Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Ther Oncolytics ; 29: 17-29, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37077714

RESUMO

Primary drug resistance and minimal residual disease are major challenges in the treatment of B cell neoplasms. Therefore, this study aimed to identify a novel treatment capable of eradicating malignant B cells and drug-resistant disease. Oncolytic viruses eradicate malignant cells by direct oncolysis and activation of anti-tumor immunity, have proven anti-cancer efficacy, and are safe and well tolerated in clinical use. Here, we demonstrate that the oncolytic virus coxsackievirus A21 can kill a range of B cell neoplasms, irrespective of an anti-viral interferon response. Moreover, CVA21 retained its capacity to kill drug-resistant B cell neoplasms, where drug resistance was induced by co-culture with tumor microenvironment support. In some cases, CVA21 efficacy was actually enhanced, in accordance with increased expression of the viral entry receptor ICAM-1. Importantly, the data confirmed preferential killing of malignant B cells and CVA21 dependence on oncogenic B cell signaling pathways. Significantly, CVA21 also activated natural killer (NK) cells to kill neoplastic B cells and drug-resistant B cells remained susceptible to NK cell-mediated lysis. Overall, these data reveal a dual mode of action of CVA21 against drug-resistant B cells and support the development of CVA21 for the treatment of B cell neoplasms.

2.
J Immunother Cancer ; 9(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33741729

RESUMO

BACKGROUND: Multiple myeloma (MM) remains an incurable disease and oncolytic viruses offer a well-tolerated addition to the therapeutic arsenal. Oncolytic reovirus has progressed to phase I clinical trials and its direct lytic potential has been extensively studied. However, to date, the role for reovirus-induced immunotherapy against MM, and the impact of the bone marrow (BM) niche, have not been reported. METHODS: This study used human peripheral blood mononuclear cells from healthy donors and in vitro co-culture of MM cells and BM stromal cells to recapitulate the resistant BM niche. Additionally, the 5TGM1-Kalw/RijHSD immunocompetent in vivo model was used to examine reovirus efficacy and characterize reovirus-induced immune responses in the BM and spleen following intravenous administration. Collectively, these in vitro and in vivo models were used to characterize the development of innate and adaptive antimyeloma immunity following reovirus treatment. RESULTS: Using the 5TGM1-Kalw/RijHSD immunocompetent in vivo model we have demonstrated that reovirus reduces both MM tumor burden and myeloma-induced bone disease. Furthermore, detailed immune characterization revealed that reovirus: (i) increased natural killer (NK) cell and CD8+ T cell numbers; (ii) activated NK cells and CD8+ T cells and (iii) upregulated effector-memory CD8+ T cells. Moreover, increased effector-memory CD8+ T cells correlated with decreased tumor burden. Next, we explored the potential for reovirus-induced immunotherapy using human co-culture models to mimic the myeloma-supportive BM niche. MM cells co-cultured with BM stromal cells displayed resistance to reovirus-induced oncolysis and bystander cytokine-killing but remained susceptible to killing by reovirus-activated NK cells and MM-specific cytotoxic T lymphocytes. CONCLUSION: These data highlight the importance of reovirus-induced immunotherapy for targeting MM cells within the BM niche and suggest that combination with agents which boost antitumor immune responses should be a priority.


Assuntos
Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Reoviridae/imunologia , Baço/imunologia , Microambiente Tumoral/imunologia , Animais , Medula Óssea/virologia , Linfócitos T CD8-Positivos/virologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/imunologia , Citotoxicidade Imunológica , Feminino , Humanos , Células Matadoras Naturais/virologia , Masculino , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/virologia , Vírus Oncolíticos/patogenicidade , Reoviridae/patogenicidade , Baço/virologia , Evasão Tumoral
3.
J Immunother Cancer ; 7(1): 164, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31262361

RESUMO

BACKGROUND: The oncolytic virus, coxsackievirus A21 (CVA21), has shown promise as a single agent in several clinical trials and is now being tested in combination with immune checkpoint blockade. Combination therapies offer the best chance of disease control; however, the design of successful combination strategies requires a deeper understanding of the mechanisms underpinning CVA21 efficacy, in particular, the role of CVA21 anti-tumor immunity. Therefore, this study aimed to examine the ability of CVA21 to induce human anti-tumor immunity, and identify the cellular mechanism responsible. METHODS: This study utilized peripheral blood mononuclear cells from i) healthy donors, ii) Acute Myeloid Leukemia (AML) patients, and iii) patients taking part in the STORM clinical trial, who received intravenous CVA21; patients receiving intravenous CVA21 were consented separately in accordance with local institutional ethics review and approval. Collectively, these blood samples were used to characterize the development of innate and adaptive anti-tumor immune responses following CVA21 treatment. RESULTS: An Initial characterization of peripheral blood mononuclear cells, collected from cancer patients following intravenous infusion of CVA21, confirmed that CVA21 activated immune effector cells in patients. Next, using hematological disease models which were sensitive (Multiple Myeloma; MM) or resistant (AML) to CVA21-direct oncolysis, we demonstrated that CVA21 stimulated potent anti-tumor immune responses, including: 1) cytokine-mediated bystander killing; 2) enhanced natural killer cell-mediated cellular cytotoxicity; and 3) priming of tumor-specific cytotoxic T lymphocytes, with specificity towards known tumor-associated antigens. Importantly, immune-mediated killing of both MM and AML, despite AML cells being resistant to CVA21-direct oncolysis, was observed. Upon further examination of the cellular mechanisms responsible for CVA21-induced anti-tumor immunity we have identified the importance of type I IFN for NK cell activation, and demonstrated that both ICAM-1 and plasmacytoid dendritic cells were key mediators of this response. CONCLUSION: This work supports the development of CVA21 as an immunotherapeutic agent for the treatment of both AML and MM. Additionally, the data presented provides an important insight into the mechanisms of CVA21-mediated immunotherapy to aid the development of clinical biomarkers to predict response and rationalize future drug combinations.


Assuntos
Enterovirus , Leucemia Mieloide Aguda/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Imunidade Adaptativa , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Humanos , Imunidade Inata , Molécula 1 de Adesão Intercelular/imunologia , Leucemia Mieloide Aguda/imunologia , Masculino , Pessoa de Meia-Idade , Linfócitos T Citotóxicos/imunologia
4.
Mol Ther ; 27(6): 1139-1152, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31053413

RESUMO

A clinical oncolytic herpes simplex virus (HSV) encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), talimogene laherparepvec, causes regression of injected and non-injected melanoma lesions in patients and is now licensed for clinical use in advanced melanoma. To date, limited data are available regarding the mechanisms of human anti-tumor immune priming, an improved understanding of which could inform the development of future combination strategies with improved efficacy. This study addressed direct oncolysis and innate and adaptive human immune-mediated effects of a closely related HSV encoding GM-CSF (HSVGM-CSF) alone and in combination with histone deacetylase inhibition. We found that HSVGM-CSF supported activation of anti-melanoma immunity via monocyte-mediated type I interferon production, which activates NK cells, and viral maturation of immature dendritic cells (iDCs) into potent antigen-presenting cells for cytotoxic T lymphocyte (CTL) priming. Addition of the histone deacetylase inhibitor valproic acid (VPA) to HSVGM-CSF treatment of tumor cells increased viral replication, viral GM-CSF production, and oncolysis and augmented the development of anti-tumor immunity. Mechanistically, VPA increased expression of activating ligands for NK cell recognition and induced expression of tumor-associated antigens, supporting innate NK cell killing and CTL priming. These data support the clinical combination of talimogene laherparepvec with histone deacetylase inhibition to enhance oncolysis and anti-tumor immunity.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Melanoma/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/metabolismo , Simplexvirus/imunologia , Neoplasias Cutâneas/terapia , Ácido Valproico/uso terapêutico , Antígenos de Neoplasias/efeitos dos fármacos , Antígenos de Neoplasias/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Produtos Biológicos/uso terapêutico , Sobrevivência Celular/genética , Células Dendríticas/imunologia , Quimioterapia Combinada , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Herpesvirus Humano 1 , Humanos , Interferon Tipo I/metabolismo , Células Matadoras Naturais/imunologia , Células MCF-7 , Melanoma/patologia , Vírus Oncolíticos/genética , Simplexvirus/genética , Neoplasias Cutâneas/patologia , Linfócitos T Citotóxicos/imunologia
5.
Cell Immunol ; 297(1): 1-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26051632

RESUMO

Multiple myeloma (MM) produces significant cellular and humoral immune defects. We have previously reported that MM induces CD4(+)CD25(+)FoxP3(+) cells (TRegs), via tumour expression of the immune checkpoint regulator, ICOS-L. We sought to define what impact the immunomodulatory drug lenalidomide, alone or with dexamethasone, has on TReg cell generation. Lenalidomide pre-treatment of MM cell lines reduced TReg generation and the concomitant TReg:TEff (CD4(+)CD25(+)FoxP3(-): effector T cells) ratio, as a consequence of reduced ICOSL transcription. Dexamethasone did not affect surface ICOS-L expression but did induce TReg cell apoptosis without impacting on TEff cell survival. Combined lenalidomide and dexamethasone significantly reduced both TReg induction and the TReg:TEff cell ratio. In vivo, serial analysis of the TReg:TEff ratio in MM patients on lenalidomide-dexamethasone therapy revealed a progressive reduction towards age-matched control values, though not complete correction. Our data demonstrate for the first time immune synergism to explain the observed immune-modulation associated with lenalidomide-dexamethasone therapy.


Assuntos
Dexametasona/uso terapêutico , Fatores Imunológicos/uso terapêutico , Ligante Coestimulador de Linfócitos T Induzíveis/biossíntese , Mieloma Múltiplo/tratamento farmacológico , Linfócitos T Reguladores/citologia , Talidomida/análogos & derivados , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dexametasona/farmacologia , Regulação para Baixo , Feminino , Humanos , Fatores Imunológicos/farmacologia , Lenalidomida , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/patologia , Linfócitos T Reguladores/imunologia , Talidomida/farmacologia , Talidomida/uso terapêutico
6.
Microbes Infect ; 17(3): 205-14, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25499189

RESUMO

Helicobacter pylori transactivates the epidermal growth factor receptor (EGFR) on gastric epithelial cells via a signalling cascade involving a disintegrin and metalloprotease 17 (ADAM17) cleavage of membrane bound heparin binding-epidermal growth factor (HB-EGF). The effects of H. pylori on ADAM17 C-terminus in epithelial cells have been examined. Total cellular ADAM17 and surface expression of ADAM17 were significantly increased by H. pylori in AGS gastric epithelial cells. These changes were associated with ADAM17 C-terminal phosphorylation at T375 and S791. AGS cells lacking the ADAM17 C-terminal domain induced significantly attenuated cleavage of HB-EGF and were also unable to upregulate HB-EGF and EGFR transcripts to the same extent as cells expressing full length ADAM17. In mitotic unstimulated AGS and ADAM17 over-expressing AGS cells, ADAM17 was highly T735 phosphorylated indicating ADAM17 T735 phosphorylation is modified during the cell cycle. In conclusion, H. pylori induced ADAM17 C-terminal T735 and/or S791 phosphorylation in gastric epithelial cells are likely to be an important trigger inducing ADAM17 activation and shedding of HB-EGF leading to EGFR transactivation. ADAM17 over-expression in gastric cancer represents a potential target for therapeutic intervention.


Assuntos
Proteínas ADAM/metabolismo , Células Epiteliais/fisiologia , Helicobacter pylori/fisiologia , Treonina/metabolismo , Proteína ADAM17 , Linhagem Celular Tumoral , Células Epiteliais/imunologia , Helicobacter pylori/patogenicidade , Humanos , Fosforilação/imunologia , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/genética , Regulação para Cima/imunologia
7.
Mol Immunol ; 58(2): 160-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24365750

RESUMO

Mutations in the RAS family of oncogenes are highly prevalent in human cancer and, amongst its manifold effects, oncogenic RAS impairs the expression of components of the antigen presentation pathway. This allows evasion of cytotoxic T lymphocytes (CTL). CTL and natural killer (NK) cells are reciprocally regulated by MHC class I molecules and any gain in CTL recognition obtained by therapeutic inactivation of oncogenic RAS may be offset by reduced NK cell activation. We have investigated the consequences of targeted inactivation of oncogenic RAS on the recognition by both CTL and NK cells. Inactivation of oncogenic RAS, either by genetic deletion or inactivation with an inducible intracellular domain antibody (iDAb), increased MHC class I expression in human colorectal cell lines. The common RAS mutations, at codons 12, 13 and 61, all inhibited antigen presentation. Although MHC class I modulates the activity of both CTL and NK cells, the enhanced MHC class I expression resulting from inactivation of mutant KRAS did not significantly affect the in vitro recognition of these cell lines by either class of cytotoxic lymphocyte. These results show that oncogenic RAS and its downstream signalling pathways modulate the antigen presentation pathway and that this inhibition is reversible. However, the magnitude of these effects was not sufficient to alter the in vitro recognition of tumour cell lines by either CTL or NK cells.


Assuntos
Anticorpos/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas ras/imunologia , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Deleção de Genes , Células HCT116 , Humanos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Neoplasias/imunologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética
8.
PLoS One ; 7(5): e35981, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666318

RESUMO

Regulatory T-cells (T(Reg) cells) are increased in patients with multiple myeloma (MM). We investigated whether MM cells could generate and/or expand T(Reg) cells as a method of immuno-surveillance avoidance. In an in vitro model, CD4(+)CD25(-)FoxP3(-) T-cells co-cultured with malignant plasma cells (primary MM cells and cell lines) induced a significant generation of CD4(+)CD25(+)FoxP3(+) inducible T(Reg) cells (tT(Reg) cells; p<0.0001), in a contact-dependent manner. tT(Reg) cells were polyclonal, demonstrated a suppressive phenotype and phenotypically, demonstrated increased FoxP3 (p = 0.0001), increased GITR (p<0.0001), increased PD1 (p = 0.003) and decreased CD62L (p = 0.007) expression compared with naturally occurring T(Reg) cells. FACS-sorted tT(Reg) cells differentiated into FoxP(+)IL-17(+) and FoxP3(-)IL-17(+) CD4(+) cells upon TCR-mediated stimulation. Blocking experiments with anti-ICOS-L MoAb resulted in a significant inhibition of tT(Reg) cell generation whereas both IL-10 & TGFß blockade did not. MM tumour cells can directly generate functional T(Reg) cells in a contact-dependent manner, mediated by ICOS/ICOS-L. These features suggest that tumour generation of T(Reg) cells may contribute to evasion of immune surveillance by the host.


Assuntos
Comunicação Celular/imunologia , Mieloma Múltiplo/patologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Apresentadoras de Antígenos/imunologia , Linhagem Celular Tumoral , Humanos , Ligante Coestimulador de Linfócitos T Induzíveis/metabolismo , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interferon gama/biossíntese , Mieloma Múltiplo/imunologia , Fenótipo , Plasmócitos/patologia , Linfócitos T Reguladores/metabolismo
10.
BMC Med Genet ; 12: 84, 2011 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-21689419

RESUMO

BACKGROUND: XPC is involved in the nucleotide excision repair of DNA damaged by carcinogens known to cause bladder cancer. Individuals homozygous for the variant allele of XPC c.1496C > T (p.Ala499Val) were shown in a large pooled analysis to have an increased bladder cancer risk, and we found two 3'UTR variants, *611T > A and c.*618A > G, to be in strong linkage disequilibrium with c.1496T. Here we determined if these two 3'UTR variants can affect mRNA stability and assessed the impact of all three variants on mRNA and protein expression. METHODS: In vitro mRNA stability assays were performed and mRNA and protein expression measured both in plasmid-based assays and in lymphocytes and lymphoblastoid cell lines from bladder and breast cancer patients. RESULTS: The two 3'UTR variants were associated with reduced protein and mRNA expression in plasmid-based assays, suggesting an effect on mRNA stability and/or transcription/translation. A near-significant reduction in XPC protein expression (p = 0.058) was detected in lymphoblastoid cell lines homozygous for these alleles but no differences in mRNA stability in these lines was found or in mRNA or protein levels in lymphocytes heterozygous for these alleles. CONCLUSION: The two 3'UTR variants may be the variants underlying the association of c.1496C > T and bladder cancer risk acting via a mechanism modulating protein expression.


Assuntos
Regiões 3' não Traduzidas , Proteínas de Ligação a DNA/genética , Neoplasias da Bexiga Urinária/genética , Alelos , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Homozigoto , Humanos , Polimorfismo de Nucleotídeo Único , Biossíntese de Proteínas , Estabilidade de RNA , RNA Mensageiro/metabolismo , Fatores de Risco , Transcrição Gênica
11.
Carcinogenesis ; 32(4): 516-21, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21273643

RESUMO

The XPC gene is involved in repair of bulky DNA adducts formed by carcinogenic metabolites and oxidative DNA damage, both known bladder cancer risk factors. Single nucleotide polymorphisms (SNPs) in XPC have been associated with increased bladder cancer risk. Recently, rarer genetic variants have been identified but it is difficult to ascertain which are of functional importance. During a mutation screen of XPC in DNA from 33 bladder tumour samples and matched blood samples, we identified five novel variants in the patients' germ line DNA. In a case-control study of 771 bladder cancer cases and 800 controls, c.905T>C (Phe302Ser), c.1177C>T (Arg393Trp), c.*156G>A [3' untranslated region (UTR)] and c.2251-37C>A (in an intronic C>G SNP site) were found to be rare variants, with a combined odds ratio of 3.1 (95% confidence interval 1.0-9.8, P=0.048) for carriage of one variant. The fifth variant was a 2% minor allele frequency SNP not associated with bladder cancer. The two non-synonymous coding variants were predicted to have functional effects using analytical algorithms; a reduced recruitment of GFP-tagged XPC plasmids containing either c.905T>C or c.1177C>T to sites of 408 nm wavelength laser-induced oxidative DNA damage was found in vitro. c.*156G>A appeared to be associated with reduced messenger RNA stability in an in vitro plasmid-based assay. Although the laser microbeam assay is relevant to a range of DNA repair genes, our 3' UTR assay based on Green fluorescent protein(GFP) has widespread applicability and could be used to assess any gene. These assays may be useful in determining which rare variants are functional, prior to large genotyping efforts.


Assuntos
Proteínas de Ligação a DNA/genética , Polimorfismo de Nucleotídeo Único , Neoplasias da Bexiga Urinária/genética , Regiões 3' não Traduzidas/genética , Estudos de Casos e Controles , Linhagem Celular Tumoral , Humanos , Mutação
12.
Biochem J ; 431(3): 423-31, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20704564

RESUMO

Cytotoxic lymphocytes eliminate infected cells and tumours via the perforin-mediated delivery of pro-apoptotic serine proteases known as granzymes. Granzyme B triggers apoptosis via the cleavage of a repertoire of cellular proteins, leading to caspase activation and mitochondrial depolarization. A simple bioinformatics strategy identified a candidate granzyme B cleavage site in the widely expressed BNIP-2 (BCL2/adenovirus E1B-19K protein-interacting protein 2). Granzyme B cleaved recombinant BNIP-2 in vitro and endogenous BNIP-2 was cleaved during the NK (natural killer) cell-mediated killing of tumour cells. Cleavage required the site identified in the bioinformatics screen and was caspase-independent. Expression of either full-length BNIP-2 or a truncated molecule mimicking the granzyme B cleaved form was pro-apoptotic and led to the caspase-dependent cleavage of BNIP-2 at a site distinct from granzyme B cleavage. Inhibition of BNIP-2 expression did not affect the susceptibility to NK cell-mediated killing. Furthermore, target cells in which BID (BH3-interacting domain death agonist) expression was inhibited also remained highly susceptible to NK cell-mediated killing, revealing redundancy in the pro-apoptotic response to human cytotoxic lymphocytes. Such redundancy reduces the opportunity for escape from apoptosis induction and maximizes the chances of immune-mediated clearance of infected cells or tumour cells.


Assuntos
Proteínas de Transporte/metabolismo , Citotoxicidade Imunológica , Granzimas/metabolismo , Células Matadoras Naturais/imunologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/imunologia , Caspases/metabolismo , Linhagem Celular , Sequência Conservada , Granzimas/imunologia , Humanos , Células Matadoras Naturais/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência , Especificidade por Substrato
13.
Mob DNA ; 1(1): 9, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20226006

RESUMO

BACKGROUND: Diversity of immunoglobulins and the T cell antigen receptors is achieved via the recombination activating gene (RAG)-mediated rearrangement of variable (V), diversity (D) and joining (J) gene segments, and this underpins the efficient recognition of a seemingly limitless array of antigens. Analysis of V(D)J recombination activity is typically performed using extrachromosomal recombination substrates that are recovered from transfected cells and selected using bacterial transformation. We have developed a two-colour fluorescence-based system that simplifies detection of both deletion and inversion joining events mediated by RAG proteins. RESULTS: This system employs two fluorescent reporter genes that differentially mark unrearranged substrates and those that have undergone RAG-mediated deletion or inversion events. The recombination products bear the hallmarks of true V(D)J recombination and activity can be detected using fluorescence microscopy or flow cytometry. Recombination events can be detected without the need for cytotoxic selection of recombination products and the system allows analysis of recombination activity using substrates integrated into the genome. CONCLUSIONS: This system will be useful in the analysis and exploitation of the V(D)J recombination machinery and suggests that similar approaches could be used to replace expression of one gene with another during lymphocyte development.

14.
Brief Funct Genomic Proteomic ; 7(1): 8-16, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18208863

RESUMO

Natural killer (NK) cells are lymphocytes with an innate ability to recognize and kill infected cells and tumour cells. Unlike B and T cells, NK cells do not express an antigen receptor. Instead, NK cells detect changes in the phenotype of the target cell surface; malignant transformation or infection resulting in the loss or gain of particular molecules that are detected by inhibitory or activating receptors on the NK cell surface. The identification and characterization of NK cells and their receptors was made possible by monoclonal antibody technology. The ease with which genes and gene products can now be identified and manipulated has accelerated our understanding of NK cell function. Furthermore, gene and protein profiling studies are beginning to refine our understanding of NK cells, their interactions with other cells and their effector mechanisms. This review illustrates some of the basic features of NK cell biology and highlights the contribution made by post-genomic technology in defining the molecular mechanisms by which NK cells identify and kill susceptible targets.


Assuntos
Células Matadoras Naturais/imunologia , Apoptose , Clonagem Molecular , Biologia Computacional , Genômica , Humanos , Ligantes , Proteômica , Receptores Imunológicos/genética
15.
Carcinogenesis ; 28(2): 356-62, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16926174

RESUMO

Nitrosated glycine derivatives react with DNA to form O6-carboxymethyl-2'-deoxyguanosine (O6-CMdG) and O6-methyl-2'-deoxyguanosine (O6-MedG) adducts concurrently. O6-CMdG is not repaired by O6-alkylguanine alkyltransferases and might be expected to lead to mutations via a similar mechanism to O6-MedG. Potassium diazoacetate (KDA) is a stable form of nitrosated glycine and its ability to induce mutations in the p53 gene in a functional yeast assay was studied. Treatment of a plasmid containing the human p53 cDNA sequence with KDA afforded readily detectable levels of O6-CMdG and O6-MedG. The treated plasmid was used to transform yeast cells and coloured colonies harbouring a p53 sequence with functional mutations were detected. Recovery of the mutated plasmids followed by DNA sequencing enabled the mutation spectrum of KDA to be characterised. The most common mutations induced by KDA were substitutions with >50% occurring at GC base pairs. In contrast to the methylating agent methylnitrosourea which gives predominantly (>80%) GC-->AT transitions, KDA produced almost equal amounts of transitions (GC-->AT) and transversions (GC-->TA and AT-->TA). This difference is probably due to a different mode of base mispairing for O6-CMdG compared with O6-MedG. The pattern of mutations induced by KDA was very similar to the patterns observed in mutated p53 in human gastrointestinal tract tumours. These results are consistent with the hypothesis that nitrosation of glycine (or glycine derivatives) may contribute to characteristic human p53 mutation profiles. This conclusion is borne out by recent observations that O6-CMdG is present in human DNA both from blood and exfoliated colorectal cells and is consistent with recent epidemiological studies that have concluded that endogenous nitrosation arising from red meat consumption is related to an increased risk of colorectal cancer.


Assuntos
Compostos Azo/farmacologia , Adutos de DNA/química , Desoxiguanosina/análogos & derivados , Neoplasias Gastrointestinais/genética , Genes p53 , Mutação , Cromatografia de Afinidade , Cromatografia Líquida de Alta Pressão , Desoxiguanosina/química , Glicina/farmacologia , Humanos , Plasmídeos
16.
J Neurochem ; 90(2): 379-91, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15228595

RESUMO

Aggresomes are associated with many neurodegenerative disorders, including Parkinson's disease, and polyglutamine disorders such as Huntington's disease. These inclusions commonly contain ubiquitylated proteins. The stage at which these proteins are ubiquitylated remains unclear. A malfunction of the ubiquitin/proteasome system (UPS) may be associated with their formation. Conversely, it may reflect an unsuccessful attempt by the cell to remove them. Previously, we demonstrated that overexpression of Parkin, a ubiquitin-protein ligase associated with autosomal recessive juvenile Parkinsonism, generates aggresome-like inclusions in UPS compromised cells. Mutations in the de-ubiquitylating enzyme, UCH-L1, cause a rare form of Parkinsonism. We now demonstrate that overexpression of UCH-L1 also forms ribbon-like aggresomes in response to proteasomal inhibition. Disease-associated mutations, which affect enzymatic activities, significantly increased the number of inclusions. UCH-L1 aggresomes co-localized with ubiquitylated proteins, HSP70, gamma-tubulin and, to a lesser extent, the 20S proteasome and the chaperone BiP. Similar to Parkin inclusions, we found UCH-L1 aggresomes to be surrounded by a tubulin rather than a vimentin cage-like structure. Furthermore, UCH-L1 aggregates with Parkin and alpha-synuclein in some, but not all inclusions, suggesting the heterogeneous nature of these inclusion bodies. This study provides additional evidence that aggregation-prone proteins are likely to recruit UPS components in an attempt to clear proteins from failing proteasomes. Furthermore, UCH-L1 accumulation is likely to play a pathological role in inclusion formation in Parkinson's disease.


Assuntos
Cisteína Endopeptidases/metabolismo , Corpos de Inclusão/metabolismo , Complexos Multienzimáticos/metabolismo , Neurônios/metabolismo , Doença de Parkinson/genética , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Animais , Células COS , Proteínas de Transporte/metabolismo , Linhagem Celular , Chlorocebus aethiops , Chaperona BiP do Retículo Endoplasmático , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Corpos de Inclusão/química , Corpos de Inclusão/patologia , Corpos de Lewy/metabolismo , Corpos de Lewy/patologia , Substâncias Macromoleculares , Chaperonas Moleculares/metabolismo , Complexos Multienzimáticos/antagonistas & inibidores , Mutagênese Sítio-Dirigida , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Doença de Parkinson/metabolismo , Testes de Precipitina , Complexo de Endopeptidases do Proteassoma , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade , Sinucleínas , Transfecção , Tubulina (Proteína)/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , alfa-Sinucleína
17.
FEBS Lett ; 554(3): 501-4, 2003 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-14623119

RESUMO

Human homologue of Drosophila ariadne (HHARI) is a RING-IBR-RING domain protein identified through its ability to bind the human ubiquitin-conjugating enzyme, UbcH7. We now demonstrate that HHARI also interacts with the eukaryotic mRNA cap binding protein, translation initiation factor 4E homologous protein (4EHP), via the N-terminal RING1 finger of HHARI. HHARI, 4EHP and UbcH7 do not form a stable heterotrimeric complex as 4EHP cannot immunoprecipitate UbcH7 even in the presence of HHARI. Overexpression of 4EHP and HHARI in mammalian cells leads to polyubiquitylation of 4EHP. By contrast, HHARI does not promote its own autoubiquitylation. Thus, by promoting the ubiquitin-mediated degradation of 4EHP, HHARI may have a role in both protein degradation and protein translation.


Assuntos
Proteínas de Transporte/metabolismo , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Ubiquitina/metabolismo , Proteínas de Transporte/química , Linhagem Celular , Humanos , Testes de Precipitina , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Homologia de Sequência de Aminoácidos , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases
18.
Mol Biol Cell ; 14(11): 4541-56, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12937272

RESUMO

Association between protein inclusions and neurodegenerative diseases, including Parkinson's and Alzheimer's diseases, and polyglutamine disorders, has been widely documented. Although ubiquitin is conjugated to many of these aggregated proteins, the 26S proteasome does not efficiently degrade them. Mutations in the ubiquitin-protein ligase Parkin are associated with autosomal recessive juvenile Parkinsonism. Although Parkin-positive inclusions are not detected in brains of autosomal recessive juvenile Parkinsonism patients, Parkin is found in Lewy bodies in sporadic disease. This suggests that loss of Parkin ligase activity via mutation, or sequestration to Lewy bodies, is a contributory factor to sporadic disease onset. We now demonstrate that decreased proteasomal activity causes formation of large, noncytotoxic inclusions within the cytoplasm of both neuronal and nonneuronal cells overexpressing Parkin. This is not a general phenomenon as there is an absence of similar inclusions when HHARI, a structural homolog of Parkin, is overexpressed. The inclusions colocalize with ubiquitin and with proteasomes. Furthermore, Parkin inclusions colocalize with gamma-tubulin, acetylated alpha-tubulin, and cause redistribution of vimentin, suggesting aggresome-like properties. Our data imply that lower proteasomal activity, previously observed in brain tissue of Parkinson's disease patients, leads to Parkin accumulation and a concomitant reduction in ligase activity, thereby promoting Lewy body formation.


Assuntos
Cisteína Endopeptidases/metabolismo , Corpos de Inclusão/enzimologia , Complexos Multienzimáticos/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células COS , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Chlorocebus aethiops , Clonagem Molecular , Cisteína Endopeptidases/efeitos dos fármacos , Cisteína Endopeptidases/genética , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Peróxido de Hidrogênio/farmacologia , Corpos de Inclusão/genética , Leupeptinas/farmacologia , Doença por Corpos de Lewy/genética , Doença por Corpos de Lewy/metabolismo , Microscopia Confocal , Complexos Multienzimáticos/efeitos dos fármacos , Complexos Multienzimáticos/genética , Pressão Osmótica/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/genética , Complexo de Endopeptidases do Proteassoma , Sorbitol/farmacologia , Tubulina (Proteína)/metabolismo , Células Tumorais Cultivadas , Tunicamicina/farmacologia , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Vimentina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA