Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 726393, 2021.
Article in English | MEDLINE | ID: mdl-34721389

ABSTRACT

RhoA of the Rho GTPase family is prenylated at its C-terminus. Prenylation of RhoA has been shown to control T helper 17 (Th17) cell-mediated colitis. By characterizing T cell-specific RhoA conditional knockout mice, we have recently shown that RhoA is required for Th2 and Th17 cell differentiation and Th2/Th17 cell-mediated allergic airway inflammation. It remains unclear whether RhoA plays a cell-intrinsic role in regulatory T (Treg) cells that suppress effector T cells such as Th2/Th17 cells to maintain immune tolerance and to promote tumor immune evasion. Here we have generated Treg cell-specific RhoA-deficient mice. We found that homozygous RhoA deletion in Treg cells led to early, fatal systemic inflammatory disorders. The autoimmune responses came from an increase in activated CD4+ and CD8+ T cells and in effector T cells including Th17, Th1 and Th2 cells. The immune activation was due to impaired Treg cell homeostasis and increased Treg cell plasticity. Interestingly, heterozygous RhoA deletion in Treg cells did not affect Treg cell homeostasis nor cause systemic autoimmunity but induced Treg cell plasticity and an increase in effector T cells. Importantly, heterozygous RhoA deletion significantly inhibited tumor growth, which was associated with tumor-infiltrating Treg cell plasticity and increased tumor-infiltrating effector T cells. Collectively, our findings suggest that graded RhoA expression in Treg cells distinguishes tumor immunity from autoimmunity and that rational targeting of RhoA in Treg cells may trigger anti-tumor T cell immunity without causing autoimmune responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , rhoA GTP-Binding Protein/deficiency , Animals , Autoimmunity , Cell Line, Tumor , Female , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Tumor Escape , rhoA GTP-Binding Protein/immunology
2.
J Neuroinflammation ; 18(1): 234, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34654444

ABSTRACT

BACKGROUND: Plenty of macrophages are recruited to the injured nerve to play key roles in the immunoreaction and engulf the debris of degenerated axons and myelin during Wallerian degeneration, thus creating a conducive microenvironment for nerve regeneration. Recently, drugs targeting the RhoA pathway have been widely used to promote peripheral axonal regeneration. However, the role of RhoA in macrophage during Wallerian degeneration and nerve regeneration after peripheral nerve injury is still unknown. Herein, we come up with the hypothesis that RhoA might influence Wallerian degeneration and nerve regeneration by affecting the migration and phagocytosis of macrophages after peripheral nerve injury. METHODS: Immunohistochemistry, Western blotting, H&E staining, and electrophysiology were performed to access the Wallerian degeneration and axonal regeneration after sciatic nerve transection and crush injury in the LyzCre+/-; RhoAflox/flox (cKO) mice or Lyz2Cre+/- (Cre) mice, regardless of sex. Macrophages' migration and phagocytosis were detected in the injured nerves and the cultured macrophages. Moreover, the expression and potential roles of ROCK and MLCK were also evaluated in the cultured macrophages. RESULTS: 1. RhoA was specifically knocked out in macrophages of the cKO mice; 2. The segmentation of axons and myelin, the axonal regeneration, and nerve conduction in the injured nerve were significantly impeded while the myoatrophy was more severe in the cKO mice compared with those in Cre mice; 3. RhoA knockout attenuated the migration and phagocytosis of macrophages in vivo and in vitro; 4. ROCK and MLCK were downregulated in the cKO macrophages while inhibition of ROCK and MLCK could weaken the migration and phagocytosis of macrophages. CONCLUSIONS: Our findings suggest that RhoA depletion in macrophages exerts a detrimental effect on Wallerian degeneration and nerve regeneration, which is most likely due to the impaired migration and phagocytosis of macrophages resulted from disrupted RhoA/ROCK/MLCK pathway. Since previous research has proved RhoA inhibition in neurons was favoring for axonal regeneration, the present study reminds us of that the cellular specificity of RhoA-targeted drugs is needed to be considered in the future application for treating peripheral nerve injury.


Subject(s)
Macrophages/metabolism , Peripheral Nerve Injuries/metabolism , Wallerian Degeneration/metabolism , Wallerian Degeneration/prevention & control , rhoA GTP-Binding Protein/deficiency , Animals , Cell Movement/physiology , Cells, Cultured , Female , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peripheral Nerve Injuries/pathology , Wallerian Degeneration/pathology , rhoA GTP-Binding Protein/genetics
3.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34101619

ABSTRACT

The small GTPase RhoA and its downstream effectors are critical regulators in the pathophysiological processes of asthma. The underlying mechanism, however, remains undetermined. Here, we generated an asthma mouse model with RhoA-conditional KO mice (Sftpc-cre;RhoAfl/fl) in type II alveolar epithelial cells (AT2) and demonstrated that AT2 cell-specific deletion of RhoA leads to exacerbation of allergen-induced airway hyperresponsiveness and airway inflammation with elevated Th2 cytokines in bronchoalveolar lavage fluid (BALF). Notably, Sftpc-cre;RhoAfl/fl mice showed a significant reduction in Tgf-ß1 levels in BALF and lung tissues, and administration of recombinant Tgf-ß1 to the mice rescued Tgf-ß1 and alleviated the increased allergic airway inflammation observed in Sftpc-cre;RhoAfl/fl mice. Using RNA sequencing technology, we identified Slc26a4 (pendrin), a transmembrane anion exchange, as the most upregulated gene in RhoA-deficient AT2 cells. The upregulation of SLC26A4 was further confirmed in AT2 cells of asthmatic patients and mouse models and in human airway epithelial cells expressing dominant-negative RHOA (RHOA-N19). SLA26A4 was also elevated in serum from asthmatic patients and negatively associated with the percentage of forced expiratory volume in 1 second (FEV1%). Furthermore, SLC26A4 inhibition promoted epithelial TGF-ß1 release and attenuated allergic airway inflammation. Our study reveals a RhoA/SLC26A4 axis in AT2 cells that functions as a protective mechanism against allergic airway inflammation.


Subject(s)
Alveolar Epithelial Cells/immunology , Asthma/immunology , Sulfate Transporters/metabolism , rhoA GTP-Binding Protein/deficiency , Alveolar Epithelial Cells/metabolism , Animals , Asthma/drug therapy , Asthma/pathology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Humans , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Ovalbumin/administration & dosage , Ovalbumin/immunology , Recombinant Proteins/administration & dosage , Symptom Flare Up , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/metabolism , rhoA GTP-Binding Protein/genetics
4.
Cell Rep ; 31(12): 107796, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579923

ABSTRACT

Nervous tissue homeostasis requires the regulation of microglia activity. Using conditional gene targeting in mice, we demonstrate that genetic ablation of the small GTPase Rhoa in adult microglia is sufficient to trigger spontaneous microglia activation, producing a neurological phenotype (including synapse and neuron loss, impairment of long-term potentiation [LTP], formation of ß-amyloid plaques, and memory deficits). Mechanistically, loss of Rhoa in microglia triggers Src activation and Src-mediated tumor necrosis factor (TNF) production, leading to excitotoxic glutamate secretion. Inhibiting Src in microglia Rhoa-deficient mice attenuates microglia dysregulation and the ensuing neurological phenotype. We also find that the Rhoa/Src signaling pathway is disrupted in microglia of the APP/PS1 mouse model of Alzheimer disease and that low doses of Aß oligomers trigger microglia neurotoxic polarization through the disruption of Rhoa-to-Src signaling. Overall, our results indicate that disturbing Rho GTPase signaling in microglia can directly cause neurodegeneration.


Subject(s)
Aging/pathology , Microglia/pathology , Nerve Degeneration/pathology , Neurons/metabolism , rhoA GTP-Binding Protein/deficiency , Aging/metabolism , Amyloid beta-Peptides/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Cell Polarity , Cell Survival , Mice, Inbred C57BL , Microglia/metabolism , Phenotype , Synapses/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
5.
Sci Rep ; 9(1): 11666, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406143

ABSTRACT

Imbalanced angiogenesis is a characteristic of several diseases. Rho GTPases regulate multiple cellular processes, such as cytoskeletal rearrangement, cell movement, microtubule dynamics, signal transduction and gene expression. Among the Rho GTPases, RhoA, Rac1 and Cdc42 are best characterized. The role of endothelial Rac1 and Cdc42 in embryonic development and retinal angiogenesis has been studied, however the role of endothelial RhoA is yet to be explored. Here, we aimed to identify the role of endothelial RhoA in endothelial cell functions, in embryonic and retinal development and explored compensatory mechanisms. In vitro, RhoA is involved in cell proliferation, migration and tube formation, triggered by the angiogenesis inducers Vascular Endothelial Growth Factor (VEGF) and Sphingosine-1 Phosphate (S1P). In vivo, through constitutive and inducible endothelial RhoA deficiency we tested the role of endothelial RhoA in embryonic development and retinal angiogenesis. Constitutive endothelial RhoA deficiency, although decreased survival, was not detrimental for embryonic development, while inducible endothelial RhoA deficiency presented only mild deficiencies in the retina. The redundant role of RhoA in vivo can be attributed to potential differences in the signaling cues regulating angiogenesis in physiological versus pathological conditions and to the alternative compensatory mechanisms that may be present in the in vivo setting.


Subject(s)
Endothelium, Vascular/metabolism , Neovascularization, Physiologic , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Embryo, Mammalian , Embryonic Development , Endothelium, Vascular/cytology , Female , Human Umbilical Vein Endothelial Cells , Humans , Lysophospholipids/metabolism , Male , Mice, Transgenic , Retinal Vessels/embryology , Retinal Vessels/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Vascular Endothelial Growth Factor A/metabolism , rhoA GTP-Binding Protein/genetics
6.
J Leukoc Biol ; 106(5): 1139-1151, 2019 11.
Article in English | MEDLINE | ID: mdl-31260596

ABSTRACT

Asthma is a heterogeneous chronic airway inflammation in which Th2 and Th17 cells are key players in its pathogenesis. We have reported that RhoA of Rho GTPases orchestrated glycolysis for Th2 cell differentiation and allergic airway inflammation by the use of a conditional RhoA-deficient mouse line. However, the role of RhoA in Th17 cells remains to be elucidated. In this study, we investigated the effects of RhoA deficiency on Th17 cells in the context of ex vivo cell culture systems and an in vivo house dust mites (HDM)-induced allergic airway inflammation. We found that RhoA deficiency inhibited Th17 differentiation and effector cytokine secretion, which was associated with the downregulations of Stat3 and Rorγt, key Th17 transcription factors. Furthermore, loss of RhoA markedly suppressed Th17 and neutrophil-involved airway inflammation induced by HDM in mice. The infiltrating inflammatory cells in the lungs and bronchoalveolar lavage (BAL) fluids were dramatically reduced in conditional RhoA-deficient mice. Th17 as well as Th2 effector cytokines were suppressed in the airways at both protein and mRNA levels. Interestingly, Y16, a specific RhoA inhibitor, was able to recapitulate the most phenotypes of RhoA genetic deletion in Th17 differentiation and allergic airway inflammation. Our data demonstrate that RhoA is a key regulator of Th17 cell differentiation and function. RhoA might serve as a potential novel therapeutic target for asthma and other inflammatory disorders.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Pyroglyphidae/immunology , Th17 Cells/immunology , rhoA GTP-Binding Protein/deficiency , Animals , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Cell Differentiation/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology , rhoA GTP-Binding Protein/immunology
7.
PLoS One ; 12(2): e0172613, 2017.
Article in English | MEDLINE | ID: mdl-28222172

ABSTRACT

OBJECTIVE: Epidermal stem cells (ESCs) play a critical role in wound healing, but the mechanism underlying ESC proliferation is not well defined. Here, we explore the effects of RhoA on ESC proliferation and the possible underlying mechanism. METHODS: Human ESCs were enriched by rapid adhesion to collagen IV. RhoA(+/+)(G14V), RhoA(-/-)(T19N) and pGFP control plasmids were transfected into human ESCs. The effect of RhoA on cell proliferation was detected by cell proliferation and DNA synthesis assays. Induction of PKN1 activity by RhoA was determined by immunoblot analysis, and the effects of PKN1 on RhoA in terms of inducing cell proliferation and cyclin D1 expression were detected using specific siRNA targeting PKN1. The effects of U-46619 (a RhoA agonist) and C3 transferase (a RhoA antagonist) on ESC proliferation were observed in vivo. RESULTS: RhoA had a positive effect on ESC proliferation, and PKN1 activity was up-regulated by the active RhoA mutant (G14V) and suppressed by RhoA T19N. Moreover, the ability of RhoA to promote ESC proliferation and DNA synthesis was interrupted by PKN1 siRNA. Additionally, cyclin D1 protein and mRNA expression levels were up-regulated by RhoA G14V, and these effects were inhibited by siRNA-mediated knock-down of PKN1. RhoA also promoted ESC proliferation via PKN in vivo. CONCLUSION: This study shows that the effect of RhoA on ESC proliferation is mediated by activation of the PKN1-cyclin D1 pathway in vitro, suggesting that RhoA may serve as a new therapeutic target for wound healing.


Subject(s)
Cyclin D1/physiology , Epidermal Cells , Epithelial Cells/metabolism , Protein Kinase C/physiology , Signal Transduction/physiology , Stem Cells/metabolism , Wound Healing , rhoA GTP-Binding Protein/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP Ribose Transferases/pharmacology , Animals , Botulinum Toxins/pharmacology , Burns/physiopathology , Burns/therapy , Carbazoles/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/biosynthesis , Cyclin D1/genetics , DNA Replication/drug effects , Epithelial Cells/cytology , Humans , Indole Alkaloids/pharmacology , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Primary Cell Culture , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA Interference , RNA, Small Interfering , Random Allocation , Stem Cells/cytology , Transfection , Wound Healing/physiology , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
8.
Circ Res ; 120(5): 799-815, 2017 Mar 03.
Article in English | MEDLINE | ID: mdl-28115390

ABSTRACT

RATIONALE: Abdominal aortic aneurysms (AAAs) are characterized by pathological remodeling of the aortic wall. Although both increased Krüppel-like factor 5 (KLF5) expression and macrophage infiltration have been implicated in vascular remodeling, the role of KLF5 in macrophage infiltration and AAA formation remains unclear. OBJECTIVE: To determine the role of KLF5 in AAA formation and macrophage infiltration into AAAs. METHODS AND RESULTS: KLF5 expression was significantly increased in human AAA tissues and in 2 mouse models of experimental AAA. Moreover, in myeloid-specific Klf5 knockout mice (myeKlf5-/- mice), macrophage infiltration, medial smooth muscle cell loss, elastin degradation, and AAA formation were markedly decreased. In cell migration and time-lapse imaging analyses, the migration of murine myeKlf5-/- macrophages was impaired, and in luciferase reporter assays, KLF5 activated Myo9b (myosin IXB) transcription by direct binding to the Myo9b promoter. In subsequent coimmunostaining studies, Myo9b was colocalized with filamentous actin, cortactin, vinculin, and Tks5 in the podosomes of phorbol 12,13-dibutyrate-treated macrophages, indicating that Myo9b participates in podosome formation. Gain- and loss-of-function experiments showed that KLF5 promoted podosome formation in macrophages by upregulating Myo9b expression. Furthermore, RhoA-GTP levels increased after KLF5 knockdown in macrophages, suggesting that KLF5 lies upstream of RhoA signaling. Finally, Myo9b expression was increased in human AAA tissues, located in macrophages, and positively correlated with AAA size. CONCLUSIONS: These data are the first to indicate that KLF5-dependent regulation of Myo9b/RhoA is required for podosome formation and macrophage migration during AAA formation, warranting consideration of the KLF5-Myo9b-RhoA pathway as a therapeutic target for AAA treatment.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/metabolism , Myosins/biosynthesis , Podosomes/metabolism , rhoA GTP-Binding Protein/biosynthesis , Animals , Cell Line , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/deficiency , Male , Mice , Mice, Knockout , Myosins/deficiency , Signal Transduction/physiology , rhoA GTP-Binding Protein/deficiency
9.
BMC Nephrol ; 17(1): 66, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27389190

ABSTRACT

BACKGROUND: Podocyte apoptosis is a major mechanism that leads to proteinuria in many kidney diseases. However, the concert mechanisms that cause podocyte apoptosis in these kidney diseases are not fully understood. RhoA is one of Rho GTPases that has been well studied and plays a key role in regulating cytoskeletal architecture. Previous study showed that insufficient RhoA could result in rat aortic smooth muscle cell apoptosis. However, whether RhoA is involved in podocyte apoptosis remains unknown. METHODS: Culture podocytes were treated with LPS, ADR or siRNA for 48 h before harvest. Subcellular immunoblotting, qRT-PCR, immunofluorescence and flow cytometry were used to exam the expression and function of RhoA or YAP in podocytes. RESULTS: We found that the expression of RhoA and its activity were significantly decreased in LPS or ADR-injured podocytes, accompanying loss of stress fibers and increased cell apoptosis. Knocking down RhoA or its downstream effector mDia expression by siRNA also caused loss of stress fibers and podocyte apoptosis. Moreover, our results further demonstrated that RhoA deficiency could reduce the mRNA and protein expression of YAP, which had been regarded as an anti-apoptosis protein in podocyte. Silenced dendrin expression significantly abolished RhoA, mDia or YAP deficiency-induced podocyte apoptosis. CONCLUSION: RhoA deficiency could disrupt podocyte cytoskeleton and induce podocyte apoptosis by inhibiting YAP/dendrin signal. RhoA/mDia/YAP/dendrin signal pathway may potentially play an important role in regulating podocyte apoptosis. Maintaining necessary RhoA would be one potent way to prevent proteinuria kidney diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Podocytes/physiology , Podocytes/ultrastructure , rhoA GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Doxorubicin/pharmacology , Formins , Gene Silencing , Lipopolysaccharides/pharmacology , Mice , Nerve Tissue Proteins/genetics , Phosphoproteins/genetics , Podocytes/drug effects , RNA, Messenger/metabolism , Signal Transduction , Stress Fibers/drug effects , Stress Fibers/ultrastructure , YAP-Signaling Proteins , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
10.
J Allergy Clin Immunol ; 137(1): 231-245.e4, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26100081

ABSTRACT

BACKGROUND: Mitochondrial metabolism is known to be important for T-cell activation. However, its involvement in effector T-cell differentiation has just begun to gain attention. Importantly, how metabolic pathways are integrated with T-cell activation and effector cell differentiation and function remains largely unknown. OBJECTIVE: We sought to test our hypothesis that RhoA GTPase orchestrates glycolysis for TH2 cell differentiation and TH2-mediated allergic airway inflammation. METHODS: Conditional RhoA-deficient mice were generated by crossing RhoA(flox/flox) mice with CD2-Cre transgenic mice. Effects of RhoA on TH2 differentiation were evaluated based on in vitro TH2-polarized culture conditions and in vivo in ovalbumin-induced allergic airway inflammation. Cytokine levels were measured by using intracellular staining and ELISA. T-cell metabolism was measured by using the Seahorse XF24 Analyzer and flow cytometry. RESULTS: Disruption of RhoA inhibited T-cell activation and TH2 differentiation in vitro and prevented the development of allergic airway inflammation in vivo, with no effect on TH1 cells. RhoA deficiency in activated T cells led to multiple defects in metabolic pathways, such as glycolysis and oxidative phosphorylation. Importantly, RhoA couples glycolysis to TH2 cell differentiation and allergic airway inflammation through regulating IL-4 receptor mRNA expression and TH2-specific signaling events. Finally, inhibition of Rho-associated protein kinase, an immediate downstream effector of RhoA, blocked TH2 differentiation and allergic airway inflammation. CONCLUSION: RhoA is a key component of the signaling cascades leading to TH2 differentiation and allergic airway inflammation at least in part through control of T-cell metabolism and the Rho-associated protein kinase pathway.


Subject(s)
Glycolysis , Respiratory Hypersensitivity/metabolism , Th2 Cells/metabolism , rhoA GTP-Binding Protein/metabolism , Allergens/immunology , Animals , Cell Differentiation , Inflammation/immunology , Inflammation/metabolism , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology , Th2 Cells/cytology , Th2 Cells/immunology , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/immunology
11.
PLoS One ; 10(6): e0127923, 2015.
Article in English | MEDLINE | ID: mdl-26030593

ABSTRACT

Numerous cellular studies have indicated that RhoA signaling is required for oncogenic Ras-induced transformation, suggesting that RhoA is a useful target in Ras induced neoplasia. However, to date very limited data exist to genetically attribute RhoA function to Ras-mediated tumorigenesis in mammalian models. In order to assess whether RhoA is required for K-Ras-induced lung cancer initiation, we utilized the K-RasG12D Lox-Stop-Lox murine lung cancer model in combination with a conditional RhoAflox/flox and RhoC-/- knockout mouse models. Deletion of the floxed Rhoa gene and expression of K-RasG12D was achieved by either CCSP-Cre or adenoviral Cre, resulting in simultaneous expression of K-RasG12D and deletion of RhoA from the murine lung. We found that deletion of RhoA, RhoC or both did not adversely affect normal lung development. Moreover, we found that deletion of either RhoA or RhoC alone did not suppress K-RasG12D induced lung adenoma initiation. Rather, deletion of RhoA alone exacerbated lung adenoma formation, whereas dual deletion of RhoA and RhoC together significantly reduced K-RasG12D induced adenoma formation. Deletion of RhoA appears to induce a compensatory mechanism that exacerbates adenoma formation. The compensatory mechanism is at least partly mediated by RhoC. This study suggests that targeting of RhoA alone may allow for compensation and a paradoxical exacerbation of neoplasia, while simultaneous targeting of both RhoA and RhoC is likely to lead to more favorable outcomes.


Subject(s)
Adenoma/genetics , Adenoma/pathology , Genes, ras/physiology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , rhoA GTP-Binding Protein/genetics , Animals , Blotting, Western , Genes, ras/genetics , Immunohistochemistry , Mice , ras Proteins/genetics , ras Proteins/metabolism , rhoA GTP-Binding Protein/deficiency , rhoC GTP-Binding Protein
12.
Nat Commun ; 6: 6725, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25857352

ABSTRACT

Histamine-induced vascular leakage is an integral component of many highly prevalent human diseases, including allergies, asthma and anaphylaxis. Yet, how histamine induces the disruption of the endothelial barrier is not well defined. By using genetically modified animal models, pharmacologic inhibitors and a synthetic biology approach, here we show that the small GTPase RhoA mediates histamine-induced vascular leakage. Histamine causes the rapid formation of focal adherens junctions, disrupting the endothelial barrier by acting on H1R Gαq-coupled receptors, which is blunted in endothelial Gαq/11 KO mice. Interfering with RhoA and ROCK function abolishes endothelial permeability, while phospholipase Cß plays a limited role. Moreover, endothelial-specific RhoA gene deletion prevents vascular leakage and passive cutaneous anaphylaxis in vivo, and ROCK inhibitors protect from lethal systemic anaphylaxis. This study supports a key role for the RhoA signalling circuitry in vascular permeability, thereby identifying novel pharmacological targets for many human diseases characterized by aberrant vascular leakage.


Subject(s)
Anaphylaxis/genetics , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Histamine/pharmacology , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Adherens Junctions/pathology , Amides/pharmacology , Anaphylaxis/chemically induced , Anaphylaxis/metabolism , Anaphylaxis/pathology , Animals , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression Regulation , Humans , Male , Mice , Mice, Knockout , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Signal Transduction , Skin/drug effects , Skin/metabolism , Skin/pathology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/deficiency
13.
J Immunol ; 193(12): 5973-82, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25398325

ABSTRACT

Thymocyte development is regulated by complex signaling pathways. How these signaling cascades are coordinated remains elusive. RhoA of the Rho family small GTPases plays an important role in actin cytoskeleton organization, cell adhesion, migration, proliferation, and survival. Nonetheless, the physiological function of RhoA in thymocyte development is not clear. By characterizing a conditional gene targeting mouse model bearing T cell deletion of RhoA, we show that RhoA critically regulates thymocyte development by coordinating multiple developmental events. RhoA gene disruption caused a strong developmental block at the pre-TCR checkpoint and during positive selection. Ablation of RhoA led to reduced DNA synthesis in CD4(-)CD8(-), CD4(+)CD8(-), and CD4(-)CD8(+) thymocytes but not in CD4(+)CD8(+) thymocytes. Instead, RhoA-deficient CD4(+)CD8(+) thymocytes showed an impaired mitosis. Furthermore, we found that abrogation of RhoA led to an increased apoptosis in all thymocyte subpopulations. Importantly, we show that the increased apoptosis was resulted from reduced pre-TCR expression and increased production of reactive oxygen species (ROS), which may be because of an enhanced mitochondrial function, as manifested by increased oxidative phosphorylation, glycolysis, mitochondrial membrane potential, and mitochondrial biogenesis in RhoA-deficient thymocytes. Restoration of pre-TCR expression or treatment of RhoA-deficient mice with a ROS scavenger N-acetylcysteine partially restored thymocyte development. These results suggest that RhoA is required for thymocyte development and indicate, to our knowledge, for the first time that fine-tuning of ROS production by RhoA, through a delicate control of metabolic circuit, may contribute to thymopoiesis.


Subject(s)
Gene Targeting , Mitochondria/genetics , Mitochondria/metabolism , Thymocytes/cytology , Thymocytes/metabolism , rhoA GTP-Binding Protein/genetics , Animals , Antigens, Surface , Apoptosis/genetics , Apoptosis/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Cell Survival/genetics , Gene Expression Profiling , Immunophenotyping , Mice , Mice, Knockout , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , V(D)J Recombination , rhoA GTP-Binding Protein/deficiency
14.
J Exp Med ; 210(11): 2371-85, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24101377

ABSTRACT

Hematopoietic progenitor cells (HPCs) are central to hematopoiesis as they provide large numbers of lineage-defined blood cells necessary to sustain blood homeostasis. They are one of the most actively cycling somatic cells, and their precise control is critical for hematopoietic homeostasis. The small GTPase RhoA is an intracellular molecular switch that integrates cytokine, chemokine, and adhesion signals to coordinate multiple context-dependent cellular processes. By using a RhoA conditional knockout mouse model, we show that RhoA deficiency causes a multilineage hematopoietic failure that is associated with defective multipotent HPCs. Interestingly, RhoA(-/-) hematopoietic stem cells retained long-term engraftment potential but failed to produce multipotent HPCs and lineage-defined blood cells. This multilineage hematopoietic failure was rescued by reconstituting wild-type RhoA into the RhoA(-/-) Lin(-)Sca-1(+)c-Kit(+) compartment. Mechanistically, RhoA regulates actomyosin signaling, cytokinesis, and programmed necrosis of the HPCs, and loss of RhoA results in a cytokinesis failure of HPCs manifested by an accumulation of multinucleated cells caused by failed abscission of the cleavage furrow after telophase. Concomitantly, the HPCs show a drastically increased death associated with increased TNF-RIP-mediated necrosis. These results show that RhoA is a critical and specific regulator of multipotent HPCs during cytokinesis and thus essential for multilineage hematopoiesis.


Subject(s)
Cytokinesis , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/pathology , rhoA GTP-Binding Protein/metabolism , Actomyosin/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Lineage/drug effects , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Cytokinesis/drug effects , Formins , HEK293 Cells , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/ultrastructure , Homeostasis/drug effects , Humans , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Necrosis , Protein Binding/drug effects , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/deficiency
15.
PLoS One ; 8(7): e69315, 2013.
Article in English | MEDLINE | ID: mdl-23935982

ABSTRACT

RhoA plays a multifaceted role in platelet biology. During platelet development, RhoA has been proposed to regulate endomitosis, proplatelet formation, and platelet release, in addition to having a role in platelet activation. These processes were previously studied using pharmacological inhibitors in vitro, which have potential drawbacks, such as non-specific inhibition or incomplete disruption of the intended target proteins. Therefore, we developed a conditional knockout mouse model utilizing the CRE-LOX strategy to ablate RhoA, specifically in megakaryocytes and in platelets to determine its role in platelet development. We demonstrated that deleting RhoA in megakaryocytes in vivo resulted in significant macrothrombocytopenia. RhoA-null megakaryocytes were larger, had higher mean ploidy, and exhibited stiff membranes with micropipette aspiration. However, in contrast to the results observed in experiments relying upon pharmacologic inhibitors, we did not observe any defects in proplatelet formation in megakaryocytes lacking RhoA. Infused RhoA-null megakaryocytes rapidly released platelets, but platelet levels rapidly plummeted within several hours. Our evidence supports the hypothesis that changes in membrane rheology caused infused RhoA-null megakaryocytes to prematurely release aberrant platelets that were unstable. These platelets were cleared quickly from circulation, which led to the macrothrombocytopenia. These observations demonstrate that RhoA is critical for maintaining normal megakaryocyte development and the production of normal platelets.


Subject(s)
Blood Platelets/enzymology , Megakaryocytes/enzymology , Ploidies , Thrombopoiesis , rhoA GTP-Binding Protein/metabolism , Animals , Cell Membrane/metabolism , Cell Size , Cytoskeleton/metabolism , Gene Deletion , Gene Targeting , Mice , Mutation/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Rheology , Thrombocytopenia/pathology , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
16.
PLoS One ; 8(6): e67015, 2013.
Article in English | MEDLINE | ID: mdl-23825607

ABSTRACT

The assembly of neuronal circuits during development requires the precise navigation of axons, which is controlled by attractive and repulsive guidance cues. In the developing spinal cord, ephrinB3 functions as a short-range repulsive cue that prevents EphA4 receptor-expressing corticospinal tract and spinal interneuron axons from crossing the midline, ensuring proper formation of locomotor circuits. Here we report that the small GTPase RhoA, a key regulator of cytoskeletal dynamics, is also required for ephrinB3/EphA4-dependent locomotor circuit formation. Deletion of RhoA from neural progenitor cells results in mice that exhibit a rabbit-like hopping gait, which phenocopies mice lacking ephrinB3 or EphA4. Consistent with this locomotor defect, we found that corticospinal tract axons and spinal interneuron projections from RhoA-deficient mice aberrantly cross the spinal cord midline. Furthermore, we determined that loss of RhoA blocks ephrinB3-induced growth cone collapse of cortical axons and disrupts ephrinB3 expression at the spinal cord midline. Collectively, our results demonstrate that RhoA is essential for the ephrinB3/EphA4-dependent assembly of cortical and spinal motor circuits that control normal locomotor behavior.


Subject(s)
Locomotion , Nerve Net/enzymology , Nerve Net/physiology , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Brain/cytology , Ephrin-B3/metabolism , Gene Knockout Techniques , Growth Cones/metabolism , Mice , Molecular Sequence Data , Nerve Net/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptor, EphA4/metabolism , Spinal Cord/cytology , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics
17.
Mol Biol Cell ; 23(20): 4008-19, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22918940

ABSTRACT

Rho GTPases share a common inhibitor, Rho guanine nucleotide dissociation inhibitor (RhoGDI), which regulates their expression levels, membrane localization, and activation state. The selective dissociation of individual Rho GTPases from RhoGDI ensures appropriate responses to cellular signals, but the underlying mechanisms are unclear. Diacylglycerol kinase ζ (DGKζ), which phosphorylates diacylglycerol to yield phosphatidic acid, selectively dissociates Rac1 by stimulating PAK1-mediated phosphorylation of RhoGDI on Ser-101/174. Similarly, phosphorylation of RhoGDI on Ser-34 by protein kinase Cα (PKCα) selectively releases RhoA. Here we show DGKζ is required for RhoA activation and Ser-34 phosphorylation, which were decreased in DGKζ-deficient fibroblasts and rescued by wild-type DGKζ or a catalytically inactive mutant. DGKζ bound directly to the C-terminus of RhoA and the regulatory arm of RhoGDI and was required for efficient interaction of PKCα and RhoA. DGKζ-null fibroblasts had condensed F-actin bundles and altered focal adhesion distribution, indicative of aberrant RhoA signaling. Two targets of the RhoA effector ROCK showed reduced phosphorylation in DGKζ-null cells. Collectively our findings suggest DGKζ functions as a scaffold to assemble a signaling complex that functions as a RhoA-selective, GDI dissociation factor. As a regulator of Rac1 and RhoA activity, DGKζ is a critical factor linking changes in lipid signaling to actin reorganization.


Subject(s)
Diacylglycerol Kinase/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Biocatalysis , Diacylglycerol Kinase/chemistry , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/enzymology , Focal Adhesions/metabolism , Mice , Models, Biological , Multiprotein Complexes/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Kinase C-alpha/metabolism , Protein Stability , Protein Structure, Tertiary , Signal Transduction , Stress Fibers/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/chemistry , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rhoA GTP-Binding Protein/deficiency
18.
Neuron ; 73(5): 911-24, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22405202

ABSTRACT

The positioning of neurons in the cerebral cortex is of crucial importance for its function as highlighted by the severe consequences of migrational disorders in patients. Here we show that genetic deletion of the small GTPase RhoA in the developing cerebral cortex results in two migrational disorders: subcortical band heterotopia (SBH), a heterotopic cortex underlying the normotopic cortex, and cobblestone lissencephaly, in which neurons protrude beyond layer I at the pial surface of the brain. Surprisingly, RhoA(-/-) neurons migrated normally when transplanted into wild-type cerebral cortex, whereas the converse was not the case. Alterations in the radial glia scaffold are demonstrated to cause these migrational defects through destabilization of both the actin and the microtubules cytoskeleton. These data not only demonstrate that RhoA is largely dispensable for migration in neurons but also showed that defects in radial glial cells, rather than neurons, can be sufficient to produce SBH.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/physiology , Neuroglia/metabolism , Neurons/physiology , rhoA GTP-Binding Protein/deficiency , Age Factors , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cell Movement , Cell Proliferation , Cerebral Cortex/embryology , Cerebral Cortex/transplantation , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , Classical Lissencephalies and Subcortical Band Heterotopias/metabolism , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , Disease Models, Animal , Electroporation , Embryo, Mammalian , Embryonic Stem Cells/physiology , Embryonic Stem Cells/transplantation , Female , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Neurons/metabolism , Neurons/ultrastructure , Pregnancy , Silver Staining , rhoA GTP-Binding Protein/genetics
19.
Proc Natl Acad Sci U S A ; 108(18): 7607-12, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502507

ABSTRACT

The organization of neural progenitors in the developing mammalian neuroepithelium is marked by cadherin-based adherens junctions. Whereas RhoA, a founding member of the small Rho GTPase family, has been shown to play important roles in epithelial adherens junctions, its physiological roles in neural development remain uncertain due to the lack of specific loss-of-function studies. Here, we show that RhoA protein accumulates at adherens junctions in the developing mouse brain and colocalizes to the cadherin-catenin complex. Conditional deletion of RhoA in midbrain and forebrain neural progenitors using Wnt1-Cre and Foxg1-Cre mice, respectively, disrupts apical adherens junctions and causes massive dysplasia of the brain. Furthermore, RhoA-deficient neural progenitor cells exhibit accelerated proliferation, reduction of cell- cycle exit, and increased expression of downstream target genes of the hedgehog pathway. Consequently, both lines of conditional RhoA-deficient embryos exhibit expansion of neural progenitor cells and exencephaly-like protrusions. These results demonstrate a critical role of RhoA in the maintenance of apical adherens junctions and the regulation of neural progenitor proliferation in the developing mammalian brain.


Subject(s)
Adherens Junctions/metabolism , Brain/embryology , Cell Proliferation , Neural Stem Cells/metabolism , rhoA GTP-Binding Protein/deficiency , Animals , Bromodeoxyuridine , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , In Situ Nick-End Labeling , Indoles , Mice , Mice, Mutant Strains , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , rhoA GTP-Binding Protein/metabolism
20.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L984-91, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19717551

ABSTRACT

Recent studies revealed an involvement of RhoA/Rho-kinase in the contraction of bronchial smooth muscle (BSM), and this pathway has now been proposed as a new target for asthma therapy. A posttranslational geranylgeranylation of RhoA is required for its activation. Thus selective inhibition of geranylgeranyltransferase may be a novel strategy for treatment of the BSM hyperresponsiveness in asthmatics. To test this hypothesis, we investigated the effect of a geranylgeranyltransferase inhibitor, GGTI-2133, on antigen-induced BSM hyperresponsiveness by using mice with experimental asthma. Mice were sensitized and repeatedly challenged with ovalbumin antigen. Animals also were treated with GGTI-2133 (5 mg/kg ip) once a day before and during the antigen inhalation period. Repeated antigen inhalation caused a BSM hyperresponsiveness to acetylcholine with the increased expressions of RhoA and the anti-farnesyl-positive 21-kDa proteins, probably geranylgeranylated RhoA. The in vivo GGTI-2133 treatments significantly inhibited BSM hyperresponsiveness induced by antigen exposure. In another series of experiments, BSM tissues isolated from the repeatedly antigen-challenged mice were cultured for 48 h in the absence or presence of GGTI-2133. Under these conditions, the putative geranylgeranylated RhoA was decreased in a GGTI-2133 concentration-dependent manner. The in vitro incubation with GGTI-2133 also inhibited BSM hyperresponsiveness induced by antigen exposure. These findings suggest that GGTI-2133 inhibits antigen-induced BSM hyperresponsiveness, probably by reducing downstream signal transduction of RhoA. Selective geranylgeranyltransferase inhibitors may be beneficial for the treatment of airway hyperresponsiveness, one of the characteristic features of allergic bronchial asthma.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Bronchial Hyperreactivity/enzymology , Bronchial Hyperreactivity/prevention & control , Enzyme Inhibitors/pharmacology , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Alkyl and Aryl Transferases/metabolism , Amides/pharmacology , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Male , Mice , Mice, Inbred BALB C , Molecular Weight , Muscle Relaxation/drug effects , Muscle, Smooth/pathology , Prenylation/drug effects , Pyridines/pharmacology , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL