Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
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.
Front Immunol ; 12: 643260, 2021.
Article in English | MEDLINE | ID: mdl-33936062

ABSTRACT

We have previously demonstrated that benzo(a)pyrene (BaP) co-exposure with dermatophagoides group 1 allergen (Der f 1) can potentiate Der f 1-induced airway inflammation. The underlying mechanism, however, remains undetermined. Here we investigated the molecular mechanisms underlying the potentiation of BaP exposure on Der f 1-induced airway inflammation in asthma. We found that BaP co-exposure potentiated Der f 1-induced TGFß1 secretion and signaling activation in human bronchial epithelial cells (HBECs) and the airways of asthma mouse model. Moreover, BaP exposure alone or co-exposure with Der f 1-induced aryl hydrocarbon receptor (AhR) activity was determined by using an AhR-dioxin-responsive element reporter plasmid. The BaP and Der f 1 co-exposure-induced TGFß1 expression and signaling activation were attenuated by either AhR antagonist CH223191 or AhR knockdown in HBECs. Furthermore, AhR knockdown led to the reduction of BaP and Der f 1 co-exposure-induced active RhoA. Inhibition of RhoA signaling with fasudil, a RhoA/ROCK inhibitor, suppressed BaP and Der f 1 co-exposure-induced TGFß1 expression and signaling activation. This was further confirmed in HBECs expressing constitutively active RhoA (RhoA-L63) or dominant-negative RhoA (RhoA-N19). Luciferase reporter assays showed prominently increased promoter activities for the AhR binding sites in the promoter region of RhoA. Inhibition of RhoA suppressed BaP and Der f 1 co-exposure-induced airway hyper-responsiveness, Th2-associated airway inflammation, and TGFß1 signaling activation in asthma. Our studies reveal a previously unidentified functional axis of AhR-RhoA in regulating TGFß1 expression and signaling activation, representing a potential therapeutic target for allergic asthma.


Subject(s)
Antigens, Dermatophagoides/toxicity , Arthropod Proteins/toxicity , Asthma , Basic Helix-Loop-Helix Transcription Factors/immunology , Benzo(a)pyrene/toxicity , Cysteine Endopeptidases/toxicity , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , Transforming Growth Factor beta1/immunology , rhoA GTP-Binding Protein/immunology , Animals , Asthma/chemically induced , Asthma/immunology , Asthma/pathology , Female , Male , Mice , Signal Transduction/drug effects , Signal Transduction/immunology
3.
mBio ; 12(2)2021 04 06.
Article in English | MEDLINE | ID: mdl-33824207

ABSTRACT

Human cytomegalovirus (HCMV) microRNAs play essential roles in latency and reactivation in CD34+ hematopoietic progenitor cells (HPCs) via regulation of viral and cellular gene expression. In the present study, we show that HCMV miR-US25-1 targets RhoA, a small GTPase required for CD34+ HPC self-renewal, proliferation, and hematopoiesis. Expression of miR-US25-1 impairs signaling through the nonmuscle myosin II light chain, which leads to a block in cytokinesis and an inhibition of proliferation. Moreover, infection with an HCMV mutant lacking miR-US25-1 resulted in increased proliferation of CD34+ HPCs and a decrease in the proportion of genome-containing cells at the end of latency culture. These observations provide a mechanism by which HCMV limits proliferation to maintain latent viral genomes in CD34+ HPCs.IMPORTANCE Each herpesvirus family establishes latency in a unique cell type. Since herpesvirus genomes are maintained as episomes, the virus needs to devise mechanisms to retain the latent genome during cell division. Alphaherpesviruses overcome this obstacle by infecting nondividing neurons, while gammaherpesviruses tether their genome to the host chromosome in dividing B cells. The betaherpesvirus human cytomegalovirus (HCMV) establishes latency in CD34+ hematopoietic progenitor cells (HPCs), but the mechanism used to maintain the viral genome is unknown. In this report, we demonstrate that HCMV miR-US25-1 downregulates expression of RhoA, a key cell cycle regulator, which results in inhibition of CD34+ HPC proliferation by blocking mitosis. Mutation of miR-US25-1 during viral infection results in enhanced cellular proliferation and a decreased frequency of genome-containing CD34+ HPCs. These results reveal a novel mechanism through which HCMV is able to regulate cell division to prevent viral genome loss during proliferation.


Subject(s)
Antigens, CD34/genetics , Cell Proliferation/genetics , Cytomegalovirus/genetics , Genome, Viral , Hematopoietic Stem Cells/physiology , Host-Pathogen Interactions , MicroRNAs/genetics , Virus Latency/genetics , rhoA GTP-Binding Protein/genetics , Antigens, CD34/immunology , Antigens, CD34/metabolism , Cytomegalovirus/pathogenicity , Down-Regulation , Gene Expression Regulation , HEK293 Cells , Humans , MicroRNAs/metabolism , Mitosis/genetics , Signal Transduction/genetics , rhoA GTP-Binding Protein/immunology
4.
J Biol Chem ; 296: 100261, 2021.
Article in English | MEDLINE | ID: mdl-33837725

ABSTRACT

GPR56 is a member of the adhesion G-protein-coupled receptor family shown to play important roles in cell adhesion, brain development, immune function, and tumorigenesis. GPR56 is highly upregulated in colorectal cancer and correlates with poor prognosis. Several studies have shown GPR56 couples to the Gα12/13 class of heterotrimeric G-proteins to promote RhoA activation. However, due to its structural complexity and lack of a high-affinity receptor-specific ligand, the complete GPR56 signaling mechanism remains largely unknown. To delineate the activation mechanism and intracellular signaling functions of GPR56, we generated a monoclonal antibody (mAb) that binds with high affinity and specificity to the extracellular domain (ECD). Using deletion mutants, we mapped the mAb binding site to the GAIN domain, which mediates membrane-proximal autoproteolytic cleavage of the ECD. We showed that GPR56 overexpression in 293T cells leads to increased phosphorylation of Src, Fak, and paxillin adhesion proteins and activation of the Gα12/13-RhoA-mediated serum response factor (SRF) pathway. Treatment with the mAb potentiated Src-Fak phosphorylation, RhoA-SRF signaling, and cell adhesion. Consistently, GPR56 knockdown in colorectal cancer cells decreased Src-Fak pathway phosphorylation and cell adhesion. Interestingly, GPR56-mediated activation of Src-Fak phosphorylation occurred independent of RhoA, yet mAb-induced potentiation of RhoA-SRF signaling was Src-dependent. Furthermore, we show that the C-terminal portion of the Serine-Threonine-Proline-rich (STP) region, adjacent to the GAIN domain, was required for Src-Fak activation. However, autoproteolytic cleavage of the ECD was dispensable. These data support a new ECD-dependent mechanism by which GPR56 functions to regulate adhesion through activation of Src-Fak signaling.


Subject(s)
Colorectal Neoplasms/genetics , Focal Adhesion Kinase 1/genetics , Receptors, G-Protein-Coupled/genetics , Serum Response Factor/genetics , rhoA GTP-Binding Protein/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinogenesis/genetics , Cell Adhesion/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Focal Adhesion Kinase 1/immunology , Gene Expression Regulation, Neoplastic/genetics , Humans , Paxillin/genetics , Paxillin/immunology , Phosphorylation/genetics , Receptors, G-Protein-Coupled/immunology , Serum Response Factor/immunology , Signal Transduction/genetics , rhoA GTP-Binding Protein/immunology , src-Family Kinases/genetics , src-Family Kinases/immunology
5.
Int Immunopharmacol ; 94: 107489, 2021 May.
Article in English | MEDLINE | ID: mdl-33774357

ABSTRACT

The objective of this study is to investigate the role of IL-38 in osteoarthritis (OA). IL-38 levels in serum and synovial fluid (SF) of patients with OA were examined to identify the correlation between IL-38 expression and OA activity and to determine its anti-inflammatory effects in IL-1ß-induced chondrocytes. A total of 75 patients with OA who underwent joint replacement surgery and 25 age- and sex-matched healthy volunteers were recruited. The levels of IL-38 in serum and SF are shown to be significant elevated in OA patients compared with that of healthy controls. Serum and SF IL-38 levels of OA patients are positively correlated with Kellgren-Lawrence (K-L) grades 2 to 3, as well as with pro-inflammatory cytokines IL-6, IL-23, and TNF-α, but are negatively correlated with the anti-inflammatory cytokine IL-10 in K-L grades 3 to 4. Furthermore, overexpression of IL-38 in vitro is shown to attenuate the expression of pro-inflammatory cytokines such as COX-2, IL-6, IL-8, IL-36Ra, IL-36α/ß/γ, iNOS, and TNF-α, as well as matrix degrading enzymes such as MMP3, MMP13, and ADAMTS5, and apoptosis-related indicators Bax/Bcl-2, cleaved caspase 3/pro-caspase 3, and cleaved caspase 9/pro-caspase 9. IL-38 overexpression also reduces expression of the signaling proteins p-p38, p-p65, p-JNK, and RhoA significantly. Taken together, our results show that expression of IL-38 is increased in OA tissues and OA rat chondrocytes, and is positively correlated with early disease activity. This increased IL-38 expression lead to the inactivation of MAPK, NF-κB, JNK, and RhoA signaling pathways, which might have impletion on OA chondrocytes apoptosis, degradation and inflammatory effect. Thus, IL-38 probably serves as a novel therapeutic target for the treatment of OA.


Subject(s)
Chondrocytes/immunology , Cytokines/immunology , Osteoarthritis/immunology , Aged , Animals , Cartilage, Articular/cytology , Cytokines/blood , Cytokines/genetics , Female , Hip Joint , Humans , Knee Joint , Male , Middle Aged , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Osteoarthritis/blood , Rats, Sprague-Dawley , Signal Transduction , rhoA GTP-Binding Protein/immunology
6.
Aging (Albany NY) ; 12(24): 24853-24871, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33361519

ABSTRACT

Duchenne Muscular Dystrophy (DMD) patients often suffer from both muscle wasting and osteoporosis. Our previous studies have revealed reduced regeneration potential in skeletal muscle and bone, concomitant with ectopic calcification of soft tissues in double knockout (dKO, dystrophin-/-; utrophin-/-) mice, a severe murine model for DMD. We found significant involvement of RhoA/ROCK (Rho-Associated Protein Kinase) signaling in mediating ectopic calcification of muscles in dKO mice. However, the cellular identity of these RhoA+ cells, and the role that RhoA plays in the chronic inflammation-associated pathologies has not been elucidated. Here, we report that CD68+ macrophages are highly prevalent at the sites of ectopic calcification of dKO mice, and that these macrophages highly express RhoA. Macrophages from dKO mice feature a shift towards a more pro-inflammatory M1 polarization and an increased expression of various senescence-associated secretory phenotype (SASP) factors that was reduced with the RhoA/ROCK inhibitor Y-27632. Further, systemic inhibition of RhoA activity in dKO mice led to reduced number of RhoA+/CD68+ cells, as well as a reduction in fibrosis and ectopic calcification. Together, these data revealed that RhoA signaling may be a key regulator of imbalanced mineralization in the dystrophic musculoskeletal system and consequently a therapeutic target for the treatment of DMD or other related muscle dystrophies.


Subject(s)
Calcinosis/metabolism , Macrophages/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Myocardium/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Calcinosis/immunology , Calcinosis/pathology , Cellular Senescence/genetics , Cellular Senescence/immunology , Disease Models, Animal , Dystrophin/genetics , Macrophages/immunology , Mice , Mice, Knockout , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/immunology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/immunology , Myocardium/pathology , Utrophin/genetics , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/immunology
7.
Cell Signal ; 70: 109599, 2020 06.
Article in English | MEDLINE | ID: mdl-32165237

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen which periodically reactivates, causing severe clinical consequences in immunosuppressed patients, organ and stem cell transplant recipients or newborn babies with congenital infections. HCMV infection stimulates the expression of several proinflammatory cytokines, which may contribute to the pathogenesis of the infection. Rho GTPases mediate cytokine expression while increasing evidence implicates them in important aspects of HCMV life cycle. Here, we studied the role of RhoA on the interleukin 11 (IL-11) release in HCMV-infected fibroblasts. Human fibroblasts, either endogenously expressing or silenced for RhoA, were infected by HCMV or UV-inactivated virus and IL-11 transcription and secretion were evaluated. We found that HCMV lytic infection increased the IL-11 levels, both in terms of transcription and translation. Both infectious and non-infectious HCMV particles were able to induce the IL-11 production. The depletion of RhoA resulted in an even higher release of IL-11, revealing the implication of this specific Rho isoform in this biological event. Finally, infection of cells in the presence of the HCMV DNA replication inhibitor, ganciclovir, significantly reduced the secretion of IL-11, strongly associating its induction with active viral DNA replication. Collectively, these data demonstrate, for the first time, a novel role of RhoA GTPase during HCMV lytic infection, regulating the activation of an immune response through IL-11.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus , Fibroblasts/metabolism , Interleukin-11/immunology , rhoA GTP-Binding Protein/immunology , Cytomegalovirus/immunology , Cytomegalovirus/physiology , Fibroblasts/pathology , HEK293 Cells , Humans
8.
Br J Haematol ; 189(1): 54-66, 2020 04.
Article in English | MEDLINE | ID: mdl-32064593

ABSTRACT

Peripheral T-cell lymphomas (PTCL) comprise a heterogeneous group of aggressive lymphoproliferative disorders almost all of which are associated with poor clinical outcomes. Angioimmunoblastic T-cell lymphoma (AITL) and some peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) have similarities to normal CD4+ T-cell subsets in their gene expression profiles. A cell of origin model is, therefore, emerging and is likely to be refined in the future. Follicular helper (Tfh) T cells are now established as the cell of origin of AITL and about 20% of PTCL-NOS. Sequencing studies have identified recurrent genetic alterations in epigenetic modifiers, T-cell receptor signalling pathway intermediates or RHOA, most commonly a specific mutation leading to RHOA G17V. While PTCL-NOS remains a diagnosis of exclusion, advances in genomics have identified subgroups expressing transcription factors TBX 21 (Th1-like origin) and GATA3 (Th2-like origin). These findings suggest new biomarkers and new therapeutic avenues including the hypomethylating agent azacytidine, or inhibitors of proximal T-cell receptor (TCR) signalling and potentially certain monoclonal antibodies. The advances over the past few years, therefore, prompt stratified medicine approaches to test biologically based treatments and determine the clinical utility of the new disease classifications.


Subject(s)
Epigenesis, Genetic/immunology , Gene Expression Regulation, Neoplastic/immunology , Lymphoma, T-Cell, Peripheral , Mutation, Missense , Neoplasm Proteins , T-Lymphocytes, Helper-Inducer/immunology , rhoA GTP-Binding Protein , Humans , Lymphoma, T-Cell, Peripheral/classification , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/immunology , Lymphoma, T-Cell, Peripheral/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/genetics , Signal Transduction/immunology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/immunology
9.
Acta Biochim Biophys Sin (Shanghai) ; 51(10): 1026-1033, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31553428

ABSTRACT

Ibuprofen, a medication in the nonsteroidal anti-inflammatory drug class, is widely used for treating inflammatory diseases such as osteoarthritis. It has been shown in recent years that ibuprofen has a strong effect on Ras homolog gene family, member A (RhoA) inhibition in multiple cell types. Our previous finding also demonstrated that interleukin-1ß (IL-1ß) increases filamentous actin (F-actin) of chondrocytes via RhoA pathway. Therefore, we hypothesized that ibuprofen may suppress the IL-1ß-induced F-actin upregulation in chondrocytes by inhibiting RhoA pathway. To this end, in this study, articular chondrocytes from New Zealand White rabbits were pretreated with 500 µM ibuprofen for 2 h, then with 10 ng/ml IL-1ß for 24 h. Results showed that pretreatment with ibuprofen inhibited the IL-1ß-induced nitric oxide (NO) and prostaglandin E2 (PGE2) production, protected the chondrocyte phenotype from IL-1ß stimulation, and inhibited the IL-1ß-induced actin remodeling via RhoA signaling modulation. In conclusion, ibuprofen showed not only anti-inflammatory function, but also RhoA inhibition in articular chondrocytes.


Subject(s)
Actins/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chondrocytes/drug effects , Ibuprofen/pharmacology , Interleukin-1beta/immunology , Animals , Cells, Cultured , Chondrocytes/immunology , Chondrocytes/pathology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Rabbits , rhoA GTP-Binding Protein/immunology
10.
Biosci Rep ; 39(7)2019 07 31.
Article in English | MEDLINE | ID: mdl-31262973

ABSTRACT

Oxymatrine (OMT) is an important quinoxaline alkaloid that has a wide range of pharmacological effects and has been shown to alleviate ulcerative colitis due to its profound anti-inflammatory effects. The RhoA/ROCK (Rho kinase) signaling pathway has been shown to be related to the pathogenesis of several autoimmune diseases; however, the specific mechanisms of RhoA/ROCK signaling in inflammatory bowel disease (IBD) remain elusive. Therefore, we sought to determine whether OMT could ameliorate acute intestinal inflammation by targeting the RhoA/ROCK signaling pathway. The potential therapeutic effect of OMT on acute intestinal inflammation and its impact on the RhoA/ROCK signaling pathway were assessed in six groups of mice treated with low, medium and high doses of OMT (25, 50 and 100 mg/kg, respectively), and an inhibitor of ROCK, Y-27632, as a positive control, after initiating dextran sodium sulfate (DSS)-induced acute intestinal inflammation. The model group and normal group were injected intraperitoneally with equal doses of PBS. Our results showed that OMT treatment could protect the integrity of the epithelial barrier, relieve oxidative stress, inhibit the expression of inflammatory mediators and pro-inflammatory cytokines, restrain the differentiation of Th17 cells and promote the differentiation of Treg cells via inhibition of the RhoA/ROCK pathway, thus providing therapeutic benefits for ulcerative colitis (UC). Therefore, inhibiting the RhoA/ROCK pathway might be a new approach that can be used in UC therapy, which deserves to be investigated further.


Subject(s)
Alkaloids/pharmacology , Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/drug therapy , Quinolizines/pharmacology , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics , Amides/pharmacology , Animals , Cell Differentiation , Cell Proliferation , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Dextran Sulfate , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Humans , Interleukins/genetics , Interleukins/immunology , Male , Mice , Mice, Inbred BALB C , Oxidative Stress , Pyridines/pharmacology , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/pathology , Treatment Outcome , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/immunology
11.
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
12.
Cancer Sci ; 110(8): 2357-2367, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31169331

ABSTRACT

Dendritic cells (DCs) are potent and specialized antigen presenting cells, which play a crucial role in initiating and amplifying both the innate and adaptive immune responses against cancer. Tumor cells can escape from immune attack by secreting suppressive cytokines that solely or cooperatively impair the immune function of DCs. However, the underlying mechanisms are not fully defined. Vascular endothelial growth factor (VEGF) has been identified as a major cytokine in the tumor microenvironment. To elucidate the effects of VEGF on the motility and immune function of mature DCs (mDCs), the cells were treated with 50 ng/mL VEGF and investigated by proteomics and molecular biological technologies. The results showed that VEGF can impair the migration capacity and immune function of mDCs through the RhoA-cofilin1 pathway mediated by the VEGF receptor 2, suggesting impaired motility of mDCs by VEGF is one of the aspects of immune escape mechanisms of tumors. It is clinically important to understand the biological behavior of DCs and the immune escape mechanisms of tumor as well as how to improve the efficiency of antitumor therapy based on DCs.


Subject(s)
Actin Depolymerizing Factors/metabolism , Cell Movement/immunology , Dendritic Cells/metabolism , Signal Transduction/immunology , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Depolymerizing Factors/immunology , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Human Umbilical Vein Endothelial Cells , Humans , Vascular Endothelial Growth Factor Receptor-2/immunology , rhoA GTP-Binding Protein/immunology
13.
Int Immunopharmacol ; 72: 74-81, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30959374

ABSTRACT

Microvesicles (MVs) have been extensively identified in various biological fluids including bronchoalveolar lavage fluid (BALF), peripheral blood and ascitic fluids. Our previous study showed that MVs are responsible for acute lung injury, but the exact mechanism underlying MVs formation remains poorly understood. In the present study, we investigate the potential role of RhoA/Rock signaling in MVs generation and the biological activity of MVs in ventilator-induced lung injury (VILI). Our results revealed that high tide ventilation induced super MVs releasing into the lung and subsequently caused lung inflammation. Strikingly, intratracheal instillation of MVs that isolated from highly ventilated mice triggered significant lung inflammation in naive mice. The MVs production is strongly correlated with lung inflammation and the upregulation of RhoA, Rock and phospho-Limk (phosphorylation of Limk is the activated form). RhoA inhibitor decreased the expression of Rock and the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Rock inhibitor also decreased the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Our data demonstrated that the production of MVs requires RhoA/Rock signaling, and VILI might be potentially prevented by targeting RhoA/Rock signaling pathway.


Subject(s)
ADP Ribose Transferases/therapeutic use , Botulinum Toxins/therapeutic use , Cell-Derived Microparticles/drug effects , Ventilator-Induced Lung Injury/drug therapy , rhoA GTP-Binding Protein/antagonists & inhibitors , Amides/therapeutic use , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Pyridines/therapeutic use , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/immunology
14.
J Allergy Clin Immunol ; 143(4): 1560-1574.e6, 2019 04.
Article in English | MEDLINE | ID: mdl-30194990

ABSTRACT

BACKGROUND: Numbers of mesenchymal stem cells (MSCs) are increased in the airways after allergen challenge. Ras homolog family member A (RhoA)/Rho-associated protein kinase 1 (ROCK) signaling is critical in determining the lineage fate of MSCs in tissue repair/remodeling. OBJECTIVES: We sought to investigate the role of RhoA/ROCK signaling in lineage commitment of MSCs during allergen-induced airway remodeling and delineate the underlying mechanisms. METHODS: Active RhoA expression in lung tissues of asthmatic patients and its role in cockroach allergen-induced airway inflammation and remodeling were investigated. RhoA/ROCK signaling-mediated MSC lineage commitment was assessed in an asthma mouse model by using MSC lineage tracing mice (nestin-Cre; ROSA26-EYFP). The role of RhoA/ROCK in MSC lineage commitment was also examined by using MSCs expressing constitutively active RhoA (RhoA-L63) or dominant negative RhoA (RhoA-N19). Downstream RhoA-regulated genes were identified by using the Stem Cell Signaling Array. RESULTS: Lung tissues from asthmatic mice showed increased expression of active RhoA when compared with those from control mice. Inhibition of RhoA/ROCK signaling with fasudil, a RhoA/ROCK inhibitor, reversed established cockroach allergen-induced airway inflammation and remodeling, as assessed based on greater collagen deposition/fibrosis. Furthermore, fasudil inhibited MSC differentiation into fibroblasts/myofibroblasts but promoted MSC differentiation into epithelial cells in asthmatic nestin-Cre; ROSA26-EYFP mice. Consistently, expression of RhoA-L63 facilitated differentiation of MSCs into fibroblasts/myofibroblasts, whereas expression of RhoA-19 switched the differentiation toward epithelial cells. The gene array identified the Wnt signaling effector lymphoid enhancer-binding factor 1 (Lef1) as the most upregulated gene in RhoA-L63-transfected MSCs. Knockdown of Lef1 induced MSC differentiation away from fibroblasts/myofibroblasts but toward epithelial cells. CONCLUSIONS: These findings uncover a previously unrecognized role of RhoA/ROCK signaling in MSC-involved airway repair/remodeling in the setting of asthma.


Subject(s)
Airway Remodeling/immunology , Asthma/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Mesenchymal Stem Cells/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Asthma/immunology , Asthma/pathology , Cell Lineage/immunology , Lymphoid Enhancer-Binding Factor 1/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/immunology
15.
Front Immunol ; 9: 1928, 2018.
Article in English | MEDLINE | ID: mdl-30233567

ABSTRACT

The Ras GTPase superfamily, including more than 100 members, plays a vital role in a number of cellular processes, such as cytoskeleton recombination, gene expression, and signaling pathway regulation. Some members of the superfamily participate in innate immunity in animals. However, there have been few studies of RhoA on this aspect. In the present study, we identified a RhoA GTPase in the shrimp Marsupenaeus japonicus and named it MjRhoA. Expression of MjRhoA was significantly upregulated in hemocytes and heart of shrimp challenged with Vibrio anguillarum. Overexpression of MjRhoA in shrimp caused the total bacterial number to decrease significantly and knockdown of MjRhoA increased the bacterial number obviously, with a consequent decline in shrimp survival. These results confirmed the antibacterial function of MjRhoA in shrimp. Further study showed that rate of phagocytosis of hemocytes was decreased in MjRhoA-knockdown shrimp. Interestingly, we observed that MjRhoA was translocated onto the hemocyte membrane at 1 h post V. anguillarum challenge. The expression levels of the ß-integrin-mediated phagocytosis markers ROCK2 and Arp2/3 declined significantly after knockdown of MjRhoA. These results suggested that the antibacterial function of MjRhoA was related to ß-integrin-mediated phagocytosis in shrimp. Our previous study identified that a C-type lectin, hFcLec4, initiated ß-integrin mediated phagocytosis after bacterial infection. Thus, knockdown of hFcLec4 and ß-integrin was performed. The results showed that the translocation of MjRhoA from the cytoplasm to membrane was inhibited and the expression level of MjRhoA was decreased, suggesting that MjRhoA participated in hFcLec4-integrin mediated phagocytosis. Therefore, our study identified a new hFcLec4-integrin-RhoA dependent phagocytosis against bacterial infection in shrimp.


Subject(s)
Arthropod Proteins/immunology , Bacterial Infections/immunology , Integrins/immunology , Penaeidae , Phagocytosis/immunology , rhoA GTP-Binding Protein/immunology , Animals , Hemocytes/immunology , Hemocytes/microbiology , Penaeidae/immunology , Penaeidae/microbiology
16.
Front Immunol ; 9: 2001, 2018.
Article in English | MEDLINE | ID: mdl-30254631

ABSTRACT

We previously identified Fam65b as an atypical inhibitor of the small G protein RhoA. Using a conditional model of a Fam65b-deficient mouse, we first show that Fam65b restricts spontaneous RhoA activation in resting T lymphocytes and regulates intranodal T cell migration in vivo. We next aimed at understanding, at the molecular level, how the brake that Fam65b exerts on RhoA can be relieved upon signaling to allow RhoA activation. Here, we show that chemokine stimulation phosphorylates Fam65b in T lymphocytes. This post-translational modification decreases the affinity of Fam65b for RhoA and favors Fam65b shuttling from the plasma membrane to the cytosol. Functionally, we show that the degree of Fam65b phosphorylation controls some cytoskeletal alterations downstream active RhoA such as actin polymerization, as well as T cell migration in vitro. Altogether, our results show that Fam65b expression and phosphorylation can finely tune the amount of active RhoA in order to favor optimal T lymphocyte motility.


Subject(s)
Carrier Proteins/immunology , Cell Movement/immunology , Membrane Proteins/immunology , Proteins/immunology , T-Lymphocytes/immunology , rho GTP-Binding Proteins/immunology , rhoA GTP-Binding Protein/immunology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/immunology , Animals , Carrier Proteins/genetics , Cell Adhesion Molecules , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation/immunology , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Proteins/genetics , T-Lymphocytes/cytology , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/genetics
17.
J Cell Mol Med ; 22(9): 4550-4554, 2018 09.
Article in English | MEDLINE | ID: mdl-29993181

ABSTRACT

Sphingosine-1-phosphate (S1P) is a crucial regulator in vascular inflammation. Our recent study found that under pathophysiological concentration in active anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV), S1P participated in MPO-ANCA-positive IgG-induced glomerular endothelial cell (GEnC) activation via a S1P receptor (S1PR)-dependent way. However, the downstream signalling pathways are not fully clear yet. In this study, we demonstrated that Rho guanosine triphosphatases (GTPases) signalling pathways, RhoA and Rac1 in particular, were implicated in MPO-ANCA-positive IgG-mediated GEnCs activation enhanced by pathophysiological concentration of S1P in AAV. These results provide mechanistic insights into vascular barrier dysfunction in AAV, which may facilitate the development of effective therapies.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/genetics , Endothelial Cells/drug effects , Immunoglobulin G/genetics , Lysophospholipids/pharmacology , Sphingosine/analogs & derivatives , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics , Antibodies, Antineutrophil Cytoplasmic/biosynthesis , Endothelial Cells/cytology , Endothelial Cells/immunology , Gene Expression Regulation , Humans , Immunoglobulin G/biosynthesis , Kidney Glomerulus/cytology , Kidney Glomerulus/immunology , Peroxidase/antagonists & inhibitors , Peroxidase/genetics , Peroxidase/immunology , Primary Cell Culture , Signal Transduction , Sphingosine/pharmacology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , rac1 GTP-Binding Protein/immunology , rhoA GTP-Binding Protein/immunology
18.
Pathog Dis ; 76(3)2018 04 01.
Article in English | MEDLINE | ID: mdl-29718184

ABSTRACT

Numerous pathogens including Clostridium difficile and Yersinia pestis have evolved toxins or effectors targeting GTPases from the RhoA subfamily (RhoA/B/C) to inhibit or hijack the host cytoskeleton dynamics. The resulting impairment of RhoA GTPases activity is sensed by the host via an innate immune complex termed the pyrin inflammasome in which caspase-1 is activated. The cascade leading to activation of the pyrin inflammasome has been recently uncovered. In this review, following a brief presentation of RhoA GTPases-modulating toxins, we present the pyrin inflammasome and its regulatory mechanisms. Furthermore, we discuss how some pathogens have developed strategies to escape detection by the pyrin inflammasome. Finally, we present five monogenic autoinflammatory diseases associated with pyrin inflammasome deregulation. The molecular insights provided by the study of these diseases and the corresponding mutations on pyrin inflammasome regulation and activation are presented.


Subject(s)
Bacterial Toxins/immunology , Enterocolitis, Pseudomembranous/immunology , Inflammasomes/immunology , Plague/immunology , Pyrin/immunology , rhoA GTP-Binding Protein/immunology , Animals , Autoimmunity , Bacterial Toxins/biosynthesis , Clostridioides difficile/immunology , Clostridioides difficile/metabolism , Clostridioides difficile/pathogenicity , Enterocolitis, Pseudomembranous/microbiology , Enterocolitis, Pseudomembranous/pathology , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/genetics , Inflammation , Isoenzymes/genetics , Isoenzymes/immunology , Plague/microbiology , Plague/pathology , Pyrin/genetics , Syndrome , Yersinia pestis/immunology , Yersinia pestis/metabolism , Yersinia pestis/pathogenicity , rhoA GTP-Binding Protein/genetics
19.
Arch Oral Biol ; 87: 151-156, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29294450

ABSTRACT

OBJECTIVE: The present study was designed to analyze the immunolocalization of proteins involved in cytoskeleton remodeling, such as moesin and Rho-A, in benign odontogenic lesions that present with expansive growth and invasive clinical behavior. MATERIALS AND METHODS: Expressions of moesin and Rho-A in odontogenic epithelium were evaluated by immunohistochemical analysis in 45 odontogenic lesions using monoclonal antibodies. RESULTS: Our results demonstrated strong membranous and cytoplasmic expressions of moesin in the epithelial cells in 66.7% and 44.4% of the odontogenic lesions, respectively. Furthermore, Rho-A expression in odontogenic epithelium was strong in the membrane and cytoplasm of 51.1% and 62.2% of the odontogenic lesions, respectively. A statistically significant correlation was found between the membranous and cytoplasmic expressions of moesin (p = 0.000) and those of Rho-A (p = 0.048) in odontogenic epithelial cells, while no statistically significant correlation was found between moesin and Rho-A expressions (p > 0.05). CONCLUSIONS: The present study confirmed the strong expressions of moesin and Rho-A by odontogenic epithelial cells, suggesting their involvement in the development of benign odontogenic lesions. However, this study has failed to detect the connection between the moesin and Rho-A interaction in expansive growth and local invasiveness of these lesions.


Subject(s)
Cytoplasm/metabolism , Cytoskeleton/metabolism , Epithelium/metabolism , Microfilament Proteins/metabolism , Odontogenic Cysts/metabolism , Odontogenic Tumors/metabolism , rhoA GTP-Binding Protein/metabolism , Adolescent , Adult , Aged , Child , Cytoplasm/immunology , Cytoskeleton/immunology , Epithelium/immunology , Female , Humans , Immunohistochemistry , Male , Microfilament Proteins/immunology , Middle Aged , Odontogenic Cysts/immunology , Odontogenic Tumors/immunology , rhoA GTP-Binding Protein/immunology
20.
Toxicon ; 149: 54-64, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29146177

ABSTRACT

Clostridium difficile and Clostridium sordellii produce related potent toxins (C. difficile toxin A (TcdA) and toxin B (TcdB), C. sordellii lethal toxin (TcsL) and hemorrhagic toxin (TcsH)) which belong to the large clostridial glucosylating toxin (LCGT) family. TcsL is the main C. sordellii toxin as most of toxigenic C. sordellii strains only synthesize this toxin. Intestinal colonization by C. difficile subsequently to unbalanced microbiota is accompanied by release of toxins which induce local tissue destruction and severe inflammatory response. TcdA and TcdB inactivate Rho-GTPases. Notably inactivation of RhoA results in the stimulation of the pyrin/ASC inflammasome, which is one of the main signaling pathways used by these toxins to trigger the inflammatory response. In contrast, TcsL induces an anti-inflammatory effect, mainly by inactivating Ras proteins which results in blockage of the cell cycle and killing of immune cells. The absence or moderate local inflammatory response allows C. sordellii spreading in deep tissues, production of toxin which is released in the general circulation and causes a toxic shock syndrome. The toxin mechanisms of pro-versus anti-inflammatory responses are discussed.


Subject(s)
Bacterial Toxins/toxicity , Clostridioides difficile/chemistry , Clostridium sordellii/chemistry , Bacterial Proteins/immunology , Bacterial Proteins/toxicity , Bacterial Toxins/immunology , Cytokines/immunology , Enterotoxins/immunology , Enterotoxins/toxicity , Humans , Inflammasomes/physiology , Models, Immunological , rhoA GTP-Binding Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL