Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Comput Biol Med ; 170: 108080, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38306776

RESUMO

Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a life-threatening syndrome induced by various diseases, including COVID-19. In the progression of ALI/ARDS, activated neutrophils play a central role by releasing various inflammatory mediators, including elastase. Sivelestat is a selective and competitive inhibitor of neutrophil elastase. Although its protective effects on attenuating ALI/ARDS have been confirmed in several models of lung injury, clinical trials have presented inconsistent results on its therapeutic efficacy. Therefore, in this report, we used a network pharmacology approach coupled with animal experimental validation to unravel the concrete therapeutic targets and biological mechanisms of sivelestat in treating ALI/ARDS. In bioinformatic analyses, we found 118 targets of sivelestat against ALI/ARDS, and identified six hub genes essential for sivelestat treatment of ALI/ARDS, namely ERBB2, GRB2, PTK2, PTPN11, ESR1, and CCND1. We also found that sivelestat targeted several genes expressed in human lung microvascular endothelial cells after lipopolysaccharide (LPS) treatment at 4 h (ICAM-1, PTGS2, RND1, BCL2A1, TNF, CA2, and ADORA2A), 8 h (ICAM-1, PTGS2, RND1, BCL2A1, MMP1, BDKRB1 and SLC40A1), and 24 h (ICAM-1). Further animal experiments showed that sivelestat was able to attenuate LPS-induced ALI by inhibiting the overexpression of ICAM-1, VCAM-1, and PTGS2 and increasing the phosphorylation of PTK2. Taken together, the bioinformatic findings and experimentative data indicate that the therapeutic effects of sivelestat against ALI/ARDS mainly focus on the early stage of ALI/ARDS by pharmacological modulation of inflammatory reaction, vascular endothelial injury, and cell apoptosis-related molecules.


Assuntos
Lesão Pulmonar Aguda , Glicina/análogos & derivados , Síndrome do Desconforto Respiratório , Sulfonamidas , Animais , Humanos , Molécula 1 de Adesão Intercelular/uso terapêutico , Células Endoteliais , Lipopolissacarídeos/uso terapêutico , Ciclo-Oxigenase 2/uso terapêutico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Pulmão , Proteínas rho de Ligação ao GTP/uso terapêutico
2.
Bioorg Med Chem ; 90: 117337, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37253305

RESUMO

Rho guanosine triphosphatases (Rho GTPases), as members of the Ras superfamily, are GDP/GTP binding proteins that behave as molecular switches for the transduction of signals from external stimuli. Rho GTPases play essential roles in a number of cellular processes including cell cycle, cell polarity as well as cell migration. The dysregulations of Rho GTPases are related with various diseases, especially with cancers. Accumulating evidence supports that Rho GTPases play important roles in cancer development and progression. Rho GTPases become potential therapeutic targets for cancer therapy. And a number of inhibitors targeting Rho GTPases have been developed. In this review, we discuss their structural features, summarize their roles in cancer, and focus on the recent progress of their inhibitors, which are beneficial for the drug discovery targeting Rho GTPases.


Assuntos
Neoplasias , Proteínas rho de Ligação ao GTP , Humanos , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/uso terapêutico , Neoplasias/tratamento farmacológico , Descoberta de Drogas , Ciclo Celular , Movimento Celular
3.
J Ovarian Res ; 16(1): 8, 2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36627652

RESUMO

BACKGROUND: Chemotherapy is among the most common treatment methods for ovarian cancer (OC). However, chemoresistance limits the effectiveness of chemotherapy and leads to treatment failure. We herein investigate the biological effect of forkhead box D3 (FOXD3) in the chemoresistance of OC cells. METHODS: Expression of FOXD3, miR-335 and disheveled-associated activator of morphogenesis 1 (DAAM1) was detected in OC cells and tissues. The regulatory network of FOXD3/miR-335/DAAM1 was validated by dual-luciferase reporter and ChIP assays in vitro. After ectopic expression and depletion experiments in carboplatin/paclitaxel (CP)-resistant (A2780CP) or sensitive (A2780S) OC cells, cell viability, colony formation and apoptosis were tested by CCK-8 assay, colony formation assay and flow cytometry respectively. Effects of FOXD3 on the chemoresistance of OC cells in vivo were evaluated in OC xenografts in nude mice. RESULTS: Overexpression of FOXD3 impaired the proliferation and chemoresistance of OC cells, which was related to the promotion of the miR-335 expression. Functionally, DAAM1 was a putative target of miR-335. Silencing of DAAM1 was responsible for the inhibition of myosin II activation, consequently leading to suppressed OC cell proliferation and chemoresistance. In vivo results further showed that FOXD3 weakened the chemoresistance of OC cells to CP. CONCLUSION: Taken together, we unveil a novel FOXD3/miR-335/DAAM1/myosin II axis that regulates the chemoresistance of OC both in vitro and in vivo.


Assuntos
Fatores de Transcrição Forkhead , MicroRNAs , Neoplasias Ovarianas , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Proteínas dos Microfilamentos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/uso terapêutico
4.
Altern Ther Health Med ; 29(2): 58-63, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36239568

RESUMO

Objective: We aimed to explore the mechanism of microRNA-936 (miR-936) targeting G protein coupled receptor 78 (GPR78) regulating chemoresistance of non-small cell lung cancer (NSCLC) by activating the Galphaq Rho GTPase pathway. Methods: We added cisplatin to DMEM medium of HCC827/cisplatin cells and adjusted the final concentration to 1 µg/mL. Cells were divided into the control group and the miR-936 transfection group. Tissue samples were divided into the normal tissue group and the NSCLC tissue group. The mRNA expression of miR-936 in tissue samples was analyzed via reverse transcription polymerase chain reaction (RT-PCR). Cell migration and invasion were detected by wound healing assay. Cell counting kit 8 (CCK-8) was used to detect the cell viability 1, 2 and 3 days after cisplatin induction. The toxicity of cisplatin was analyzed by flow cytometry. The targeting relationship between miR-936 and GPR78 was detected by luciferase reporter gene assay. The regulation of miR-936 on GPR78/Rho GTPase was analyzed by Western blot. Results: The expression of miR-936 in NSCLC was lower than in normal tissues (P < .05). The number of cell migrations and invasions in the miR-936 transfection group was lower than in the control group (P < .05). The cell viability in the miR-936 transfection group was lower than in the control group on the 1st, 2nd and 3rd day (P < .05). With the increase in cisplatin concentration, the apoptosis rate of cells increased in a dependent manner (P < .05). Compared with GPR78 Mut, overexpression of miR-936 inhibited the luciferase activity of GPR78 WT 3'- UTR (P < .05). The expression of GPR78, RhoA, Rac1 and ABCB1 protein in the miR-936 transfection group was lower than in the control group (P < .05). The expression of GPR78 protein in the inhibitor+miR-936 transfection group was lower than in the inhibitor+control group (P < .05). Conclusion: miR-936 targets GPR78 and improves the sensitivity of NSCLC cells to cisplatin via the Galphaq Rho GTPase pathway.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Cisplatino/farmacologia , Cisplatino/metabolismo , Cisplatino/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/farmacologia , Proteínas rho de Ligação ao GTP/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Luciferases/metabolismo , Luciferases/farmacologia , Luciferases/uso terapêutico , Proliferação de Células , Linhagem Celular Tumoral
5.
Cytokine Growth Factor Rev ; 67: 11-24, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35934612

RESUMO

Breast cancer (BC) is the most frequently diagnosed cancer among women in all the populations of the world. Although the BC mortality rate has declined, resistance to treatment is still a significant challenge for patient survival. Various cellular signaling pathways, such as Wnt and Rho/GTPase have been linked to the development, migration, and metastasis of BC, and also in treatment resistance mechanisms. Some studies have shown an association between two important cellular pathways, Wnt and Rho/GTPase, in cytoskeleton activation and cancer invasion. However, their involvement in BC has received little attention. This review summarizes the Wnt and Rho/GTPases signaling pathway functions, and also the crosstalk between these pathways in the progression, metastasis, and drug resistance mechanisms in BC. Considering the signaling pathways involved in BC tumorigenesis, future studies will need to investigate possible molecular interventions and new opportunities for the development of personalized strategies for BC treatment in order to improve overall outcomes.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/terapia , Carcinogênese , Movimento Celular , Feminino , Humanos , Transdução de Sinais , Via de Sinalização Wnt/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/uso terapêutico
6.
Environ Sci Pollut Res Int ; 29(12): 17417-17424, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34665416

RESUMO

The RhoA gene showed an important genotypic association with nicotine dependence and smoking initiation. The current study aims to investigate the effect of the Rho GTPase inhibitor ML141 in the progression of nicotine dependence in a mice model of precipitated nicotine withdrawal syndrome by mecamylamine.The experimental procedure involved administration of 2.5 mg/kg nicotine dissolved in normal saline subcutaneously (s.c) four times a day consecutively for 7 days and last single dose in the morning on 8th day. ML-141 was dissolved in dimethyl sulfoxide (DMSO) and was administered daily with nicotine as corrective treatment at a dose of 1,5 and 10 mg/kg (p < 0.05). An injection of 3 mg/kg of mecamylamine intraperitoneal (ip) was given an hour later than the last nicotine dose on the day 8 to precipitate withdrawal of nicotine and withdrawal severity was assessed by measuring hyperalgesia, piloerection, jumping frequency, tremors, and withdrawal severity score (WSS). Various behavioural changes such as hyperalgesia, piloerection, jumping frequency, and tremors were monitored and WSS was calculated. ML-141 a selective Rho GTPase inhibitor was found to show dose-dependent effect on all these parameters. Inhibition of Rho GTPase was found to reduce the severity of withdrawal syndrome; therefore, it can be concluded that Rho GTPase would serve as a suitable biological target by regulating the reward system in brain and could be used as new target for drug discovery.


Assuntos
Síndrome de Abstinência a Substâncias , Tabagismo , Animais , Mecamilamina/farmacologia , Mecamilamina/uso terapêutico , Camundongos , Nicotina , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Proteínas rho de Ligação ao GTP/uso terapêutico
7.
Mil Med Res ; 7(1): 48, 2020 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-33038921

RESUMO

BACKGROUND: p53 and DIRAS3 are tumor suppressors that are frequently silenced in tumors. In this study, we sought to determine whether the concurrent re-expression of p53 and DIRAS3 could effectively induce head and neck squamous cell carcinoma (HNSCC) cell death. METHODS: CAL-27 and SCC-25 cells were treated with Ad-DIRAS3 and rAd-p53 to induce re-expression of DIRAS3 and p53 respectively. The effects of DIRAS3 and p53 re-expression on the growth and apoptosis of HNSCC cells were examined by TUNEL assay, flow cytometric analysis and MTT. The effects of DIRAS3 and p53 re-expression on Akt phosphorylation, oncogene expression, and the interaction of 4E-BP1 with eIF4E were determined by real-time PCR, Western blotting and immunoprecipitation analysis. The ability of DIRAS3 and p53 re-expression to induce autophagy was evaluated by transmission electron microscopy, LC3 fluorescence microscopy and Western blotting. The effects of DIRAS3 and p53 re-expression on HNSCC growth were evaluated by using an orthotopic xenograft mouse model. RESULTS: TUNEL assay and flow cytometric analysis showed that the concurrent re-expression of DIRAS3 and p53 significantly induced apoptosis (P < 0.001). MTT and flow cytometric analysis revealed that DIRAS3 and p53 re-expression significantly inhibited proliferation and induced cell cycle arrest (P < 0.001). Mechanistically, the concurrent re-expression of DIRAS3 and p53 down-regulated signal transducer and activation of transcription 3 (STAT3) and up-regulated p21WAF1/CIP1 and Bax (P < 0.001). DIRAS3 and p53 re-expression also inhibited Akt phosphorylation, increased the interaction of eIF4E with 4E-BP1, and reduced the expression of c-Myc, cyclin D1, vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), epidermal growth factor receptor (EGFR) and Bcl-2 (P < 0.001). Moreover, the concurrent re-expression of DIRAS3 and p53 increased the percentage of cells with GFP-LC3 puncta compared with that in cells treated with control adenovirus (50.00% ± 4.55% vs. 4.67% ± 1.25%, P < 0.001). LC3 fluorescence microscopy and Western blotting further showed that DIRAS3 and p53 re-expression significantly promoted autophagic activity but also inhibited autophagic flux, resulting in overall impaired autophagy. Finally, the concurrent re-expression of DIRAS3 and p53 significantly decreased the tumor volume compared with the control group in a HNSCC xenograft mouse model [(3.12 ± 0.75) mm3 vs. (189.02 ± 17.54) mm3, P < 0.001]. CONCLUSIONS: The concurrent re-expression of DIRAS3 and p53 is a more effective approach to HNSCC treatment than current treatment strategies.


Assuntos
Autofagia/genética , Fragmentos de Peptídeos/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/complicações , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Proteína Supressora de Tumor p53/farmacologia , Proteínas rho de Ligação ao GTP/farmacologia , Animais , Apoptose/genética , Células Cultivadas , Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço/fisiopatologia , Proteína Supressora de Tumor p53/uso terapêutico , Proteínas rho de Ligação ao GTP/uso terapêutico
8.
F1000Res ; 82019.
Artigo em Inglês | MEDLINE | ID: mdl-31723415

RESUMO

Podocytes, or glomerular epithelial cells, form the final layer in the glomerular capillary wall of the kidney. Along with the glomerular basement membrane and glomerular endothelial cells, they make up the glomerular filtration barrier which allows the passage of water and small molecules and, in healthy individuals, prevents the passage of albumin and other key proteins. The podocyte is a specialised and terminally differentiated cell with a specific cell morphology that is largely dependent on a highly dynamic underlying cytoskeletal network and that is essential for maintaining glomerular function and integrity in healthy kidneys. The RhoGTPases (RhoA, Rac1 and Cdc42), which act as molecular switches that regulate actin dynamics, are known to play a crucial role in maintaining the cytoskeletal and molecular integrity of the podocyte foot processes in a dynamic manner. Recently, novel protein interaction networks that regulate the RhoGTPases in the podocyte and that are altered by disease have been discovered. This review will discuss these networks and their potential as novel therapeutic targets in nephrotic syndrome. It will also discuss the evidence that they are direct targets for (a) steroids, the first-line agents for the treatment of nephrotic syndrome, and (b) certain kinase inhibitors used in cancer treatment, leading to nephrotoxicity.


Assuntos
Nefropatias , Síndrome Nefrótica , Podócitos , Proteínas rho de Ligação ao GTP , Citoesqueleto , Células Endoteliais , Humanos , Síndrome Nefrótica/tratamento farmacológico , Proteínas rho de Ligação ao GTP/fisiologia , Proteínas rho de Ligação ao GTP/uso terapêutico
9.
Clin Cancer Res ; 21(1): 24-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25336694

RESUMO

RHO GTPases, members of the RAS superfamily of small GTPases, are adhesion and growth factor-activated molecular switches that play important roles in tumor development and progression. When activated, RHO-family GTPases such as RAC1, CDC42, and RHOA, transmit signals by recruiting a variety of effector proteins, including the protein kinases PAK, ACK, MLK, MRCK, and ROCK. Genetically induced loss of RHO function impedes transformation by a number of oncogenic stimuli, leading to an interest in developing small-molecule inhibitors that either target RHO GTPases directly, or that target their downstream protein kinase effectors. Although inhibitors of RHO GTPases and their downstream signaling kinases have not yet been widely adopted for clinical use, their potential value as cancer therapeutics continues to facilitate pharmaceutical research and development and is a promising therapeutic strategy.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Fosfotransferases/genética , Proteínas rho de Ligação ao GTP/genética , Carcinogênese , Humanos , Terapia de Alvo Molecular , Neoplasias/patologia , Fosfotransferases/antagonistas & inibidores , Fosfotransferases/uso terapêutico , Transdução de Sinais/genética , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/uso terapêutico
10.
PLoS One ; 6(1): e16135, 2011 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-21283639

RESUMO

BACKGROUND: Spinal cord injury (SCI) often results in permanent functional loss. This physical trauma leads to secondary events, such as the deposition of inhibitory chondroitin sulfate proteoglycan (CSPG) within astroglial scar tissue at the lesion. METHODOLOGY/PRINCIPAL FINDINGS: We examined whether local delivery of constitutively active (CA) Rho GTPases, Cdc42 and Rac1 to the lesion site alleviated CSPG-mediated inhibition of regenerating axons. A dorsal over-hemisection lesion was created in the rat spinal cord and the resulting cavity was conformally filled with an in situ gelling hydrogel combined with lipid microtubes that slowly released constitutively active (CA) Cdc42, Rac1, or Brain-derived neurotrophic factor (BDNF). Treatment with BDNF, CA-Cdc42, or CA-Rac1 reduced the number of GFAP-positive astrocytes, as well as CSPG deposition, at the interface of the implanted hydrogel and host tissue. Neurofilament 160kDa positively stained axons traversed the glial scar extensively, entering the hydrogel-filled cavity in the treatments with BDNF and CA-Rho GTPases. The treated animals had a higher percentage of axons from the corticospinal tract that traversed the CSPG-rich regions located proximal to the lesion site. CONCLUSION: Local delivery of CA-Cdc42, CA-Rac1, and BDNF may have a significant therapeutic role in overcoming CSPG-mediated regenerative failure after SCI.


Assuntos
Axônios/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Proteoglicanas de Sulfatos de Condroitina/fisiologia , Traumatismos da Medula Espinal/terapia , Regeneração da Medula Espinal/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/administração & dosagem , Animais , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Ratos , Proteína cdc42 de Ligação ao GTP/administração & dosagem , Proteínas rac1 de Ligação ao GTP/administração & dosagem , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/uso terapêutico
11.
Medicina (B Aires) ; 70(6): 555-64, 2010.
Artigo em Espanhol | MEDLINE | ID: mdl-21163747

RESUMO

Rho GTPases are a key protein family controlling the transduction of external signals to cytoplasmatic and nuclear effectors. In the last few years, the development of genetic and pharmacological tools has allowed a more precise definition of the specific roles of Rho GTPases. The aim of this review is to describe the cellular functions regulated by these proteins with focus on the molecular mechanism involved. We also address the role of Rho GTPases in the development of different human diseases such as cancer. Finally, we describe different experimental therapeutic strategies with Rho GTPases as molecular targets.


Assuntos
Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Proteínas rho de Ligação ao GTP/uso terapêutico , Antineoplásicos/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Humanos , Proteínas rho de Ligação ao GTP/fisiologia
12.
Medicina (B.Aires) ; 70(6): 555-564, dic. 2010. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-633805

RESUMO

Las Rho GTPasas son una familia de proteínas clave en la transmisión de señales provenientes del exterior celular hacia efectores intracelulares tanto citoplasmáticos como nucleares. En los últimos año ha habido un desarrollo vertiginoso de múltiples herramientas genéticas y farmacológicas, lo que ha permitido establecer de manera mucho más precisa las funciones específicas de estas proteínas. El objetivo de la presente revisión es hacer foco en las múltiples funciones celulares reguladas por las Rho GTPasas, describiendo en detalle el mecanismo molecular involucrado. Se discute además la participación de estas proteínas en diversas enfermedades humanas haciendo énfasis en su vinculación con el cáncer. Por último, se hace una actualización detallada sobre las estrategias terapéuticas en experimentación que tienen a las Rho GTPasas como blancos moleculares.


Rho GTPases are a key protein family controlling the transduction of external signals to cytoplasmatic and nuclear effectors. In the last few years, the development of genetic and pharmacological tools has allowed a more precise definition of the specific roles of Rho GTPases. The aim of this review is to describe the cellular functions regulated by these proteins with focus on the molecular mechanism involved. We also address the role of Rho GTPases in the development of different human diseases such as cancer. Finally, we describe different experimental therapeutic strategies with Rho GTPases as molecular targets.


Assuntos
Humanos , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Proteínas rho de Ligação ao GTP/uso terapêutico , Antineoplásicos/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Proteínas rho de Ligação ao GTP/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...