Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.651
Filtrar
1.
Biochem Pharmacol ; 222: 116049, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38342347

RESUMO

We previously showed that digitoxin inhibits angiogenesis and cancer cell proliferation and migration and these effects were associated to protein tyrosine kinase 2 (FAK) inhibition. Considering the interactions between FAK and Rho GTPases regulating cell cytoskeleton and movement, we investigated the involvement of RhoA and Rac1 in the antiangiogenic effect of digitoxin. Phalloidin staining of human umbilical vein endothelial cells (HUVECs) showed the formation of stress fibers in cells treated with 10 nM digitoxin. By Rhotekin- and Pak1- pull down assays, detecting the GTP-bound form of GTPases, we observed that digitoxin (10-25 nM) induced sustained (0.5-6 h) RhoA activation with no effect on Rac1. Furthermore, inhibition of HUVEC migration and capillary-like tube formation by digitoxin was counteracted by hindering RhoA-ROCK axis with RhoA silencing or Y-27632 treatment. Digitoxin did not decrease p190RhoGAP phosphorylation at Tyr1105 (a site targeted by FAK), suggesting that RhoA activation was independent from FAK inhibition. Because increasing evidence points to a redox regulation of RhoA, we measured intracellular ROS and found that digitoxin treatment enhanced ROS levels in a concentration-dependent manner (1-25 nM). Notably, the flavoprotein inhibitor DPI or the pan-NADPH oxidase (NOX) inhibitor VAS-2870 antagonized both ROS increase and RhoA activation by digitoxin. Our results provide evidence that inhibition of HUVEC migration and tube formation by digitoxin is dependent on ROS production by endothelial NOX, which leads to the activation of RhoA/ROCK pathway. Digitoxin effects on proteins regulating cytoskeletal organization and cell motility could have a wider impact on cancer progression, beyond the antiangiogenic activity.


Assuntos
Digitoxina , NADPH Oxidases , Humanos , Espécies Reativas de Oxigênio/metabolismo , Digitoxina/farmacologia , Células Endoteliais da Veia Umbilical Humana , Quinase 1 de Adesão Focal/metabolismo , Fosforilação , Movimento Celular , NADPH Oxidases/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
2.
Curr Mol Pharmacol ; 17: e18761429274883, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38389417

RESUMO

Guanine nucleotide exchange factor H1 (GEF-H1) is a unique protein modulated by the GDP/GTP exchange. As a regulator of the Rho-GTPase family, GEF-H1 can be activated through a microtubule-depended mechanism and phosphorylation regulation, enabling it to perform various pivotal biological functions across multiple cellular activities. These include the regulation of Rho-GTPase, cytoskeleton formation, cellular barrier, cell cycle, mitosis, cell differentiation, and vesicle trafficking. Recent studies have revealed its crucial effect on the tumor microenvironment (TME) components, promoting tumor initiation and progress. Consequently, an in-depth exploration of GEF-H1's biological roles and association with tumors holds promise for its potential as a valuable molecular target in tumor treatment.


Assuntos
Neoplasias , Proteína rhoA de Ligação ao GTP , Humanos , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Microtúbulos/metabolismo , Proteínas , Neoplasias/metabolismo , Microambiente Tumoral
3.
Discov Med ; 36(180): 190-198, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38273759

RESUMO

BACKGROUND: Colorectal cancer (CRC) is a common malignancy with high morbidity and mortality. To improve CMC prognosis, research must identify safe and effective natural drugs that improve the proliferation, migration, and epithelial mesenchymal transition (EMT) processes of CRC. The purpose of this paper is to understand how cichoric acid (CA) impacts CRC proliferation, metastasis, and EMT of CRC by adjusting the Ras homolog family member A (RhoA)/RHO-associated coiled coil protein kinase (ROCK) pathway. METHODS: Human Colon Cancer Cells (HCT116) cells were randomly divided into Control (blank medium treatment), low concentration CA (CA-L), medium concentration CA (CA-M), high concentration CA (CA-H), and high-concentration CA+RhoA activator U46619 (CA-H+U46619) groups. Cell proliferation, migration and invasion, and apoptosis were evaluated with cell counting kit-8 (CCK-8) assay, transwell assay, and flow cytometry, respectively. The expression of RhoA, ROCK, and EMT-associated proteins were detected by Western Blot. The CRC transplanted tumor model of nude mice was constructed, and the mice were grouped into low-dose CA (CA-Low, 15 mg/kg CA), high-dose CA (CA-High, 30 mg/kg CA), high-dose CA+RhoA activator U46619 (CA-High+U46619, 30 mg/kg CA+10 mM U46619), and Model groups at random, with 12 mice in each group. Tumor volume, mass, and inhibition rate were measured and calculated, and the pathological changes of tumor in nude mice were detected by hematoxylin-eosin (HE) staining. RESULTS: Compared with Control, the optical density of cells at 450 nm (OD450) value (48 h, 72 h), cell migration number, cell invasion number, RhoA, ROCK1, N-cadherin, vimentin protein expression levels of HCT116 cells were reduced in CA-M and CA-H groups; however, E-cadherin level and apoptosis rate were increased (p < 0.05). In the CA-High group, we observed a significant decrease (p < 0.05) in both tumor volume and mass in nude mice. Additionally, the tumor tissue cells exhibited better organization, reduced size, reduced tumor and vascular tissue hyperplasia, and decreased infiltration of inflammatory cells. U46619 decreased the retardation of CA on the proliferation, EMT, and migration of CRC tumor cells as well as the growth of transplanted CRC tumors in nude mice. CONCLUSIONS: CA may reduce CRC migration, proliferation, and EMT by inhibiting the activation of the RhoA/ROCK signaling pathway.


Assuntos
Ácidos Cafeicos , Neoplasias Colorretais , Succinatos , Proteína rhoA de Ligação ao GTP , Humanos , Animais , Camundongos , Camundongos Nus , Linhagem Celular Tumoral , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/farmacologia , Proteína rhoA de Ligação ao GTP/uso terapêutico , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/uso terapêutico , Transdução de Sinais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Proliferação de Células , Movimento Celular , Quinases Associadas a rho/metabolismo
4.
Phytomedicine ; 124: 155268, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38176265

RESUMO

BACKGROUND: Obesity has emerged as a global epidemic. Recent research has indicated that diet-induced obesity can be prevented by promoting lacteal junction zippering. Berberine, which is derived from natural plants, is found to be promising in weight reduction, but the underlying mechanism remains unspecified. PURPOSE: To determine whether berberine protects against obesity by regulating the lacteal junction and to explore potential molecular mechanisms. METHODS: Following the induction of the diet-induced obese (DIO) model, mice were administered low and high doses of berberine for 4 weeks. Indicators associated with insulin resistance and lipid metabolism were examined. Various methods, such as Oil Red O staining, transmission electron microscopy imaging, confocal imaging and others were used to observe the effects of berberine on lipid absorption and the lacteal junction. In vitro, human dermal lymphatic endothelial cells (HDLECs) were used to investigate the effect of berberine on LEC junctions. Western Blot and immunostaining were applied to determine the expression levels of relevant molecules. RESULTS: Both low and high doses of berberine reduced body weight in DIO mice without appetite suppression and ameliorated glucolipid metabolism disorders. We also found that the weight loss effect of berberine might contribute to the inhibition of small intestinal lipid absorption. The possible mechanism was related to the promotion of lacteal junction zippering via suppressing the ras homolog gene family member A (RhoA)/Rho-associated kinase (ROCK) signaling pathway. In vitro, berberine also promoted the formation of stable mature junctions in HDLECs, involving the same signaling pathway. CONCLUSION: Berberine could promote lacteal junction zippering and ameliorate diet-induced obesity through the RhoA/ROCK signaling pathway.


Assuntos
Berberina , Camundongos , Humanos , Animais , Berberina/farmacologia , Células Endoteliais/metabolismo , Transdução de Sinais , Obesidade/tratamento farmacológico , Dieta , Lipídeos , Proteína rhoA de Ligação ao GTP/metabolismo
5.
Cell Signal ; 114: 110978, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37972801

RESUMO

Spinal cord injury (SCI) is a devastating disorder and a leading cause of disability in adults worldwide. Multiple studies have reported the upregulation of programmed cell death 1 (PD-1) following SCI. However, the underlying mechanism of PD-1 deficiency in SCI is not well established. Therefore, we aimed to investigate the role and potential mechanism of PD-1 in SCI pathogenesis. PD-1 Knockout (KO) SCI mouse model was established, and PD-1 expression was evaluated in tissue samples by western blot assay. We then used a series of function gain-and-loss assays to determine the role of PD-1 in SCI pathogenesis. Moreover, mechanistic assays were performed to explore the association between PD-1, neuron-glia antigen-2 (NG2) glia cells, and miR-23b-5p and then investigated the involved signaling pathway. Results illustrated that PD-1 deficiency enhanced the inflammatory response, neuron loss, and functional impairment induced by SCI. We found that NG2 glia depletion aggravated inflammation, reduced neural survival, and suppressed locomotor recovery in murine SCI model. Further analysis indicated that NG2+ cells were increased in the spinal cord of SCI mice, and PD-1 deficiency increased the number of NG2+ cells by activating the Nogo receptor/ras homolog family member A/Rho kinase (NgR/RhoA/ROCK) signaling. Mechanistically, miR-23b-5p was identified as the negative regulator of PD-1 in NG2 glia. MiR-23b-5p deficiency reduced the expression of inflammatory cytokines, enhanced neural survival, and promoted locomotor recovery in SCI mice, which was counteracted by PD-1 deficiency. In conclusion, PD-1 deficiency exacerbates SCI in vivo by regulating reprogramming of NG2 glia and activating the NgR/RhoA/ROCK signaling.


Assuntos
MicroRNAs , Receptor de Morte Celular Programada 1 , Traumatismos da Medula Espinal , Animais , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Neuroglia/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Transdução de Sinais , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Receptores Nogo/metabolismo , Quinases Associadas a rho/metabolismo
6.
Biophys J ; 123(1): 57-67, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-37978802

RESUMO

Rho-specific guanine nucleotide dissociation inhibitors (RhoGDIs) play a crucial role in the regulation of Rho family GTPases. They act as negative regulators that prevent the activation of Rho GTPases by forming complexes with the inactive GDP-bound state of GTPase. Release of Rho GTPase from the RhoGDI-bound complex is necessary for Rho GTPase activation. Biochemical studies provide evidence of a "phosphorylation code," where phosphorylation of some specific residues of RhoGDI selectively releases its GTPase partner (RhoA, Rac1, Cdc42, etc.). This work attempts to understand the molecular mechanism behind this specific phosphorylation-induced reduction in binding affinity. Using several microseconds long atomistic molecular dynamics simulations of the wild-type and phosphorylated states of the RhoA-RhoGDI complex, we propose a molecular-interaction-based mechanistic model for the dissociation of the complex. Phosphorylation induces major structural changes, particularly in the positively charged polybasic region (PBR) of RhoA and the negatively charged N-terminal region of RhoGDI that contribute most to the binding affinity. Molecular mechanics Poisson-Boltzmann surface area binding energy calculations show a significant weakening of interaction on phosphorylation at the RhoA-specific site of RhoGDI. In contrast, phosphorylation at a Rac1-specific site does not affect the overall binding affinity significantly, which confirms the presence of a phosphorylation code. RhoA-specific phosphorylation leads to a reduction in the number of contacts between the PBR of RhoA and the N-terminal region of RhoGDI, which manifests a reduction of the binding affinity. Using hydrogen bond occupancy analysis and energetic perturbation network, we propose a mechanistic model for the allosteric response, i.e., long-range signal propagation from the site of phosphorylation to the PBR and buried geranylgeranyl group in the form of rearrangement and rewiring of hydrogen bonds and salt bridges. Our results highlight the crucial role of specific electrostatic interactions in manifestation of the phosphorylation code.


Assuntos
Inibidores de Dissociação do Nucleotídeo Guanina , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho , Inibidores da Dissociação do Nucleotídeo Guanina rho-Específico/metabolismo , Fosforilação , Inibidores de Dissociação do Nucleotídeo Guanina/química , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Inibidor alfa de Dissociação do Nucleotídeo Guanina rho/metabolismo , Ligação Proteica , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
7.
Cell Biochem Biophys ; 82(1): 127-137, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37801199

RESUMO

LAMB3, a major extracellular matrix and basal membrane component, is involved in wound healing. We aimed to understand its role in Asherman's syndrome (AS), which is associated with infertility, by using bioinformatics analysis and cultured endometrial stromal cells (ESCs). MRNAs extracted from tissues obtained from control subjects and patients with severe intrauterine adhesion were sequenced and subjected to bioinformatics analysis and the RhoA/ROCK1/MYL9 pathway was implicated and this subsequently studied using cultured primary ESCs. The effects of overexpression and knockdown and activation and inhibition of LAMB3 on the mesenchymal to myofibroblastic phenotypic transformation of ECCs were assessed using PCR and western blot analysis. Phalloidin was used to localize the actin cytoskeletal proteins. Silencing of LAMB3 reversed the TGF-ß-induced ESC myofibroblast phenotype conversion, whereas overexpression of LAMB3 promoted this process. Activation and silencing of LAMB3 led to remodeling of the ESC cytoskeleton. Overexpression and silencing of LAMB3 caused activation and inhibition of ESCs, respectively. Y-27632 and LPA reversed the activation and inhibition of the RhoA/ROCK1/MYL9 pathway after overexpression and silencing, respectively. These results suggest that LAMB3 can regulate ESC fibrosis transformation and cytoskeleton remodeling via the RhoA/ROCK1/MYL9 pathway. This study provides a potential new target for gene therapy and drug intervention of AS.


Assuntos
Citoesqueleto , Quinases Associadas a rho , Humanos , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Actinas/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Cadeias Leves de Miosina/metabolismo
8.
Biomed Pharmacother ; 170: 115983, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38134633

RESUMO

ARHGEF17 encodes the protein RhoGEF17, which is highly expressed in vascular endothelial cells. It is a guanine nucleotide exchange factor (GEF) that accelerates the exchange of GDP with GTP on many small GTPases through its Dbl homology (DH) domain, enabling the activation of Rho-GTPases such as RhoA, RhoB, and RhoC. Rho GTPase-regulated changes in the actin cytoskeleton and cell adhesion kinetics are the main mechanisms mediating many endothelial cell (EC) alterations, including cell morphology, migration, and division changes, which profoundly affect EC barrier function. This review focuses on ARHGEF17 expression, activation and biological functions in ECs, linking its regulation of cellular morphology, migration, mitosis and other cellular behaviors to disease onset and progression. Understanding ARHGEF17 mechanisms of action will contribute to the design of therapeutic approaches targeting RhoGEF17, a potential drug target for the treatment of various endothelium-related diseases, Such as vascular inflammation, carcinogenesis and transendothelial metastasis of tumors.


Assuntos
Células Endoteliais , Neoplasias , Humanos , Células Endoteliais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Neoplasias/tratamento farmacológico , Endotélio
9.
Mol Biol Cell ; 35(1): br1, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37910204

RESUMO

Fibroblasts migrate discontinuously by generating transient leading-edge protrusions and irregular, abrupt retractions of a narrow trailing edge. In contrast, keratinocytes migrate persistently and directionally via a single, stable, broad protrusion paired with a stable trailing-edge. The Rho GTPases Rac1, Cdc42 and RhoA are key regulators of cell protrusions and retractions. However, how these molecules mediate cell-type specific migration modes is still poorly understood. In fibroblasts, all three Rho proteins are active at the leading edge, suggesting short-range coordination of protrusive Rac1 and Cdc42 signals with RhoA retraction signals. Here, we show that Cdc42 was surprisingly active in the trailing-edge of migrating keratinocytes. Elevated Cdc42 activity colocalized with the effectors MRCK and N-WASP suggesting that Cdc42 controls both myosin activation and actin polymerization in the back. Indeed, Cdc42 was required to maintain the highly dynamic contractile acto-myosin retrograde flow at the trailing edge of keratinocytes, and its depletion induced ectopic protrusions in the back, leading to decreased migration directionality. These findings suggest that Cdc42 is required to stabilize the dynamic cytoskeletal polarization in keratinocytes, to enable persistent, directional migration.


Assuntos
Movimento Celular , Queratinócitos , Proteína cdc42 de Ligação ao GTP , Proteínas rho de Ligação ao GTP , Proteína cdc42 de Ligação ao GTP/metabolismo , Fibroblastos/metabolismo , Queratinócitos/fisiologia , Miosinas/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Humanos
10.
Curr Biol ; 34(2): 245-259.e8, 2024 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-38096821

RESUMO

In animals, cells often move as collectives to shape organs, close wounds, or-in the case of disease-metastasize. To accomplish this, cells need to generate force to propel themselves forward. The motility of singly migrating cells is driven largely by an interplay between Rho GTPase signaling and the actin network. Whether cells migrating as collectives use the same machinery for motility is unclear. Using the zebrafish posterior lateral line primordium as a model for collective cell migration, we find that active RhoA and myosin II cluster on the basal sides of the primordium cells and are required for primordium motility. Positive and negative feedbacks cause RhoA and myosin II activities to pulse. These pulses of RhoA signaling stimulate actin polymerization at the tip of the protrusions and myosin-II-dependent actin flow and protrusion retraction at the base of the protrusions and deform the basement membrane underneath the migrating primordium. This suggests that RhoA-induced actin flow on the basal sides of the cells constitutes the motor that pulls the primordium forward, a scenario that likely underlies collective migration in other contexts.


Assuntos
Actinas , Peixe-Zebra , Animais , Actinas/metabolismo , Peixe-Zebra/metabolismo , Polimerização , Movimento Celular , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas do Citoesqueleto/metabolismo , Miosina Tipo II/metabolismo
11.
Sci Signal ; 16(816): eadk9171, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-38113334

RESUMO

Oncogenic small guanosine triphosphatases (GTPases) are often characterized by a limited set of activating mutations that affect their intrinsic biochemical function, but RHOA-which is frequently mutated in gastric cancer-appears not to have read the instruction manual. Having previously characterized the Y42C RHOA mutation in gastric cancer, in this issue of Science Signaling, Schaefer et al. take on the slightly less common L57V mutation and find that individual RHOA mutations can have different and unpredictable signaling outcomes.


Assuntos
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Mutação , Transdução de Sinais/genética , Mutação com Ganho de Função , GTP Fosfo-Hidrolases/genética , Proteína rhoA de Ligação ao GTP/metabolismo
12.
BMC Biol ; 21(1): 240, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37907898

RESUMO

BACKGROUND: PFTK1/Eip63E is a member of the cyclin-dependent kinases (CDKs) family and plays an important role in normal cell cycle progression. Eip63E expresses primarily in postnatal and adult nervous system in Drosophila melanogaster but its role in CNS development remains unknown. We sought to understand the function of Eip63E in the CNS by studying the fly ventral nerve cord during development. RESULTS: Our results demonstrate that Eip63E regulates axogenesis in neurons and its deficiency leads to neuronal defects. Functional interaction studies performed using the same system identify an interaction between Eip63E and the small GTPase Rho1. Furthermore, deficiency of Eip63E homolog in mice, PFTK1, in a newly generated PFTK1 knockout mice results in increased axonal outgrowth confirming that the developmental defects observed in the fly model are due to defects in axogenesis. Importantly, RhoA phosphorylation and activity are affected by PFTK1 in primary neuronal cultures. We report that GDP-bound inactive RhoA is a substrate of PFTK1 and PFTK1 phosphorylation is required for RhoA activity. CONCLUSIONS: In conclusion, our work establishes an unreported neuronal role of PFTK1 in axon development mediated by phosphorylation and activation of GDP-bound RhoA. The results presented add to our understanding of the role of Cdks in the maintenance of RhoA-mediated axon growth and its impact on CNS development and axonal regeneration.


Assuntos
Quinases Ciclina-Dependentes , Drosophila melanogaster , Animais , Camundongos , Ciclo Celular , Quinases Ciclina-Dependentes/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Neurônios/metabolismo , Fosforilação , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Int J Mol Sci ; 24(21)2023 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-37958606

RESUMO

Schizophrenia is one of the most serious psychiatric disorders and is characterized by reductions in both brain volume and spine density in the frontal cortex. RhoA belongs to the RAS homolog (Rho) family and plays critical roles in neuronal development and structural plasticity via Rho-kinase. RhoA activity is regulated by GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). Several variants in GAPs and GEFs associated with RhoA have been reported to be significantly associated with schizophrenia. Moreover, several mouse models carrying schizophrenia-associated gene variants involved in RhoA/Rho-kinase signaling have been developed. In this review, we summarize clinical evidence showing that variants in genes regulating RhoA activity are associated with schizophrenia. In the last half of the review, we discuss preclinical evidence indicating that RhoA/Rho-kinase is a potential therapeutic target of schizophrenia. In particular, Rho-kinase inhibitors exhibit anti-psychotic-like effects not only in Arhgap10 S490P/NHEJ mice, but also in pharmacologic models of schizophrenia (methamphetamine- and MK-801-treated mice). Accordingly, we propose that Rho-kinase inhibitors may have antipsychotic effects and reduce cognitive deficits in schizophrenia despite the presence or absence of genetic variants in small GTPase signaling pathways.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Esquizofrenia , Humanos , Camundongos , Animais , Quinases Associadas a rho/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Genômica
14.
Biochem Pharmacol ; 218: 115899, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37907138

RESUMO

Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.


Assuntos
Sistema Cardiovascular , Quinases Associadas a rho , Actinas/metabolismo , Sistema Cardiovascular/metabolismo , Proteínas rho de Ligação ao GTP , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Transdução de Sinais/fisiologia , Humanos
15.
Int Immunopharmacol ; 124(Pt B): 110992, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37806106

RESUMO

Sepsis begins with vascular endothelial barrier breakdown and causes widespread organ failure. Protease-activated receptor 1 (PAR1) is an important target for modulating vascular endothelial permeability; however, little research has been undertaken in sepsis, and its putative molecular mechanism remains unknown. The vascular endothelial permeability was examined by detecting FITC-dextran flux. F-actin was examined by immunofluorescence (IF). PAR1, ERM phosphorylation, and RhoA/ROCK signaling pathway expression in lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs) line were examined by IF and Western blot. To develop the sepsis model, cecal ligation and puncture (CLP) were conducted. The PAR1 inhibitor SCH79797 was utilized to inhibit PAR1 expression in vivo. Vascular permeability in main organs weres measured by Evans blue dye extravasation. The pathological changes in main organs were examined by HE staining. The expression of PAR1, ERM phosphorylation, and the RhoA/ROCK signaling pathway was examined using IF, immunohistochemical and WB in CLP mice. In vitro, in response to LPS stimulation of HUVECs, PAR1 mediated the phosphorylation of ERM, promoted F-actin rearrangement, and increased endothelial hyperpermeability, all of which were prevented by inhibiting PAR1 or RhoA. Additionally, inhibiting PAR1 expression reduced RhoA and ROCK expression. In vivo, we showed that inhibiting PAR1 expression will reduce ezrin/radixin/moesin (ERM) phosphorylation to relieve vascular endothelial barrier dysfunction and thereby ameliorate multiorgan dysfunction syndrome (MODS) in CLP-induced septic mice. This study revealed that PAR1-mediated phosphorylation of ERM induced endothelial barrier dysfunction, which in turn led to MODS in sepsis, and that the RhoA/ROCK signaling pathway underlay these effects.


Assuntos
Receptor PAR-1 , Sepse , Humanos , Camundongos , Animais , Receptor PAR-1/metabolismo , Actinas/metabolismo , Fosforilação , Lipopolissacarídeos/farmacologia , Transdução de Sinais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Sepse/metabolismo , Quinases Associadas a rho/metabolismo , Permeabilidade Capilar
16.
JCI Insight ; 8(19)2023 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-37811653

RESUMO

Transglutaminase 2 (TGase2) has been shown to contribute to the mesangial IgA1 deposition in a humanized mouse model of IgA nephropathy (IgAN), but the mechanism is not fully understood. In this study, we found that inhibition of TGase2 activity could dramatically decrease the amount of polymeric IgA1 (pIgA1) isolated from patients with IgAN that interacts with human mesangial cells (HMC). TGase2 was expressed both in the cytosol and on the membrane of HMC. Upon treatment with pIgA1, there were more TGase2 recruited to the membrane. Using a cell model of mesangial deposition of pIgA1, we identified 253 potential TGase2-associated proteins in the cytosolic fraction and observed a higher concentration of cellular vesicles and increased expression of Ras homolog family member A (RhoA) in HMC after pIgA1 stimulation. Both the amount of pIgA1 deposited on HMC and membrane TGase2 level were decreased by inhibition of the vesicle trafficking pathway. Mechanistically, TGase2 was found to be coprecipitated with RhoA in the cellular vesicles. Membrane TGase2 expression was greatly increased by overexpression of RhoA, while it was reduced by knockdown of RhoA. Our in vitro approach demonstrated that TGase2 was transported from the cytosol to the membrane through a RhoA-mediated vesicle-trafficking pathway that can facilitate pIgA1 interaction with mesangium in IgAN.


Assuntos
Glomerulonefrite por IGA , Animais , Camundongos , Humanos , Glomerulonefrite por IGA/metabolismo , Imunoglobulina A/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase , Proteína rhoA de Ligação ao GTP/metabolismo , Mesângio Glomerular/metabolismo , Polímeros
17.
Curr Alzheimer Res ; 20(6): 420-430, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37723951

RESUMO

BACKGROUND: Studies have found synaptic plasticity damage to be an early marker of Alzheimer's disease (AD). RhoA/ROCK pathway is involved in the regulation of synaptic plasticity. Acupuncture can significantly improve the cognitive state of AD. OBJECTIVE: We aimed to use modern biological technology to detect the changes in synaptic plasticity and RhoA/ROCK pathway in SAMP8 mice, as well as the intervention effect of acupuncture. METHODS: Morris water maze and electrophysiological techniques were used in vivo to detect the changes in spatial memory and LTP of mice. Golgi Cox staining and CASEVIEWER2.1 software were used to quantitatively analyze the changes in the morphology and number of dendritic spines in the hippocampus of mice. The activity of RhoA and ROCK2 in the hippocampus of mice was detected, respectively, by pull-down technique and ELISA. WB technique was used to detect the protein expression of ROCK2 and phosphorylation level of MLC2, LIMK2, and CRMP2 in the hippocampus of mice. RESULTS: The neurobehavior and synaptic plasticity of 8-month-old SAMP8 mice were found to be significantly impaired. Acupuncture could improve the spatial learning and memory ability of SAMP8 mice, and partially prevent the reduction in the number of spines on the secondary branches of the apical dendrites in the hippocampus and the attenuation of LTP. The RhoA/ROCK pathway was significantly activated in the hippocampus of 8-month-old SAMP8 mice, and acupuncture had an inhibitory effect on it. CONCLUSION: Acupuncture can improve synaptic plasticity by inhibiting the abnormal activation of the RhoA/ROCK pathway, and improve the spatial learning and memory ability of AD, so as to achieve the purpose of treating AD.


Assuntos
Terapia por Acupuntura , Doença de Alzheimer , Plasticidade Neuronal , Animais , Camundongos , Doença de Alzheimer/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/uso terapêutico , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/uso terapêutico
18.
J Alzheimers Dis ; 95(4): 1643-1656, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37718806

RESUMO

BACKGROUND: RhoA signaling is widely reported to be dysregulated in Alzheimer's disease (AD), but its therapeutic targeting demonstrated mixed outcomes. We hypothesize that the activation and inactivation states of RhoA and LIMK are different in the cortex and in subregions of hippocampus along the rostral-caudal dimensions. OBJECTIVE: We intended to elucidate the plane and spatial dependent RhoA signaling in association with AD. METHODS: We applied antibody pRhoA that recognizes an inactive state of RhoA (S188 phosphorylation) and antibody pLIMK against an active state of LIMK (T508 phosphorylation) to investigate RhoA signaling in wildtype (WT) and triple transgenic AD (3xTg-AD) mouse model. We prepared serial sections from the rostral to caudal coronal planes of the entire mouse brain followed by immunofluorescence staining with pRhoA and pLIMK antibodies. RESULTS: Both pRhoA and pLIMK elicited a shift of expression pattern from rostral to caudal planes. Additionally, pRhoA demonstrated dynamic redistribution between the nucleus and cytoplasm. pLIMK did not show such nucleus and cytoplasm redistribution but the expression level was changed from rostral to caudal planes. At some planes, pRhoA showed an increasing trend in expression in the cortex but a decreasing trend in the dentate gyrus of the 3xTg-AD mouse hippocampus. pLIMK tends to decrease in the cortex but increase in the dentate gyrus of 3xTg-AD mouse hippocampus. CONCLUSIONS: RhoA activation is dysregulated in both human and mouse AD brains, and the RhoA-LIMK signaling axis reveals spatial dysregulation along the rostral-caudal plane dimensions.


Assuntos
Doença de Alzheimer , Animais , Humanos , Camundongos , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Córtex Cerebral/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Camundongos Transgênicos , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Quinases Lim/metabolismo
19.
Eur J Cell Biol ; 102(4): 151355, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37639782

RESUMO

Small GTPases are highly regulated proteins that control essential signaling pathways through the activity of their effector proteins. Among the RHOA subfamily, RHOB regulates peculiar functions that could be associated with the control of the endocytic trafficking of signaling proteins. Here, we used an optimized assay based on tripartite split-GFP complementation to localize GTPase-effector complexes with high-resolution. The detection of RHOB interaction with the Rhotekin Rho binding domain (RBD) that specifically recognizes the active GTP-bound GTPase, is performed in vitro by the concomitant addition of recombinant GFP1-9 and a GFP nanobody. Analysis of RHOB-RBD complexes localization profiles combined with immunostaining and live cell imaging indicated a serum-dependent reorganization of the endosomal and membrane pool of active RHOB. We further applied this technology to the detection of RHO-effector complexes that highlighted their subcellular localization with high resolution among the different cellular compartments.


Assuntos
Transdução de Sinais , Proteína rhoB de Ligação ao GTP , Proteína rhoB de Ligação ao GTP/genética , Proteína rhoB de Ligação ao GTP/química , Proteína rhoB de Ligação ao GTP/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Membrana Celular/metabolismo , Guanosina Trifosfato/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
20.
Cells ; 12(13)2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-37443829

RESUMO

Glomerular disease due to podocyte malfunction is a major factor in the pathogenesis of chronic kidney disease. Identification of podocyte-specific signaling pathways is therefore a prerequisite to characterizing relevant disease pathways and developing novel treatment approaches. Here, we employed loss of function studies for EPB41L5 (Yurt) as a central podocyte gene to generate a cell type-specific disease model. Loss of Yurt in fly nephrocytes caused protein uptake and slit diaphragm defects. Transcriptomic and proteomic analysis of human EPB41L5 knockout podocytes demonstrated impaired mechanotransduction via the YAP/TAZ signaling pathway. Further analysis of specific inhibition of the YAP/TAZ-TEAD transcription factor complex by TEADi led to the identification of ARGHAP29 as an EPB41L5 and YAP/TAZ-dependently expressed podocyte RhoGAP. Knockdown of ARHGAP29 caused increased RhoA activation, defective lamellipodia formation, and increased maturation of integrin adhesion complexes, explaining similar phenotypes caused by loss of EPB41L5 and TEADi expression in podocytes. Detection of increased levels of ARHGAP29 in early disease stages of human glomerular disease implies a novel negative feedback loop for mechanotransductive RhoA-YAP/TAZ signaling in podocyte physiology and disease.


Assuntos
Podócitos , Humanos , Podócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Sinalização YAP , Mecanotransdução Celular , Integrinas/metabolismo , Proteômica , Proteína rhoA de Ligação ao GTP/metabolismo , Transdução de Sinais , Proteínas Ativadoras de GTPase/metabolismo , Proteínas de Membrana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...