Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
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
2.
Cells ; 9(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992982

ABSTRACT

Epac1 (exchange protein activated by cAMP) stabilizes the endothelial barrier, but detailed studies are limited by the side effects of pharmacological Epac1 modulators and transient transfections. Here, we compare the key properties of barriers between endothelial cells derived from wild-type (WT) and Epac1-knockout (KO) mice myocardium. We found that KO cell layers, unlike WT layers, had low and cAMP-insensitive trans-endothelial resistance (TER). They also had fragmented VE-cadherin staining despite having augmented cAMP levels and increased protein expression of Rap1, Rac1, RhoA, and VE-cadherin. The simultaneous direct activation of Rac1 and RhoA by CN04 compensated Epac1 loss, since TER was increased. In KO-cells, inhibition of Rac1 activity had no additional effect on TER, suggesting that other mechanisms compensate the inhibition of the Rac1 function to preserve barrier properties. In summary, Epac1 is crucial for baseline and cAMP-mediated barrier stabilization through mechanisms that are at least partially independent of Rac1.


Subject(s)
Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/genetics , Myocardium/metabolism , Neuropeptides/genetics , rac1 GTP-Binding Protein/genetics , rap1 GTP-Binding Proteins/drug effects , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Membrane Permeability/drug effects , Cyclic AMP/genetics , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Knockout , Myocardium/pathology , Neuropeptides/agonists , Signal Transduction/genetics , Transcriptional Activation/drug effects , rac1 GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/genetics
3.
Int Immunopharmacol ; 78: 106066, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31835087

ABSTRACT

Intestinal barrier dysfunction is a trigger for sepsis progression. NLRP3 inflammasome and RhoA contribute to sepsis and intestinal inflammation. The current study aimed to explore the effects of Astragaloside IV (AS-IV), a bioactive compound from Astragalus membranaceus, on sepsis-caused intestinal barrier dysfunction and whether NLRP3 inflammasome and RhoA are involved. Septic mice modeled by cecal ligation and puncture (CLP) operation were administered with 3 mg/kg AS-IV intravenously. AS-IV decreased mortality, cytokines release, I-FABP secretion, intestinal histological score and barrier permeability, and increased tight junction (TJ) expression in intestine in CLP model. Also, in Caco-2 cells subjected to lipopolysaccharide (LPS), 200 µg/mL AS-IV co-incubation reduced cytokines levels and enhanced in vitro gut barrier function without cytotoxicity. Subsequently, NLRP3 inflammasome and RhoA were highly activated both in intestinal tissue in vivo and in Caco-2 cells in vitro, both of which were significantly suppressed by AS-IV treatment. In addition, the benefits of AS-IV on Caco-2 monolayer barrier were largely counteracted by RhoA agonist CN03 and NLRP3 gene overexpression, respectively. Furthermore, LPS-induced NLRP3 inflammasome activation was abrogated by RhoA inhibitor C3 exoenzyme. However, NLRP3 knockdown by siRNA hardly affected RhoA activation in Caco-2 cells. These data suggest that AS-IV protects intestinal epithelium from sepsis-induced barrier dysfunction via inhibiting RhoA/NLRP3 inflammasome signal pathway.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Inflammasomes/drug effects , Intestinal Mucosa/drug effects , Saponins/administration & dosage , Sepsis/drug therapy , Signal Transduction/drug effects , Triterpenes/administration & dosage , ADP Ribose Transferases/pharmacology , Animals , Astragalus propinquus/chemistry , Botulinum Toxins/pharmacology , Caco-2 Cells , Disease Models, Animal , Gene Knockdown Techniques , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Injections, Intravenous , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Permeability/drug effects , RNA, Small Interfering/metabolism , Sepsis/complications , Sepsis/immunology , Signal Transduction/immunology , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
4.
Exp Cell Res ; 360(2): 113-124, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28865751

ABSTRACT

De-differentiation comprises a major drawback for the use of autologous chondrocytes in cartilage repair. Here, we investigate the role of RhoA and canonical Wnt signaling in chondrocyte phenotype. Chondrocyte de-differentiation is accompanied by an upregulation and nuclear localization of RhoA. Effectors of canonical Wnt signaling including ß-catenin and YAP/TAZ are upregulated in de-differentiating chondrocytes in a Rho-dependent manner. Inhibition of Rho activation with C3 transferase inhibits nuclear localization of RhoA, induces expression of chondrogenic markers on 2D and enhances the chondrogenic effect of 3D culturing. Upregulation of chondrogenic markers by Rho inhibition is accompanied by loss of canonical Wnt signaling markers in 3D or on 2D whereas treatment of chondrocytes with Wnt-3a abrogates this effect. However, induction of canonical Wnt signaling inhibits chondrogenic markers on 2D but enhances chondrogenic re-differentiation on 2D with C3 transferase or in 3D. These data provide insights on the context-dependent role of RhoA and Wnt signaling in de-differentiation and on mechanisms to induce chondrogenic markers for therapeutic approaches.


Subject(s)
Cell Dedifferentiation , Cell Nucleus/metabolism , Chondrocytes/physiology , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Botulinum Toxins/pharmacology , Cattle , Cell Dedifferentiation/drug effects , Cell Nucleus/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Phenotype , Protein Transport/drug effects , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Wnt Signaling Pathway/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors
5.
Thromb Haemost ; 116(3): 506-16, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27345948

ABSTRACT

Leukemia-Associated RhoGEF (LARG) is highly expressed in platelets, which are essential for maintaining normal haemostasis. We studied the function of LARG in murine and human megakaryocytes and platelets with Larg knockout (KO), shRNA-mediated knockdown and small molecule-mediated inhibition. We found that LARG is important for human, but not murine, megakaryocyte maturation. Larg KO mice exhibit macrothrombocytopenia, internal bleeding in the ovaries and prolonged bleeding times. KO platelets have impaired aggregation, α-granule release and integrin α2bß3 activation in response to thrombin and thromboxane, but not to ADP. The same agonist-specific reductions in platelet aggregation occur in human platelets treated with a LARG inhibitor. Larg KO platelets have reduced RhoA activation and myosin light chain phosphorylation, suggesting that Larg plays an agonist-specific role in platelet signal transduction. Using two different in vivo assays, Larg KO mice are protected from in vivo thrombus formation. Together, these results establish that LARG regulates human megakaryocyte maturation, and is critical for platelet function in both humans and mice.


Subject(s)
Blood Platelets/metabolism , Rho Guanine Nucleotide Exchange Factors/blood , rho GTP-Binding Proteins/blood , rhoA GTP-Binding Protein/blood , Animals , Bleeding Time , Blood Platelets/drug effects , Gene Knockdown Techniques , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Light Chains/blood , Platelet Function Tests , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombin/metabolism , Thrombin/pharmacology , Thrombopoiesis/genetics , Thrombopoiesis/physiology , Thromboxanes/blood , Thromboxanes/pharmacology , rho GTP-Binding Proteins/agonists , rhoA GTP-Binding Protein/agonists
6.
Hum Reprod ; 31(4): 723-33, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26874360

ABSTRACT

STUDY QUESTION: Is phosphatase of regenerating liver-3 (PRL-3) associated with increased motility of endometriotic cells from endometrioma? SUMMARY ANSWER: Elevated PRL-3 promotes cytoskeleton reorganization, cell migration and invasion of endometrial stromal cells (ESCs) from endometrioma. WHAT IS KNOWN ALREADY: Overexpression of PRL-3 is associated with cancer cell migration, invasion and metastatic phenotype. STUDY DESIGN, SIZE, DURATION: Primary human ESCs were isolated from eutopic endometrium of women without endometriosis (EuCo, n = 10), with histologically proven endometrioma (EuEM, n = 19) and from the cyst wall of ovarian endometriosis (OvEM, n = 26). PARTICIPANTS/MATERIALS, SETTING, METHODS: The expression of PRL-3 in ESCs derived from EuCo, EuEM and OvEM at different phases of menstrual cycle were compared. The protein and mRNA levels of PRL-3 were examined by western blot and RT-qPCR, respectively. ESCs from OvEM were transfected with/without short hairpin RNA (shRNA) or small interfering RNA (siRNA). Additionally, a plasmid-mediated delivery system was used to achieve PRL-3 overexpression in ESCs from EuEM. The cellular distribution of F-actin and α-tubulin were examined by immunocytochemistry. Cell motility was evaluated by a transwell migration/invasion assay. MAIN RESULTS AND THE ROLE OF CHANCE: The protein and mRNA levels of PRL-3 are significantly elevated in ESCs from OvEM compared with EuCo and EuEM. The expression of PRL-3 was not altered between proliferative phase and secretory phase in ESCs from all groups. Knockdown of PRL-3 significantly modified the distribution of F-actin and α-tubulin cytoskeleton, inhibited cell migration and invasion. Endogenous inhibition of PRL-3 attenuated the expression of Ras homolog gene family members A and C (RhoA, RhoC), Rho-associated coiled-coil-containing protein kinase 1 (ROCK1) and matrix metalloproteinase (MMP) 9, but not MMP2 in ESCs from OvEM. Additionally, overexpression of PRL-3 in ESCs from EuEM up-regulates cell migration and invasion, and increases the expression of RhoA, RhoC, ROCK1 and MMP9. LIMITATIONS, REASONS FOR CAUTION: Lack of in vivo animal studies is the major limitation of our report. Our results should be further confirmed in a larger cohort of patients and extended to include eutopic and ectopic endometrium from patients with peritoneal endometriosis at different stages of the disease. WIDER IMPLICATIONS OF THE FINDINGS: Our study describes that elevated expression of PRL-3 contributes to the cell motility of ESCs from endometrioma. The results emphasize the importance of metastatic-related factor PRL-3 in the pathogenesis of endometrioma. STUDY FUNDING/COMPETING INTEREST: This work was supported by National Natural Science Foundation of China (No. 81170546) and Zhejiang Medicine Science and Technology Projects (No. Y13H040003). The authors declare no conflict of interest.


Subject(s)
Cytoskeleton/metabolism , Endometriosis/metabolism , Endometrium/metabolism , Enzyme Induction , Neoplasm Proteins/metabolism , Ovarian Diseases/metabolism , Protein Tyrosine Phosphatases/metabolism , Stromal Cells/metabolism , Cell Movement , Cells, Cultured , Cytoskeleton/pathology , Endometriosis/pathology , Endometrium/cytology , Endometrium/pathology , Female , Gene Expression Regulation, Enzymologic , Humans , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Menstrual Cycle/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Ovarian Diseases/pathology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , RNA Interference , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stromal Cells/cytology , Stromal Cells/pathology , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/chemistry , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , rhoC GTP-Binding Protein
7.
Lab Invest ; 95(10): 1145-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26146960

ABSTRACT

Hepatocellular carcinoma (HCC) carries a poor prognosis with no effective treatment available other than liver transplantation for selected patients. Vascular invasion of HCC is one of the most important negative predictor of survival. As the regulation of invasion of HCC cells is not well understood, our aim was to study the mechanisms by which galectin 3, a ß-galactosidase-binding lectin mediates HCC cell migration. HCC was induced by N-diethylnitrosamine in wild-type and galectin 3(-/-) mice, and tumor formation, histology, and tumor cell invasion were assessed. The galectin 3(-/-) mice developed significantly smaller tumor burden with a less invasive phenotype than the wild-type animals. Galectin 3 was upregulated in the wild-type HCC tumor tissue, but not in the surrounding parenchyma. Galectin 3 expression in HCC was induced by NF-κB transactivation as determined by chromatin immunoprecipitation assays. In vitro studies assessed the pro-migratory effects of galectin 3. The migration of hepatoma cells was significantly decreased after transfection by the galectin 3 siRNA and also after using the Rho kinase inhibitor Y-27632. The reorganization of the actin cytoskeleton, RhoA GTPase activity and the phosphorylation of MLC2 (myosin light chain 2) were decreased in the galectin 3 siRNA-transfected cells. In addition, in vitro and in vivo evidence showed that galectin 3 deficiency reduced hepatoma cell proliferation and increased their apoptosis rate. In conclusion, galectin 3 is an important lectin that is induced in HCC cells, and promotes hepatoma cell motility and invasion by an autocrine pathway. Targeting galectin 3 therefore could be an important novel treatment strategy to halt disease progression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Galectin 3/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Myosin-Light-Chain Kinase/metabolism , Neoplasm Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cardiac Myosins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Activation/drug effects , Galectin 3/antagonists & inhibitors , Galectin 3/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Liver/drug effects , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/chemistry , Neoplasm Invasiveness , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
8.
PLoS One ; 9(9): e109055, 2014.
Article in English | MEDLINE | ID: mdl-25268128

ABSTRACT

MAZ51 is an indolinone-based molecule originally synthesized as a selective inhibitor of vascular endothelial growth factor receptor (VEGFR)-3 tyrosine kinase. This study shows that exposure of two glioma cell lines, rat C6 and human U251MG, to MAZ51 caused dramatic shape changes, including the retraction of cellular protrusions and cell rounding. These changes were caused by the clustering and aggregation of actin filaments and microtubules. MAZ51 also induced G2/M phase cell cycle arrest. This led to an inhibition of cellular proliferation, without triggering significant cell death. These alterations induced by MAZ51 occurred with similar dose- and time-dependent patterns. Treatment of glioma cells with MAZ51 resulted in increased levels of phosphorylated GSK3ß through the activation of Akt, as well as increased levels of active RhoA. Interestingly, MAZ51 did not affect the morphology and cell cycle patterns of rat primary cortical astrocytes, suggesting it selectively targeted transformed cells. Immunoprecipitation-western blot analyses indicated that MAZ51 did not decrease, but rather increased, tyrosine phosphorylation of VEGFR-3. To confirm this unanticipated result, several additional experiments were conducted. Enhancing VEGFR-3 phosphorylation by treatment of glioma cells with VEGF-C affected neither cytoskeleton arrangements nor cell cycle patterns. In addition, the knockdown of VEGFR-3 in glioma cells did not cause morphological or cytoskeletal alterations. Furthermore, treatment of VEGFR-3-silenced cells with MAZ51 caused the same alterations of cell shape and cytoskeletal arrangements as that observed in control cells. These data indicate that MAZ51 causes cytoskeletal alterations and G2/M cell cycle arrest in glioma cells. These effects are mediated through phosphorylation of Akt/GSK3ß and activation of RhoA. The anti-proliferative activity of MAZ51 does not require the inhibition of VEGFR-3 phosphorylation, suggesting that it is a potential candidate for further clinical investigation for treatment of gliomas, although the precise mechanism(s) underlying its effects remain to be determined.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/genetics , Indoles/pharmacology , Naphthalenes/pharmacology , Neuroglia/drug effects , Proto-Oncogene Proteins c-akt/genetics , rhoA GTP-Binding Protein/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/ultrastructure , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Microtubules/drug effects , Microtubules/ultrastructure , Neuroglia/metabolism , Neuroglia/pathology , Neuroglia/ultrastructure , Phosphorylation , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Vascular Endothelial Growth Factor C/pharmacology , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/metabolism
9.
Cell Commun Signal ; 12: 24, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24690323

ABSTRACT

BACKGROUND: Cellular plasticity confers cancer cells the ability to adapt to microenvironmental changes, a fundamental requirement for tumour progression and metastasis. The epithelial to mesenchymal transition (EMT) is a transcriptional programme associated with increased cell motility and stemness. Besides EMT, the mesenchymal to amoeboid transition (MAT) has been described during tumour progression but to date, little is known about its transcriptional control and involvement in stemness. The aim of this manuscript is to investigate (i) the transcriptional profile associated with the MAT programme and (ii) to study whether MAT acquisition in melanoma cancer cells correlates with clonogenic potential to promote tumour growth. RESULTS: By using a multidisciplinary approach, we identified four different treatments able to induce MAT in melanoma cells: EphA2 overexpression, Rac1 functional inhibition using its RacN17 dominant negative mutant, stimulation with Ilomastat or treatment with the RhoA activator Calpeptin. First, gene expression profiling identified the transcriptional pathways associated with MAT, independently of the stimulus that induces the MAT programme. Notably, gene sets associated with the repression of mesenchymal traits, decrease in the secretion of extracellular matrix components as well as increase of cellular stemness positively correlate with MAT. Second, the link between MAT and stemness has been investigated in vitro by analysing stemness markers and clonogenic potential of melanoma cells undergoing MAT. Finally, the link between MAT inducing treatments and tumour initiating capability has been validated in vivo. CONCLUSION: Taken together, our results demonstrate that MAT programme in melanoma is characterised by increased stemness and clonogenic features of cancer cells, thus sustaining tumour progression. Furthermore, these data suggest that stemness is not an exclusive feature of cells undergoing EMT, but more generally is associated with an increase in cellular plasticity of cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Cell Movement , Dipeptides/pharmacology , Humans , Melanoma/pathology , Neoplastic Stem Cells/physiology , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Transcription, Genetic , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
10.
Shock ; 40(1): 49-58, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23635848

ABSTRACT

The aim of this present study was to examine changes in RhoA protein levels and the role in RhoA in lymphatic contractility and reactivity after hemorrhagic shock. Levels of RhoA and phospho-RhoA in lymphatic tissue isolated from hemorrhagic shock rats were measured, and the contractility and reactivity to substance P of lymphatics isolated from control rats and rats subjected to shock 0.5 and 2 h were determined with an isolated lymphatic perfusion system at a transmural pressure of 3 cmH2O. At the same time, lymphatics isolated from rats subjected to shock 0.5 and 2 h were incubated with agonists and antagonists of RhoA/Rho kinase signaling. Contractile frequency, end-diastolic and end-systolic diameter, and passive diameter were recorded and used to calculate lymphatic tonic index, contractile amplitude, and fractional pump flow. After stimulation with a gradient of substance P, the differences between the preadministration and postadministration values of contractile frequency, contractile amplitude, tonic index, and fractional pump flow were calculated to further assess lymphatic reactivity. RhoA protein levels were significantly increased at 0.5 h after shock but decreased at 2 and 3 h after shock; p-Rho levels were initially increased after shock and subsequently decreased. The contractility and reactivity of 0.5-h-shocked lymphatics were significantly reduced by the RhoA antagonist C3 transferase and the Rho kinase antagonist Y-27632. The RhoA agonist U-46619 increased the contractility and reactivity of 2-h-shocked lymphatics, whereas Y-27632 suppressed the effect of U-46619. Okadaic acid, an inhibitor of myosin light-chain phosphatase, had no effect on the contractility of 2-h-shocked lymphatics, but improved lymphatic reactivity. These results suggest that RhoA is involved in the modulation of lymphatic pump function during hemorrhagic shock and that its effects may be mediated by Rho kinase and MLCP.


Subject(s)
Lymphatic Vessels/metabolism , Shock, Hemorrhagic/metabolism , rhoA GTP-Binding Protein/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , In Vitro Techniques , Male , Rats , Rats, Wistar , Shock, Hemorrhagic/physiopathology , rhoA GTP-Binding Protein/agonists
11.
Cell Death Dis ; 4: e568, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23559002

ABSTRACT

Although statins are known to inhibit proliferation and induce death in a number of cancer cell types, the mechanisms through which downregulation of the mevalonate (MVA) pathway activates death signaling remain poorly understood. Here we set out to unravel the signaling networks downstream of the MVA pathway that mediate the death-inducing activity of simvastatin. Consistent with previous reports, exogenously added geranylgeranylpyrophosphate, but not farnesylpyrophosphate, prevented simvastatin's growth-inhibitory effect, thereby suggesting the involvement of geranylgeranylated proteins such as Rho GTPases in the anticancer activity of simvastatin. Indeed, simvastatin treatment led to increased levels of unprenylated Ras homolog gene family, member A (RhoA), Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division cycle 42 (Cdc42). Intriguingly, instead of inhibiting the functions of Rho GTPases as was expected with loss of prenylation, simvastatin caused a paradoxical increase in the GTP-bound forms of RhoA, Rac1 and Cdc42. Furthermore, simvastatin disrupted the binding of Rho GTPases with the cytosolic inhibitor Rho GDIα, which provides a potential mechanism for GTP loading of the cytosolic Rho GTPases. We also show that the unprenylated RhoA- and Rac1-GTP retained at least part of their functional activities, as evidenced by the increase in intracellular superoxide production and JNK activation in response to simvastatin. Notably, blocking superoxide production attenuated JNK activation as well as cell death induced by simvastatin. Finally, we provide evidence for the involvement of the B-cell lymphoma protein 2 family, Bcl-2-interacting mediator (Bim), in a JNK-dependent manner, in the apoptosis-inducing activity of simvastatin. Taken together, our data highlight the critical role of non-canonical regulation of Rho GTPases and involvement of downstream superoxide-mediated activation of JNK pathway in the anticancer activity of simvastatin, which would have potential clinical implications.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Signal Transduction/drug effects , Simvastatin/pharmacology , rac1 GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/agonists , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mevalonic Acid/metabolism , Prenylation , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Superoxides/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
12.
Mol Endocrinol ; 26(5): 716-35, 2012 May.
Article in English | MEDLINE | ID: mdl-22456196

ABSTRACT

Recently, we have identified serum response factor (SRF) as a mediator of clinically relevant androgen receptor (AR) action in prostate cancer (PCa). Genes that rely on SRF for androgen responsiveness represent a small fraction of androgen-regulated genes, but distinguish benign from malignant prostate, correlate with aggressive disease, and are associated with biochemical recurrence. Thus, understanding the mechanism(s) by which SRF conveys androgen regulation to its target genes may provide novel opportunities to target clinically relevant androgen signaling. Here, we show that the small GTPase ras homolog family member A (RhoA) mediates androgen-responsiveness of more than half of SRF target genes. Interference with expression of RhoA, activity of the RhoA effector Rho-associated coiled-coil containing protein kinase 1 (ROCK), and actin polymerization necessary for nuclear translocation of the SRF cofactor megakaryocytic acute leukemia (MAL) prevented full androgen regulation of SRF target genes. Androgen treatment induced RhoA activation, increased the nuclear content of MAL, and led to MAL recruitment to the promoter of the SRF target gene FHL2. In clinical specimens RhoA expression was higher in PCa cells than benign prostate cells, and elevated RhoA expression levels were associated with aggressive disease features and decreased disease-free survival after radical prostatectomy. Overexpression of RhoA markedly increased the androgen-responsiveness of select SRF target genes, in a manner that depends on its GTPase activity. The use of isogenic cell lines and a xenograft model that mimics the transition from androgen-stimulated to castration-recurrent PCa indicated that RhoA levels are not altered during disease progression, suggesting that RhoA expression levels in the primary tumor determine disease aggressiveness. Androgen-responsiveness of SRF target genes in castration-recurrent PCa cells continued to rely on AR, RhoA, SRF, and MAL and the presence of intact SRF binding sites. Silencing of RhoA, use of Rho-associated coiled-coil containing protein kinase 1 inhibitors, or an inhibitor of SRF-MAL interaction attenuated (androgen-regulated) cell viability and blunted PCa cell migration. Taken together, these studies demonstrate that the RhoA signaling axis mediates clinically relevant AR action in PCa.


Subject(s)
Androgens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism , Androgens/adverse effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Nude , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic/drug effects , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Receptors, Androgen/genetics , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Trans-Activators , Transcription Factors/genetics , Transcription Factors/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/genetics
13.
Toxicol Lett ; 205(3): 250-6, 2011 Sep 10.
Article in English | MEDLINE | ID: mdl-21708234

ABSTRACT

A close link between arsenic exposure and hypertension has been well-established through many epidemiological reports, yet the mechanism underlying it remains unclear. Here we report that nanomolar concentrations of monomethylarsonous acid (MMA(III)), a toxic trivalent methylated arsenic metabolite, can potentiate agonist-induced vasoconstriction and pressor responses. In freshly isolated rat aortic ring, exposure to nanomolar MMA(III) (100-500 nM) potentiated phenylephrine (PE)-induced vasoconstriction while at higher concentrations (≥2.5 µM), suppression of vasoconstriction and apoptosis of vascular smooth muscle were observed. Potentiation of agonist-induced vasoconstriction was also observed with other contractile agonists and it was retained in endothelium-denuded aortic rings, suggesting that these events are agonist-independent and smooth muscle cell dependent. Interestingly, exposure to MMA(III) resulted in increased myosin light chain phosphorylation while PE-induced Ca2+ influx was not affected, reflecting that Ca2+ sensitization is involved. In line with this, MMA(III) enhanced agonist-induced activation of small GTPase RhoA, a key contributor to Ca2+ sensitization. Of note, treatment of MMA(III) to rats induced significantly higher pressor responses in vivo, demonstrating that this event can occur in vivo indeed. We believe that RhoA-mediated Ca2+ sensitization and the resultant potentiation of vasoconstriction by MMA(III) may shed light on arsenic-associated hypertension.


Subject(s)
Hypertension/chemically induced , Organometallic Compounds/toxicity , Pressoreceptors/drug effects , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Apoptosis/drug effects , Arsenic Poisoning/physiopathology , Arsenicals/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hypertension/etiology , In Vitro Techniques , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myosin Light Chains/metabolism , Osmolar Concentration , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/antagonists & inhibitors , Vasoconstrictor Agents/chemistry , Vasoconstrictor Agents/pharmacology , rhoA GTP-Binding Protein/agonists
14.
J Cell Physiol ; 226(8): 2159-69, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21520068

ABSTRACT

Tensile load is known to regulate the osteogenesis of mesenchymal stem cells (MSCs) and osteogenic progenitors; therefore it is widely used in clinical treatment and tissue engineering. Meanwhile, in vitro, both published studies and our lab data demonstrate that the application of intermittent tensile loading which stimulates cells several minutes or hours each day for several days has promoted the osteogenic differentiation of MSCs. Whereas, for clinic trails, it is important to know accurately how and how long mechanical tension should be applied. Hence, it is necessary to investigate different kinds of mechanical tension on osteogenesis of MSCs. Until now, during the osteogenesis, there has been no research on the effect of continuous cyclic mechanical tension (CCMT) which provides continuous stimulation throughout the study period. We firstly figure out CCMT inhibiting the expression of osteogenic genes such as key transcription factor Runx2. It is known that RhoA regulates cell differentiation in response to mechanical stimuli. MAPK signaling acts as a downstream effector of RhoA. So, we ask in MSCs, if CCMT regulates the osteogenic master gene Runx2 through RhoA-ERK1/2 pathway. And then, we find out there is a decrease in RhoA activity after CCMT stimulation. Pre-treatment of CCMT-loaded MSCs with LPA, a RhoA activator, restores ALP activity and significantly rescues Runx2 expression, while pre-treatment with C3 toxin, a RhoA inhibitor, further decreases the activity of ALP and down-regulates the expression of Runx2. Following results indicate that the inhibition of Runx2 expression after CCMT stimulation is mediated by RhoA-ERK1/2 pathway.


Subject(s)
Core Binding Factor Alpha 1 Subunit/biosynthesis , Mesenchymal Stem Cells/metabolism , Stress, Mechanical , ADP Ribose Transferases/pharmacology , Adult , Alkaline Phosphatase/analysis , Botulinum Toxins/pharmacology , Cells, Cultured , Humans , Lysophospholipids/pharmacology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors
15.
J Pharmacol Sci ; 115(2): 135-43, 2011.
Article in English | MEDLINE | ID: mdl-21258176

ABSTRACT

Protease-activated receptor 1 (PAR1) that can be activated by serine proteinases such as thrombin has been demonstrated to contribute to the development of cardiac remodeling and hypertrophy after myocardial injury. Here, we investigated the mechanisms by which PAR1 leads to hypertrophic cardiomyocyte growth using cultured rat neonatal ventricular myocytes. PAR1 stimulation with thrombin (1 U/ml) or a synthetic agonist peptide (TFLLR-NH(2), 50 µM) for 48 h induced an increase in cell size and myofibril formation associated with BNP (brain natriuretic peptide) production. This actin reorganization assessed by fluorescein isothiocyanate (FITC)-conjugated phalloidin staining appeared at 1 h after PAR1 stimulation, and this response was reduced by a protein kinase C (PKC) inhibitor, chelerythrine, inhibitors of Rho (simvastatin) and Rho-associated kinase (ROCK) (Y-27632), but not by pertussis toxin (PTX). By Western blot analysis, translocation of PKCα or PKCε from the cytosol to membrane fractions was observed in cells stimulated with thrombin or TFLLR-NH(2) for 2 - 5 min. In addition, PAR1 stimulation for 3 - 5 min increased the level of active RhoA. Furthermore, inhibitors of PKC and ROCK and Rho abrogated PAR1-mediated increase in cell size. Depletion of PKCα or PKCε by specific small interfering RNA also suppressed both actin reorganization and cell growth. These results suggest that PAR1 stimulation of cardiomyocytes induces cell hypertrophy with actin cytoskeletal reorganization through activation of PKCα and PKCε isoforms and RhoA via PTX-insensitive G proteins.


Subject(s)
Actins/metabolism , Myocytes, Cardiac/physiology , Oligopeptides/pharmacology , Protein Kinase C/metabolism , Receptor, PAR-1/metabolism , Thrombin/pharmacology , rhoA GTP-Binding Protein/metabolism , Animals , Cell Size/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Myocytes, Cardiac/cytology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Protein Kinase C-epsilon/genetics , Protein Kinase C-epsilon/metabolism , RNA, Small Interfering , Rats , Receptor, PAR-1/antagonists & inhibitors , Signal Transduction , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors
16.
J Biol Chem ; 284(40): 27315-26, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19654316

ABSTRACT

Pericellular proteolysis by membrane-type 1 matrix metalloproteinase (MT1-MMP) plays a pivotal role in tumor cell invasion. Localization of MT1-MMP at the invasion front of cells, e.g. on lamellipodia and invadopodia, has to be regulated in coordination with reorganization of the actin cytoskeleton. However, little is known about how such invasion-related actin structures are regulated at the sites where MT1-MMP localizes. During analysis of MT1-MMP-associated proteins, we identified a heretofore uncharacterized protein. This protein, which we call p27RF-Rho, enhances activation of RhoA by releasing it from inhibition by p27(kip1) and thereby regulates actin structures. p27(kip1) is a well known cell cycle regulator in the nucleus. In contrast, cytoplasmic p27(kip1) has been demonstrated to bind GDP-RhoA and inhibit GDP-GTP exchange mediated by guanine nucleotide exchange factors. p27RF-Rho binds p27(kip1) and prevents p27(kip1) from binding to RhoA, thereby freeing the latter for activation. Knockdown of p27RF-Rho expression renders cells resistant to RhoA activation stimuli, whereas overexpression of p27RF-Rho sensitizes cells to such stimulation. p27RF-Rho exhibits a punctate distribution in invasive human tumor cell lines. Stimulation of the cells with lysophosphatidic acid induces activation of RhoA and induces the formation of punctate actin structures within foci of p27RF-Rho localization. Some of the punctate actin structures co-localize with MT1-MMP and cortactin. Down-regulation of p27RF-Rho prevents both redistribution of actin into the punctate structures and tumor cell invasion. Thus, p27RF-Rho is a new potential target for cancer therapy development.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Laminin/metabolism , Matrix Metalloproteinase 14/metabolism , Proteoglycans/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Conserved Sequence , Cyclin-Dependent Kinase Inhibitor p27/pharmacology , Drug Combinations , Enzyme Activation , Gene Expression Regulation , Guanosine Triphosphate/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Matrix Metalloproteinase 2/metabolism , Substrate Specificity , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/antagonists & inhibitors
17.
Neurochem Res ; 34(5): 859-66, 2009 May.
Article in English | MEDLINE | ID: mdl-18770030

ABSTRACT

Proteasome inhibition has been observed in many neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Here, the effect of proteasome inhibition on the morphology of cultured rat cortical astrocytes was investigated. Increasing evidence suggests that the function of astrocytes is related closely to its morphology. Lactacystin, a specific inhibitor of the 20S proteasome, can induce astrocytes stellation in a dose dependent manner and reorganize the cytoskeleton of astrocytes. Furthermore, decreased levels of expression of Rho A, total Akt, and Phospho-Akt were found in the process of astrocytes stellation and lysophosphatidic acid, an activator of Rho A, can largely reverse the astrocytes stellation caused by lactacystin. This suggests that proteasome inhibition in astrocytes could stabilize signals of morphological changes that might be processed through Rho and Akt signaling cascade. Our results suggest that proteasome inhibition might function as a factor regulating astrocytes morphology in some pathophysiological conditions.


Subject(s)
Acetylcysteine/analogs & derivatives , Astrocytes/drug effects , Cerebral Cortex/cytology , Proteasome Inhibitors , Acetylcysteine/pharmacology , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Cell Survival/drug effects , Cells, Cultured , Lysophospholipids/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Rats , Signal Transduction , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/physiology
18.
J Cardiovasc Pharmacol ; 51(4): 388-95, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18427282

ABSTRACT

The effect of atorvastatin on adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) expression and cholesterol efflux remains controversial. In an effort to clarify this issue, ABCA1 expression and apolipoprotein AI (apoAI)-mediated cholesterol efflux after atorvastatin treatment were investigated in THP-1 macrophages. Atorvastatin from 2 microM to 40 microM dose-dependently inhibited ABCA1 expression in human monocyte-derived macrophages and phorbol 12-myristate 13-acetate (PMA)-stimulated THP-1 monocytes. ApoAI-mediated cholesterol efflux was reduced in PMA-stimulated THP-1 cells treated with atorvastatin, this effect was abolished with acetylated low-density lipoprotein (LDL) pretreatment. Atorvastatin treatment also dose-dependently reduced liver X receptor alpha (LXRalpha) expression and Rho activation. Rho activation by farnysylpyophosphate (FPP) and lysophosphatidic acid (LPA) did not salvage, but further depressed, the cholesterol efflux and ABCA1 expression in the presence of atorvastatin. Without atorvastatin, Rho activation by mevalonate, FPP, and LPA diminished apoAI-mediated cholesterol efflux, and Rho activation by GTPgammaS also decreased ABCA1 messenger ribonucleic acid (mRNA) by 16%. Furthermore, Rho inhibition by C3 exoenzyme increased ABCA1 mRNA by 48% despite a 17% decrease in apoAI-mediated cholesterol efflux. LXRalpha agonists (T01901317 and 22(R)-hydroxycholesterol) prevented any reductions in cholesterol efflux or ABCA1 expression associated with atorvastatin treatment. Furthermore, Western blot analysis demonstrated the reciprocal inhibition of Rho and LXRalpha. In conclusion, atorvastatin decreases ABCA1 expression in noncholesterol-loaded macrophages in an LXRalpha- but not Rho-dependent pathway; this effect can be compromised after acetylated LDL cholesterol loading.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism , DNA-Binding Proteins/physiology , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Macrophages/drug effects , Pyrroles/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Atorvastatin , Cell Line , Cholesterol/biosynthesis , DNA-Binding Proteins/agonists , Dose-Response Relationship, Drug , Foam Cells/drug effects , Foam Cells/metabolism , Gene Expression Regulation , Humans , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophage Activation , Macrophages/metabolism , Orphan Nuclear Receptors , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Tetradecanoylphorbol Acetate/pharmacology , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/metabolism
19.
Glia ; 55(14): 1498-507, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17705198

ABSTRACT

The mature peripheral nervous system (PNS) generally shows better regeneration of injured axons as opposed to the central nervous system (CNS). However, complete functional recovery is rarely achieved even in the PNS although morphologically good axonal regeneration often occurs. This mainly results from aberrant reinnervation due to extensive branching of cut axons with consequent failure of synchronized movements of the muscles. Myelin-associated glycoprotein (MAG), a well-characterized molecule existing both in the CNS and PNS myelin, is considered to be a potent inhibitor of axonal regeneration especially in the CNS. In the present study, we investigated whether MAG has any effects not only on axonal elongation, but also on axonal branching. We show herein that MAG minimized branching of the peripheral axons both in vitro and in vivo via activation of RhoA. Furthermore, after sciatic nerve transection in rats, focal and temporary application of MAG to the lesion dramatically enhanced the functional recovery. Using double retrograde labeling and preoperative/postoperative labeling of spinal neurons, reduced hyperinnervation and improved accuracy of target reinnervation was confirmed, respectively. In conclusion, as MAG significantly improves the quality of axonal regeneration, it can be used as a new therapeutic approach for peripheral nerve repair with possible focal and temporary application.


Subject(s)
Axons/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration/physiology , Recovery of Function/physiology , Sciatic Nerve/metabolism , Sciatic Neuropathy/metabolism , Animals , Axons/drug effects , Axons/ultrastructure , Biomarkers/analysis , Biomarkers/metabolism , Cells, Cultured , Denervation , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Growth Cones/drug effects , Growth Cones/metabolism , Growth Cones/ultrastructure , Mice , Mice, Transgenic , Myelin-Associated Glycoprotein/pharmacology , Myelin-Associated Glycoprotein/therapeutic use , Nerve Regeneration/drug effects , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Sciatic Nerve/drug effects , Sciatic Nerve/physiopathology , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/physiopathology , Treatment Outcome , rhoA GTP-Binding Protein/agonists , rhoA GTP-Binding Protein/metabolism
20.
Exp Cell Res ; 312(8): 1361-70, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16448650

ABSTRACT

In chick embryo fibroblasts, the mRNA for extracellular matrix protein tenascin-C is induced 2-fold by cyclic strain (10%, 0.3 Hz, 6 h). This response is attenuated by inhibiting Rho-dependent kinase (ROCK). The RhoA/ROCK signaling pathway is primarily involved in actin dynamics. Here, we demonstrate its crucial importance in regulating tenascin-C expression. Cyclic strain stimulated RhoA activation and induced fibroblast contraction. Chemical activators of RhoA synergistically enhanced the effects of cyclic strain on cell contractility. Interestingly, tenascin-C mRNA levels perfectly matched the extent of RhoA/ROCK-mediated actin contraction. First, RhoA activation by thrombin, lysophosphatidic acid, or colchicine induced tenascin-C mRNA to a similar extent as strain. Second, RhoA activating drugs in combination with cyclic strain caused a super-induction (4- to 5-fold) of tenascin-C mRNA, which was again suppressed by ROCK inhibition. Third, disruption of the actin cytoskeleton with latrunculin A abolished induction of tenascin-C mRNA by chemical RhoA activators in combination with cyclic strain. Lastly, we found that myosin II activity is required for tenascin-C induction by cyclic strain. We conclude that RhoA/ROCK-controlled actin contractility has a mechanosensory function in fibroblasts that correlates directly with tenascin-C gene expression. Previous RhoA/ROCK activation, either by chemical or mechanical signals, might render fibroblasts more sensitive to external tensile stress, e.g., during wound healing.


Subject(s)
Actins/metabolism , Connective Tissue/metabolism , Fibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Tenascin/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Chick Embryo , Connective Tissue/ultrastructure , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fibroblasts/ultrastructure , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/physiology , Mice , Microtubules/drug effects , Microtubules/metabolism , Myosin Type II/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Stress, Mechanical , Tenascin/genetics , Tensile Strength/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Wound Healing/drug effects , Wound Healing/physiology , rho-Associated Kinases , rhoA GTP-Binding Protein/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...