Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
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.
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
5.
Cells ; 10(9)2021 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-34571970

RESUMEN

Current treatment options for patients infected with hepatitis B virus (HBV) are suboptimal, because the approved drugs rarely induce cure due to the persistence of the viral DNA genome in the nucleus of infected hepatocytes, and are associated with either severe side effects (pegylated interferon-alpha) or require life-long administration (nucleos(t)ide analogs). We report here the evaluation of the safety and therapeutic efficacy of a novel, humanized antibody (hzVSF) in the woodchuck model of HBV infection. hzVSF has been shown to act as a viral entry inhibitor, most likely by suppressing vimentin-mediated endocytosis of virions. Targeting the increased vimentin expression on liver cells by hzVSF after infection with HBV or woodchuck hepatitis virus (WHV) was demonstrated initially. Thereafter, hzVSF safety was assessed in eight woodchucks naïve for WHV infection. Antiviral efficacy of hzVSF was evaluated subsequently in 24 chronic WHV carrier woodchucks by monotreatment with three ascending doses and in combination with tenofovir alafenamide fumarate (TAF). Consistent with the proposed blocking of WHV reinfection, intravenous hzVSF administration for 12 weeks resulted in a modest but transient reduction of viral replication and associated liver inflammation. In combination with oral TAF dosing, the antiviral effect of hzVSF was enhanced and sustained in half of the woodchucks with an antibody response to viral proteins. Thus, hzVSF safely but modestly alters chronic WHV infection in woodchucks; however, as a combination partner to TAF, its antiviral efficacy is markedly increased. The results of this preclinical study support future evaluation of this novel anti-HBV drug in patients.


Asunto(s)
Alanina/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Antivirales/farmacología , Virus de la Hepatitis B de la Marmota/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Hígado/efectos de los fármacos , Tenofovir/análogos & derivados , Vimentina/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Quimioterapia Combinada , Endocitosis/efectos de los fármacos , Células Hep G2 , Hepatitis B/metabolismo , Hepatitis B/virología , Virus de la Hepatitis B de la Marmota/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Hígado/metabolismo , Hígado/virología , Marmota , Tenofovir/farmacología , Vimentina/metabolismo , Carga Viral , Replicación Viral/efectos de los fármacos
6.
Cancer Lett ; 520: 400-408, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34332039

RESUMEN

Epithelial mesenchymal transition (EMT) is highly correlated with metastasis during cancer development. Although previous studies have revealed that ISO is able to inhibit cancer cell invasion and stem-cell properties, little is known about the effects of ISO on EMT markers. The present study explores the potential regulation of ISO on EMT, leading to the inhibition of migration and invasion of bladder cancer cells. We found that ISO inhibited Vimentin, one of the EMT markers, in the invasive bladder cancer cell lines U5637 and T24T. ISO reduced Vimentin protein level by increasing the expression of METTL14. On the other hand, ISO upregulated the METTL14 mRNA by activating the transcription factor FOXO3a. The results demonstrate that ISO inhibits invasion by affecting the EMT marker and offer a novel insight into understanding the upregulation of METTL14 by ISO.


Asunto(s)
Proteína Forkhead Box O3/genética , Metiltransferasas/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Vimentina/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Transducción de Señal/efectos de los fármacos , Estilbenos/farmacología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Vimentina/antagonistas & inhibidores
7.
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
8.
Eur J Med Chem ; 214: 113188, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33550185

RESUMEN

Herein, we describe the design, synthesis and structure-activity relationships of a series of novel s-triazine compounds can induce methuotic phenotype in various types of cancer cells. (E)-1-(4-Chlorophenyl)-3-(4-((4-morpholino-6-styryl-1,3,5-triazine-2-yl)amino)phenyl)urea, compound V6, exhibited a striking methuotic phenotype with a minimal effective concentration of less than 10 nM in U87 glioblastoma cells. Based on structure-activity relationship studies, we designed and synthesized an active probe P1 that retained the full potential of V6 in inducing the methuotic phenotype in U87 glioblastoma cells. Using this probe following affinity-based proteomic profiling strategy, we identified vimentin as the specific target protein of compound V6. Molecular docking revealed that V6 can form hydrogen bonds with vimentin at 273R and 276Y in its rod domain.


Asunto(s)
Antineoplásicos/farmacología , Triazinas/farmacología , Vimentina/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Fenotipo , Relación Estructura-Actividad , Triazinas/síntesis química , Triazinas/química , Células Tumorales Cultivadas , Vimentina/genética , Vimentina/metabolismo
9.
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
10.
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
11.
Sci Rep ; 10(1): 6657, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313093

RESUMEN

As a potential drug/gene delivery system, the ultrasound-targeted microbubble destruction (UTMD) system can be used as a vehicle as well as increasing the permeability of biological barriers to enhance the effect of tumor treatment. However, the effect of UTMD in the tumor EMT process is unknown. In this study, we aimed to investigate the potential and mechanism of UTMD induced oxidative stress in inhibiting EMT of breast cancer. Human breast MDA231 cells were treated with microbubble (MB), ultrasound (US) and UTMD, respectively. The generation of oxidative stress, the levels of miR-200c, ZEB1 and vimentin, and the numbers of migratory cells were evaluated quantitatively and qualitatively by the measurement of intracellular reactive oxygen species (ROS), qRT-PCR, western blot assay, and transwell assay. Then, to evaluate the role of UTMD-induced oxidative stress and miR-200c in the epithelial-mesenchymal transition (EMT) inhibition, the ROS scavenger N-acetyl-L-cysteine (NAC) and miR-200c inhibitor were used before UTMD treatment. We found that UTMD induced oxidative stress, upregulated the expression of miR-200c, downregulated the expression of ZEB1 and vimentin and suppressed the MDA231 cell migration. The addition of NAC and miR-200c inhibitor had an opposite impact on the expression of miR-200c and ZEB1, thus hindered the effects of UTMD on MDA231 cells EMT. In conclusion, UTMD can inhibit the EMT characteristics of MDA231 cells. The mechanism may be related to the regulation of the miR-200c/ZEB1 axis through the generation of ROS induced by UTMD, which may provide a new strategy to prevent the tumor cells EMT under UTMD treatment.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Interferente Pequeño/metabolismo , Vimentina/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Acetilcisteína/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de la radiación , Femenino , Depuradores de Radicales Libres/farmacología , Técnicas de Transferencia de Gen , Humanos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Microburbujas , Estrés Oxidativo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Ondas Ultrasónicas , Vimentina/antagonistas & inhibidores , Vimentina/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/antagonistas & inhibidores , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
13.
Clin Mol Hepatol ; 26(1): 45-53, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31564085

RESUMEN

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is the most common liver cancer with high mortality rate in patients suffering from liver diseases. The drug of choice used in advanced-stage of HCC is sorafenib. However, adaptive resistance has been observed in HCC patients undergoing long-term sorafenib treatment, lowering its effectiveness. Hence, it is important to overcome drug resistance to improve overall management of HCC. Here, we have identified a candidate biomarker for sorafenib resistance in a HCC model cell line, HepG2. METHODS: Initially, comparative proteomic profiling of parental HepG2 [HepG2 (P)] and sorafenib-resistant HepG2 [HepG2 (R)] cells was performed via MALDI (matrix-assisted laser desorption/ionization) which revealed the deregulation of vimentin in HepG2 (R) cells. Gene and protein level expression of vimentin was also observed through quantitative real-time polymerase chain reaction (qRT PCR) and fluorescence-activated cell sorting (FACS), respectively. Furthermore, withaferin A was used to study regulation of vimentin expression and its significance in sorafenib resistance. RESULTS: Both gene and protein level of vimentin expression was found to be downregulated in HepG2 (R) in comparison to HepG2 (P). Interestingly, the study demonstrated that withaferin A further lowered the expression of vimentin in HepG2 (R) cells in a dose-dependent manner. Also, inhibition of vimentin lowered ABCG2 expression and decreased cell viability in parental as well as sorafenib resistant HepG2 cells. CONCLUSION: Hence, our study for the first time highlighted the probable therapeutic potential of vimentin in sorafenib resistant HepG2, a HCC model cell line.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Sorafenib/farmacología , Vimentina/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Vimentina/antagonistas & inhibidores , Witanólidos/farmacología
14.
Oncogene ; 38(4): 455-468, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30111817

RESUMEN

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) is a major advance in treating NSCLC with EGFR-activating mutations. However, acquired resistance, due partially to secondary mutations limits their use. Here we report that NSCLC cells with acquired resistance to gefitinib or osimertinib (AZD9291) exhibit EMT features, with a decrease in E-cadherin, and increases in vimentin and stemness, without possessing any EGFR secondary mutations. Knockdown of E-cadherin in parental cells increased gefitinib resistance and stemness, while knockdown of vimentin in resistant cells resulted in opposite effects. Src activation and Hakai upregulation were found in gefitinib-resistant cells. Knockdown of Hakai elevated E-cadherin expression, attenuated stemness, and resensitized the cells to gefitinib. Clinical cancer specimens with acquired gefitinib resistance also showed a decrease in E-cadherin and an increase in Hakai expression. The dual HDAC and HMGR inhibitor JMF3086 inhibited the Src/Hakai and Hakai/E-cadherin interaction to reverse E-cadherin expression, and attenuated vimentin and stemness to restore gefitinib sensitivity. The EMT features of AZD9291-resistant H1975 cells were related to the upregulation of Zeb1. Both gefitinib and AZD9291 sensitivity was restored by JMF3086 through reversing EMT. Our study not only revealed a common mechanism of EMT in both gefitinib and AZD9291 resistance beyond EGFR mutations per se, but also provides a new strategy to overcome it.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/fisiología , Gefitinib/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de Neoplasias/fisiología , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Acrilamidas , Compuestos de Anilina , Animales , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/antagonistas & inhibidores , Cadherinas/biosíntesis , Cadherinas/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Ensayos de Selección de Medicamentos Antitumorales , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Organismos Libres de Patógenos Específicos , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Vimentina/antagonistas & inhibidores , Vimentina/biosíntesis , Vimentina/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/biosíntesis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
15.
Cancer Lett ; 433: 176-185, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29991446

RESUMEN

Glioblastoma multiforme (GBM) is the most prevalent and aggressive brain tumor. The current standard therapy, which includes radiation and chemotherapy, is frequently ineffective partially because of drug resistance and poor penetration of the blood-brain barrier. Reducing resistance and increasing sensitivity to chemotherapy may improve outcomes. Glioma stem cells (GSCs) are a source of relapse and chemoresistance in GBM; sensitization of GSCs to temozoliomide (TMZ), the primary chemotherapeutic agent used to treat GBM, is therefore integral for therapeutic efficacy. We previously discovered a unique tumor-specific target, cell surface vimentin (CSV), on patient-derived GSCs. In this study, we found that the anti-CSV monoclonal antibody 86C efficiently increased GSC sensitivity to TMZ. The combination TMZ+86C induced significantly greater antitumor effects than TMZ alone in eight of 12 GSC lines. TMZ+86C-sensitive GSCs had higher CSV expression overall and faster CSV resurfacing among CSV- GSCs compared with TMZ+86C-resistant GSCs. Finally, TMZ+86C increased apoptosis of tumor cells and prolonged survival compared with either drug alone in GBM mouse models. The combination of TMZ+86C represents a promising strategy to reverse GSC chemoresistance.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Temozolomida/administración & dosificación , Vimentina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Glioblastoma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Hematol Oncol ; 11(1): 85, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925404

RESUMEN

BACKGROUND: Inhibition of metastasis of head and neck squamous cell carcinoma (HNSCC) is one of the most important challenges in cancer treatment. Src, a non-receptor tyrosine kinase, has been implicated as a key promoter in tumor progression and metastasis of HNSCC. However, Src therapy for HNSCC is limited by lack of efficient in vivo delivery and underlying mechanisms remain elusive. METHODS: Src knockdown cells were achieved by lentiviral-mediated interference. Cell migration and invasion were examined by wound healing and Transwell assays. Protein levels were determined by Western blot and/or immunohistochemistry. The Src inhibitor saracatinib was loaded into self-assembling nanoparticles by the solvent evaporation method. An experimental metastasis mouse model was generated to investigate the drug efficacy in metastasis. RESULTS: Blockade of Src kinase activity by saracatinib effectively suppressed invasion and metastasis of HNSCC. Mechanistic assessment of the drug effects in HNSCC cells showed that saracatinib induced suppression of Src-dependent invasion/metastasis through downregulating the expression levels of Vimentin and Snail proteins. In tests in mice, saracatinib loaded into the novel multifunctional nanoparticles exhibited superior effects on suppression of HNSCC metastasis compared with the free drug, which is mainly attributed to highly specific and efficient tumor-targeted drug delivery system. CONCLUSIONS: These findings and advances are of great importance to the development of Src-targeted nanomedicine as a more effective therapy for metastatic HNSCC.


Asunto(s)
Benzodioxoles/uso terapéutico , Neoplasias de Cabeza y Cuello/patología , Metástasis de la Neoplasia/tratamiento farmacológico , Quinazolinas/uso terapéutico , Familia-src Quinasas/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Benzodioxoles/administración & dosificación , Modelos Animales de Enfermedad , Portadores de Fármacos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Ratones , Nanomedicina/métodos , Nanopartículas/uso terapéutico , Quinazolinas/administración & dosificación , Factores de Transcripción de la Familia Snail/antagonistas & inhibidores , Vimentina/antagonistas & inhibidores
17.
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
18.
Phytomedicine ; 41: 67-73, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29519321

RESUMEN

BACKGROUND: Use of pharmaceutical agent for breast cancer chemotherapy is an interesting method that induces cells death by different way, such as apoptosis. Parthenolide is the main compound in feverfew that has been used to cure migraine and rheumatoid arthritis for long time. Parthenolide has been predominately investigated as inducer of apoptosis in human cancer cells. PURPOSE: We examined the expression of vimentin and Elongation factor α - 1 as breast cancer biomarkers in MCF7 cells exposure to Parthenolide. METHOD: In this study, we investigated the antitumor mechanism of Parthenolide on the human breast cancer cell line MCF7, using SEM, flow cytometry and proteomics techniques. RESULT: Comparative proteome analyses are shown Elongation factor1-α and vimentin was suppressed in response to Parthenolide treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Factor 1 de Elongación Peptídica/metabolismo , Sesquiterpenos/farmacología , Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Factor 1 de Elongación Peptídica/antagonistas & inhibidores , Tanacetum parthenium/química , Vimentina/antagonistas & inhibidores , Vimentina/metabolismo
19.
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
20.
Appl Immunohistochem Mol Morphol ; 26(5): 337-344, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-27556820

RESUMEN

Glioblastoma multiforme (GBM) has a high recurrence and mortality rate. Because of a poor understanding of the mechanism for this disease, treatment regimens have remained limited. Vimentin, one of the major cytoskeletal proteins, is associated with cellular structure. However, the function of vimentin in GBM is still undefined. In the present study, we investigated the expression level of vimentin in 179 GBM tissues using immunohistochemistry. We found that the vimentin expression level was associated with the time to progression (P=0.029). A Kaplan-Meier analysis revealed that patients with high vimentin expression had a significantly shorter overall survival (P=0.0002) and progression-free survival (P=0.0001) compared with those with low expression. Furthermore, in vitro experiments showed that withaferin-A, a chemical inhibitor of vimentin, could inhibit GBM cell migration and invasion activity when its concentrations were <0.5 µM, and higher concentrations of withaferin-A could decrease the viability of U251and U87 cells significantly. In conclusion, our results indicated that vimentin may play an important role in the progression of GBM.


Asunto(s)
Neoplasias Encefálicas/diagnóstico , Glioblastoma/diagnóstico , Vimentina/metabolismo , Adulto , Anciano , Neoplasias Encefálicas/mortalidad , Carcinogénesis , Línea Celular Tumoral , Movimiento Celular , Citoesqueleto/metabolismo , Femenino , Glioblastoma/mortalidad , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Análisis de Supervivencia , Vimentina/antagonistas & inhibidores , Witanólidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...