Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Anticancer Drugs ; 27(10): 979-87, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27548349

RESUMO

Worldwide, cervix carcinoma is among the most dangerous cancer types, and novel therapies are under development. Cancer treatments are often hampered because of lack of specificity. The chicken anemia virus-derived apoptin induces apoptosis selectively in tumor cells and leaves normal cells unharmed. Here, we have carried out in-vitro and in-vivo studies on the cytotoxic effect of apoptin in a cervix carcinoma model. Apoptin was fused to the protein transduction domain 4 (PTD4), enabling delivery of the fusion protein across cellular membranes. PTD4-apoptin protein is located in the nuclei of human cervical carcinoma HeLa cells and in the cytoplasm of normal cells L02. By MTT and flow cytometry analysis, we have proven that PTD4-apoptin protein induced apoptosis in the cervical carcinoma cells. PTD4-apoptin enhanced the level of active executioner caspase-3. Neither caspase-3 activation nor apoptin-induced accumulation of the mitochondrial outer-membrane protein Mfn-2 was affected by ectopic Bcl-2 expression. In contrast, apoptin-mediated AKT activation was inhibited by Bcl-2. In vivo, cervix carcinoma xenografts were treated for 7 days with PTD4-apoptin protein. The PTD4-apoptin treatment induced a decrease in the cervix carcinoma, whereas the PTD4-GFP protein-treated controls expanded significantly. TUNEL analysis showed that PTD4-apoptin protein induced apoptosis in cervix carcinoma cells, in contrast to the control PTD-GFP-treated ones. Our results indicate that apoptin is a potential anticancer agent for treating cervix carcinoma.


Assuntos
Proteínas Recombinantes de Fusão/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Células HeLa , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Distribuição Aleatória , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Adv Exp Med Biol ; 818: 39-59, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25001531

RESUMO

The chicken anemia virus derived protein apoptin harbors cancer-selective cell killing characteristics, essentially based on phosphorylation-mediated nuclear transfer in cancer cells and efficient cytoplasmic degradation in normal cells. Here, we describe a growing set of preclinical experiments underlying the promises of the anti-cancer potential of apoptin. Various non-replicative oncolytic viral vector systems have revealed the safety and efficacy of apoptin. In addition, apoptin enhanced the oncolytic potential of adenovirus, parvovirus and Newcastle disease virus vectors. Intratumoral injection of attenuated Salmonella typhimurium bacterial strains and plasmid-based systems expressing apoptin resulted in significant tumor regression. In-vitro and in-vivo experiments showed that recombinant membrane-transferring PTD4- or TAT-apoptin proteins have potential as a future anticancer therapeutics. In xenografted hepatoma and melanoma mouse models PTD4-apoptin protein entered both cancer and normal cells, but only killed cancer cells. Combinatorial treatment of PTD4-apoptin with various (chemo)therapeutic compounds revealed an additive or even synergistic effect, reducing the side effects of the single (chemo)therapeutic treatment. Degradable polymeric nanocapsules harboring MBP-apoptin fusion-protein induced tumor-selective cell killing in-vitro and in-vivo and revealed the potential of polymer-apoptin protein vehicles as an anticancer agent.Besides its direct use as an anticancer therapeutic, apoptin research has also generated novel possibilities for drug design. The nuclear location domains of apoptin are attractive tools for targeting therapeutic compounds into the nucleus of cancer cells. Identification of cancer-related processes targeted by apoptin can potentially generate novel drug targets. Recent breakthroughs important for clinical applications are reported inferring apoptin-based clinical trials as a feasible reality.


Assuntos
Proteínas do Capsídeo/metabolismo , Carcinoma Hepatocelular , Terapia Genética , Neoplasias Hepáticas , Melanoma , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Proteínas do Capsídeo/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Desenho de Fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Melanoma/terapia , Camundongos
3.
Biochem Biophys Res Commun ; 422(1): 169-73, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22575449

RESUMO

The small viral protein apoptin is capable of inducing apoptosis selectively in human tumor cells. In normal cells apoptin localizes in the cytoplasm where it forms aggregates, becomes epitope-shielded and eventually degraded. By inhibiting the proteasome activity with the chemical inhibitors bortezomib and Ada-Ahx(3)L(3)VS apoptin levels can be stabilized in normal cells similar to the tumor suppressor p53 protein. In contrast, proteasome inhibition in tumor cells did not affect the apoptin stability while it still stabilized p53 levels. Apparently, apoptin is degraded by proteasomal activity in normal human cells, a process that no longer takes place in tumor cells. This loss of proteasomal susceptibility appears to be specific for apoptin.


Assuntos
Proteínas do Capsídeo/metabolismo , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Fibroblastos/metabolismo , Humanos , Mutação , Fosforilação , Estabilidade Proteica , Proteólise , Pirazinas/farmacologia , Proteína Supressora de Tumor p53/metabolismo
4.
Anal Biochem ; 421(1): 68-74, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22080040

RESUMO

Apoptin, a protein derived from chicken anemia virus (CAV), induces apoptosis selectively in human tumor cells as compared with normal cells. This activity depends on phosphorylation and relocation of apoptin to the nucleus of cancer cells. Here, we describe an in vitro kinase assay that allows the biochemical characterization of apoptin kinase activity in tumor cells. The kinase phosphorylates apoptin in a strictly ATP-dependent fashion and in a broad salt range. The kinase activity is present constitutively in both cytoplasm and nucleus of various human tumor cells. Q-column chromatography showed that both cytoplasmic and nuclear fractions have identical fractionation characteristics, suggesting that the same kinase is present in both cellular compartments. Kinase activity derived from positive Q-column fractions bound to amylose-maltose-binding protein (MBP)-apoptin and could be eluted with ATP only in the presence of the cofactor Mg(2+). Apparently, unphosphorylated apoptin interacts with the kinase and is released only after phosphorylation has occurred, proving that our assay recognizes the genuine apoptin kinase. This is further corroborated by the finding that apoptin is phosphorylated in vitro at positions Thr108 and Thr107, in concert with earlier in vivo observations. Our assay excludes cyclin-dependent kinase 2 (CDK2) and protein kinase C beta (PKC-ß), previously nominated by two separate studies as being the genuine apoptin kinase.


Assuntos
Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/farmacologia , Neoplasias/metabolismo , Proteínas Quinases/análise , Transporte Ativo do Núcleo Celular , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Quinase 2 Dependente de Ciclina/metabolismo , Citoplasma/enzimologia , Células HeLa , Humanos , Técnicas In Vitro , Células Jurkat , Cinética , Espectrometria de Massas , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fosforilação , Proteína Quinase C/metabolismo , Proteínas Quinases/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Treonina/química
5.
Eur J Pharmacol ; 654(1): 17-25, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21184754

RESUMO

PTD4-apoptin protein enters cells and harbors tumor-selective cell death activity. Dacarbazine is the mainstay of treatment for malignant melanoma. In this study, we investigated the cytotoxic effect of PTD4-apoptin protein and/or dacarbazine in mouse B16-F1 and human A875 and SK-MEL-5 melanoma cells in vitro and by means of a mouse B16-F1 melanoma model in vivo. PTD4-apoptin protein inhibits the growth of B16-F1, A875 and SK-MEL-5 melanoma cells in a dose-dependent manner, but not in normal human cell lines WI-38 and L-02. PTD4-apoptin combined with dacarbazine revealed a synergistic cytotoxic effect (coefficient of drug interaction<1) in all three different tumor cell lines. In vivo, PTD4-apoptin protein and dacarbazine alone effectively inhibited the growth of B16-F1 melanoma in C57BL/6 mice. Strikingly, combined PTD4-apoptin/dacarbazine treatment significantly increased the antitumor effect in comparison to the single treatments. As important, a combined PTD4-apoptin/dacarbazine treatment with a 50% reduction of dacarbazine revealed similar antitumor activities, without detectable hematologic side effects. A combined PTD4-apoptin/dacarbazine treatment represents a promising novel efficient and safe anticancer strategy.


Assuntos
Proteínas do Capsídeo/farmacologia , Dacarbazina/farmacologia , Melanoma/tratamento farmacológico , Proteínas Recombinantes de Fusão/química , Animais , Antineoplásicos Alquilantes/farmacologia , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/química , Linhagem Celular , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Humanos , Melanoma/patologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL
6.
Nucleic Acids Res ; 38(22): 8277-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20693527

RESUMO

Programmed ribosomal frameshifting is a translational recoding mechanism commonly used by RNA viruses to express two or more proteins from a single mRNA at a fixed ratio. An essential element in this process is the presence of an RNA secondary structure, such as a pseudoknot or a hairpin, located downstream of the slippery sequence. Here, we have tested the efficiency of RNA oligonucleotides annealing downstream of the slippery sequence to induce frameshifting in vitro. Maximal frameshifting was observed with oligonucleotides of 12-18 nt. Antisense oligonucleotides bearing locked nucleic acid (LNA) modifications also proved to be efficient frameshift-stimulators in contrast to DNA oligonucleotides. The number, sequence and location of LNA bases in an otherwise DNA oligonucleotide have to be carefully manipulated to obtain optimal levels of frameshifting. Our data favor a model in which RNA stability at the entrance of the ribosomal tunnel is the major determinant of stimulating slippage rather than a specific three-dimensional structure of the stimulating RNA element.


Assuntos
Mudança da Fase de Leitura do Gene Ribossômico , Oligonucleotídeos Antissenso/química , Oligonucleotídeos/química , Oligodesoxirribonucleotídeos/química , Termodinâmica
7.
Basic Clin Pharmacol Toxicol ; 106(2): 130-4, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19874285

RESUMO

Treatment of head and neck cancers is still rather poor and worldwide new treatment options are sought. Sensitizing radioresistant tumours by combining irradiation with other therapeutics to induce apoptosis are widely investigated. We examined whether chicken anaemia virus-derived apoptin protein would have a beneficial effect on irradiation of radiosensitive SCC61 and radioresistant SQD9 human head and neck squamous carcinoma cell lines. In both cell lines, concurrent exposure to irradiation and apoptin resulted in analysed mitochondrial cytochrome c release and in cleavage of caspase-3, whereas irradiation alone of SQD9 cells under identical conditions did not. Moreover, in comparison with the irradiation, only the synchronized treatment of apoptin and irradiation resulted in increased cell death in especially the radioresistant SQD9 cells, as measured by means of a colony survival assay. Our data reveal that apoptin treatment represents an effective way for enhancing radiotherapy of tumours responding poorly to radiotherapy.


Assuntos
Proteínas do Capsídeo/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Radiossensibilizantes/farmacologia , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/radioterapia , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 3/efeitos da radiação , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Terapia Combinada , Citocromos c/efeitos dos fármacos , Citocromos c/metabolismo , Citocromos c/efeitos da radiação , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/radioterapia , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação
8.
Eur J Pharmacol ; 625(1-3): 165-73, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19836376

RESUMO

All human cells have a genetic program that upon activation will cause cell death, named apoptosis. Cancer cells can grow due to unbalances in proliferation, cell cycle regulation and their apoptosis machinery: genomic mutations resulting in non-functional pro-apoptosis proteins or over-expression of anti-apoptosis proteins form the basis of tumor formation. Surprisingly, lessons learned from viruses show that cancer cannot be regarded simply as the opposite of apoptosis. For instance, adenovirus can only transform cells when both its anti- and pro-apoptotic proteins are produced. Oncolytic viruses are known to replicate selectively in tumor cells resulting in cell death. Proteins derived from viruses, i.e. chicken anemia virus (CAV)-derived apoptosis-inducing protein (apoptin), adenovirus early region 4 open reading frame (E4orf4) and parvovirus-H1 derived non-structural protein 1 (NS1), the human alpha-lactalbumin made lethal to tumor cells (HAMLET), which is present in human milk or the human cytokines melanoma differentiation-associated gene-7 (mda-7) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) have all the ability to induce tumor-selective apoptosis. The tumor-selective apoptosis-inducing proteins seem to interact with transforming survival processes, which can become redirected by these proteins into cell death. Transformation-related processes have been identified, which seem to be crucial for the tumor-selectively killing activity of these proteins. For instance, the transformation-related protein phosphatase 2A (PP2A) plays a role in the induction of tumor-selective apoptosis. The proteins mda-7, TRAIL and HAMLET are already successfully tested in first clinical trials. Proteins harboring tumor-selective apoptosis characteristics represent, therefore, a therapeutic potential and a tool for unraveling tumor-related processes. Fundamental molecular and (pre)clinical therapeutic studies of the various tumor-selective apoptosis-inducing proteins apoptin, E4orf4, HAMLET, mda-7, NS1, TRAIL and related proteins will be discussed.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Proteínas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias/fisiopatologia , Vírus Oncolíticos/metabolismo , Proteínas Virais/farmacologia
9.
J Mol Histol ; 40(3): 177-81, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19685146

RESUMO

Despite recent advances, the prognosis of oral squamous cell carcinoma is still poor. Therapeutic options such as radiotherapy, chemotherapy, surgery and the novel treatment option gene therapy are being investigated in animal models. Diverse models have been studied to induce oral squamous cell carcinomas. The carcinogenic 4-nitroquinoline-1-oxide (4NQO) model has proven to be successful although until now it is unknown at what time point the established tumor is a representative squamous cell carcinoma and has a suitable volume for scientific treatment. For this end we applied 4NQO 3 times a week during 16 weeks and measured the volume of tumor tissue each week until the end of the experiment at 40 weeks. Concurrent histopathology at different time points up to the end of the experiment revealed that all mice bearing oral tumors were diagnosed with squamous cell carcinoma. Immunohistochemistry with markers cyclin D1 and E-cadherin revealed that the generated mouse oral tumors showed strong similarities with the described immunopathology in human oral tumors. The 4NQO model is a suitable alternative for preclinical gene therapy experiments with primary oral tumors. Future survey of therapeutic options in the carcinogenic 4NQO model should be conducted around 40 weeks after the start of the treatment.


Assuntos
Carcinoma de Células Escamosas/patologia , Modelos Animais de Doenças , Neoplasias Bucais/patologia , 4-Nitroquinolina-1-Óxido/química , Animais , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Ciclina D1/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos CBA , Neoplasias Bucais/metabolismo , Língua/patologia , Carga Tumoral
10.
Int J Cancer ; 124(12): 2973-81, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19326452

RESUMO

Apoptin protein harbors tumor-selective cell death activity, which makes it a potential anticancer therapy candidate. This study reports an apoptin therapy approach based on protein transduction domain 4 (PTD4)-mediated transduction of recombinant apoptin protein. In vitro, the PTD4-apoptin fusion protein is located in the nucleus and induces cell death in, e.g., human hepatocarcinoma HepG2 cells. In normal human L-02 hepatocytes, PTD4-apoptin protein retained mainly cytoplasmic and did not induce detectable levels of cell death, illustrating that the PTD4 domain does not affect apoptin's tumor-selective characteristics. In vivo, liver, cervix and gastric carcinoma xenografts treated with PTD4-apoptin protein for 6 days via the tumor epidermis exhibited a significant tumor growth inhibition because of apoptin-mediated cell death. In addition, treatment of human hepatocarcinoma xenografts during 3 weeks showed that PTD4-apoptin protein has significant anticancer activity, whereas control treatment with PTD4-enhanced green fluorescence protein or saline did not. Cell death and disruption of the tumor integrity were apparent in the PTD4-apoptin transduced xenografted tumors. As important, although PTD4-apoptin protein could be detected in the epidermal tissue covering the subcutaneous tumor tissue and in several organs, such as liver and brain, of the treated mice, no tissue disruption or signs of cell death could be detected. Our in vivo data reveal that apoptin protein delivery constitutes a novel powerful and safe anticancer therapy.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas do Capsídeo/metabolismo , Carcinoma Hepatocelular/terapia , Proteínas de Transporte/metabolismo , Neoplasias Hepáticas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Proteínas do Capsídeo/genética , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/genética , Núcleo Celular/metabolismo , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sinais de Localização Nuclear , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/terapia , Frações Subcelulares , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/terapia
11.
Cancer Biol Ther ; 7(9): 1368-73, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18708764

RESUMO

Apoptin, a chicken anemia virus-derived protein, induces apoptosis in various tumor cell lines and xenografted tumors. Its apoptotic activity is not hampered by tumor-suppressor p53 mutations or overexpression of anti-apoptosis proteins Bcl-2 or Bcl-x(L). We report for the first time the effects of apoptin expression in primary oral tumors, induced by the carcinogen 4-Nitroquinoline- 1-oxide in immunocompetent mice. In vivo a significant amount of primary oral tumor cells expressing apoptin cells underwent apoptosis, whereas synthesis of the LacZ control product did not. Ectopical expression of apoptin in passage 1 cell cultures derived from these oral tumors also resulted in apoptin-induced. Both in-vivo and in-vitro treated cells underwent apoptosis via the activation of caspase-3. The fact that apoptin induces apoptosis in primary squamous cell carcinoma cells indicates that apoptin is a potential therapeutic agent for treatment of head and neck squamous cell carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/farmacologia , Carcinoma de Células Escamosas/metabolismo , Neoplasias Bucais/metabolismo , 4-Nitroquinolina-1-Óxido/toxicidade , Adenoviridae/genética , Animais , Carcinógenos/toxicidade , Carcinoma de Células Escamosas/induzido quimicamente , Carcinoma de Células Escamosas/patologia , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes/metabolismo , Vetores Genéticos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos CBA , Neoplasias Bucais/induzido quimicamente , Neoplasias Bucais/genética , Projetos Piloto , Plasmídeos , Distribuição Aleatória , Transfecção , Carga Tumoral , Células Tumorais Cultivadas
12.
Mini Rev Med Chem ; 7(11): 1155-65, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18045219

RESUMO

Viruses can produce viral oncoproteins that drive multiple genetic alterations as the consequence of neoplastic transformation. Viral proteins encoded by onco-related viruses such as polyomavirus SV40 or Epstein-Barr virus are involved in cellular processes resulting in imbalance between proliferation and cell death, knowledge of which continues to be crucial for combating cancer. On the other hand, viruses also generate viral components that, from a cold viral protein, can become a tumor-selective killer by sensing cellular tumorigenic hallmarks. For instance, the avian virus derived apoptin protein has been proven to induce tumor-regression in various pre-clinical animal models without showing detectable side effects. In particular, apoptin-interacting protein partners such as components of the anaphase promoting complex were identified as potential anticancer drug targets. The adenovirus-derived protein E4orf4, another viral protein with tumor-specific apoptosis characteristics, has been proven to interact with the tumor-suppressor protein phosphatase 2A. This review aims to describe recent studies with representative viral elements that have contributed to our understanding of critical tumorigenic processes and have conferred an impact on the development of novel anti-cancer therapies.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Proteínas do Capsídeo/fisiologia , Elementos Facilitadores Genéticos , Neoplasias/tratamento farmacológico , Vírus 40 dos Símios/fisiologia , Proteínas Virais/fisiologia , Animais , Proteínas do Capsídeo/efeitos dos fármacos , Proteínas do Capsídeo/farmacologia , Humanos , Proteína Fosfatase 2/metabolismo , Transdução de Sinais , Proteínas Virais/efeitos dos fármacos , Proteínas Virais/metabolismo
13.
Basic Clin Pharmacol Toxicol ; 100(2): 127-31, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17244262

RESUMO

Gene therapy experiments in animal models have shown that apoptin expression results in tumour regression without any significant side effects. Therefore, apoptin is regarded as a potential anticancer drug for clinical applications. In this study, we analysed whether chemotherapeutic agents combined with apoptin treatment could result in enhanced cytotoxicity in human tumour cell cultures. Combined treatment with recombinant adenovirus AdAptVP3 expressing apoptin and etoposide clearly showed an additive cytotoxic effect on human osteosarcoma U2OS cells. Paclitaxel treatment combined with apoptin expression significantly inhibited the survival of p53-positive human osteosarcoma U2OS and non-small lung carcinoma A549 cells, p53-negative human osteosarcoma Saos-2 cells and p53-mutant human prostate cancer Du145 cells, already at low doses of the chemotherapeutic agent. Our results indicate that the cytotoxicity-enhancing action by the tumour-specific apoptin in combination with chemotherapeutic agents might offer an effective and safe antitumour therapeutics.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Proteínas do Capsídeo/biossíntese , Etoposídeo/farmacologia , Paclitaxel/farmacologia , Adenoviridae/genética , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA Viral/genética , Sinergismo Farmacológico , Vetores Genéticos , Humanos , Transfecção , beta-Galactosidase/metabolismo
14.
J Virol ; 78(18): 9965-76, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331730

RESUMO

Apoptin, a viral death protein derived from chicken anemia virus, displays a number of tumor-specific behaviors. In particular, apoptin is phosphorylated, translocates to the nucleus, and induces apoptosis specifically in tumor or transformed cells, whereas it is nonphosphorylated and remains primarily inactive in the cytoplasm of nontransformed normal cells. Here, we show that in normal cells apoptin can also be activated by the transient transforming signals conferred by ectopically expressed simian virus 40 (SV40) large T antigen (LT), which rapidly induces apoptin's phosphorylation, nuclear accumulation, and the ability to induce apoptosis. Further analyses with mutants of LT showed that the minimum domain capable of inducing all three of apoptin's tumor-specific properties resided in the N-terminal J domain, a sequence which is largely shared by SV40 small t antigen (st). Interestingly, the J domain in st, which lacks its own nuclear localization signal (NLS), required nuclear localization to activate apoptin. These results reveal the existence of a cellular pathway shared by conditions of transient transformation and the stable cancerous or precancerous state, and they support a model whereby a transient transforming signal confers on apoptin both the upstream activity of phosphorylation and the downstream activity of nuclear accumulation and apoptosis induction. Such a pathway may reflect a general lesion contributing to human cancers.


Assuntos
Antígenos Transformantes de Poliomavirus/fisiologia , Proteínas do Capsídeo/fisiologia , Vírus 40 dos Símios/fisiologia , Sequência de Aminoácidos , Animais , Antígenos Transformantes de Poliomavirus/química , Antígenos Transformantes de Poliomavirus/genética , Apoptose , Linhagem Celular , Núcleo Celular/virologia , Transformação Celular Neoplásica , Transformação Celular Viral , Vírus da Anemia da Galinha/patogenicidade , Vírus da Anemia da Galinha/fisiologia , Humanos , Camundongos , Mutação , Estrutura Terciária de Proteína , Vírus 40 dos Símios/genética , Vírus 40 dos Símios/patogenicidade
15.
J Gen Virol ; 85(Pt 6): 1445-1450, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15166427

RESUMO

TT virus (TTV) is widespread among the global population. Its pathogenic nature is still unclear but TTV seems to be more prevalent in cases of hepatitis than in healthy individuals. TTV harbours similarities to chicken anaemia virus (CAV). Here, the apoptotic potential of a putative TTV-derived 105 aa protein and of the main apoptosis-inducing agent of CAV, Apoptin, is compared. As the putative protein induced apoptosis in various human hepatocellular carcinoma (HCC) cell lines, it was named TTV-derived apoptosis-inducing protein (TAIP). The apoptotic activity of TAIP in HCC lines was comparable with that of Apoptin. Conversely, unlike Apoptin, TAIP induced only low-level apoptosis in several non-HCC human cancer cell lines. The data suggest that TAIP acts in a different way to Apoptin as it is selective to a certain degree for HCC lines. This activity of TAIP, coupled with the heterogeneity of TTV isolates, may help to explain the variable reports of TTV pathogenicity.


Assuntos
Apoptose , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Torque teno virus/patogenicidade , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Células COS , Proteínas do Capsídeo/fisiologia , Linhagem Celular Tumoral , Humanos , Dados de Sequência Molecular
16.
Int J Cancer ; 109(1): 38-42, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14735465

RESUMO

Nonfunctional p53 and especially upregulation of Bcl-x(L) result in advanced disease and poor prognosis of patients suffering head and neck squamous cell carcinoma (HNSCC). Aberrancies of Bcl-x(L) and/or p53 in HNSCC lead to inability of anticancer drugs to induce apoptosis. Bcl-x(L) and/or mutated p53 inhibit the apoptotic process by preventing the mitochondrial release of cytochrome c and/or activation of execution caspases. Here, we report that expression of the avian virus-derived apoptin protein resulted in induction of apoptosis in the HNSCC-derived cell line UMSSC-14B despite the presence of nonfunctional p53. Apoptin activated the execution caspase 3 and induced the release of mitochondrial cytochrome c. Upregulation of Bcl-x(L) in UMSCC-14B cells did not interfere with the apoptin-induced apoptosis, whereas it clearly negatively affected the p53-induced one. Bcl-x(L) significantly decreased the p53-induced cytochrome c release, but not the apoptin-triggered one. Our data demonstrate that apoptin induces apoptosis independent of Bcl-x(L) and p53 and may constitute a potential therapeutic agent for treatment of HNSCC.


Assuntos
Apoptose , Proteínas do Capsídeo/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/farmacologia , Western Blotting , Caspase 3 , Caspases/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Genes p53 , Humanos , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Mutação , Plasmídeos/metabolismo , Prognóstico , Frações Subcelulares/metabolismo , Fatores de Tempo , Transfecção , Regulação para Cima , Proteína bcl-X
17.
Vet Microbiol ; 98(2): 89-94, 2004 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-14741120

RESUMO

In 1990, the chicken anemia virus (CAV) genome was cloned by us and proven to be representative for CAV isolates worldwide. This genome contains unique promoter/enhancer replication elements and genes. Upon infection of its target cells, CAV replicates via a double-stranded (ds) DNA intermediate. From this ds CAV molecule, a single mRNA is transcribed, which encodes for three distinct proteins VP1, VP2, and VP3 or apoptin. Its capsid contains only the VP1 protein. However, for the production of the neutralizing epitope, co-synthesis of VP1 and VP2 are needed. CAV genomes with mutations in the 12 bp insert of the promoter/enhancer region were shown to produce immunogenic functional CAV particles. Mutations in these and other regulatory elements of CAV might also decrease its virus load resulting in a reduced pathogenic effect. CAV causes fatal cytopathogenic effects in e.g. chicken thymocytes via apoptosis. Under in vitro conditions, CAV replicates only in transformed chicken cell lines, which indicates that at least a part of the CAV life-cycle requires transformed-like cellular events. In these transformed cell lines, the synthesis of the apoptin protein alone mimics the CAV-induced apoptosis, whereas the VP2 protein also harbors some apoptotic activity. Extensive studies on apoptin resulted in the characterization of domains essential for its apoptotic activity and nuclear localization, which seems to be related with its ability to induce apoptosis. Therefore, both VP2 and apoptin are of interest in reducing the pathogenicity of CAV infections. A series of biomedical studies on apoptin have been carried out in human cell systems, which are informative about the mechanism of CAV-induced apoptosis in chicken (transformed) cells. Synthesis of apoptin alone induces apoptosis in various human transformed and/or tumorigenic cell lines, but not in normal human diploid cells. A striking difference in the cellular localization of apoptin was observed in human normal diploid cells versus tumor cells. In all tumor cells, apoptin is located mainly in the heterochromatic regions of the nucleus, whereas in normal cells it is present in peri-nuclear structures. Apoptin contains a bipartite nuclear localization signal, and one domain that resemble a nuclear export signal. Elucidation of parts of the apoptin-induced apoptotic pathway revealed unique characteristics: apoptin-induced apoptosis is independent of the tumor suppressor p53. The anti-apoptotic protein Bcl-2 does not inhibit but even accelerates apoptin-induced apoptosis in tumor cells, whereas over expression of Bcl-2 in normal cells has no effect on the apoptin activity. Upstream caspases are not involved, whereas downstream caspase 3 is, but seems not to be essential. A number of novel proteins were shown to interact with apoptin in transformed cells. Future studies of apoptin, VP2 and related cellular proteins in chicken cells will unravel the regulatory aspects of CAV-induced apoptosis.


Assuntos
Apoptose/fisiologia , Vírus da Anemia da Galinha/fisiologia , Galinhas , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/virologia , Animais , Infecções por Circoviridae/imunologia , Infecções por Circoviridae/patologia , Infecções por Circoviridae/virologia , Genoma Viral , Doenças das Aves Domésticas/patologia
18.
J Mol Med (Berl) ; 82(1): 56-63, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14647920

RESUMO

Biliary tract cancer, or cholangiocarcinoma, has a poor prognosis. Resection is the only curative treatment, but only a minority of patients are eligible. Chemotherapy and gamma-irradiation are merely palliative, as they are unable to remove the malignancy completely. The chicken anemia virus-derived protein apoptin induces apoptosis in a wide range of human tumor cells and is not hindered by mutations inactivating p53 or by overexpression of Bcl-2, changes known to frustrate chemotherapy and radiation therapy. We examined whether apoptin kills human biliary tract cancer cells. Expression of apoptin by means of plasmids caused extensive cell death in three independent cholangiocarcinoma cell lines, CC-LP, CC-SW, and Mz-ChA-1, regardless of their oncogenic mutations, which included inactivated p16 and p53 and the disruption of the transforming growth factor beta signaling pathway. In vitro delivery of apoptin by an adenoviral vector completely eradicated cholangiocarcinoma cells. Moreover, coexpression of the broad-spectrum caspase inhibitor p35 with apoptin only delayed the induced cell death. Changes in nuclear morphology still occurred early after transfection, and nuclei eventually disintegrated, suggesting that apoptin-induced cell death in these cells is not blocked by mutations in either the initiation or execution phase of apoptosis. The efficient induction of cell death by apoptin in cholangiocarcinoma cell lines makes apoptin an attractive candidate for molecular therapy of biliary tract cancer.


Assuntos
Neoplasias do Sistema Biliar/metabolismo , Proteínas do Capsídeo/metabolismo , Morte Celular/fisiologia , Colangiocarcinoma/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Neoplasias do Sistema Biliar/patologia , Neoplasias do Sistema Biliar/terapia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/uso terapêutico , Inibidores de Caspase , Linhagem Celular Tumoral , Vírus da Anemia da Galinha/metabolismo , Colangiocarcinoma/patologia , Colangiocarcinoma/terapia , Vetores Genéticos , Humanos , Proteína Supressora de Tumor p53/metabolismo , Proteínas Virais/metabolismo
19.
Eur J Biochem ; 270(17): 3619-27, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12919326

RESUMO

Recombinant, bacterially expressed apoptin protein induces apoptosis in human tumour cell lines but not in normal cells, mimicking the behaviour of ectopically expressed apoptin. Recombinant apoptin is isolated exclusively as a highly stable multimeric complex of 30-40 monomers, with little, if any, alpha-helical and beta-sheet structure. Despite its apparent disorder, multimeric apoptin is biologically active. Here, we present evidence that most of the apoptin moieties within the complex may well share a similar conformation. Furthermore, the multimer has extensive and uniform hydrophobic patches and conformationally stable domains. Only a small fraction of apoptin subunits can exchange between multimers under physiologically relevant conditions. These results prompt a model in which the apoptin multimer has a highly stable core of nonexchangeable subunits to which exchangeable subunits are attached through hydrophobic interactions. In combination with previous findings, our results lead us to propose that the stable core of apoptin is the biologically relevant structure.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Substituição de Aminoácidos , Naftalenossulfonato de Anilina/metabolismo , Sítios de Ligação , Proteínas do Capsídeo/genética , Neoplasias do Colo/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Transferência Ressonante de Energia de Fluorescência , Humanos , Interações Hidrofóbicas e Hidrofílicas , Peso Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Titulometria , Células Tumorais Cultivadas
20.
Exp Cell Res ; 289(1): 36-46, 2003 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-12941602

RESUMO

Apoptin, a protein derived from chicken anemia virus, induces apoptosis in human transformed or tumor cells but not in normal cells. When produced in bacteria as a recombinant fusion with maltose-binding protein (MBP-Apoptin), Apoptin forms a distinct, stable multimeric complex that is remarkably homogeneous and uniform. Here, using cytoplasmic microinjection, we showed that recombinant MBP-Apoptin multimers retained the characteristics of the ectopically expressed wild-type Apoptin; namely, the complexes translocated to the nucleus of tumor cells and induced apoptosis, whereas they remained in the cytoplasm of normal, primary cells and exerted no apparent toxic effect. In normal cells, MBP-Apoptin formed increasingly large, organelle-sized globular bodies with time postinjection and eventually lost the ability to be detected by immunofluorescence analysis. Costaining with an acidotrophic marker indicated that these globular structures did not correspond to lysosomes. Immunoprecipitation studies showed that MBP-Apoptin remained fully antibody-accessible regardless of buffer stringency when microinjected into tumor cells. In contrast, MBP-Apoptin in normal cells was only recoverable under stringent lysis conditions, whereas under milder conditions they became fully shielded with time on two epitopes spanning the entire protein. Further biochemical analysis showed that the long-term fate of Apoptin protein aggregates in normal cells was their eventual elimination. Our results provide the first example of a tumor-specific apoptosis-inducing aggregate that is essentially sequestered by factors or conditions present in the cytoplasm of healthy, nontransformed cells. This characteristic should reveal more about the cellular interactions of this viral protein as well as further enhance its safety as a potential tumor-specific therapeutic agent.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas do Capsídeo/uso terapêutico , Neoplasias/tratamento farmacológico , Antineoplásicos/imunologia , Apoptose/fisiologia , Proteínas do Capsídeo/imunologia , Proteínas de Transporte/farmacologia , Proteínas de Transporte/uso terapêutico , Epitopos/imunologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Substâncias Macromoleculares , Proteínas Ligantes de Maltose , Neoplasias/imunologia , Neoplasias/metabolismo , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...