Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 410
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 98(7): e0060624, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38809020

RESUMEN

Rabies virus (RABV) is highly lethal and triggers severe neurological symptoms. The neuropathogenic mechanism remains poorly understood. Ras-related C3 botulinum toxin substrate 1 (Rac1) is a Rho-GTPase that is involved in actin remodeling and has been reported to be closely associated with neuronal dysfunction. In this study, by means of a combination of pharmacological inhibitors, small interfering RNA, and specific dominant-negatives, we characterize the crucial roles of dynamic actin and the regulatory function of Rac1 in RABV infection, dominantly in the viral entry phase. The data show that the RABV phosphoprotein interacts with Rac1. RABV phosphoprotein suppress Rac1 activity and impedes downstream Pak1-Limk1-Cofilin1 signaling, leading to the disruption of F-actin-based structure formation. In early viral infection, the EGFR-Rac1-signaling pathway undergoes a biphasic change, which is first upregulated and subsequently downregulated, corresponding to the RABV entry-induced remodeling pattern of F-actin. Taken together, our findings demonstrate for the first time the role played by the Rac1 signaling pathway in RABV infection and may provide a clue for an explanation for the etiology of rabies neurological pathogenesis.IMPORTANCEThough neuronal dysfunction is predominant in fatal rabies, the detailed mechanism by which rabies virus (RABV) infection causes neurological symptoms remains in question. The actin cytoskeleton is involved in numerous viruses infection and plays a crucial role in maintaining neurological function. The cytoskeletal disruption is closely associated with abnormal nervous symptoms and induces neurogenic diseases. In this study, we show that RABV infection led to the rearrangement of the cytoskeleton as well as the biphasic kinetics of the Rac1 signal transduction. These results help elucidate the mechanism that causes the aberrant neuronal processes by RABV infection and may shed light on therapeutic development aimed at ameliorating neurological disorders.


Asunto(s)
Citoesqueleto de Actina , Actinas , Virus de la Rabia , Transducción de Señal , Proteína de Unión al GTP rac1 , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Humanos , Citoesqueleto de Actina/metabolismo , Animales , Virus de la Rabia/fisiología , Actinas/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/genética , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/genética , Quinasas Lim/metabolismo , Quinasas Lim/genética , Internalización del Virus , Rabia/metabolismo , Rabia/virología , Línea Celular , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Factores Despolimerizantes de la Actina/metabolismo
2.
Mol Cell ; 62(3): 397-408, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27153537

RESUMEN

Cofilin/actin-depolymerizing factor (ADF) proteins are critical nodes that relay signals from protein kinase cascades to the actin cytoskeleton, in particular through site-specific phosphorylation at residue Ser3. This is important for regulation of the roles of cofilin in severing and stabilizing actin filaments. Consequently, cofilin/ADF Ser3 phosphorylation is tightly controlled as an almost exclusive substrate for LIM kinases. Here we determine the LIMK1:cofilin-1 co-crystal structure. We find an interface that is distinct from canonical kinase-substrate interactions. We validate this previously unobserved mechanism for high-fidelity kinase-substrate recognition by in vitro kinase assays, examination of cofilin phosphorylation in mammalian cells, and functional analysis in S. cerevisiae. The interface is conserved across all LIM kinases. Remarkably, we also observe both pre- and postphosphotransfer states in the same crystal lattice. This study therefore provides a molecular understanding of how kinase-substrate recognition acts as a gatekeeper to regulate actin cytoskeletal dynamics.


Asunto(s)
Cofilina 1/metabolismo , Quinasas Lim/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Sitios de Unión , Dominio Catalítico , Cofilina 1/química , Cofilina 1/genética , Cristalografía por Rayos X , Células HEK293 , Humanos , Quinasas Lim/química , Quinasas Lim/genética , Modelos Moleculares , Mutación , Fosforilación , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Serina , Relación Estructura-Actividad , Especificidad por Sustrato , Transfección
3.
Biochemistry (Mosc) ; 89(3): 393-406, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38648760

RESUMEN

Courtship suppression is a behavioral adaptation of the fruit fly. When majority of the females in a fly population are fertilized and non-receptive for mating, a male, after a series of failed attempts, decreases its courtship activity towards all females, saving its energy and reproductive resources. The time of courtship decrease depends on both duration of unsuccessful courtship and genetically determined features of the male nervous system. Thereby, courtship suppression paradigm can be used for studying molecular mechanisms of learning and memory. p-Cofilin, a component of the actin remodeling signaling cascade and product of LIM-kinase 1 (LIMK1), regulates Drosophila melanogaster forgetting in olfactory learning paradigm. Previously, we have shown that limk1 suppression in the specific types of nervous cells differently affects fly courtship memory. Here, we used Gal4 > UAS system to induce limk1 overexpression in the same types of neurons. limk1 activation in the mushroom body, glia, and fruitless neurons decreased learning index compared to the control strain or the strain with limk1 knockdown. In cholinergic and dopaminergic/serotoninergic neurons, both overexpression and knockdown of limk1 impaired Drosophila short-term memory. Thus, proper balance of the limk1 activity is crucial for normal cognitive activity of the fruit fly.


Asunto(s)
Cortejo , Proteínas de Drosophila , Drosophila melanogaster , Quinasas Lim , Memoria , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Masculino , Quinasas Lim/metabolismo , Quinasas Lim/genética , Femenino , Cuerpos Pedunculados/metabolismo , Cuerpos Pedunculados/fisiología , Conducta Sexual Animal
4.
Ecotoxicol Environ Saf ; 272: 116110, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38364763

RESUMEN

OBJECTIVE: We here explored whether perinatal nonylphenol (NP) exposure causes myocardial fibrosis (MF) during adulthood in offspring rats and determined the role of the TGF-ß1/LIMK1 signaling pathway in NP-induced fibrosis in cardiac fibroblasts (CFs). METHODS AND RESULTS: Histopathology revealed increased collagen deposition and altered fiber arrangement in the NP and isoproterenol hydrochloride (ISO) groups compared with the blank group. Systolic and diastolic functions were impaired. Western blotting and qRT-PCR demonstrated that the expression of central myofibrosis-related proteins (collagens Ι and ΙΙΙ, MMP2, MMP9, TGF-ß1, α-SMA, IL-1ß, and TGF-ß1) and genes (Collagen Ι, Collagen ΙΙΙ, TGF-ß1, and α-SMA mRNA) was upregulated in the NP and ISO groups compared with the blank group. The mRNA-seq analysis indicated differential expression of TGF-ß1 signaling pathway-associated genes and proteins. Fibrosis-related protein and gene expression increased in the CFs stimulated with the recombinant human TGF-ß1 and NP, which was consistent with the results of animal experiments. According to the immunofluorescence analysis and western blotting, NP exposure activated the TGF-ß1/LIMK1 signaling pathway whose action mechanism in NP-induced CFs was further validated using the LIMK1 inhibitor (BMS-5). The inhibitor modulated the TGF-ß1/LIMK1 signaling pathway and suppressed the NP-induced increase in fibrosis-related protein expression in the CFs. Thus, the aforementioned pathway is involved in NP-induced fibrosis. CONCLUSION: We here provide the first evidence that perinatal NP exposure causes myocardial fibrosis in growing male rat pups and reveal the molecular mechanism and functional role of the TGF-ß1/LIMK1 signaling pathway in this process.


Asunto(s)
Cardiomiopatías , Fenoles , Factor de Crecimiento Transformador beta1 , Humanos , Ratas , Masculino , Animales , Adulto , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Cardiomiopatías/metabolismo , Colágeno/metabolismo , Transducción de Señal , Fibrosis , ARN Mensajero/metabolismo , Fibroblastos , Miocardio/metabolismo , Quinasas Lim/metabolismo
5.
Am J Respir Cell Mol Biol ; 68(4): 417-429, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36662576

RESUMEN

TAS2Rs (bitter taste receptors) are GPCRs (G protein-coupled receptors) expressed on human airway smooth muscle (HASM) cells; when activated by receptor agonists they evoke marked airway relaxation. In both taste and HASM cells, TAS2Rs activate a canonical Gßγ-mediated stimulation of Ca2+ release from intracellular stores by activation of PLCß (phospholipase Cß). Alone, this [Ca2+]i signaling does not readily account for relaxation, particularly since bronchoconstrictive agonists acting at Gq-coupled receptors also increase [Ca2+]i. We established that TAS2R14 activation in HASM promotes relaxation through F-actin (filamentous actin) severing. This destabilization of actin was from agonist-promoted activation (dephosphorylation) of cofilin, which was pertussis toxin sensitive. Cofilin dephosphorylation was due to TAS2R-mediated deactivation of LIM domain kinase. The link between early receptor action and the distal cofilin dephosphorylation was found to be the polarity protein partitioning defective 3 (Par3), a known binding partner with PLCß that inhibits LIM kinase. The physiologic relevance of this pathway was assessed using knock-downs of cofilin and Par3 in HASM cells and in human precision-cut lung slices. Relaxation by TAS2R14 agonists was ablated with knock-down of either protein as assessed by magnetic twisting cytometry in isolated cells or intact airways in the slices. Blocking [Ca2+]i release by TAS2R14 inhibited agonist-promoted cofilin dephosphorylation, confirming a role for [Ca2+]i in actin-modifying pathways. These results further elucidate the mechanistic basis of TAS2R-mediated HASM relaxation and point toward nodal points that may act as asthma or chronic obstructive pulmonary disease response modifiers or additional targets for novel bronchodilators.


Asunto(s)
Actinas , Asma , Receptores Acoplados a Proteínas G , Humanos , Actinas/metabolismo , Asma/metabolismo , Quinasas Lim/metabolismo , Pulmón/metabolismo , Relajación Muscular/fisiología , Receptores Acoplados a Proteínas G/metabolismo
6.
Environ Toxicol ; 38(5): 1038-1052, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36722453

RESUMEN

Metastasis is a leading cause to treatment failure in hepatocellular carcinoma (HCC) patients. Exosomes act as pivotal mediators in communication between different cells and exert effects on recipient cells by delivering bioactive cargoes, such as microRNAs (miRNAs). MiRNAs function in multiple steps of HCC development, including metastasis. MiR-374c-5p was previously identified as a tumor suppressor in some malignancies, while the current knowledge of its role in HCC metastasis is still limited. Herein, miR-374c-5p was found to be downregulated in HCC cell lines and clinical samples, and positively related with favorable prognosis in HCC patients. MiR-374c-5p transferred by exosomes derived from bone marrow mesenchymal stem cell (BMSC) suppressed migration, invasion and proliferation of HCC cells. LIMK1 was verified as downstream target gene of miR-374c-5p. Knockdown of LIMK1 reduced invasion, migration and proliferation of HCC cells, whereas overexpression functioned oppositely. The miR-374c-5p/LIMK1 axis suppressed epithelial-mesenchymal transition (EMT) by inactivating Wnt/ß-catenin pathway. In addition, miR-374c-5p was downregulated and LIMK1 upregulated in TGF-ß1 induced EMT. This EMT model could be reversed by LIMK1 silencing or miR-374c-5p overexpression. These results suggest that exo-miR-374c-5p suppresses EMT via targeting LIMK1-Wnt/ß-catenin axis and the axis is involved in TGF-ß1 induced metastasis of HCC, thereby identifying miR-374c-5p as a potential target for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Células Madre Mesenquimatosas , MicroARNs , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Proliferación Celular/genética , Quinasas Lim/genética , Quinasas Lim/metabolismo
7.
Environ Toxicol ; 38(5): 1063-1077, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36793247

RESUMEN

Leukemia is a type of disease in which hematopoietic stem cells proliferate clonally at the genetic level. We discovered previously by high-resolution mass spectrometry that diallyl disulfide (DADS), which is one of the effective ingredients of garlic, reduces the performance of RhoGDI2 from APL HL-60 cells. Although RhoGDI2 is oversubscribed in several cancer categories, the effect of RhoGDI2 in HL-60 cells has remained unexplained. We aimed to investigate the influence of RhoGDI2 on DADS-induced differentiation of HL-60 cells to elucidate the association among the effect of inhibition or over-expression of RhoGDI2 with HL-60 cell polarization, migration and invasion, which is important for establishing a novel generation of inducers to elicit leukemia cell polarization. Co-transfection with RhoGDI2-targeted miRNAs apparently decreases the malignant biological behavior of cells and upregulates cytopenias in DADS-treated HL-60 cell lines, which increases CD11b and decreases CD33 and mRNA levels of Rac1, PAK1 and LIMK1. Meanwhile, we generated HL-60 cell lines with high-expressing RhoGDI2. The proliferation, migration and invasion capacity of such cells were significantly increased by the treated with DADS, while the reduction capacity of the cells was decreased. There was a reduction in CD11b and an increase in CD33 production, as well as an increase in the mRNA levels of Rac1, PAK1 and LIMK1. It also confirmed that inhibition of RhoGDI2 attenuates the EMT cascade via the Rac1/Pak1/LIMK1 pathway, thereby inhibiting the malignant biological behavior of HL-60 cells. Thus, we considered that inhibition of RhoGDI2 expression might be a new therapeutic direction for the treatment of human promyelocytic leukemia. The anti-cancer property of DADS against HL-60 leukemia cells might be regulated by RhoGDI2 through the Rac1-Pak1-LIMK1 pathway, which provides new evidence for DADS as a clinical anti-cancer medicine.


Asunto(s)
Leucemia , Inhibidor beta de Disociación del Nucleótido Guanina rho , Humanos , Compuestos Alílicos/farmacología , Diferenciación Celular/efectos de los fármacos , Disulfuros/farmacología , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Leucemia/metabolismo , Leucemia/patología , Quinasas Lim/genética , Quinasas Lim/metabolismo , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/farmacología , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/farmacología , Inhibidor beta de Disociación del Nucleótido Guanina rho/efectos de los fármacos , Inhibidor beta de Disociación del Nucleótido Guanina rho/metabolismo , ARN Mensajero , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
8.
J Cell Physiol ; 237(7): 3012-3029, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35567755

RESUMEN

This study provides comprehensive mechanistic evidence for the role of clusterin, a stress-response secretory chaperone protein, in the modulation of intraocular pressure (IOP) by regulating the trabecular meshwork (TM) actin cytoskeleton and the extracellular matrix (ECM). The pathological stressors on TM known to elevate IOP significantly lowered clusterin protein levels indicating stress-related clusterin function loss. Small interfering RNA-mediated clusterin loss in human TM cells in vitro induced actin polymerization and stabilization via protein kinase D1, serine/threonine-protein kinase N2 (PRK2), and LIM kinase 1 (LIMK1), and the recruitment and activation of adhesome proteins including paxillin, vinculin, and integrin αV and ß5. A complete loss of clusterin as seen in clusterin knockout mice (Clu-/- ) led to significant IOP elevation at postnatal Day 70. Contrarily, constitutive clusterin expression using adenovirus (AdCLU) in HTM cells resulted in the loss of actin polymerization via decreased PRK2, and LIMK1 and negative regulation of integrin αV and ß5. Furthermore, we found that AdCLU treatment in HTM cells significantly decreased the ECM protein expression and distribution by significantly increasing matrix metalloprotease 2 (MMP2) activity and lowering the levels of pro-fibrotic proteins such as transforming growth factor-ß2 (TGFß2), thrombospondin-1 (TSP-1), and plasminogen activator inhibitor-1 (PAI-1). Finally, we found that HTM cells supplemented with recombinant human clusterin attenuated the pro-fibrotic effects of TGFß2. For the first time this study demonstrates the importance of clusterin in the regulation of TM actin cytoskeleton - ECM interactions and the maintenance of IOP, thus making clusterin an interesting target to reverse elevated IOP.


Asunto(s)
Presión Intraocular , Malla Trabecular , Actinas/metabolismo , Animales , Células Cultivadas , Clusterina/genética , Clusterina/metabolismo , Clusterina/farmacología , Matriz Extracelular/metabolismo , Humanos , Integrina alfaV/metabolismo , Integrina alfaV/farmacología , Quinasas Lim/metabolismo , Ratones , Polimerizacion , Factor de Crecimiento Transformador beta2/farmacología
9.
EMBO J ; 37(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29496740

RESUMEN

Protein aggregation is a hallmark of diverse neurodegenerative diseases. Multiple lines of evidence have revealed that protein aggregates can penetrate inside cells and spread like prions. How such aggregates enter cells remains elusive. Through a focused siRNA screen targeting genes involved in membrane trafficking, we discovered that mutant SOD1 aggregates, like viruses, exploit cofilin-1 to remodel cortical actin and enter cells. Upstream of cofilin-1, signalling from the RHO GTPase and the ROCK1 and LIMK1 kinases controls cofilin-1 activity to remodel actin and modulate aggregate entry. In the spinal cord of symptomatic SOD1G93A transgenic mice, cofilin-1 phosphorylation is increased and actin dynamics altered. Importantly, the RHO to cofilin-1 signalling pathway also modulates entry of tau and α-synuclein aggregates. Our results identify a common host cell signalling pathway that diverse protein aggregates exploit to remodel actin and enter cells.


Asunto(s)
Cofilina 1/metabolismo , Agregado de Proteínas , Proteínas de Unión al GTP rho/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Quinasas Lim/metabolismo , Ratones Transgénicos , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Médula Espinal/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
10.
Development ; 146(3)2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30696709

RESUMEN

Cytoskeletal dynamics are involved in multiple cellular processes during oocyte meiosis, including spindle organization, actin-based spindle migration and polar body extrusion. Here, we report that the vesicle trafficking protein Rab23, a GTPase, drives the motor protein Kif17, and that this is important for spindle organization and actin dynamics during mouse oocyte meiosis. GTP-bound Rab23 accumulated at the spindle and promoted migration of Kif17 to the spindle poles. Depletion of Rab23 or Kif17 caused polar body extrusion failure. Further analysis showed that depletion of Rab23/Kif17 perturbed spindle formation and chromosome alignment, possibly by affecting tubulin acetylation. Kif17 regulated tubulin acetylation by associating with αTAT and Sirt2, and depletion of Kif17 altered expression of these proteins. Moreover, depletion of Kif17 decreased the level of cytoplasmic actin, which abrogated spindle migration to the cortex. The tail domain of Kif17 associated with constituents of the RhoA-ROCK-LIMK-cofilin pathway to modulate assembly of actin filaments. Taken together, our results demonstrate that the Rab23-Kif17-cargo complex regulates tubulin acetylation for spindle organization and drives actin-mediated spindle migration during meiosis.


Asunto(s)
Cinesinas/metabolismo , Meiosis/fisiología , Oocitos/metabolismo , Huso Acromático/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Acetilación , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Animales , Cinesinas/genética , Quinasas Lim/genética , Quinasas Lim/metabolismo , Ratones , Oocitos/citología , Transducción de Señal/fisiología , Sirtuina 2/genética , Sirtuina 2/metabolismo , Huso Acromático/genética , Tubulina (Proteína)/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
11.
Connect Tissue Res ; 63(5): 475-484, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35019797

RESUMEN

PURPOSE: The etiology of adolescent idiopathic scoliosis (AIS) remains unclear. The chondrogenic differentiation of mesenchymal stem cells (MSCs) is important in AIS, and the Ras homolog gene family member A (RHOA) is associated with chondrogenesis. The purpose of this study was to explore the effect of RHOA on the chondrogenic differentiation of MSCs in AIS. METHODS: We isolated MSCs from patients with AIS (AIS MSCs) and individuals without AIS (control MSCs). The inhibitor Y27632 was used to inhibit the function of RHOA/ROCK signaling, and plasmid-based overexpression and siRNA-mediated knockdown were used to manipulate RHOA expression. CCK-8 was used to detect cell viability. The phosphorylation levels of LIMK1, MLC2 and cofilin were detected by Western blotting. The mRNA expression of aggrecan, SOX9, and COL2A1 were confirmed using RT-PCR. Immunofluorescence was used to analyze F-actin and collagen II. Alcian blue staining was performed to assess the secretion of glycosaminoglycans (GAGs). RESULTS: We found that RHOA was significantly upregulated in AIS MSCs, and the phosphorylation levels of LIMK1, MLC2, and cofilin were increased. The mRNA expressions of aggrecan, SOX9, and COL2A1 were notably reduced in AIS MSCs. However, these effects were abolished by Y27632 treatment and RHOA knockdown in AIS MSCs. In addition, RHOA knockdown in AIS MSCs increased the content of collagen II and GAGs. RHOA overexpression in the control MSCs markedly activated the RHOA/ROCK signaling and decreased the expression of aggrecan, SOX9, and COL2A1, F-actin, and GAGs. CONCLUSION: RHOA regulates the chondrogenic differentiation ability of MSCs in AIS via the RHOA/ROCK signaling pathway and this regulation may involve SOX9.


Asunto(s)
Células Madre Mesenquimatosas , Escoliosis , Proteína de Unión al GTP rhoA , Factores Despolimerizantes de la Actina/metabolismo , Factores Despolimerizantes de la Actina/farmacología , Actinas/metabolismo , Actinas/farmacología , Adolescente , Agrecanos/metabolismo , Agrecanos/farmacología , Diferenciación Celular , Células Cultivadas , Condrogénesis , Colágeno/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Quinasas Lim/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/metabolismo , Escoliosis/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
12.
PLoS Comput Biol ; 17(11): e1009171, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34843456

RESUMEN

Predictive approaches such as virtual screening have been used in drug discovery with the objective of reducing developmental time and costs. Current machine learning and network-based approaches have issues related to generalization, usability, or model interpretability, especially due to the complexity of target proteins' structure/function, and bias in system training datasets. Here, we propose a new method "DRUIDom" (DRUg Interacting Domain prediction) to identify bio-interactions between drug candidate compounds and targets by utilizing the domain modularity of proteins, to overcome problems associated with current approaches. DRUIDom is composed of two methodological steps. First, ligands/compounds are statistically mapped to structural domains of their target proteins, with the aim of identifying their interactions. As such, other proteins containing the same mapped domain or domain pair become new candidate targets for the corresponding compounds. Next, a million-scale dataset of small molecule compounds, including those mapped to domains in the previous step, are clustered based on their molecular similarities, and their domain associations are propagated to other compounds within the same clusters. Experimentally verified bioactivity data points, obtained from public databases, are meticulously filtered to construct datasets of active/interacting and inactive/non-interacting drug/compound-target pairs (~2.9M data points), and used as training data for calculating parameters of compound-domain mappings, which led to 27,032 high-confidence associations between 250 domains and 8,165 compounds, and a finalized output of ~5 million new compound-protein interactions. DRUIDom is experimentally validated by syntheses and bioactivity analyses of compounds predicted to target LIM-kinase proteins, which play critical roles in the regulation of cell motility, cell cycle progression, and differentiation through actin filament dynamics. We showed that LIMK-inhibitor-2 and its derivatives significantly block the cancer cell migration through inhibition of LIMK phosphorylation and the downstream protein cofilin. One of the derivative compounds (LIMKi-2d) was identified as a promising candidate due to its action on resistant Mahlavu liver cancer cells. The results demonstrated that DRUIDom can be exploited to identify drug candidate compounds for intended targets and to predict new target proteins based on the defined compound-domain relationships. Datasets, results, and the source code of DRUIDom are fully-available at: https://github.com/cansyl/DRUIDom.


Asunto(s)
Quinasas Lim/antagonistas & inhibidores , Quinasas Lim/química , Factores Despolimerizantes de la Actina/química , Factores Despolimerizantes de la Actina/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Biología Computacional , Simulación por Computador , Desarrollo de Medicamentos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Humanos , Técnicas In Vitro , Ligandos , Quinasas Lim/metabolismo , Aprendizaje Automático , Simulación del Acoplamiento Molecular , Invasividad Neoplásica/prevención & control , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Farmacología en Red/estadística & datos numéricos , Fosforilación/efectos de los fármacos , Dominios Proteicos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Interfaz Usuario-Computador
13.
Bioorg Med Chem Lett ; 61: 128614, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35151865

RESUMEN

High rates of recurrence and treatment resistance in the most common malignant adult brain cancer, glioblastoma (GBM), suggest that monotherapies are not sufficiently effective. Combination therapies are increasingly pursued, but the possibility of adverse drug-drug interactions may preclude clinical implementation. Developing single molecules with multiple targets is a feasible alternative strategy to identify effective and tolerable pharmacotherapies for GBM. Here, we report the development of a novel, first-in-class, dual aurora and lim kinase inhibitor termed F114. Aurora kinases and lim kinases are involved in neoplastic cell division and cell motility, respectively. Due to the importance of these cellular functions, inhibitors of aurora kinases and lim kinases are being pursued separately as anti-cancer therapies. Using in vitro and ex vivo models of GBM, we found that F114 inhibits GBM proliferation and invasion. These results establish F114 as a promising new scaffold for dual aurora/lim kinase inhibitors that may be used in future drug development efforts for GBM, and potentially other cancers.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa B/antagonistas & inhibidores , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Quinasas Lim/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Aurora Quinasa A/metabolismo , Aurora Quinasa B/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Quinasas Lim/metabolismo , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
14.
J Cell Mol Med ; 25(12): 5560-5571, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33982869

RESUMEN

Lung cancer is the leading cause of cancer-related deaths. LIM domain kinase (LIMK) 1 is a member of serine/threonine kinase family and highly expressed in various cancers. Luteolin, a polyphenolic plant flavonoid, has been reported to suppress tumour proliferation through inducing apoptosis and autophagy via MAPK activation in glioma. However, the mechanism of luteolin on suppressing lung cancer growth is still unclear. We found that luteolin targeted LIMK1 from the in silico screening and significantly inhibited the LIMK1 kinase activity, which was confirmed with pull-down binding assay and computational docking models. Treatment with luteolin inhibited lung cancer cells anchorage-independent colony growth and induced apoptosis and cell cycle arrest at G1 phase. Luteolin also decreased the expression of cyclin D1 and increased the levels of cleaved caspase-3 by down-regulating LIMK1 signalling related targets, including p-LIMK and p-cofilin. Furthermore, luteolin suppressed the lung cancer patient-derived xenograft tumour growth by decreasing Ki-67, p-LIMK and p-cofilin expression in vivo. Taken together, these results provide insight into the mechanism that underlies the anticancer effects of luteolin on lung cancer, which involved in down-regulation of LIMK1 and its interaction with cofilin. It also provides valuable evidence for translation towards lung cancer clinical trials with luteolin.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Quinasas Lim/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Luteolina/farmacología , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Técnicas In Vitro , Quinasas Lim/genética , Quinasas Lim/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Br J Cancer ; 124(5): 995-1008, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33311589

RESUMEN

BACKGROUND: SPOP, an E3 ubiquitin ligase adaptor, can act either as a tumour suppressor or a tumour promoter. In prostate cancer (PCa), it inhibits tumorigenesis by degrading several oncogenic substrates. SPOP is the most altered gene in PCa (~15%), which renders it ineffective, promoting cancer. The remaining PCa tumours, which retain WT-SPOP, still progress to castration-resistant (CRPC) stage, indicating that other critical mechanisms exist for downregulating SPOP. SPOP is reduced in ~94% of WT-SPOP-bearing prostate tumours; however, no molecular mechanism is known for its downregulation. METHODS: SPOP was identified as a direct target of LIMK2 using an innovative technique. The reciprocal relationship between SPOP and LIMK2 and its consequences on oncogenicity were analysed using a variety of biochemical assays. To probe this relationship in vivo, xenograft studies were conducted. RESULTS: LIMK2 degrades SPOP by direct phosphorylation at three sites. SPOP promotes LIMK2's ubiquitylation, creating a feedback loop. SPOP's degradation stabilises AR, ARv7 and c-Myc promoting oncogenicity. Phospho-resistant SPOP completely suppresses tumorigenesis in vivo, indicating that LIMK2-mediated SPOP degradation is a key event in PCa progression. CONCLUSIONS: While genomically altered SPOP-bearing tumours require gene therapy, uncovering LIMK2-SPOP relationship provides a powerful opportunity to retain WT-SPOP by inhibiting LIMK2, thereby halting disease progression.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Quinasas Lim/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Proteínas Represoras/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Humanos , Quinasas Lim/genética , Masculino , Ratones , Ratones Desnudos , Mutación , Proteínas Nucleares/genética , Fosforilación , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Proteínas Represoras/genética , Células Tumorales Cultivadas , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
16.
FASEB J ; 34(8): 10948-10965, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32598099

RESUMEN

Nogo-A is a key inhibitory molecule to axon regeneration, and plays diverse roles in other pathological conditions, such as stroke, schizophrenia, and neurodegenerative diseases. Nogo-66 and Nogo-Δ20 fragments are two known functional domains of Nogo-A, which act through the Nogo-66 receptor (NgR1) and sphingosine-1-phosphate receptor 2 (S1PR2), respectively. Here, we reported a new functional domain of Nogo-A, Nogo-A aa 846-861, was identified in the Nogo-A-specific segment that promotes complete Freund's adjuvant (CFA)-induced inflammatory pain. Intrathecal injection of its antagonist peptide 846-861PE or the specific antibody attenuated the CFA-induced inflammatory heat hyperalgesia. The 846-861 PE reduced the content of transient receptor potential vanilloid subfamily member 1 (TRPV1) in dorsal root ganglia (DRG) and decreased the response of DRG neurons to capsaicin. These effects were accompanied by a reduction in LIMK/cofilin phosphorylation and actin polymerization. GST pull-down and fluorescence resonance energy transfer (FRET) assays both showed that Nogo-A aa 846-861 bound to NgR1. Moreover, we demonstrated that Nogo-A aa 846-861 inhibited neurite outgrowth from cortical neurons and DRG explants. We concluded that Nogo-A aa 846-861 is a novel ligand of NgR1, which activates the downstream signaling pathways that inhibit axon growth and promote inflammatory pain.


Asunto(s)
Inflamación/metabolismo , Regeneración Nerviosa/fisiología , Neuritas/metabolismo , Proyección Neuronal/fisiología , Proteínas Nogo/metabolismo , Receptor Nogo 1/metabolismo , Dolor/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Animales , Axones/metabolismo , Axones/fisiología , Línea Celular , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/patología , Inflamación/patología , Quinasas Lim/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Neuritas/patología , Neurogénesis/fisiología , Neuronas/metabolismo , Neuronas/patología , Dolor/patología , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPV/metabolismo
17.
Exp Cell Res ; 388(1): 111822, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31926945

RESUMEN

Actin filaments are highly dynamic structures involved in many cellular processes including cell-to-cell/substrate association and cell motility. The actin cytoskeleton is tightly regulated by actin-binding proteins, which include the members of the ADF (actin-depolymerizing factor)/cofilin family. The members of the LIM kinase family of proteins (LIMK1 and 2) regulate actin dynamics by controlling the binding affinity of ADF/cofilin towards actin. LIMK2 has two major splice variants, LMK2a and LIMK2b. We have generated mice lacking LIMK2a expression (LIMK2a KO), to study its specific role in the regulation of the actin cytoskeleton. The LIMK2a KO mice showed a significant prolonged bleeding complication upon injuries compared to wild type mice. This prolonged bleeding prompted us to check the expression of the LIMK2 protein in platelets as it was previously suggested that it is not expressed in platelets. We showed that human and mouse express LIMK2 in platelets and using our LIMK2a KO mice we have identified a potential key role for LIMK2 in platelet functions including platelet spreading, aggregation and thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Quinasas Lim/metabolismo , Agregación Plaquetaria , Citoesqueleto de Actina/metabolismo , Animales , Plaquetas/fisiología , Células Cultivadas , Humanos , Quinasas Lim/genética , Ratones , Ratones Endogámicos C57BL
18.
Chem Pharm Bull (Tokyo) ; 69(4): 325-332, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790078

RESUMEN

Chemoresistance is one of the main factors of treatment failure of cervical cancer (CC). Here, we intended to discover the role and mechanism of miR-509-5p in the paclitaxel chemoresistance of CC cells. RT-PCR was conducted to verify miR-509-3p expression. HCC94 and C-33A paclitaxel-resistant CC cell models were constructed. Additionally, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry were performed to verify the viability and apoptosis of HCC94 and C-33A cells after upregulating miR-509-3p. Besides, the downstream target of miR-509-3p was analyzed by bioinformatics, and the targeted relationship between miR-509-3p and RAC1 was identified by the dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Further, the expression of apoptotic proteins (Bcl2, Bax, and Caspase3) and the RAC1/PAK1/LIMK1/Cofilin pathway was monitored by Western blot. The result showed that upregulating miR-509-3p markedly inhibited the viability and promoted the apoptosis of CC cells. On the other hand, miR-509-3p was distinctly downregulated in paclitaxel-resistant HCC94 and C-33A cells (vs. normal cells). The transfection of miR-509-3p mimics notably increased their sensitivity to paclitaxel. Meanwhile, RAC1 was found as the potential target of miR-509-3p in bioinformatics analysis. Moreover, the RAC1/p21 (RAC1) activated kinase 1 (PAK1)/LIM kinase 1 (LIMK1)/Cofilin pathway was significantly activated in paclitaxel-resistant HCC94 and C-33A cells, while miR-509-3p overexpression significantly inactivated this pathway. Additionally, downregulation of RAC1 also partly reversed the paclitaxel-resistance of CC cells and inhibited PAK1/LIMK1/Cofilin. All in all, miR-509-3p enhances the apoptosis and chemosensitivity of CC cells by regulating the RAC1/PAK1/LIMK1/Cofilin pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Resistencia a Antineoplásicos , MicroARNs/genética , Paclitaxel/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cofilina 1/genética , Cofilina 1/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Quinasas Lim/genética , Quinasas Lim/metabolismo , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Neoplasias del Cuello Uterino/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
19.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445542

RESUMEN

Endoglin (Eng) is an endothelial cell (EC) transmembrane glycoprotein involved in adhesion and angiogenesis. Eng mutations result in vessel abnormalities as observed in hereditary hemorrhagic telangiectasia of type 1. The role of Eng was investigated in endothelial functions and permeability under inflammatory conditions, focusing on the actin dynamic signaling pathway. Endothelial Colony-Forming Cells (ECFC) from human cord blood and mouse lung/aortic EC (MLEC, MAEC) from Eng+/+ and Eng+/- mice were used. ECFC silenced for Eng with Eng-siRNA and ctr-siRNA were used to test tubulogenesis and permeability +/- TNFα and +/- LIM kinase inhibitors (LIMKi). In silico modeling of TNFα-Eng interactions was carried out from PDB IDs 5HZW and 5HZV. Calcium ions (Ca2+) flux was studied by Oregon Green 488 in epifluorescence microscopy. Levels of cofilin phosphorylation and tubulin post-translational modifications were evaluated by Western blot. F-actin and actin-tubulin distribution/co-localization were evaluated in cells by confocal microscopy. Eng silencing in ECFCs resulted in a decrease of cell sprouting by 50 ± 15% (p < 0.05) and an increase in pseudo-tube width (41 ± 4.5%; p < 0.001) compared to control. Upon TNFα stimulation, ECFC Eng-siRNA displayed a significant higher permeability compared to ctr-siRNA (p < 0.01), which is associated to a higher Ca2+ mobilization (p < 0.01). Computational analysis suggested that Eng mitigated TNFα activity. F-actin polymerization was significantly increased in ECFC Eng-siRNA, MAEC+/-, and MLEC+/- compared to controls (p < 0.001, p < 0.01, and p < 0.01, respectively) as well as actin/tubulin distribution (p < 0.01). Furthermore, the inactive form of cofilin (P-cofilin at Ser3) was significantly decreased by 36.7 ± 4.8% in ECFC Eng-siRNA compared to ctr-siRNA (p < 0.001). Interestingly, LIMKi reproduced the absence of Eng on TNFα-induced ECFC-increased permeability. Our data suggest that Eng plays a critical role in the homeostasis regulation of endothelial cells under inflammatory conditions (TNFα), and loss of Eng influences ECFC-related permeability through the LIMK/cofilin/actin rearrangement-signaling pathway.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Permeabilidad de la Membrana Celular , Endoglina/metabolismo , Células Endoteliales/patología , Inflamación/patología , Quinasas Lim/metabolismo , Neovascularización Patológica/patología , Factores Despolimerizantes de la Actina/genética , Animales , Endoglina/genética , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Quinasas Lim/genética , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo
20.
Zhonghua Gan Zang Bing Za Zhi ; 29(5): 427-432, 2021 May 20.
Artículo en Zh | MEDLINE | ID: mdl-34107579

RESUMEN

Objective: To study LIM kinase 1 (LIMK1) expressional condition, and its regulatory effects on the proliferation and metastasis of hepatocellular carcinoma cells and tissues. Methods: The online database starBase v3.0 and GEPIA were used to analyze the LIMK1 expression in hepatocellular carcinoma cells and normal liver tissues, and then the relevant survival analysis was performed. LIMK1 expression in hepatocellular carcinoma cell line was analyzed by Western blot. Hep3B and Huh7 cells were transiently transfected after LIMK1 protein expression was down-regulated by small interfering RNA (siRNA). LIMK1 effects on the proliferation of Hep3B and Huh7 cells were observed by MTT assay and colony formation assay. Transwell assay was used to detect the change in metastatic ability of hepatocellular carcinoma cell after the down-regulation of LIMK1 expression. Western blot was used to detect the changes of related indexes in the process of epithelial mesenchymal transition after the down-regulation of LIMK1 expression. Data were analyzed by one-way ANOVA. Results: The expression level of LIMK1 in liver cancer tissues was significantly higher than that of normal liver tissues, and was related with prognosis (P ​< 0.01). Furthermore, LIMK1 expression in HCC cell lines was significantly higher than that of immortalized liver L02 cells (P < 0.05). Functional correlated experiment showed that the proliferation and metastatic ability of liver cancer cells were significantly inhibited after LIMK1 expression down-regulation (P < 0.05). Simultaneously, LIMK1 was also involved in the process of epithelial-mesenchymal transition. Conclusion: LIMK1 was overexpressed in HCC tissues and cells, and may regulate the proliferation and metastasis of HCC cells and participate in epithelial-mesenchymal transition process.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Quinasas Lim/genética , Quinasas Lim/metabolismo , Neoplasias Hepáticas/genética , Invasividad Neoplásica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA