Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 43(1): 363-377, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38012357

RESUMEN

This comprehensive review explores vimentin as a pivotal therapeutic target in cancer treatment, with a primary focus on mitigating metastasis and overcoming drug resistance. Vimentin, a key player in cancer progression, is intricately involved in processes such as epithelial-to-mesenchymal transition (EMT) and resistance mechanisms to standard cancer therapies. The review delves into diverse vimentin inhibition strategies. Precision tools, including antibodies and nanobodies, selectively neutralize vimentin's pro-tumorigenic effects. DNA and RNA aptamers disrupt vimentin-associated signaling pathways through their adaptable binding properties. Innovative approaches, such as vimentin-targeted vaccines and microRNAs (miRNAs), harness the immune system and post-transcriptional regulation to combat vimentin-expressing cancer cells. By dissecting vimentin inhibition strategies across these categories, this review provides a comprehensive overview of anti-vimentin therapeutics in cancer treatment. It underscores the growing recognition of vimentin as a pivotal therapeutic target in cancer and presents a diverse array of inhibitors, including antibodies, nanobodies, DNA and RNA aptamers, vaccines, and miRNAs. These multifaceted approaches hold substantial promise for tackling metastasis and overcoming drug resistance, collectively presenting new avenues for enhanced cancer therapy.


Asunto(s)
Aptámeros de Nucleótidos , MicroARNs , Anticuerpos de Dominio Único , Vacunas , Humanos , Aptámeros de Nucleótidos/farmacología , Aptámeros de Nucleótidos/uso terapéutico , Resistencia a Medicamentos , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , Metástasis de la Neoplasia , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/uso terapéutico , Vacunas/farmacología , Vacunas/uso terapéutico , Vimentina/antagonistas & inhibidores , Vimentina/genética , Vimentina/metabolismo
2.
Mol Ther ; 31(10): 3015-3033, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37641404

RESUMEN

Pirfenidone and nintedanib are only anti-pulmonary fibrosis (PF) drugs approved by the FDA. However, they are not target specific, and unable to modify the disease status. Therefore, it is still desirable to discover more effective agents against PF. Vimentin (VIM) plays key roles in tissue regeneration and wound healing, but its molecular mechanism remains unknown. In this work, we demonstrated that atractylodinol (ATD) significantly inhibits TGF-ß1-induced epithelial-mesenchymal transition and fibroblast-to-myofibroblast transition in vitro. ATD also reduces bleomycin-induced lung injury and fibrosis in mice models. Mechanistically, ATD inhibited TGF-ß receptor I recycling by binding to VIM (KD = 454 nM) and inducing the formation of filamentous aggregates. In conclusion, we proved that ATD (derived from Atractylodes lancea) modified PF by targeting VIM and inhibiting the TGF-ß/Smad signaling pathway. Therefore, VIM is a druggable target and ATD is a proper drug candidate against PF. We prove a novel VIM function that TGF-ß receptor I recycling. These findings paved the way to develop new targeted therapeutics against PF.


Asunto(s)
Fibrosis Pulmonar , Animales , Ratones , Bleomicina , Transición Epitelial-Mesenquimal , Pulmón/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/prevención & control , Receptor Tipo I de Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/antagonistas & inhibidores , Vimentina/metabolismo
3.
Small ; 18(6): e2105640, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34866333

RESUMEN

Infection of human cells by pathogens, including SARS-CoV-2, typically proceeds by cell surface binding to a crucial receptor. The primary receptor for SARS-CoV-2 is the angiotensin-converting enzyme 2 (ACE2), yet new studies reveal the importance of additional extracellular co-receptors that mediate binding and host cell invasion by SARS-CoV-2. Vimentin is an intermediate filament protein that is increasingly recognized as being present on the extracellular surface of a subset of cell types, where it can bind to and facilitate pathogens' cellular uptake. Biophysical and cell infection studies are done to determine whether vimentin might bind SARS-CoV-2 and facilitate its uptake. Dynamic light scattering shows that vimentin binds to pseudovirus coated with the SARS-CoV-2 spike protein, and antibodies against vimentin block in vitro SARS-CoV-2 pseudovirus infection of ACE2-expressing cells. The results are consistent with a model in which extracellular vimentin acts as a co-receptor for SARS-CoV-2 spike protein with a binding affinity less than that of the spike protein with ACE2. Extracellular vimentin may thus serve as a critical component of the SARS-CoV-2 spike protein-ACE2 complex in mediating SARS-CoV-2 cell entry, and vimentin-targeting agents may yield new therapeutic strategies for preventing and slowing SARS-CoV-2 infection.


Asunto(s)
Unión Proteica , SARS-CoV-2 , Vimentina , Anticuerpos/farmacología , COVID-19 , Humanos , Glicoproteína de la Espiga del Coronavirus , Vimentina/antagonistas & inhibidores , Vimentina/metabolismo
4.
J Biol Chem ; 295(44): 15013-15028, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32855235

RESUMEN

The antimalarial agents artemisinins inhibit cytomegalovirus (CMV) in vitro and in vivo, but their target(s) has been elusive. Using a biotin-labeled artemisinin, we identified the intermediate filament protein vimentin as an artemisinin target, validated by detailed biochemical and biological assays. We provide insights into the dynamic and unique modulation of vimentin, depending on the stage of human CMV (HCMV) replication. In vitro, HCMV entry and viral progeny are reduced in vimentin-deficient fibroblasts, compared with control cells. Similarly, mouse CMV (MCMV) replication in vimentin knockout mice is significantly reduced compared with controls in vivo, confirming the requirement of vimentin for establishment of infection. Early after HCMV infection of human foreskin fibroblasts vimentin level is stable, but as infection proceeds, vimentin is destabilized, concurrent with its phosphorylation and virus-induced calpain activity. Intriguingly, in vimentin-overexpressing cells, HCMV infection is reduced compared with control cells. Binding of artesunate, an artemisinin monomer, to vimentin prevents virus-induced vimentin degradation, decreasing vimentin phosphorylation at Ser-55 and Ser-83 and resisting calpain digestion. In vimentin-deficient fibroblasts, the anti-HCMV activity of artesunate is reduced compared with controls. In summary, an intact and stable vimentin network is important for the initiation of HCMV replication but hinders its completion. Artesunate binding to vimentin early during infection stabilizes it and antagonizes subsequent HCMV-mediated vimentin destabilization, thus suppressing HCMV replication. Our target discovery should enable the identification of vimentin-binding sites and compound moieties for binding.


Asunto(s)
Antivirales/farmacología , Artemisininas/farmacología , Citomegalovirus/efectos de los fármacos , Vimentina/antagonistas & inhibidores , Antivirales/química , Antivirales/metabolismo , Artemisininas/química , Artemisininas/metabolismo , Artesunato/farmacología , Sitios de Unión , Calpaína/metabolismo , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/metabolismo , Reposicionamiento de Medicamentos , Humanos , Espectrometría de Masas , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Vimentina/genética , Vimentina/metabolismo , Replicación Viral/efectos de los fármacos , Witanólidos/farmacología
5.
Mol Pharm ; 18(11): 4140-4147, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34657437

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with poor prognosis. Evidence has shown that vimentin is a key regulator of lung fibrogenesis. 99mTc-labeled N-acetylglucosamine-polyethyleneimine (NAG-PEI), a vimentin-targeting radiotracer, was used for the early diagnosis of IPF, and NAG-PEI was also used as a therapeutic small interfering RNA (siRNA) delivery vector for the treatment of IPF in this study. Single-photon emission-computed tomography (SPECT) imaging of bleomycin (BM)- and silica-induced IPF mice with 99mTc-labeled NAG-PEI was performed to visualize pulmonary fibrosis and monitor the treatment efficiency of siRNA-loaded NAG-PEI, lipopolysaccharide (LPS, a tolerogenic adjuvant), or zymosan (ZYM, an immunostimulant). The lung uptakes of 99mTc-NAG-PEI in the BM- and silica-induced IPF mice were clearly and directly correlated with IPF progression. The lung uptake of 99mTc-NAG-PEI in the NAG-PEI/TGF-ß1-siRNA treatment group or LPS treatment group was evidently lower than that in the control group, while the lung uptake of 99mTc-NAG-PEI was significantly higher in the ZYM treatment group compared to that in the control group. These results demonstrate that NAG-PEI is a potent MicroSPECT imaging-guided theranostic platform for IPF diagnosis and therapy.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , ARN Interferente Pequeño/administración & dosificación , Radiofármacos/administración & dosificación , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Acetilglucosamina/administración & dosificación , Acetilglucosamina/química , Animales , Biodiversidad , Bleomicina/administración & dosificación , Bleomicina/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/patología , Pulmón/diagnóstico por imagen , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Polietileneimina/administración & dosificación , Polietileneimina/química , ARN Interferente Pequeño/genética , Radiofármacos/química , Radiofármacos/farmacocinética , Dióxido de Silicio/administración & dosificación , Dióxido de Silicio/toxicidad , Tecnecio , Tomografía Computarizada de Emisión de Fotón Único , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo
6.
Cell Biochem Funct ; 39(6): 813-820, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34128234

RESUMEN

Prostate cancer (PCa) is the second most prevalent cancer in men worldwide. Most cases of death from PCa are due to metastasis. Early stages of metastasis are mediated by epithelial-mesenchymal transition (EMT) process through which cancer cells acquire motility and invasive characteristics. Thus, more potent and novel therapeutic strategies must be designed based on the inhibition of EMT or metastasis. Herein, we employ a co-culture system to evaluate the anti-EMT effects of human amniotic mesenchymal stromal cells (hAMSCs) on LNCaP PCa cells. The RNA of treated (sample) and untreated cancer cells (control) and whole-cell lysates of related cells were prepared and analysed through quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, respectively. Based on the results, the expression of vimentin, Snail and Zeb1 in LNCaP cells decreased and the expression of E-cadherin increased after treatment with hAMSCs. Furthermore, induction of the cellular apoptosis in LNCaP cells was detected. The anti-cancer activity of conditioned medium from hAMSCs was shown using hanging drop technique (a 3D cell culture model). Our findings support the idea that stem cells can be considered as a novel therapeutic approach to inhibit prostate cancer cells. SIGNIFICANCE OF THE STUDY: The anti-tumour activity of hAMSCs on LNCaP prostate cancer cells using 2D and 3D cell culture models via induction of apoptosis, suppression of EMT process and down-regulation of EGFR was shown. The results of the present study support this idea that hAMSCs may be a potent therapeutic tool to suppress tumour growth in LNCaP prostate cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/antagonistas & inhibidores , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Regulación hacia Abajo/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Células Tumorales Cultivadas , Vimentina/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
7.
Biochem Biophys Res Commun ; 533(3): 338-345, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-32958256

RESUMEN

We have previously demonstrated that the peptide mimicking small extracellular ring domain of CD82 (CD82EC1-mP) could inhibit tumor cell motility and metastasis. However, its acting mechanism is not understood. Here, we reported that the cell motility-inhibitory function of CD82EC1-mP was involved in the downregulation of epithelial-mesenchymal transition (EMT). Both vimentin and E-cadherin are EMT makers. We found that CD82EC1-mP could inhibit the expression of vimentin, but promot the expression of E-cadherin, suggesting that CD82EC1-mP suppressed EMT. Hippo/YAP and Wnt/ß-catenin are both key signal pathways that regulate the EMT process. The futher studies showed that CD82EC1-mP couled activate GSK3ß, promote the phosphorylation of ß-catenin, and inhibit the ß-catenin nuclear location. Moreover, CD82EC1-mP couled activate Hipoo kinase cascade, promote the phosphorylation of YAP, and inhibit the YAP nuclear location. These results suggested that CD82EC1-mP inhibited invation and matestasis via inhibiting EMT through downregulating Wnt pathway and upregulating Hippo pathway.


Asunto(s)
Antineoplásicos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Proteína Kangai-1/genética , Péptidos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Vía de Señalización Wnt/efectos de los fármacos , Células A549 , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/síntesis química , Cadherinas/agonistas , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Vía de Señalización Hippo , Humanos , Proteína Kangai-1/antagonistas & inhibidores , Proteína Kangai-1/química , Proteína Kangai-1/metabolismo , Imitación Molecular , Células PC-3 , Péptidos/síntesis química , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vimentina/antagonistas & inhibidores , Vimentina/genética , Vimentina/metabolismo , Proteínas Señalizadoras YAP , beta Catenina/genética , beta Catenina/metabolismo
8.
FASEB J ; 32(5): 2841-2854, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29401610

RESUMEN

Vimentin is a cytoskeletal intermediate filament protein that is expressed in mesenchymal cells and cancer cells during the epithelial-mesenchymal transition. The goal of this study was to identify vimentin-targeting small molecules by using the Tocriscreen library of 1120 biochemically active compounds. We monitored vimentin filament reorganization and bundling in adrenal carcinoma SW13 vimentin-positive (SW13-vim+) cells via indirect immunofluorescence. The screen identified 18 pharmacologically diverse hits that included 2 statins-simvastatin and mevastatin. Simvastatin induced vimentin reorganization within 15-30 min and significant perinuclear bundling within 60 min (IC50 = 6.7 nM). Early filament reorganization coincided with increased vimentin solubility. Mevastatin produced similar effects at >1 µM, whereas the structurally related pravastatin and lovastatin did not affect vimentin. In vitro vimentin filament assembly assays revealed a direct targeting mechanism, as determined biochemically and by electron microscopy. In SW13-vim+ cells, simvastatin, but not pravastatin, reduced total cell numbers (IC50 = 48.1 nM) and promoted apoptosis after 24 h. In contrast, SW13-vim- cell viability was unaffected by simvastatin, unless vimentin was ectopically expressed. Simvastatin similarly targeted vimentin filaments and induced cell death in MDA-MB-231 (vim+), but lacked effect in MCF7 (vim-) breast cancer cells. In conclusion, this study identified vimentin as a direct molecular target that mediates simvastatin-induced cell death in 2 different cancer cell lines.-Trogden, K. P., Battaglia, R. A., Kabiraj, P., Madden, V. J., Herrmann, H., Snider, N. T. An image-based small-molecule screen identifies vimentin as a pharmacologically relevant target of simvastatin in cancer cells.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Simvastatina/farmacología , Vimentina/metabolismo , Neoplasias de las Glándulas Suprarrenales/metabolismo , Neoplasias de las Glándulas Suprarrenales/ultraestructura , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/ultraestructura , Muerte Celular , Femenino , Humanos , Lovastatina/análogos & derivados , Lovastatina/farmacología , Células MCF-7 , Microscopía Fluorescente , Proteínas de Neoplasias/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores
9.
Gene Ther ; 25(5): 321-330, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29925853

RESUMEN

Recent researches have reported that long noncoding RNA (lncRNA) five prime to Xist (FTX) plays a crucial role in the initiation and progression of cancers. In the current study, the clinical significance and functional roles of lncRNA FTX in colorectal cancer (CRC) progression were investigated. A significant increase of lncRNA FTX expression in CRC tissue and cell lines was observed. Overexpression of lncRNA FTX was significantly associated with the bigger tumor diameter, the advanced TNM stage, the lymph node, and distant metastasis, and also predicted poor prognosis of patients with CRC. Functional analyses demonstrated that knockdown of lncRNA FTX markedly inhibited CRC cell proliferation, migration, and invasion in vitro. Mechanistically, FTX directly interacted with miR-215 and suppressed miR-215 expression. FTX also bind to vimentin and reduced its phosphorylation level on Ser83 in CRC cells. Finally, using siRNAs against lncRNA FTX could dramatically inhibit CRC growth and distant metastasis in vivo. Taken together, our data demonstrated an oncogenic role of lncRNA FTX in CRC tumorigenesis and progression via interaction with miR-215 and vimentin. Then, a promising therapeutic target for CRC was provided.


Asunto(s)
Neoplasias Colorrectales/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Vimentina/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Fosforilación , ARN Largo no Codificante/genética , Vimentina/antagonistas & inhibidores , Vimentina/genética
10.
Br J Cancer ; 118(2): 233-247, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29123267

RESUMEN

BACKGROUND: Bactericidal/Permeability-increasing-fold-containing family B member 1 (BPIFB1, previously termed LPLUNC1) is highly expressed in the nasopharynx, significantly downregulated in nasopharyngeal carcinoma (NPC), and associated with prognosis in NPC patients. Because metastasis represents the primary cause of NPC-related death, we explored the role of BPIFB1 in NPC migration and invasion. METHODS: The role of BPIFB1 in NPC metastasis was investigated in vitro and in vivo. A co-immunoprecipitation assay coupled with mass spectrometry was used to identify BPIFB1-binding proteins. Additionally, western blotting, immunofluorescence, and immunohistochemistry allowed assessment of the molecular mechanisms associated with BPIFB1-specific metastatic inhibition via vitronectin (VTN) and vimentin (VIM) interactions. RESULTS: Our results showed that BPIFB1 expression markedly inhibited NPC cell migration, invasion, and lung-metastatic abilities. Additionally, identification of two BPIFB1-interacting proteins, VTN and VIM, showed that BPIFB1 reduced VTN expression and the formation of a VTN-integrin αV complex in NPC cells, leading to inhibition of the FAK/Src/ERK signalling pathway. Moreover, BPIFB1 attenuated NPC cell migration and invasion by inhibiting VTN- or VIM-induced epithelial-mesenchymal transition. CONCLUSIONS: This study represents the first demonstration of BPIFB1 function in NPC migration, invasion, and lung metastasis. Our findings indicate that re-expression of BPIFB1 might represent a useful strategy for preventing and treating NPC.


Asunto(s)
Autoantígenos/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas/metabolismo , Vimentina/metabolismo , Vitronectina/metabolismo , Animales , Autoantígenos/biosíntesis , Línea Celular Tumoral , Movimiento Celular/fisiología , Proteínas de Unión a Ácidos Grasos , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Desnudos , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Vimentina/antagonistas & inhibidores , Vimentina/biosíntesis , Vitronectina/antagonistas & inhibidores , Vitronectina/biosíntesis , Familia-src Quinasas/metabolismo
11.
Nat Chem Biol ; 10(11): 924-926, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25218743

RESUMEN

The tumor suppressor protein prostate apoptosis response-4 (Par-4), which is secreted by normal cells, selectively induces apoptosis in cancer cells. We identified a 3-arylquinoline derivative, designated Arylquin 1, as a potent Par-4 secretagogue in cell cultures and mice. Mechanistically, Arylquin 1 binds vimentin, displaces Par-4 from vimentin for secretion and triggers the efficient paracrine apoptosis of diverse cancer cells. Thus, targeting vimentin with Par-4 secretagogues efficiently induces paracrine apoptosis of tumor cells.


Asunto(s)
Aminoquinolinas/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias/patología , Vimentina/antagonistas & inhibidores , Aminoquinolinas/administración & dosificación , Aminoquinolinas/química , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Comunicación Paracrina/efectos de los fármacos , Vimentina/química
12.
Mol Pharmacol ; 87(3): 378-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25488183

RESUMEN

Gefitinib is widely used for the treatment of lung cancer in patients with sensitizing epidermal growth factor receptor mutations, but patients tend to develop resistance after an average of 10 months. Low molecular weight heparins, such as enoxaparin, potently inhibit experimental metastasis. This study aimed to determine the potential of combined enoxaparin and gefitinib (enoxaparin + gefitinib) treatment to inhibit tumor resistance to gefitinib both in vitro and in vivo. A549 and H1975 cell migration was analyzed in wound closure and Transwell assays. Akt and extracellular signal-related kinase 1/2 signaling pathways were identified, and a proteomics analysis was conducted using SDS-PAGE/liquid chromatography-tandem mass spectrometry analysis. Molecular interaction networks were visualized using the Cytoscape bioinformatics platform. Protein expression of dedicator of cytokinesis 1 (DOCK1) and cytoskeleton intermediate filament vimentin were identified using an enzyme-linked immunosorbent assay, Western blot, and small interfering RNA transfection of A549 cells. In xenograft A549-luc-C8 tumors in nude mice, enoxaparin + gefitinib inhibited tumor growth and reduced lung colony formation compared with gefitinib alone. Furthermore, the combination had stronger inhibitory effects on cell migration than either agent used individually. Additional enoxaparin administration resulted in better effective inhibition of Akt activity compared with gefitinib alone. Proteomics and network analysis implicated DOCK1 as the key node molecule. Western blot verified the effective inhibition of the expression of DOCK1 and vimentin phosphorylation by enoxaparin + gefitinib compared with gefitinib alone. DOCK1 knockdown confirmed its role in cell migration, Akt expression, and vimentin phosphorylation. Our data indicate that enoxaparin sensitizes gefitinib antitumor and antimigration activity in lung cancer by suppressing DOCK1 expression, Akt activity, and vimentin phosphorylation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Enoxaparina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/administración & dosificación , Vimentina/metabolismo , Proteínas de Unión al GTP rac/biosíntesis , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Gefitinib , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rac/antagonistas & inhibidores
13.
J Biol Chem ; 289(10): 6960-6968, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24474691

RESUMEN

Nuclear reprogramming of somatic cells can be induced by oocyte factors. Despite numerous attempts, the factors responsible for successful nuclear reprogramming remain elusive. In the present study, we found that porcine oocytes with the first polar body collected at 42 h of in vitro maturation had a stronger ability to support early development of cloned embryos than porcine oocytes with the first polar body collected at 33 h of in vitro maturation. To explore the key reprogramming factors responsible for the difference, we compared proteome signatures of the two groups of oocytes. 18 differentially expressed proteins between these two groups of oocytes were discovered by mass spectrometry (MS). Among these proteins, we especially focused on vimentin (VIM). A certain amount of VIM protein was stored in oocytes and accumulated during oocyte maturation, and maternal VIM was specifically incorporated into transferred somatic nuclei during nuclear reprogramming. When maternal VIM function was inhibited by anti-VIM antibody, the rate of cloned embryos developing to blastocysts was significantly lower than that of IgG antibody-injected embryos and non-injected embryos (12.24 versus 22.57 and 21.10%; p < 0.05), but the development of in vitro fertilization and parthenogenetic activation embryos was not affected. Furthermore, we found that DNA double strand breaks dramatically increased and that the p53 pathway was activated in cloned embryos when VIM function was inhibited. This study demonstrates that maternal VIM, as a genomic protector, is crucial for nuclear reprogramming in porcine cloned embryos.


Asunto(s)
Reprogramación Celular , Clonación de Organismos , Oocitos/fisiología , Porcinos/embriología , Vimentina/metabolismo , Animales , Blastocisto/metabolismo , Blastocisto/fisiología , Desarrollo Embrionario , Femenino , Técnicas de Transferencia Nuclear , Oocitos/metabolismo , Cuerpos Polares/metabolismo , Cuerpos Polares/fisiología , Porcinos/genética , Porcinos/metabolismo , Vimentina/antagonistas & inhibidores
14.
Infect Immun ; 83(2): 625-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25422262

RESUMEN

"Mycobacterium avium subsp. hominissuis" is an opportunistic environmental pathogen that causes respiratory illness in immunocompromised patients, such as those with cystic fibrosis as well as other chronic respiratory diseases. Currently, there is no efficient approach to prevent or treat M. avium subsp. hominissuis infection in the lungs. During initial colonization of the airways, M. avium subsp. hominissuis forms microaggregates composed of 3 to 20 bacteria on human respiratory epithelial cells, which provides an environment for phenotypic changes leading to efficient mucosal invasion in vitro and in vivo. DNA microarray analysis was employed to identify genes associated with the microaggregate phenotype. The gene encoding microaggregate-binding protein 1 (MBP-1) (MAV_3013) is highly expressed during microaggregate formation. When expressed in noninvasive Mycobacterium smegmatis, MBP-1 increased the ability of the bacteria to bind to HEp-2 epithelial cells. Using anti-MBP-1 immune serum, microaggregate binding to HEp-2 cells was significantly reduced. By far-Western blotting, and verified by coimmunoprecipitation, we observed that MBP-1 interacts with the host cytoskeletal protein vimentin. As visualized by confocal microscopy, microaggregates, as well as MBP-1, induced vimentin polymerization at the site of bacterium-host cell contact. Binding of microaggregates to HEp-2 cells was inhibited by treatment with an antivimentin antibody, suggesting that MBP-1 expression is important for M. avium subsp. hominissuis adherence to the host cell. MBP-1 immune serum significantly inhibited M. avium subsp. hominissuis infection throughout the respiratory tracts of mice. This study characterizes a pathogenic mechanism utilized by M. avium subsp. hominissuis to bind and invade the host respiratory epithelium, suggesting new potential targets for the development of antivirulence therapy.


Asunto(s)
Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Células Epiteliales/microbiología , Mycobacterium avium/patogenicidad , Mucosa Respiratoria/microbiología , Animales , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Células Epiteliales/citología , Femenino , Humanos , Sueros Inmunes/inmunología , Sueros Inmunes/farmacología , Huésped Inmunocomprometido , Ratones , Ratones Endogámicos C57BL , Infecciones por Mycobacterium/microbiología , Infecciones por Mycobacterium/patología , Mycobacterium avium/genética , Mycobacterium avium/inmunología , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Mucosa Respiratoria/citología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/patología , Vimentina/antagonistas & inhibidores , Vimentina/inmunología , Vimentina/metabolismo
15.
Biochem Biophys Res Commun ; 467(2): 310-5, 2015 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-26435505

RESUMEN

Recently, lysine-specific demethylase 1 (LSD1) was identified as the first histone demethylase. LSD1 interacted with androgen receptor (AR) and promoted androgen-dependent transcription of target genes, such as PSA, by ligand-induced demethylation of mono- and dimethylated histone H3 at Lys 9 (H3K9). Meanwhile, the phenomenon of epithelial-mesenchymal transition (EMT) had received considerable attention in tumor recurrence and metastasis. This study examined the effect of Pargyline (an inhibitor of LSD1) on the process of EMT in vitro and in vivo. SCID mice were injected subcutaneously with LNCap cells. Pargyline was given intraperitoneally or not after castration (implemented with Bilateral orchidectomy), then PSA levels in serum and tumor were determined to assess time to androgen-independent progression. The results showed that LSD1 expression was up-regulated when PCa progressed to Castration Resistant Prostate Cancer (CRPC). Pargyline reduced LNCap cells migration and invasion ability, and inhibited the process of EMT by up-regulating expression of E-cadherin, and down-regulating expressions of N-cadherin and Vimentin in vitro and in vivo. Although, Pargyline did not change the level of AR, it reduced PSA expression both in vitro and in vivo. Furthermore, Pargyline delayed prostate cancer transition from androgen-dependent to androgen-independent state (CRPC). These findings indicated that inhibition of LSD1 might be a promise adjunctive therapy with androgen deprivation therapy (ADT) for locally advanced or metastatic prostate cancer.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Histona Demetilasas/antagonistas & inhibidores , Pargilina/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Animales , Antihipertensivos/farmacología , Cadherinas/agonistas , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Reposicionamiento de Medicamentos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Antígeno Prostático Específico/antagonistas & inhibidores , Antígeno Prostático Específico/genética , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Vimentina/antagonistas & inhibidores , Vimentina/genética , Vimentina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Biochem Biophys Res Commun ; 443(1): 1-6, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24269590

RESUMEN

Cell motility and cell stiffness are closely related to metastatic activity of cancer cells. (-)-Epigallocatechin gallate (EGCG) has been shown to inhibit spontaneous metastasis of melanoma cell line into the lungs of mice, so we studied the effects of EGCG on cell motility, cell stiffness, and expression of vimentin and Slug, which are molecular phenotypes of epithelial-mesenchymal transition (EMT). Treatments of human non-small cell lung cancer cell lines H1299 and Lu99 with 50 and 100 µM EGCG reduced cell motility to 67.5% and 43.7% in H1299, and 71.7% and 31.5% in Lu99, respectively in in vitro wound healing assay. Studies on cell stiffness using atomic force microscope (AFM) revealed that treatment with 50 µM EGCG increased Young's modulus of H1299 from 1.24 to 2.25 kPa and that of Lu99 from 1.29 to 2.28 kPa, showing a 2-fold increase in cell stiffness, i.e. rigid elasticity of cell membrane. Furthermore, treatment with 50 µM EGCG inhibited high expression of vimentin and Slug in the cells at a leading edge of scratch. Methyl-ß-cyclodextrin, a reagent to deplete cholesterol in plasma membrane, showed inhibition of EMT phenotypes similar that by EGCG, suggesting that EGCG induces inhibition of EMT phenotypes by alteration of membrane organization.


Asunto(s)
Antineoplásicos/farmacología , Catequina/análogos & derivados , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Metástasis de la Neoplasia/prevención & control , Factores de Transcripción/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores , Animales , Catequina/farmacología , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Colesterol/metabolismo , Módulo de Elasticidad/efectos de los fármacos , Humanos , Ratones , Microscopía de Fuerza Atómica , Factores de Transcripción de la Familia Snail , beta-Ciclodextrinas/farmacología
18.
Nutr Cancer ; 66(2): 285-94, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24447182

RESUMEN

Metastasis and chemoresistance represent two detrimental events that greatly hinder the outcome for those suffering with head and neck squamous cell carcinoma (HNSCC). Herein, we investigated benzyl isothiocyanate's (BITC) ability to inhibit HNSCC migration and invasion and enhance chemotherapy. Our data suggests that treatment with BITC 1) induced significant reductions in the viability of multiple HNSCC cell lines tested (HN12, HN8, and HN30) after 24 and 48 h, 2) decreased migration and invasion of the HN12 cells in a dose dependent manner, and 3) inhibited expression and altered localization of the epithelial-mesenchymal transition (EMT) marker, vimentin. We also observed that a pretreatment of BITC followed by cisplatin treatment 1) induced a greater decrease in HN12, HN30, and HN8 cell viability and total cell count than either treatment alone and 2) significantly increased apoptosis when compared to either treatment alone. Taken together these data suggest that BITC has the capacity to inhibit processes involved in metastasis and enhance the effectiveness of chemotherapy. Consequently, the results indicate that further investigation, including in vivo studies, are warranted.


Asunto(s)
Carcinoma de Células Escamosas/patología , Movimiento Celular/efectos de los fármacos , Cisplatino/farmacología , Neoplasias de Cabeza y Cuello/patología , Isotiocianatos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Vimentina/antagonistas & inhibidores , Vimentina/genética , Vimentina/metabolismo
19.
Eur J Nutr ; 53(3): 843-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24092501

RESUMEN

PURPOSE: D,L-Sulforaphane (SFN) is a promising chemopreventive agent with in vivo efficacy against prostate cancer in experimental rodents. This study was undertaken to determine the role of vimentin and plasminogen activator inhibitor-1 (PAI-1) in anticancer effects of SFN. METHODS: Effect of SFN on levels of different proteins was determined by Western blotting or immunofluorescence microscopy. RNA interference of vimentin and PAI-1 was achieved by transient transfection. Apoptosis was quantified by flow cytometry. Transwell chambers were used to determine cell migration. RESULTS: Exposure of PC-3 and DU145 human prostate cancer cells to SFN resulted in induction of vimentin protein, which was accompanied by down-regulation of E-cadherin protein expression. The SFN-mediated induction of vimentin was also observed in a normal human prostate epithelial cell line. RNA interference of vimentin did not have any appreciable effect on early or late apoptosis resulting from SFN exposure. On the other hand, SFN-mediated inhibition of PC-3 and DU145 cell migration was significantly augmented by knockdown of the vimentin protein. Knockdown of vimentin itself was inhibitory against cell migration. The SFN-treated cells also exhibited induction of PAI-1, which is an endogenous inhibitor of urokinase-type plasminogen activator system. Similar to vimentin, PAI-1 knockdown resulted in a modest augmentation of PC-3 cell migration inhibition by SFN. Tumors from SFN-treated transgenic adenocarcinoma of mouse prostate mice showed a 1.7-fold increase in vimentin protein level compared with control tumors. CONCLUSION: The present study indicates that vimentin and PAI-1 inductions confer modest protection against SFN-mediated inhibition of prostate cancer cell migration.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Anticarcinógenos/farmacología , Isotiocianatos/farmacología , Inhibidor 1 de Activador Plasminogénico/agonistas , Neoplasias de la Próstata/tratamiento farmacológico , Regulación hacia Arriba/efectos de los fármacos , Vimentina/agonistas , Adenocarcinoma/metabolismo , Adenocarcinoma/prevención & control , Animales , Anticarcinógenos/uso terapéutico , Apoptosis/efectos de los fármacos , Cadherinas/antagonistas & inhibidores , Cadherinas/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Isotiocianatos/uso terapéutico , Masculino , Ratones Transgénicos , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Inhibidor 1 de Activador Plasminogénico/química , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Próstata/efectos de los fármacos , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/prevención & control , Interferencia de ARN , Sulfóxidos , Vimentina/antagonistas & inhibidores , Vimentina/genética , Vimentina/metabolismo
20.
Carcinogenesis ; 34(11): 2548-57, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24001454

RESUMEN

Slug and Vimentin genes play a critical role in regulating epithelial-mesenchymal transition (EMT) via downregulation of epithelial markers and upregulation of mesenchymal markers. The present study evaluated the clinical significance of Slug and Vimentin expression as potential disease biomarkers in colorectal cancer (CRC). At first, the biological role of Slug in CRC was assessed by RNA interference in CRC cell lines to assess tumor progression, invasion and migration. Next, we analyzed Slug and Vimentin expression in surgical tissue specimens from 181 CRC patients (Cohort 1) by quantitative real-time reverse transcription-PCR and 208 patients (Cohort 2) by immunohistochemistry. Knockdown of Slug using small interfering RNA in CRC cell lines resulted in inhibition of EMT, reduced cell proliferation, invasion and migration in CRC cells. Interestingly, Slug and Vimentin expression in cancer tissues was significantly higher in patients with higher T stage, lymph node involvement, liver metastasis and advanced tumor node metastasis stages. A significant correlation was observed between Slug and Vimentin expression in CRC (messenger RNA: ρ = 0.546, protein: ρ = 0.405), and increased expression of Slug and Vimentin was significantly associated with poor prognosis. Furthermore, increased expression of Slug emerged as an independent prognostic factor and a predictive marker of lymph node metastasis in CRC patients. Our data provide novel evidence for the biological and clinical significance of Slug and Vimentin expression as potential predictive biomarkers for identifying patients with lymph node metastasis or poor prognosis in CRC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/patología , Factores de Transcripción/metabolismo , Vimentina/metabolismo , Anciano , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Movimiento Celular , Proliferación Celular , Estudios de Cohortes , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Estadificación de Neoplasias , Pronóstico , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Tasa de Supervivencia , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Células Tumorales Cultivadas , Vimentina/antagonistas & inhibidores , Vimentina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA