Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 96: 129528, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37852422

RESUMO

Malignant melanoma has an aggressive nature and a high metastatic propensity resulting in the highest mortality rate of any skin cancer. In this study, we synthesized 18F-labeled procainamide (PCA) for detection of melanoma using positron emission tomography (PET), and evaluated its biological characteristics. The non-decay-corrected radiochemical yield of 18F-PCA was 10-15% and its in vitro stability was over 98% for 2 h. At 1 h, cellular uptake of 18F-PCA was 3.8-fold higher in a group with the presence of l-tyrosine than in a non-l-tyrosine-treated group. Furthermore, 18F-PCA permitted visualization of B16F10 (mouse melanoma) xenografts on microPET after intravenous injection, and was retained in the tumor for 60 min, with a high tumor-to-liver uptake ratio. 18F-PCA showed specific melanoma uptake in primary lesions with a high melanin targeting ability in small animal models. 18F-PCA may have potential as a PET imaging agent for direct melanoma detection.


Assuntos
Melanoma , Neoplasias Cutâneas , Animais , Camundongos , Humanos , Procainamida , Melanoma/diagnóstico por imagem , Melanoma/patologia , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos , Linhagem Celular Tumoral , Radioisótopos de Flúor , Melanoma Maligno Cutâneo
2.
Sci Rep ; 11(1): 24430, 2021 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-34952915

RESUMO

Bacteria-mediated cancer-targeted therapy is a novel experimental strategy for the treatment of cancers. Bacteria can be engineered to overcome a major challenge of existing therapeutics by differentiating between malignant and healthy tissue. A prerequisite for further development and study of engineered bacteria is a suitable imaging concept which allows bacterial visualization in tissue and monitoring bacterial targeting and proliferation. Optoacoustics (OA) is an evolving technology allowing whole-tumor imaging and thereby direct observation of bacterial colonization in tumor regions. However, bacterial detection using OA is currently hampered by the lack of endogenous contrast or suitable transgene fluorescent labels. Here, we demonstrate improved visualization of cancer-targeting bacteria using OA imaging and E. coli engineered to express tyrosinase, which uses L-tyrosine as the substrate to produce the strong optoacoustic probe melanin in the tumor microenvironment. Tumors of animals injected with tyrosinase-expressing E. coli showed strong melanin signals, allowing to resolve bacterial growth in the tumor over time using multispectral OA tomography (MSOT). MSOT imaging of melanin accumulation in tumors was confirmed by melanin and E. coli staining. Our results demonstrate that using tyrosinase-expressing E. coli enables non-invasive, longitudinal monitoring of bacterial targeting and proliferation in cancer using MSOT.


Assuntos
Neoplasias do Colo/terapia , Escherichia coli/metabolismo , Monofenol Mono-Oxigenase/uso terapêutico , Técnicas Fotoacústicas/métodos , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos BALB C
3.
J Nucl Med ; 62(7): 956-960, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33509975

RESUMO

Surface-exposed calreticulin (ecto-CRT) is a well-known "eat-me" signal exhibited by dying cells that contributes to their recognition and destruction by the immune system. We assessed the use of a CRT-specific binding peptide for imaging ecto-CRT during immunogenic cell death and its utility for early prediction of treatment response. Methods: A synthetic CRT-specific peptide, KLGFFKR (CRTpep), was labeled with fluorescein isothiocyanate or 18F, and the characteristics of ecto-CRT were evaluated in a colon cancer cell line in vitro and in vivo. Results: In vitro flow cytometry, immunofluorescence staining, and in vivo small-animal PET imaging results showed that CRTpep detected preapoptotic cells treated with immunogenic drugs or radiation but not those treated with the nonimmunogenic drug or a nontherapeutic dose of immunogenic drug. Conclusion: The present results indicate that the CRT-specific peptide would enable the prediction of therapeutic response, thereby facilitating early decisions on continuation or discontinuation of immunogenic treatment.


Assuntos
Morte Celular Imunogênica , Antineoplásicos , Detecção Precoce de Câncer , Humanos , Neoplasias
4.
Theranostics ; 10(6): 2509-2521, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194816

RESUMO

Photoacoustic imaging is gaining great attention in the medical world due to its significant potential for clinical translation. Light excitation in the second near-infrared (NIR-II) window (1000-1350 nm) has resolution and penetration depth suitable for several clinical applications. However, the significant challenge exists for clinical translation because of the absence of notable intrinsic chromophores in this clinically significant optical range to generate diagnostic images. Methods: We present newly developed a biocompatible nickel dithiolene-based polymeric nanoparticle (NiPNP), which have a strong and sharp absorption peak at 1064 nm, as a photoacoustic contrast agent to boost specific absorbance in the NIR-II window for in vivo deep tissue imaging. Results: We confirm the enhanced PA signal by NiPNP's strong light absorption in the NIR-II window (287% higher than that of NIR-I) and deep tissue imaging capability (~5.1 cm) through in vitro experiment. We have successfully acquired diagnostic-quality in vivo photoacoustic images in deep tissue (~3.4 cm) of sentinel lymph nodes, gastrointestinal tracts, and bladders of live rats by using clinically viable imaging system. Conclusions: Our results prove that with strong absorption in the NIR-II window and with deeper imaging depth, the clinical translation of photoacoustic imaging with NiPNP is feasible for preclinical studies and thus would facilitate further clinical investigations.


Assuntos
Nanopartículas/química , Níquel/química , Técnicas Fotoacústicas/métodos , Polímeros/química , Enxofre/química , Ultrassonografia/métodos , Animais , Meios de Contraste/química , Feminino , Trato Gastrointestinal/diagnóstico por imagem , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão/métodos , Compostos Organometálicos/química , Ratos , Linfonodo Sentinela/diagnóstico por imagem , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Bexiga Urinária/diagnóstico por imagem
5.
J Nucl Med ; 60(7): 924-929, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30552204

RESUMO

Malignant melanoma is an aggressive and serious form of skin cancer, with prognosis and treatment outcome depending heavily on the clinical stage of the disease at the time of diagnosis. Here, we synthesized a novel 18F-labeled benzamide derivative to target melanoma and then evaluated its biologic characteristics in small-animal models. Methods:N-(2-(dimethylamino)ethyl)-4-18F-fluorobenzamide (18F-DMFB) was synthesized by reaction of N-succinimidyl 4-18F-fluorobenzoate with N,N-dimethylethylenediamine. The binding affinity of 18F-DMFB was measured in B16F10 (mouse melanoma) cells with or without l-tyrosine. Small-animal PET imaging with 18F-DMFB was performed on B16F10 xenograft and metastasis mouse models. Results: The overall non-decay-corrected radiochemical yield of 18F-DMFB was approximately 10%-15%. Uptake of 18F-DMFB was melanin-specific, as cellular uptake in B16F10 increased more than 18-fold in the presence of l-tyrosine. Biodistribution studies revealed that 18F-DMFB accumulated, and was retained, in B16F10 xenografts for 120 min (10, 30, 60, and 120 min: 9.24, 10.80, 13.0, and 10.59 percentage injected dose/g, respectively) after radiotracer injection. Liver uptake of 18F-DMFB decreased from 10 to 120 min and showed fast clearance (10, 30, 60, and 120 min: 11.19, 5.7, 2.47, and 0.4 percentage injected dose/g). Furthermore, 18F-DMFB allowed visualization of metastatic lesions immediately after injection and was retained in lesions for over 60 min, with a high tumor-to-background ratio. Conclusion:18F-DMFB demonstrated a high melanin-targeting ability and tumor-specific tumor uptake in both primary and metastatic lesions in animal models bearing malignant melanoma. 18F-DMFB may be a potential PET imaging agent for melanoma.


Assuntos
Benzamidas/metabolismo , Melanoma/diagnóstico por imagem , Sondas Moleculares/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Neoplasias Cutâneas/diagnóstico por imagem , Animais , Benzamidas/farmacocinética , Transporte Biológico , Linhagem Celular Tumoral , Estabilidade de Medicamentos , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Sondas Moleculares/farmacocinética , Metástase Neoplásica , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Distribuição Tecidual
7.
Bioorg Med Chem Lett ; 28(5): 915-921, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29422388

RESUMO

Carbonic anhydrase IX is overexpressed in many solid tumors including hypoxic tumors and is a potential target for cancer therapy and diagnosis. Reported imaging agents targeting CA-IX are successful mostly in clear cell renal carcinoma as SKRC-52 and no candidate was approved yet in clinical trials for imaging of CA-IX. To validate CA-IX as a valid target for imaging of hypoxic tumor, we designed and synthesized novel [18F]-PET tracer (1) based on acetazolamide which is one of the well-known CA-IX inhibitors and performed imaging study in CA-IX expressing hypoxic tumor model as 4T1 and HT-29 in vivo models other than SKRC-52. [18F]-acetazolamide (1) was found to be insufficient for the specific accumulation in CA-IX expressing tumor. This study might be useful to understand in vivo behavior of acetazolamide PET tracer and can contribute to the development of successful PET imaging agents targeting CA-IX in future. Additional study is needed to understand the mechanism of poor targeting of CA-IX, as if CA-IX is not reliable as a sole target for imaging of CA-IX expressing hypoxic solid tumors.


Assuntos
Acetazolamida/química , Anidrase Carbônica IX/análise , Inibidores da Anidrase Carbônica/química , Carcinoma de Células Renais/enzimologia , Neoplasias Renais/enzimologia , Tomografia por Emissão de Pósitrons , Acetazolamida/síntese química , Acetazolamida/farmacocinética , Animais , Anidrase Carbônica IX/biossíntese , Anidrase Carbônica IX/metabolismo , Inibidores da Anidrase Carbônica/síntese química , Inibidores da Anidrase Carbônica/farmacocinética , Carcinoma de Células Renais/diagnóstico , Radioisótopos de Flúor , Humanos , Neoplasias Renais/diagnóstico , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/diagnóstico , Neoplasias Experimentais/enzimologia , Distribuição Tecidual
8.
J Nucl Med ; 59(2): 340-346, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28916621

RESUMO

The epidermal growth factor receptor (EGFR) is a member of the erbB family of receptors and is overexpressed in many tumor types. A repebody is a newly designed nonantibody protein scaffold for tumor targeting that contains leucine-rich repeat modules. In this study, 3 64Cu-labeled anti-EGFR repebodies with different chelators were synthesized, and their biologic characteristics were assessed in cultured cells and tumor-bearing mice. Methods: Repebodies were synthesized with the chelators 2-(p-isothiocyanatobenzyl)-1,4,7-triazacyclononane-N,N',N,″-triacetic acid trihydrochloride ([p-SCN-Bn]-NOTA), 2,2',2″-(10-(2-(2,5-dioxopyrrolidin-1-yloxy)-2-oxoethyl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl) triacetic acid (DOTA-N-hydroxysuccinimide ester), or 1-(p-isothiocyanatobenzyl)diethylenetriamine pentaacetic acid trihydrochloride ([p-SCN-Bn]-DTPA) in 1.0 M NaHCO3 buffer (pH 9.2) for 24 h. Purified NOTA-, DOTA-, and DTPA-conjugated repebody were radiolabeled with 64Cu in 0.1 M NH4OAc buffer (pH 5.5). To compare the EGFR-binding affinities of the repebodies, cellular uptake studies were performed with the human non-small cell lung cancer cell line H1650 (high expression of EGFR) and the human colon adenocarcinoma cell line SW620 (low expression of EGFR). Biodistribution and small-animal PET imaging studies were performed using H1650 tumor-bearing mice. Results: Radiochemical yields of the 64Cu-labeled repebodies were approximately 70%-80%. Cellular uptake of the NOTA-, DOTA-, and DTPA-repebodies was over 4-fold higher in H1650 cells than in SW620 cells at 1 h. The 3 repebodies had accumulated specifically in H1650 tumor-bearing nude mice by 1 h after intravenous injection and were retained for over 24 h, as measured by the percentage injected dose per gram of tissue (%ID/g). Tumor uptake of all repebodies increased from 1 to 6 h (at 1 h, 6.28, 8.46, and 6.91 %ID/g for NOTA-, DOTA-, and DTPA-repebody, respectively; at 6 h, 9.4, 8.28, and 10.1 %ID/g, respectively). H1650 tumors were clearly visible after injection of each repebody, with high tumor-to-background ratios (at 1 h, 3.43, 4.89, and 2.38 for NOTA-, DOTA-, and DTPA-repebody, respectively; at 6 h, 3.05, 4.36, and 2.08; at 24 h, 3.81, 4.58, and 2.86). Conclusion: The 3 64Cu-repebody complexes demonstrated specific and rapid uptake in EGFR-expressing tumors within 1 h and may have potential as novel EGFR imaging agents for PET.


Assuntos
Anticorpos/química , Radioisótopos de Cobre , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Animais , Anticorpos/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Humanos , Marcação por Isótopo , Camundongos , Distribuição Tecidual
9.
Theranostics ; 7(10): 2620-2633, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28819451

RESUMO

The accurate detection of disease-related biomarkers is crucial for the early diagnosis and management of disease in personalized medicine. Here, we present a molecular imaging of human epidermal growth factor receptor (EGFR)-expressing malignant tumors using an EGFR-specific repebody composed of leucine-rich repeat (LRR) modules. The repebody was labeled with either a fluorescent dye or radioisotope, and used for imaging of EGFR-expressing malignant tumors using an optical method and positron emission tomography. Our approach enabled visualization of the status of EGFR expression, allowing quantitative evaluation in whole tumors, which correlated well with the EGFR expression levels in mouse or patients-derived colon cancers. The present approach can be effectively used for the accurate detection of EGFR-expressing cancers, assisting in the development of a tool for detecting other disease biomarkers.


Assuntos
Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/patologia , Receptores ErbB/análise , Imagem Molecular/métodos , Animais , Humanos , Proteínas de Repetições Ricas em Leucina , Camundongos , Imagem Óptica/métodos , Tomografia por Emissão de Pósitrons/métodos , Proteínas/metabolismo
10.
Theranostics ; 5(12): 1328-42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26516371

RESUMO

Although strains of attenuated Salmonella typhimurium and wild-type Escherichia coli show similar tumor-targeting capacities, only S. typhimurium significantly suppresses tumor growth in mice. The aim of the present study was to examine bacteria-mediated immune responses by conducting comparative analyses of the cytokine profiles and immune cell populations within tumor tissues colonized by E. coli or attenuated Salmonellae. CT26 tumor-bearing mice were treated with two different bacterial strains: S. typhimurium defective in ppGpp synthesis (ΔppGpp Salmonellae) or wild-type E. coli MG1655. Cytokine profiles and immune cell populations in tumor tissue colonized by these two bacterial strains were examined at two time points based on the pattern of tumor growth after ΔppGpp Salmonellae treatment: 1) when tumor growth was suppressed ('suppression stage') and 2) when they began to re-grow ('re-growing stage'). The levels of IL-1ß and TNF-α were markedly increased in tumors colonized by ΔppGpp Salmonellae. This increase was associated with tumor regression; the levels of both IL-1ß and TNF-α returned to normal level when the tumors started to re-grow. To identify the immune cells primarily responsible for Salmonellae-mediated tumor suppression, we examined the major cell types that produce IL-1ß and TNF-α. We found that macrophages and dendritic cells were the main producers of TNF-α and IL-1ß. Inhibiting IL-1ß production in Salmonellae-treated mice restored tumor growth, whereas tumor growth was suppressed for longer by local administration of recombinant IL-1ß or TNF-α in conjunction with Salmonella therapy. These findings suggested that IL-1ß and TNF-α play important roles in Salmonella-mediated cancer therapy. A better understanding of host immune responses in Salmonella therapy may increase the success of a given drug, particularly when various strategies are combined with bacteriotherapy.


Assuntos
Terapia Biológica/métodos , Interleucina-1beta/análise , Neoplasias/patologia , Neoplasias/terapia , Salmonella typhimurium/imunologia , Animais , Células Dendríticas/imunologia , Modelos Animais de Doenças , Escherichia coli/imunologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/análise
11.
Angew Chem Int Ed Engl ; 54(41): 12020-4, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26315561

RESUMO

Targeted therapy based on protein-drug conjugates has attracted significant attention owing to its high efficacy and low side effects. However, efficient and stable drug conjugation to a protein binder remains a challenge. Herein, a chemoenzymatic method to generate highly stable and homogenous drug conjugates with high efficiency is presented. The approach comprises the insertion of the CaaX sequence at the C-terminal end of the protein binder, prenylation using farnesyltransferase, and drug conjugation through an oxime ligation reaction. MMAF and an EGFR-specific repebody are used as the antitumor agent and protein binder, respectively. The method enables the precisely controlled synthesis of repebody-drug conjugates with high yield and homogeneity. The utility of this approach is illustrated by the notable stability of the repebody-drug conjugates in human plasma, negligible off-target effects, and a remarkable antitumor activity in vivo. The present method can be widely used for generating highly homogeneous and stable PDCs for targeted therapy.


Assuntos
Antineoplásicos/química , Receptores ErbB/metabolismo , Oligopeptídeos/química , Oximas/química , Proteínas/química , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Farnesiltranstransferase/metabolismo , Humanos , Camundongos Nus , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Oligopeptídeos/metabolismo , Oligopeptídeos/uso terapêutico , Oximas/metabolismo , Ligação Proteica , Prenilação de Proteína , Proteínas/metabolismo , Proteínas/uso terapêutico
12.
Mol Ther Oncolytics ; 2: 15007, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27119104

RESUMO

Bacteria can be engineered to deliver anticancer proteins to tumors via a controlled expression system that maximizes the concentration of the therapeutic agent in the tumor. L-asparaginase (L-ASNase), which primarily converts asparagine to aspartate, is an anticancer protein used to treat acute lymphoblastic leukemia. In this study, Salmonellae were engineered to express L-ASNase selectively within tumor tissues using the inducible araBAD promoter system of Escherichia coli. Antitumor efficacy of the engineered bacteria was demonstrated in vivo in solid malignancies. This result demonstrates the merit of bacteria as cancer drug delivery vehicles to administer cancer-starving proteins such as L-ASNase to be effective selectively within the microenvironment of cancer tissue.

13.
PLoS One ; 8(3): e60511, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555984

RESUMO

During the last decade, an increasing number of papers have described the use of various genera of bacteria, including E. coli and S. typhimurium, in the treatment of cancer. This is primarily due to the facts that not only are these bacteria capable of accumulating in the tumor mass, but they can also be engineered to deliver specific therapeutic proteins directly to the tumor site. However, a major obstacle exists in that bacteria because the plasmid carrying the therapeutic gene is not needed for bacterial survival, these plasmids are often lost from the bacteria. Here, we report the development of a balanced-lethal host-vector system based on deletion of the glmS gene in E. coli and S. typhimurium. This system takes advantage of the phenotype of the GlmS(-) mutant, which undergoes lysis in animal systems that lack the nutrients required for proliferation of the mutant bacteria, D-glucosamine (GlcN) or N-acetyl-D-glucosamine (GlcNAc), components necessary for peptidoglycan synthesis. We demonstrate that plasmids carrying a glmS gene (GlmS(+)p) complemented the phenotype of the GlmS(-) mutant, and that GlmS(+) p was maintained faithfully both in vitro and in an animal system in the absence of selection pressure. This was further verified by bioluminescent signals from GlmS (+)pLux carried in bacteria that accumulated in grafted tumor tissue in a mouse model. The signal was up to several hundred-fold stronger than that from the control plasmid, pLux, due to faithful maintenance of the plasmid. We believe this system will allow to package a therapeutic gene onto an expression plasmid for bacterial delivery to the tumor site without subsequent loss of plasmid expression as well as to quantify bioluminescent bacteria using in vivo imaging by providing a direct correlation between photon flux and bacterial number.


Assuntos
Proteínas de Bactérias/genética , Escherichia coli/genética , Vetores Genéticos/genética , Neoplasias/genética , Neoplasias/microbiologia , Plasmídeos/genética , Salmonella typhimurium/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Deleção de Genes , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/terapia , Plasmídeos/administração & dosagem
14.
J Microbiol ; 50(3): 502-10, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22752915

RESUMO

The use of bacteria has contributed to recent advances in targeted cancer therapy especially for its tumor-specific accumulation and proliferation. In this study, we investigated the molecular events following bacterial therapy using an attenuated Salmonella Typhimurium defective in ppGpp synthesis (ΔppGpp), by analyzing those proteins differentially expressed in tumor tissues from treated and untreated mice. CT26 murine colon cancer cells were implanted in BALB/c mice and allowed to form tumors. The tumor-bearing mice were treated with the attenuated Salmonella Typhimurium. Tumor tissues were analyzed by 2D-PAGE. Fourteen differentially expressed proteins were identified by mass spectrometry. The analysis revealed that cytoskeletal components, including vimentin, drebrin-like protein, and tropomyosin-alpha 3, were decreased while serum proteins related to heme or iron metabolism, including transferrin, hemopexin, and haptoglobin were increased. Subsequent studies revealed that the decrease in cytoskeletal components occurred at the transcriptional level and that the increase in heme and iron metabolism proteins occurred in liver. Most interestingly, the same pattern of increased expression of transferrin, hemopexin, and haptoglobin was observed following radiotherapy at the dosage of 14 Gy.


Assuntos
Terapia Biológica/métodos , Neoplasias do Colo/terapia , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade , Animais , Neoplasias do Colo/química , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Proteoma/análise
15.
J Cell Sci ; 118(Pt 2): 313-22, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15615777

RESUMO

The Wnt family of proteins regulates development and cell growth. We identified Wnt3a-based regulatory mechanisms for cell proliferation in NIH3T3 fibroblast cells. The degree of Wnt3a-induced proliferation was reduced by beta-catenin small interfering RNA (siRNA) and extracellular signal-regulated kinase (ERK) siRNA, indicating that both the ERK and Wnt/beta-catenin pathways are involved in Wnt3a-induced proliferation. Wnt3a immediately and transiently activated the Raf-1-MEK-ERK cascade in a manner distinct from that of the beta-catenin increase seen in cells treated with Wnt3a. Wnt3a-induced ERK activation was maintained even though basal ERK activities were reduced by beta-catenin siRNA, indicating that Wnt3a may activate the ERK pathway independently of beta-catenin. The ERK pathway was however, activated by beta-catenin transfection, which was abolished by co-transfection with dominant-negative Tcf-4. Therefore, ERK pathway activation by Wnt signaling could occur at multiple levels, including beta-catenin-independent direct signaling resulting from a Wnt3a and beta-catenin/Tcf-4-dependent post gene transcriptional event. Wnt3a stimulated the G1 to S phase cell cycle progression. This stimulation was reduced by the ERK pathway inhibitor, indicating that Wnt3a promotes proliferation by stimulating the ERK pathway. Wnt3a therefore stimulates the proliferation of fibroblast cells, at least in part, via activation of the ERK and Wnt/beta-catenin pathways.


Assuntos
Proteínas do Citoesqueleto/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas/metabolismo , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proteínas do Citoesqueleto/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fase G1/efeitos dos fármacos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células NIH 3T3 , Proteínas/farmacologia , RNA Interferente Pequeno/metabolismo , Fase S/efeitos dos fármacos , Transativadores/efeitos dos fármacos , Proteínas Wnt , Proteína Wnt3 , Proteína Wnt3A , beta Catenina , Quinases raf/efeitos dos fármacos , Quinases raf/metabolismo
16.
Br J Pharmacol ; 137(5): 597-607, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12381673

RESUMO

Zinc is an important trace element in the body and is involved in both the proliferation and growth arrest of many kinds of cells including colorectal epithelial cells. The aim of this study was to identify the molecular mechanism of the growth regulation of colorectal cancer cells by extracellular zinc. Zinc-stimulated activation of the mitogen-activated protein kinase (MAPK) cascade was measured by immunoblotting and Elk-1 dependent trans-reporter gene expression, and zinc-stimulated p21(Cip/WAF1) activation by immunoblotting, Northern blot analysis and immunochemistry. Cell proliferation was measured by thymidine and bromodeoxyuridine (BrdU) incorporation. By treating colorectal cancer cells with 100 microM ZnCl2, MAPKs were activated in two different phases, the initial weak activation occurred within 5 min and this was followed by a stronger and more prolonged activation. Zinc concomitantly activated Raf-1-MEK-MAPK kinases, and induced Elk-1 dependent trans-reporter gene expression. Prolonged activation of MAPKs by 100 microM of ZnCl2 resulted in the induction and nuclear localization of p21(Cip/WAF1) and was related to the inhibition of both thymidine and BrdU incorporations. These results not only suggest the presence of a mechanism for p21(Cip/WAF1) dependent negative regulation of colorectal cancer cell growth by zinc but also suggest potential usage of zinc to control the growth of colorectal cancer cells.


Assuntos
Cloretos/fisiologia , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/prevenção & controle , Ciclinas/metabolismo , Espaço Extracelular/fisiologia , Inibidores do Crescimento/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Divisão Celular/efeitos dos fármacos , Cloretos/farmacologia , Neoplasias Colorretais/patologia , Inibidor de Quinase Dependente de Ciclina p21 , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Inibidores do Crescimento/farmacologia , Células HT29 , Humanos , RNA Mensageiro/biossíntese , Compostos de Zinco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA