reverse the anti-myogenic effects of mDia1ΔN3, while non-interacting regions do not. Our results suggest that Prohibitin2 sequesters mDia1, dampens its anti-myogenic activity and fine-tunes RhoA-mDia1 signalling to promote differentiation. Overall, we report that mDia1 is multi-functional signalling effector whose anti-myogenic activity is modulated by a differentiation-dependent interactome. The data have been deposited to the ProteomeXchange with identifier PXD012257.


Subject(s)
Formins/physiology , Promoter Regions, Genetic , Repressor Proteins/physiology , rhoA GTP-Binding Protein/physiology , Animals , Cell Differentiation , Cytoplasm/metabolism , HEK293 Cells , Humans , Mice , Muscle Development , Muscle Fibers, Skeletal/metabolism , Mutation , MyoD Protein/metabolism , Myogenin/metabolism , Prohibitins , Protein Binding , Protein Domains , Signal Transduction , Two-Hybrid System Techniques
16.
Am J Physiol Cell Physiol ; 317(3): C457-C465, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31216194

ABSTRACT

Cell motility and migration play critical roles in various physiological processes and disease states. Here, we show that the BBBsome, a macromolecule composed of eight Bardet-Biedl syndrome (BBS) proteins including BBS1, is a critical determinant of cell migration and wound healing. Fibroblast cells derived from mice or humans harboring a homozygous missense mutation (BBS1M390R/M390R) that disrupt the BBSome exhibit defects in migration and wound healing. Furthermore, we demonstrate that BBS1M390R/M390R mice have significantly delayed wound closure. In line with this, we provide data suggesting that BBS1M390R/M390R fibroblasts have impaired platelet-derived growth factor-AA (PDGF) receptor-α signaling, a key regulator of directional cell migration acting as a chemoattractant during postnatal migration responses such as wound healing. In addition, we show that BBS1M390R/M390R fibroblasts have upregulated RhoA expression and activity. The relevance of RhoA upregulation is demonstrated by the ability of RhoA-kinase inhibitor Y27632 to partially rescue the migration defect of BBS1M390R/M390R fibroblasts cells. We also show that accumulation of RhoA protein in BBS1M390R/M390R fibroblasts cells is associated with reduction and inactivation of the ubiquitin ligase Cullin-3. Consistent with this, Cullin-3 inhibition with MLN4924 is sufficient to reduce migration of normal fibroblasts. These data implicate the BBSome in cell motility and tissue repair through a mechanism that involves PDGF receptor signaling and Cullin-3-mediated control of RhoA.


Subject(s)
Bardet-Biedl Syndrome , Cell Movement/physiology , Cullin Proteins/physiology , Microtubule-Associated Proteins/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology , Animals , Bardet-Biedl Syndrome/genetics , Cell Movement/drug effects , Cells, Cultured , Cullin Proteins/antagonists & inhibitors , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gene Knock-In Techniques/methods , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyrimidines/pharmacology , rhoA GTP-Binding Protein/antagonists & inhibitors
17.
Acta Neuropathol ; 138(2): 275-293, 2019 08.
Article in English | MEDLINE | ID: mdl-31062076

ABSTRACT

Glioblastomas strongly invade the brain by infiltrating into the white matter along myelinated nerve fiber tracts even though the myelin protein Nogo-A prevents cell migration by activating inhibitory RhoA signaling. The mechanisms behind this long-known phenomenon remained elusive so far, precluding a targeted therapeutic intervention. This study demonstrates that the prevalent activation of AKT in gliomas increases the ER protein-folding capacity and enables tumor cells to utilize a side effect of RhoA activation: the perturbation of the IRE1α-mediated decay of SPARC mRNA. Once translation is initiated, glioblastoma cells rapidly secrete SPARC to block Nogo-A from inhibiting migration via RhoA. By advanced ultramicroscopy for studying single-cell invasion in whole, undissected mouse brains, we show that gliomas require SPARC for invading into white matter structures. SPARC depletion reduces tumor dissemination that significantly prolongs survival and improves response to cytostatic therapy. Our finding of a novel RhoA-IRE1 axis provides a druggable target for interfering with SPARC production and underscores its therapeutic value.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplasm Proteins/physiology , Nogo Proteins/biosynthesis , Osteonectin/biosynthesis , Protein Biosynthesis , White Matter/pathology , rhoA GTP-Binding Protein/physiology , Animals , Binding, Competitive , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasm Invasiveness , Nogo Proteins/genetics , Osteonectin/genetics , Protein Domains , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Sphingosine-1-Phosphate Receptors/physiology , Tumor Cells, Cultured , White Matter/metabolism
18.
Biol Psychiatry ; 85(12): 1001-1010, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30955841

ABSTRACT

BACKGROUND: Nucleus accumbens dopamine 1 receptor medium spiny neurons (D1-MSNs) play a critical role in the development of depression-like behavior in mice. Social defeat stress causes dendritic morphological changes on this MSN subtype through expression and activation of early growth response 3 (EGR3) and the Rho guanosine triphosphatase RhoA. However, it is unknown how RhoA inhibition affects electrophysiological properties underlying stress-induced susceptibility. METHODS: A novel RhoA-specific inhibitor, Rhosin, was used to inhibit RhoA activity following chronic social defeat stress. Whole-cell electrophysiological recordings of D1-MSNs were performed to assess synaptic and intrinsic consequences of Rhosin treatment on stressed mice. Additionally, recorded cells were filled and analyzed for their morphological properties. RESULTS: We found that RhoA inhibition prevents both D1-MSN hyperexcitability and reduced excitatory input to D1-MSNs caused by social defeat stress. Nucleus accumbens-specific RhoA inhibition is capable of blocking susceptibility caused by D1-MSN EGR3 expression. Lastly, we found that Rhosin enhances spine density, which correlates with D1-MSN excitability, without affecting overall dendritic branching. CONCLUSIONS: These findings demonstrate that pharmacological inhibition of RhoA during stress drives an enhancement of total spine number in a subset of nucleus accumbens neurons that prevents stress-related electrophysiological deficits and promotes stress resiliency.


Subject(s)
Neuronal Plasticity , Neurons/physiology , Nucleus Accumbens/physiology , Organic Chemicals/administration & dosage , Receptors, Dopamine D1/physiology , Resilience, Psychological , Stress, Psychological/physiopathology , rhoA GTP-Binding Protein/physiology , Animals , Early Growth Response Protein 3/metabolism , Male , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Resilience, Psychological/drug effects , rhoA GTP-Binding Protein/antagonists & inhibitors
19.
Small GTPases ; 10(6): 466-484, 2019 11.
Article in English | MEDLINE | ID: mdl-28949796

ABSTRACT

RhoGTPases are known regulators of intracellular actin dynamics that are important for maintaining endothelial barrier function. RhoA is most extensively studied as a key regulator of endothelial barrier function, however the function of the 2 highly homologous family-members (> 88%) RhoB and RhoC in endothelial barrier function is still poorly understood. This study aimed to determine whether RhoA, RhoB and RhoC have overlapping or distinct roles in barrier function and permeability in resting and activated endothelium. By using primary endothelial cells in combination with siRNA transfection to establish individual, double or triple knockdown of the RhoA/B/C RhoGTPases, we found that RhoB, but not RhoA or RhoC, is in resting endothelium a negative regulator of permeability. Loss of RhoB accounted for an accumulation of VE-cadherin at cell-cell contacts. Thrombin-induced loss of endothelial integrity is mediated primarily by RhoA and RhoB. Combined loss of RhoA/B showed decreased phosphorylation of Myosin Light Chain and increased expression of VE-cadherin at cell-cell contacts after thrombin stimulation. RhoC contributes to the Rac1-dependent restoration of endothelial barrier function. In summary, this study shows that these highly homologous RhoGTPases differentially control the dynamics of endothelial barrier function.


Subject(s)
Human Umbilical Vein Endothelial Cells/physiology , rhoA GTP-Binding Protein/physiology , rhoB GTP-Binding Protein/physiology , rhoC GTP-Binding Protein/physiology , Actins/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Communication , Cells, Cultured , Humans , RNA, Small Interfering , Thrombin/pharmacology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/genetics , rhoC GTP-Binding Protein/genetics
20.
Small GTPases ; 10(1): 26-32, 2019 01.
Article in English | MEDLINE | ID: mdl-28125332

ABSTRACT

In this commentary we discuss a paper we published recently on the activities of the GTPase RhoA during neural differentiation of murine embryonic stem cells, and relate our findings to previous studies. We narrate how we found that RhoA impedes neural differentiation by inhibiting the production as well as the secretion of noggin, a soluble factor that antagonizes bone morphogenetic protein. We discuss how the questions we tried to address shaped the study, and how embryonic stem cells isolated from a genetically modified mouse model devoid of Syx, a RhoA-specific guanine exchange factor, were used to address them. We detail several signaling pathways downstream of RhoA that are hindered by the absence of Syx, and obstructed by retinoic acid, resulting in an increase of noggin production; we explain how the lower RhoA activity and, consequently, the sparser peri-junctional stress fibers in Syx-/- cells facilitated noggin secretion; and we report unpublished results showing that pharmacological inhibition of RhoA accelerates the neuronal differentiation of human embryonic stem cells. Finally, we identify signaling mechanisms in our recent study that warrant further study, and speculate on the possibility of manipulating RhoA signaling in combination with other pathways to drive the differentiation of neuronal subtypes.


Subject(s)
Embryonic Stem Cells/cytology , Neurogenesis , rhoA GTP-Binding Protein/physiology , Animals , Carrier Proteins/physiology , Embryonic Stem Cells/physiology , Humans , Smad1 Protein/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...