Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.178
Filtrer
1.
J Cell Sci ; 137(18)2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39258310

RÉSUMÉ

Desmosomes play a crucial role in maintaining tissue barrier integrity, particularly in mechanically stressed tissues. The assembly of desmosomes is regulated by the cytoskeleton and its regulators, and desmosomes also function as a central hub for regulating F-actin. However, the specific mechanisms underlying the crosstalk between desmosomes and F-actin remain unclear. Here, we identified that ARHGAP32, a Rho GTPase-activating protein, is located in desmosomes through its interaction with desmoplakin (DSP) via its GAB2-interacting domain (GAB2-ID). We confirmed that ARHGAP32 is required for desmosomal organization, maturation and length regulation. Notably, loss of ARHGAP32 increased formation of F-actin stress fibers and phosphorylation of the regulatory myosin light chain Myl9 at T18/S19. Inhibition of ROCK activity in ARHGAP32-knockout (KO) cells effectively restored desmosomal organization and the integrity of epithelial cell sheets. Moreover, loss of DSP impaired desmosomal ARHGAP32 location and led to decreased actomyosin contractility. ARHGAP32 with a deletion of the GAB2-ID domain showed enhanced association with RhoA in the cytosol and failed to rescue the desmosomal organization in ARHGAP32-KO cells. Collectively, our study unveils that ARHGAP32 associates with and regulates desmosomes by interacting with DSP. This interaction potentially facilitates the crosstalk between desmosomes and F-actin.


Sujet(s)
Desmoplakines , Desmosomes , Protéines d'activation de la GTPase , Desmosomes/métabolisme , Humains , Protéines d'activation de la GTPase/métabolisme , Protéines d'activation de la GTPase/génétique , Desmoplakines/métabolisme , Desmoplakines/génétique , Animaux , Actines/métabolisme , Liaison aux protéines , Protéine G RhoA/métabolisme , Chiens , Phosphorylation , Cellules rénales canines Madin-Darby , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Chaînes légères de myosine/métabolisme , Chaînes légères de myosine/génétique
2.
Sci Rep ; 14(1): 22539, 2024 09 29.
Article de Anglais | MEDLINE | ID: mdl-39341923

RÉSUMÉ

Cerebral vasospasm (CVS) is an important contributor to delayed cerebral ischemia following aneurysmal subarachnoid hemorrhage (aSAH), leading to high morbidity and long-term disability. While several microRNAs (miRNAs) have been implicated in vasospasm, the underlying mechanisms for CVS remain poorly understood. Our study aims to identify miRNAs that may contribute to the development of CVS. Whole-blood samples were obtained during or outside of vasospasm from aSAH patients whose maximal vasospasm was moderate or severe. MiRNAs were isolated from serial whole-blood samples, and miRNA sequencing was performed. Differentially expressed miRNAs were identified and the expression levels in patients' samples were verified using real-time qPCR. The biological functions of identified miRNA were evaluated in human brain endothelial cells (HBECs). MiRNA profiling revealed significant upregulation of miR-148b-3p in patients during CVS. We demonstrated that miR-148b-3p directly targeted and decreased the expression of ROCK1, affecting cell proliferation, migration, and invasion of HBECs through the ROCK-LIMK-Cofilin pathway. We propose that the upregulation of miRNA-148b-3p plays a role in the development of CVS by regulating actin cytoskeletal dynamics in HBECs, which is crucial for vascular function. Our study highlights miR-148b-3p as a potential diagnostic marker as well as therapeutic target for CVS following aSAH.


Sujet(s)
Cellules endothéliales , Analyse de profil d'expression de gènes , microARN , Hémorragie meningée , Vasospasme intracrânien , rho-Associated Kinases , microARN/génétique , microARN/métabolisme , Humains , Hémorragie meningée/génétique , Hémorragie meningée/complications , Hémorragie meningée/métabolisme , Vasospasme intracrânien/génétique , Vasospasme intracrânien/métabolisme , Vasospasme intracrânien/étiologie , Adulte d'âge moyen , Femelle , Mâle , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Cellules endothéliales/métabolisme , Prolifération cellulaire , Mouvement cellulaire/génétique , Sujet âgé , Adulte , Régulation de l'expression des gènes
3.
Mol Biol Rep ; 51(1): 950, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39222158

RÉSUMÉ

BACKGROUND: Hepatic fibrosis, a prevalent chronic liver condition, involves excessive extracellular matrix production associated with aberrant wound healing. Hepatic stellate cells (HSCs) play a pivotal role in liver fibrosis, activated by inflammatory factors such as sphingosine 1-phosphate (S1P). Despite S1P's involvement in fibrosis, its specific role and downstream pathway in HSCs remain controversial. METHODS: In this study, we investigated the regulatory role of S1P/S1P receptor (S1PR) in Hippo-YAP activation in both LX-2 cell lines and primary HSCs. Real-time PCR, western blot, pharmacological inhibitors, siRNAs, and Rho activity assays were adopted to address the molecular mechanisms of S1P mediated YAP activation. RESULTS: Serum and exogenous S1P significantly increased the expression of YAP target genes in HSCs. Pharmacologic inhibitors and siRNA-mediated knockdowns of S1P receptors showed S1P receptor 2 (S1PR2) as the primary mediator for S1P-induced CTGF expression in HSCs. Results using siRNA-mediated knockdown, Verteporfin, and Phospho-Tag immunoblots showed that S1P-S1PR2 signaling effectively suppressed the Hippo kinases cascade, thereby activating YAP. Furthermore, S1P increased RhoA activities in cells and ROCK inhibitors effectively blocked CTGF induction. Cytoskeletal-perturbing reagents were shown to greatly modulate CTGF induction, suggesting the important role of actin cytoskeleton in S1P-induced YAP activation. Exogeneous S1P treatment was enough to increase the expression of COL1A1 and α-SMA, that were blocked by YAP specific inhibitor. CONCLUSIONS: Our data demonstrate that S1P/S1PR2-Src-RhoA-ROCK axis leads to Hippo-YAP activation, resulting in the up-regulation of CTGF, COL1A1 and α-SMA expression in HSCs. Therefore, S1PR2 may represent a potential therapeutic target for hepatic fibrosis.


Sujet(s)
Facteur de croissance du tissu conjonctif , Cellules étoilées du foie , Lysophospholipides , Transduction du signal , Sphingosine , Facteurs de transcription , Protéines de signalisation YAP , rho-Associated Kinases , Protéine G RhoA , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Facteur de croissance du tissu conjonctif/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Lysophospholipides/métabolisme , Lysophospholipides/pharmacologie , Humains , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Sphingosine/analogues et dérivés , Sphingosine/métabolisme , Protéines de signalisation YAP/métabolisme , Protéine G RhoA/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Récepteurs de la sphingosine-1-phosphate/métabolisme , Récepteurs de la sphingosine-1-phosphate/génétique , Lignée cellulaire , Cirrhose du foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/anatomopathologie , src-Family kinases/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Récepteurs aux lysosphingolipides/métabolisme , Récepteurs aux lysosphingolipides/génétique , Collagène de type I/métabolisme , Collagène de type I/génétique , Voie de signalisation Hippo
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(9): 974-981, 2024.
Article de Chinois | MEDLINE | ID: mdl-39267514

RÉSUMÉ

OBJECTIVES: To investigate the effects and molecular mechanisms of inhibition of the Ras homolog gene (Rho)/Rho-associated coiled-coil forming protein kinase (ROCK) pathway on the proliferation and migration of airway smooth muscle cells involving myocardin (MYOCD). METHODS: Human airway smooth muscle cells were infected with the adenoviral vector Ad-ZsGreen-shRNA-hROCK1 in vitro. The cells were randomly divided into four groups: ROCK1 gene silencing control (shNC) group, shNC + arachidonic acid (AA, Rho/ROCK pathway activator) group, ROCK1 gene silencing (shROCK1) group, and shROCK1 + AA group (n=3 each). Quantitative real-time polymerase chain reaction and Western blot were used to detect the expression levels of ROCK1 and MYOCD mRNA and protein. ELISA was employed to measure the levels of globular actin and filamentous actin, while immunofluorescent staining and scratch assays were utilized to assess cell proliferation and migration. RESULTS: Compared to the shNC + AA group, the shROCK1 + AA group exhibited decreased levels of ROCK1 and MYOCD mRNA and protein expression, reduced expression levels of globular actin and filamentous actin, and diminished cell proliferation and migration capabilities (P<0.05). CONCLUSIONS: Inhibition of the Rho/ROCK pathway suppresses the proliferation and migration of airway smooth muscle cells, which may be associated with the downregulation of MYOCD.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Myocytes du muscle lisse , Transduction du signal , Transactivateurs , rho-Associated Kinases , rho-Associated Kinases/métabolisme , rho-Associated Kinases/physiologie , rho-Associated Kinases/génétique , Humains , Myocytes du muscle lisse/physiologie , Myocytes du muscle lisse/métabolisme , Cellules cultivées , Transactivateurs/génétique , Transactivateurs/physiologie , Transactivateurs/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/physiologie , Protéines nucléaires/métabolisme , Protéines G rho/physiologie , Protéines G rho/génétique , Protéines G rho/métabolisme
5.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273383

RÉSUMÉ

Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.


Sujet(s)
Tumeurs , Myosine non-musculaire de type IIA , Humains , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Myosine non-musculaire de type IIA/métabolisme , Myosine non-musculaire de type IIA/génétique , Animaux , Chaînes lourdes de myosine/métabolisme , Chaînes lourdes de myosine/génétique , Transduction du signal , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique
6.
Ren Fail ; 46(2): 2396455, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39229866

RÉSUMÉ

Renal fibrosis is a long-term and progressively worsening condition that impacts kidney function during aging and in the context of chronic kidney disease (CKD). CKD and renal fibrosis affect approximately 10% of the global population and are prevalent in about half of individuals over the age of 70. Despite ongoing research, the mechanisms underlying renal fibrosis are still not well understood, and there is currently a lack of effective treatments available. In the present study, we demonstrated a significant increase of circPWWP2A in renal tubular cells both in vivo and in vitro models of renal fibrosis. Suppressing circPWWP2A has the potential to reduce mitochondrial dysfunction and the production of mitochondrial reactive oxygen species (mtROS), ultimately leading to the inhibition of renal fibrosis. Whereas, supplementation of circPWWP2A led to more serve mitochondrial dysfunction, mtROS production and renal fibrosis. Mechanistically, we found the expression of circPWWP2A was negatively correlated with the expression of miR-182. And we further confirmed miR-182 was the direct target of circPWWP2A by dual-luciferase reporter assay and RIP assay. Then, we found miR-182 suppressed the expression of ROCK1 in both in vitro and in vivo models of renal fibrosis. Luciferase microRNA target reporter assay further indicated ROCK1 as a direct target of miR-182. Knockdown of ROCK1 inhibits renal fibrosis and mitochondrial dysfunction, suggesting ROCK1 not only served as an injurious role in mitochondrial homeostasis but also a pro-fibrotic factor in CKD. Taking together, our findings suggest that circPWWP2A may promote renal interstitial fibrosis by modulating miR-182/ROCK1-mediated mitochondrial dysfunction.


Sujet(s)
Fibrose , Rein , microARN , ARN circulaire , Insuffisance rénale chronique , rho-Associated Kinases , microARN/métabolisme , microARN/génétique , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Animaux , ARN circulaire/génétique , ARN circulaire/métabolisme , Souris , Rein/anatomopathologie , Rein/métabolisme , Mâle , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Humains , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL
7.
Sci Signal ; 17(853): eado9852, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39255336

RÉSUMÉ

Structural plasticity of dendritic spines in the nucleus accumbens (NAc) is crucial for learning from aversive experiences. Activation of NMDA receptors (NMDARs) stimulates Ca2+-dependent signaling that leads to changes in the actin cytoskeleton, mediated by the Rho family of GTPases, resulting in postsynaptic remodeling essential for learning. We investigated how phosphorylation events downstream of NMDAR activation drive the changes in synaptic morphology that underlie aversive learning. Large-scale phosphoproteomic analyses of protein kinase targets in mouse striatal/accumbal slices revealed that NMDAR activation resulted in the phosphorylation of 194 proteins, including RhoA regulators such as ARHGEF2 and ARHGAP21. Phosphorylation of ARHGEF2 by the Ca2+-dependent protein kinase CaMKII enhanced its RhoGEF activity, thereby activating RhoA and its downstream effector Rho-associated kinase (ROCK/Rho-kinase). Further phosphoproteomic analysis identified 221 ROCK targets, including the postsynaptic scaffolding protein SHANK3, which is crucial for its interaction with NMDARs and other postsynaptic scaffolding proteins. ROCK-mediated phosphorylation of SHANK3 in the NAc was essential for spine growth and aversive learning. These findings demonstrate that NMDAR activation initiates a phosphorylation cascade crucial for learning and memory.


Sujet(s)
Protéines de tissu nerveux , Plasticité neuronale , Protéome , Récepteurs du N-méthyl-D-aspartate , Animaux , Récepteurs du N-méthyl-D-aspartate/métabolisme , Plasticité neuronale/physiologie , Souris , Phosphorylation , Protéome/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Mâle , Transduction du signal , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Souris de lignée C57BL , Phosphoprotéines/métabolisme , Phosphoprotéines/génétique , Apprentissage/physiologie , Apprentissage par évitement/physiologie , Rho guanine nucleotide exchange factors/métabolisme , Rho guanine nucleotide exchange factors/génétique , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Synapses/métabolisme , Protéine G RhoA/métabolisme , Épines dendritiques/métabolisme
8.
Elife ; 122024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39298260

RÉSUMÉ

Arpin was discovered as an inhibitor of the Arp2/3 complex localized at the lamellipodial tip of fibroblasts, where it regulated migration steering. Recently, we showed that arpin stabilizes the epithelial barrier in an Arp2/3-dependent manner. However, the expression and functions of arpin in endothelial cells (EC) have not yet been described. Arpin mRNA and protein are expressed in EC and downregulated by pro-inflammatory cytokines. Arpin depletion in Human Umbilical Vein Endothelial Cells causes the formation of actomyosin stress fibers leading to increased permeability in an Arp2/3-independent manner. Instead, inhibitors of ROCK1 and ZIPK, kinases involved in the generation of stress fibers, normalize the loss-of-arpin effects on actin filaments and permeability. Arpin-deficient mice are viable but show a characteristic vascular phenotype in the lung including edema, microhemorrhage, and vascular congestion, increased F-actin levels, and vascular permeability. Our data show that, apart from being an Arp2/3 inhibitor, arpin is also a regulator of actomyosin contractility and endothelial barrier integrity.


Sujet(s)
Actomyosine , Perméabilité capillaire , Cellules endothéliales de la veine ombilicale humaine , Animaux , Humains , Actomyosine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Souris , Serpines/métabolisme , Serpines/génétique , Souris knockout , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Complexe Arp-2-3/métabolisme , Complexe Arp-2-3/génétique , Fibres de stress/métabolisme , Cellules endothéliales/métabolisme , Protéines de transport
9.
J Cancer Res Clin Oncol ; 150(9): 424, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39297944

RÉSUMÉ

PURPOSE: This study aimed to investigate the effect of MYO3B on endometrial cancer (EC) proliferation and invasion. METHODS: The expression of MYO3B in EC tissues and cells was analyzed using TCGA database, immunohistochemical staining, real-time PCR, and western blot (WB). Cell proliferation was detected by CCK8, Annexin V-APC/PI flow cytometry was used to detect apoptosis, intracellular calcium ion (Ca2+) was detected by flow cytometry with Fluo-4 AM fluorescent probe, cell migration by scratch assay, and cell invasion by Transwell assay, and the expression of proteins related to Ca2+ homeostasis and RhoA/ROCK1 signaling pathway was detected by WB and immunofluorescence staining. RESULTS: The expression of MYO3B was an influential factor in EC recurrence, and the expression of MYO3B was significantly up-regulated in EC tissues and cells, but down-regulated in KLE cells, and MYO3B knockdown inhibited the proliferation, migration, and invasion ability of EC cells and promoted apoptosis, suggesting that MYO3B plays a tumor-promoting role in EC. Furthermore, MYO3B knockdown decreased Ca2+ concentration in EC cells and the RhoA/ROCK1 signaling pathway was inhibited, and the effect of MYO3B knockdown on RhoA/ROCK1 signaling was reversed by treatment with the Calmodulin agonist CALP-2, and the effects of MYO3B knockdown on cell proliferation, migration, and invasion were reversed after treatment with the RhoA agonist U-46,619. CONCLUSION: MYO3B promotes the proliferation and migration of endometrial cancer cells via Ca2+-RhoA/ROCK1 signaling pathway. High expression of MYO3B may be a biomarker for EC metastasis.


Sujet(s)
Calcium , Prolifération cellulaire , Évolution de la maladie , Tumeurs de l'endomètre , Transduction du signal , rho-Associated Kinases , Protéine G RhoA , Humains , Femelle , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/génétique , Protéine G RhoA/métabolisme , Calcium/métabolisme , Mouvement cellulaire , Apoptose , Lignée cellulaire tumorale , Invasion tumorale
10.
Gen Physiol Biophys ; 43(5): 399-409, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39140680

RÉSUMÉ

Acute lung injury (ALI) is a significant health condition with notable rates of morbidity and mortality globally. Long non-coding ribose nucleic acids (lncRNAs) play vital roles in mitigating various inflammation-related diseases, including ALI. The study aimed to investigate the functional role and molecular mechanisms of lncRNA SNHG1 on ALI in lipopolysaccharide (LPS)-treated A549 cells and in LPS-induced ALI mice. The expression of SNHG1 was initially examined in LPS-treated A549 cells. We further demonstrated the critical function of SNHG1 through various cellular assessments following SNHG1 knockdown, including cell counting kit (CCK)-8 assay, flow cytometry analysis, as well as enzyme-linked immunosorbent assay (ELISA). Reducing SNHG1 levels hindered the negative effects of LPS on cell viability, apoptosis, and inflammation. Moreover, SNHG1 acted as a negative regulator for miR-199a-3p, which targeted downstream ROCK2. Depletion of miR-199a-3p or enhanced expression of ROCK2 abolished the protective effects of SNHG1 knockdown on LPS-induced apoptosis and inflammation. Consistently, silencing SNHG1 alleviated LPS-induced lung injury in mice, demonstrating its potential therapeutic benefits in managing ALI. Overall, this study sheds light on the role of SNHG1 in modulating inflammation and apoptosis in ALI through the miR-199a-3p/ROCK2 pathway, offering new insights for the treatment of this condition.


Sujet(s)
Lésion pulmonaire aigüe , Techniques de knock-down de gènes , Lipopolysaccharides , microARN , ARN long non codant , rho-Associated Kinases , ARN long non codant/génétique , ARN long non codant/métabolisme , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/génétique , microARN/métabolisme , microARN/génétique , Animaux , Souris , Humains , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Mâle , Cellules A549 , Transduction du signal , Souris de lignée C57BL , Apoptose/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE