Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Virol ; 88(11): 6345-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672029

RESUMO

UNLABELLED: Kaposi's sarcoma-associated herpesvirus-encoded viral FLICE inhibitory protein (vFLIP) K13 was originally believed to protect virally infected cells against death receptor-induced apoptosis by interfering with caspase 8/FLICE activation. Subsequent studies revealed that K13 also activates the NF-κB pathway by binding to the NEMO/inhibitor of NF-κB (IκB) kinase gamma (IKKγ) subunit of an IKK complex and uses this pathway to modulate the expression of genes involved in cellular survival, proliferation, and the inflammatory response. However, it is not clear if K13 can also induce gene expression independently of NEMO/IKKγ. The minimum region of NEMO that is sufficient for supporting K13-induced NF-κB has not been delineated. Furthermore, the contribution of NEMO and NF-κB to the protective effect of K13 against death receptor-induced apoptosis remains to be determined. In this study, we used microarray analysis on K13-expressing wild-type and NEMO-deficient cells to demonstrate that NEMO is required for modulation of K13-induced genes. Reconstitution of NEMO-null cells revealed that the N-terminal 251 amino acid residues of NEMO are sufficient for supporting K13-induced NF-κB but fail to support tumor necrosis factor alpha (TNF-α)-induced NF-κB. K13 failed to protect NEMO-null cells against TNF-α-induced cell death but protected those reconstituted with the NEMO mutant truncated to include only the N-terminal 251 amino acid residues [the NEMO(1-251) mutant]. Taken collectively, our results demonstrate that NEMO is required for modulation of K13-induced genes and the N-terminal 251 amino acids of NEMO are sufficient for supporting K13-induced NF-κB. Finally, the ability of K13 to protect against TNF-α-induced cell death is critically dependent on its ability to interact with NEMO and activate NF-κB. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus-encoded vFLIP K13 is believed to protect virally infected cells against death receptor-induced apoptosis and to activate the NF-κB pathway by binding to adaptor protein NEMO/IKKγ. However, whether K13 can also induce gene expression independently of NEMO and the minimum region of NEMO that is sufficient for supporting K13-induced NF-κB remain to be delineated. Furthermore, the contribution of NEMO and NF-κB to the protective effect of K13 against death receptor-induced apoptosis is not clear. We demonstrate that NEMO is required for modulation of K13-induced genes and its N-terminal 251 amino acids are sufficient for supporting K13-induced NF-κB. The ability of K13 to protect against TNF-α-induced cell death is critically dependent on its ability to interact with NEMO and activate NF-κB. Our results suggest that K13-based gene therapy approaches may have utility for the treatment of patients with NEMO mutations and immunodeficiency.


Assuntos
Apoptose/genética , Regulação Viral da Expressão Gênica/genética , Quinase I-kappa B/metabolismo , Receptores de Morte Celular/metabolismo , Proteínas Virais/metabolismo , Animais , Western Blotting , Primers do DNA/genética , Fibroblastos , Células HEK293 , Humanos , Células Jurkat , Luciferases , Camundongos , Análise em Microsséries , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
J Virol ; 87(4): 2242-52, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23236068

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) has been linked to the development of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease (MCD). We have characterized the role of KSHV-encoded viral FLICE inhibitory protein (vFLIP) K13 in the modulation of anti-IgM-induced growth arrest and apoptosis in B cells. We demonstrate that K13 protects WEHI 231, an immature B-cell line, against anti-IgM-induced growth arrest and apoptosis. The protective effect of K13 was associated with the activation of the NF-κB pathway and was deficient in a mutant K13 with three alanine substitutions at positions 58 to 60 (K13-58AAA) and a structural homolog, vFLIP E8, both of which lack NF-κB activity. K13 upregulated the expression of NF-κB subunit RelB and blocked the anti-IgM-induced decline in c-Myc and rise in p27(Kip1) that have been associated with growth arrest and apoptosis. K13 also upregulated the expression of Mcl-1, an antiapoptotic member of the Bcl2 family. Finally, K13 protected the mature B-cell line Ramos against anti-IgM-induced apoptosis through NF-κB activation. Inhibition of anti-IgM-induced apoptosis by K13 may contribute to the development of KSHV-associated lymphoproliferative disorders.


Assuntos
Apoptose , Linfócitos B/imunologia , Linfócitos B/virologia , Herpesvirus Humano 8/patogenicidade , Interações Hospedeiro-Patógeno , Fator de Transcrição RelB/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Camundongos , Proteína de Sequência 1 de Leucemia de Células Mieloides , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
3.
J Biol Chem ; 286(32): 27988-97, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21705340

RESUMO

Myeloma cells are dependent on IL6 for their survival and proliferation during the early stages of disease, and independence from IL6 is associated with disease progression. The role of the NF-κB pathway in the IL6-independent growth of myeloma cells has not been studied. Because human herpesvirus 8-encoded K13 selectively activates the NF-κB pathway, we have used it as a molecular tool to examine the ability of the NF-κB pathway to confer IL6 independence on murine plasmacytomas. We demonstrated that ectopic expression of K13, but not its NF-κB-defective mutant or a structural homolog, protected plasmacytomas against IL6 withdrawal-induced apoptosis and resulted in emergence of IL6-independent clones that could proliferate long-term in vitro in the absence of IL6 and form abdominal plasmacytomas with visceral involvement when injected intraperitoneally into syngeneic mice. These IL6-independent clones were dependent on NF-κB activity for their survival and proliferation but were resistant to dexamethasone and INCB018424, a selective Janus kinase 1/2 inhibitor. Ectopic expression of human T cell leukemia virus 1-encoded Tax protein, which resembles K13 in inducing constitutive NF-κB activation, similarly protected plasmacytoma cells against IL6 withdrawal-induced apoptosis. Although K13 is known to up-regulate IL6 gene expression, its protective effect was not due to induction of endogenous IL6 production but instead was associated with sustained expression of several antiapoptotic members of the Bcl2 family upon IL6 withdrawal. Collectively, these results demonstrate that NF-κB activation cannot only promote the emergence of IL6 independence during myeloma progression but can also confer resistance to dexamethasone and INCB018424.


Assuntos
Antineoplásicos Hormonais/farmacologia , Dexametasona/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Interleucina-6/metabolismo , MAP Quinase Quinase 7/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , NF-kappa B/metabolismo , Plasmocitoma/metabolismo , Pirazóis/farmacologia , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Interleucina-6/genética , Interleucina-6/farmacologia , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , Camundongos , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Mutação , NF-kappa B/genética , Transplante de Neoplasias , Nitrilas , Plasmocitoma/tratamento farmacológico , Plasmocitoma/genética , Plasmocitoma/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pirimidinas , Transplante Isogênico
4.
J Biol Chem ; 286(24): 21555-64, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21531730

RESUMO

Expression of A20, a negative regulator of the NF-κB pathway, is frequently lost in several subtypes of Hodgkin and non-Hodgkin lymphoma. We report that A20 is expressed in Kaposi sarcoma-associated herpesvirus (KSHV)-infected primary effusion lymphoma cell lines, and its expression correlates closely with the expression of KSHV-encoded viral FLICE inhibitory protein K13. Ectopic expression of K13 induced A20 expression through NF-κB-mediated activation of A20 promoter. In turn, A20 blocked K13-induced NF-κB activity and up-regulation of proinflammatory cytokines CCL20 and IL-8 in a negative feedback fashion. Both the N-terminal deubiquitinating domain and the C-terminal zinc finger domain of A20 were involved in the inhibition of K13-induced NF-κB activity. Overexpression of A20 blocked K13-induced IκBα phosphorylation, NF-κB nuclear translocation, and cellular transformation. Consistent with the above, K13-induced IκBα phosphorylation and NF-κB transcriptional activation were enhanced in A20-deficient cells. Finally, A20 was found to interact physically with K13. Taken collectively, these results demonstrate that K13 is a key determinant of A20 expression in KSHV-infected cells, and A20 is a key negative regulator of K13-induced NF-κB activity. A20 might serve to control the inflammatory response to KSHV infection and protect KSHV-infected cells from apoptosis.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Regulação da Expressão Gênica , Herpesvirus Humano 8/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Virais/metabolismo , Apoptose , Linhagem Celular , Quimiocina CCL20/metabolismo , Proteínas de Ligação a DNA , Humanos , Inflamação , Interleucina-8/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
5.
Blood ; 113(22): 5660-8, 2009 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-19324905

RESUMO

Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is the etiologic agent of Kaposi sarcoma (KS), an angioproliferative lesion characterized by dramatic angiogenesis and inflammatory infiltration. In this study, we report that expression of chemokine CCL20, a potent chemoattractant of dendritic cells and lymphocytes, is strongly induced in cultured cells either by KSHV infection or on ectopic expression of viral FLICE inhibitory protein K13. This induction is caused by transcriptional activation of CCL20 gene, which is mediated by binding of the p65, p50, and c-Rel subunits of the transcription factor nuclear factor-kappaB (NF-kappaB) to an atypical NF-kappaB-binding site present in the CCL20 gene promoter. The CCL20 gene induction is defective in K13 mutants that lack NF-kappaB activity, and can be blocked by specific genetic and pharmacologic inhibitors of the NF-kappaB pathway. CCR6, the specific receptor for CCL20, is also induced in cultured cells either by KSHV infection or on K13 expression. Finally, expression of CCL20 and CCR6 is increased in clinical samples of KS. These results suggest that KSHV and K13-mediated induction of CCL20 and CCR6 may contribute to the recruitment of dendritic cells and lymphocytes into the KS lesions, and to tumor growth and metastases.


Assuntos
Quimiocina CCL20/genética , Herpesvirus Humano 8/fisiologia , NF-kappa B/metabolismo , Proteínas Virais/fisiologia , Sítios de Ligação , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/antagonistas & inibidores , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Células Cultivadas , Quimiocina CCL20/metabolismo , Células Dendríticas/patologia , Herpesvirus Humano 8/genética , Humanos , Células K562 , Linfócitos/patologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , RNA Interferente Pequeno/farmacologia , Receptores CCR6/genética , Receptores CCR6/metabolismo , Sarcoma de Kaposi/imunologia , Sarcoma de Kaposi/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/genética
6.
Sci Rep ; 11(1): 10002, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976304

RESUMO

Cytotoxicity assays are essential for the testing and development of novel immunotherapies for the treatment of cancer. We recently described a novel cytotoxicity assay, termed the Matador assay, which was based on marine luciferases and their engineered derivatives. In this study, we describe the development of a new cytotoxicity assay termed 'Matador-Glo assay' which takes advantage of a thermostable variant of Click Beetle Luciferase (Luc146-1H2). Matador-Glo assay utilizes Luc146-1H2 and D-luciferin as the luciferase-substrate pair for luminescence detection. The assay involves ectopic over-expression of Luc146-1H2 in the cytosol of target cells of interest. Upon damage to the membrane integrity, the Luc146-1H2 is either released from the dead and dying cells or its activity is preferentially measured in dead and dying cells. We demonstrate that this assay is simple, fast, specific, sensitive, cost-efficient, and not labor-intensive. We further demonstrate that the Matador-Glo assay can be combined with the marine luciferase-based Matador assay to develop a dual luciferase assay for cell death detection. Finally, we demonstrate that the Luc146-1H2 expressing target cells can also be used for in vivo bioluminescence imaging applications.


Assuntos
Benzotiazóis , Besouros/enzimologia , Testes Imunológicos de Citotoxicidade , Luciferases , Animais , Humanos , Células K562 , Camundongos Endogâmicos NOD , Camundongos SCID
7.
Sci Rep ; 10(1): 5712, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32235878

RESUMO

Primary effusion lymphoma (PEL) is a subtype of non-Hodgkin lymphoma associated with infection by Kaposi sarcoma-associated herpes virus (KSHV). PEL is an aggressive disease with extremely poor prognosis when treated with conventional chemotherapy. Narciclasine, a natural product present in Amaryllidaceae family of flowering plants including daffodils, belongs to a class of molecules termed 'isocarbostyril alkaloid'. We have found that narciclasine displays preferential cytotoxicity towards PEL at low nanomolar concentrations and is approximately 10 and 100-fold more potent than its structural analogs lycoricidine and lycorine, respectively. Narciclasine arrested cell-cycle progression at the G1 phase and induced apoptosis in PEL, which is accompanied by activation of caspase-3/7, cleavage of PARP and increase in the surface expression of Annexin-V. Although narciclasine treatment resulted in a marked decrease in the expression of MYC and its direct target genes,time-course experiments revealed that MYC is not a direct target of narciclasine. Narciclasine treatment neither induces the expression of KSHV-RTA/ORF50 nor the production of infectious KSHV virions in PEL. Finally, narciclasine provides dramatic survival advantages to mice in two distinct mouse xenograft models of PEL. In conclusion, our results suggest that narciclasine could be a promising agent for the treatment of PEL.


Assuntos
Alcaloides de Amaryllidaceae/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Linfoma de Efusão Primária/tratamento farmacológico , Fenantridinas/farmacologia , Extratos Vegetais/farmacologia , Alcaloides de Amaryllidaceae/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Linfoma de Efusão Primária/patologia , Camundongos , Fenantridinas/uso terapêutico , Extratos Vegetais/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 10(1): 2318, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32047180

RESUMO

Success of immunotherapeutic approaches using genetically engineered antibodies and T cells modified with chimeric antigen receptors (CARs) depends, among other things, on the selection of antigen binding domains with desirable expression and binding characteristics. We developed a luciferase-based assay, termed Malibu-Glo Assay, which streamlines the process of optimization of an antigen binding domain with desirable properties and allows the sensitive detection of tumor antigens. The assay involves a recombinant immunoconjugate, termed Malibu-Glo reagent, comprising an immunoglobulin or a non-immunoglobulin based antigen binding domain genetically linked to a marine luciferase. Malibu-Glo reagent can be conveniently produced in mammalian cells as a secreted protein that retains the functional activity of both the antigen binding domain and the luciferase. Moreover, crude supernatant containing the secreted Malibu-Glo reagent can directly be used for detection of cell surface antigens obviating the laborious steps of protein purification and labeling. We further demonstrate the utility of Malibu-Glo assay for the selection of optimal single chain fragment variables (scFvs) with desired affinity characteristics for incorporation into CARs. In summary, Malibu-Glo assay is a fast, simple, sensitive, specific and economical assay for antigen detection with multiple applications in the fields of antibody engineering, antibody humanization and CAR-T cell therapy.


Assuntos
Organismos Aquáticos/enzimologia , Engenharia Genética/métodos , Luciferases/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Anticorpos de Cadeia Única/imunologia , Animais , Humanos , Luciferases/genética , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Proteínas Recombinantes de Fusão/genética , Anticorpos de Cadeia Única/genética
9.
Sci Rep ; 9(1): 1957, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760795

RESUMO

Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy has produced dramatic responses in hematologic malignancies. One of the challenges in the field is the lack of a simple assay for the detection of CARs on the surface of immune effector cells. In this study, we describe a novel luciferase-based assay, termed Topanga Assay, for the detection of CAR expression. The assay utilizes a recombinant fusion protein, called Topanga reagent, generated by joining the extra-cellular domain of a CAR-target in frame with one of the marine luciferases or their engineered derivatives. The assay involves incubation of CAR expressing cells with the Topanga reagent, a few washes and measurement of luminescence. The assay can detect CARs comprising either immunoglobulin- or non-immunoglobulin-based antigen binding domains. We further demonstrate that addition of epitope tags to the Topanga reagent not only allows its convenient one step purification but also extends its use for detection of CAR cells using flow cytometry. However, crude supernatant containing the secreted Topanga reagent can be directly used in both luminescence and flow-cytometry based assays without prior protein purification. Our results demonstrate that the Topanga assay is a highly sensitive, specific, convenient, economical and versatile assay for the detection of CARs.


Assuntos
Imunoterapia Adotiva/métodos , Luciferases/metabolismo , Receptores de Antígenos Quiméricos/análise , Linhagem Celular , Citometria de Fluxo/métodos , Humanos , Linfócitos/metabolismo , Receptores de Antígenos/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Linfócitos T/imunologia
10.
Sci Rep ; 8(1): 199, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29317736

RESUMO

A simple, accurate, sensitive and robust assay that can rapidly and specifically measure the death of target cells would have applications in many areas of biomedicine and particularly for the development of novel cellular- and immune-therapeutics. In this study, we describe a novel cytotoxicity assay, termed the Matador assay, which takes advantage of the extreme brightness, stability and glow-like characteristics of recently discovered novel marine luciferases and their engineered derivatives. The assay involves expression of a luciferase of interest in target cells in a manner so that it is preferentially retained within the healthy cells but is either released from dead and dying cells or whose activity can be preferentially measured in dead and dying cells. We demonstrate that this assay is highly sensitive, specific, rapid, and can be performed in a single-step manner without the need for any expensive equipment. We further validate this assay by demonstrating its ability to detect cytotoxicity induced by several cellular and immune-therapeutic agents including antibodies, natural killer cells, chimeric antigen receptor expressing T cells and a bispecific T cell engager.


Assuntos
Luciferases/metabolismo , Testes de Toxicidade/métodos , Linhagem Celular Tumoral , Células Cultivadas , Células HEK293 , Humanos , Luciferases/genética
11.
Cancer Res ; 62(20): 5897-901, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12384554

RESUMO

Methylation of the promoter regions of CpG-rich sites in genes is the major mechanism for the silencing of many genes in tumors. Methylation of the key apoptosis-related gene caspase 8 (CASP8) has been reported in some childhood tumors and in neuroendocrine lung tumors. We examined the methylation status of 181 pediatric tumors and found frequent methylation in rhabdomyosarcomas (83%), medulloblastomas (81%), retinoblastomas (59%), and neuroblastomas (52%). Methylation frequencies were low in Wilms' tumors (19%) and absent in hepatoblastomas, acute leukemias, osteosarcomas, Ewing's sarcomas, and ganglioneuromas and in normal tissues. Methylation of CASP8 and the tumor suppressor gene RASSF1A were highly significantly correlated in all tumor types by both the chi(2) and the Fisher's exact tests (P < 0.0001 for both tests). Because the region of the gene examined by us and others is not located in the promoter region and lacks features of a CpG island, we explored the relationship between methylation and gene silencing in detail using 23 pediatric tumor cell lines. Studies included relating the methylation of the region to gene expression at mRNA and protein levels, enzymatic assays of gene function, clonal analysis of PCR amplicons of the region, and exposure to a demethylating agent. These studies indicated that methylation correlated with the loss of gene function in most cases; however, other mechanisms of gene inactivation were present in some cases. Posttranscriptional inactivation of the closely related gene caspase 10 was present in many cell lines. Our results suggest that deregulation of the death receptor pathway to apoptosis is frequent in many types of pediatric tumors and their cell lines.


Assuntos
Azacitidina/análogos & derivados , Caspases/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias/enzimologia , Proteínas Supressoras de Tumor , Azacitidina/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Caspase 10 , Caspase 8 , Caspase 9 , Caspases/genética , Criança , Metilação de DNA , Decitabina , Inibidores Enzimáticos/farmacologia , Amplificação de Genes , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Genes myc , Humanos , Ácidos Hidroxâmicos/farmacologia , Proteínas de Neoplasias/genética , Neoplasias/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Células Tumorais Cultivadas
12.
Oncogene ; 22(36): 5712-5, 2003 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-12944921

RESUMO

Post-transcriptional gene silencing by small interfering RNAs (siRNAs) is rapidly becoming a powerful tool for genetic analysis of mammalian cells. Delivery of siRNA into mammalian cells is usually achieved via the transfection of double-stranded oligonucleotides or plasmids encoding RNA polymerase III promoter-driven small hairpin RNA. Recently, retroviral vectors have been used for siRNA delivery, which overcome the problem of poor transfection efficiency seen with the plasmid-based systems. However, retroviral vectors have several limitations, such as the need for active cell division for gene transduction, oncogenic potential, low titers and gene silencing. In this report, we have adapted a commercially available adenoassociated virus (AAV) vector for siRNA delivery into mammalian cells. We demonstrate the ability of this modified vector to deliver efficiently siRNA into HeLa S3 cells and downregulate p53 and caspase 8 expression. Our results suggest that AAV-based vectors are efficient vectors for the delivery of siRNA into mammalian cells. Based on the known ability of these vectors to infect both dividing and nondividing cells, their use as a therapeutic tool for the delivery of siRNA deserves further study.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos , RNA Interferente Pequeno/administração & dosagem , Caspase 8 , Caspase 9 , Caspases/genética , Genes p53
13.
Oncogene ; 21(55): 8510-4, 2002 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-12466971

RESUMO

We have previously reported that the key apoptosis related gene caspase 8 (CASP8) is frequently silenced in small cell lung cancer (SCLC) tumors and cell lines usually, but not always, by aberrant promoter methylation. Because CASP8 is a key component of the death-inducing signaling complex (DISC) when specific death receptors (including DR4, DR5, FAS) are activated by their specific ligands (TRAIL/FASL), we examined expression of the components of the DISC complex in lung cancer cell lines. MYC family members are frequently amplified (MYC+ve) in SCLC, and MYC is a potent inducer of apoptosis. We examined 34 SCLC lines (12 of which were MYC+ve) and 22 NSCLC lines. CASP8 gene expression was frequently lost (79%) at message and protein levels in SCLC but not in non-SCLC (NSCLC). MYC amplification was present in 45% of SCLC cell lines, which had lost CASP8 expression, but not in any of the CASP8 positive lines. The frequency of CASP8 loss was significantly higher in MYC+ve SCLC compared to MYC-ve SCLC or in NSCLC. Analyses of other DISC components showed significantly higher rates of loss of expression of CASP10, DR5, FAS and FASL in SCLC compared to NSCLC. The loss of expression of proapoptotic DISC components was significantly higher in MYC+ve SCLC cell lines and these lines were completely resistant to TRAIL. Expression of CASP10 (a caspase closely related to CASP8) was frequently absent at the protein level in both SCLC and NSCLC lines. Expression of c-FLIP (proteolytically inactive homolog of CASP8) was inversely related to expression of CASP8. Our major conclusions are: (a) The death receptor pathway is differently inactivated at multiple levels in lung cancer cell lines; and (b) MYC amplification in SCLC is associated with inactivation of most components of the DISC complex, with resistance to TRAIL and with expression of c-FLIP. These findings may have considerable clinical and therapeutic implications.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes myc , Neoplasias Pulmonares/genética , Receptores do Fator de Necrose Tumoral/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma de Células Pequenas/genética , Caspase 8 , Caspase 9 , Caspases/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte , Amplificação de Genes , Deleção de Genes , Humanos , Células Tumorais Cultivadas
14.
Retrovirology ; 2: 9, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15713234

RESUMO

BACKGROUND: The nuclear transcription factor NF-kappaB binds to the HIV-1 long terminal repeat (LTR) and is a key regulator of HIV-1 gene expression in cells latently infected with this virus. In this report, we have analyzed the ability of Kaposi's sarcoma associate herpes virus (KSHV, also known as Human Herpes virus 8)-encoded viral FLIP (Fas-associated death domain-like IL-1 beta-converting enzyme inhibitory protein) K13 to activate the HIV-1 LTR. RESULTS: We present evidence that vFLIP K13 activates HIV-1 LTR via the activation of the classical NF-kappaB pathway involving c-Rel, p65 and p50 subunits. K13-induced HIV-1 LTR transcriptional activation requires the cooperative interaction of all three components of the IKK complex and can be effectively blocked by inhibitors of the classical NF-kappaB pathway. K13 mutants that lacked the ability to activate the NF-kappaB pathway also failed to activate the HIV-1 LTR. K13 could effectively activate a HIV-1 LTR reporter construct lacking the Tat binding site but failed to activate a construct lacking the NF-kappaB binding sites. However, coexpression of HIV-1 Tat with K13 led to synergistic activation of HIV-1 LTR. Finally, K13 differentially activated HIV-1 LTRs derived from different strains of HIV-1, which correlated with their responsiveness to NF-kappaB pathway. CONCLUSIONS: Our results suggest that concomitant infection with KSHV/HHV8 may stimulate HIV-1 LTR via vFLIP K13-induced classical NF-kappaB pathway which cooperates with HIV-1 Tat protein.


Assuntos
Produtos do Gene tat/metabolismo , Repetição Terminal Longa de HIV/fisiologia , NF-kappa B/metabolismo , Ativação Transcricional , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Regulação Viral da Expressão Gênica , Repetição Terminal Longa de HIV/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Células Jurkat , Camundongos , Transcrição Gênica , Proteínas Virais/genética
15.
Cancer Biol Ther ; 4(1): 77-82, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15662128

RESUMO

The ubiquitin-proteasome pathway is responsible for degrading many critical regulatory proteins involved in immune and inflammatory responses, control of cell growth and apoptosis. Recently, proteasome inhibitors have emerged as promising new therapeutic agents in hematological malignancies. Here we show that Bortezomib (PS-341), a proteasome-inhibitor, inhibits cellular proliferation and induces apoptosis in cell lines derived from Primary Effusion Lymphoma (PEL), a subtype of non-Hodgkin's lymphoma associated with infection by human herpes virus 8 (HHV-8). Bortezomib demonstrated more cytotoxicity against PEL cells than against cell lines derived from multiple myeloma, a disease for which is in current clinical use. Apoptosis induced by Bortezomib was associated with inhibition of the classical and alternative NF-kappaB pathways, upregulation of p53, p21 and p27 and activation of caspase cascade. Finally, treatment of PEL cells with Bortezomib exerted a synergistic or additive cytotoxic effect in combination with chemotherapeutic drugs or TRAIL. Taken together, these findings suggest that Bortezomib represents a promising agent for the treatment of PEL.


Assuntos
Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Linfoma não Hodgkin/patologia , Pirazinas/farmacologia , Apoptose , Bortezomib , Infecções por Herpesviridae/complicações , Herpesvirus Humano 8/patogenicidade , Humanos , Linfoma não Hodgkin/virologia , Células Tumorais Cultivadas
16.
Cancer Biol Ther ; 2(2): 206-10, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12750565

RESUMO

Silencing of gene expression by small interfering RNAs (siRNAs) is rapidly becoming a powerful tool for genetic analysis of mammalian cells. Although in the original studies expression of siRNA in mammalian cells was achieved via the transfection of double stranded oligonucleotides, subsequent studies described the use of plasmids to achieve long-term and stable expression of siRNA. Recently, several groups have described the use of retroviral vectors for siRNA delivery. However, retroviral vectors require active cell division for gene transfer and also suffer from the problem of gene-silencing. In this report we have modified a commercially available self-inactivating lentiviral vector for the delivery of siRNA into mammalian cells. We demonstrate the ability of this modified vector to efficiently transfer siRNA into HeLa S3 cells and downregulate p53 expression. Our results suggest that lentiviruses are efficient vectors for delivery of siRNA into mammalian cells. Based on the known ability of these vectors to infect both dividing and non-dividing cells and achieve long-term multilineage gene expression, their use as a therapeutic tool for the delivery of siRNA deserves further study.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Vetores Genéticos , Lentivirus/genética , RNA Interferente Pequeno/genética , Primers do DNA/química , Regulação para Baixo , Inativação Gênica , Células HeLa , Humanos , RNA Interferente Pequeno/metabolismo , Transfecção , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Cancer Biol Ther ; 1(6): 652-60, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12642689

RESUMO

MRIT (Mach-related inducer of toxicity)/cFLIP (cellular FADD like interlukin1-beta converting enzyme inhibitory protein) is a proteolytically inactive structural homologue of caspase-8, which is known to protect against death receptors-induced apoptosis by blocking the activation of caspase-8. We have observed that exogenous expression of MRITalpha1/cFLIP(L) isoform also protects against cell death induced by a diverse group of chemotherapeutic drugs with different mechanisms of action, including doxorubicin, etoposide, cytosine arabinoside, daunorubicin, chlorambucil and cisplatin. However, MRITalpha1/cFLIP(L) failed to protect against apoptosis induced by paclitaxel and vincristine, two microtubule-damaging agents. Although MRITalpha1/cFLIP(L) protects against chemotherapy-induced apoptosis in both solid tumor and hematopoietic cell lines, this effect was more pronounced in the former. MRITalpha1/cFLIP(L) expression is decreased during drug-induced apoptosis and exogenous expression of MRITalpha1/cFLIP(L) delays the activation of caspase-8 and -3 during drug- induced apoptosis. These results suggest that MRITalpha1/cFLIPL may be an important determinant of both death receptor- and chemotherapy-induced apoptosis and strategies aimed at downregulating its expression deserve further study as a way to overcome multidrug resistance to cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte/fisiologia , Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias/prevenção & controle , Western Blotting , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Citoproteção/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Neoplasias/patologia , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas
18.
Clin Cancer Res ; 19(18): 5016-26, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23881928

RESUMO

PURPOSE: Kaposi sarcoma-associated herpes virus (KSHV)-associated primary effusion lymphomas (PEL) have extremely poor prognosis when treated with conventional chemotherapy. KSHV-encoded viral FLICE-inhibitory protein (vFLIP) K13 binds to the IkappaB kinase (IKK) complex to constitutively activate the NF-κB pathway, which has been shown to be essential for the survival and proliferation of PEL cells. The molecular chaperone HSP90 is a component of the IKK complex and is required for its activity. EXPERIMENTAL DESIGN: We have analyzed the effect of HSP90 inhibitors on the survival and proliferation of PEL cells and on the activity of the NF-κB pathway. RESULTS: We show that BIIB021, a purine scaffold-based orally administrable HSP90 inhibitor, shows preferential cytotoxicity toward PEL cells as compared with non-PEL cells. The cytotoxic effect of BIIB021 against PEL was associated with induction of cell-cycle arrest and apoptosis. BIIB021 blocked the expression of a number of cellular proteins involved in the regulation of cell cycle and apoptosis. BIIB021 also blocked constitutive NF-κB activity present in PEL cells, in part, by blocking the interaction of vFLIP K13 with the IKK complex subunits. In a xenograft model of PEL, BIIB021 significantly reduced tumor growth. CONCLUSION: BIIB021 blocks constitutive NF-κB activity in PEL and shows preferential antitumor activity against PEL in vitro and in vivo. BIIB021 may be a promising agent for treatment of PEL.


Assuntos
Adenina/análogos & derivados , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Humano 8/patogenicidade , Linfoma de Efusão Primária/tratamento farmacológico , NF-kappa B/antagonistas & inibidores , Piridinas/farmacologia , Proteínas Virais/antagonistas & inibidores , Adenina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Caspases/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/metabolismo , Humanos , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/virologia , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Purinas/química , Células Tumorais Cultivadas , Proteínas Virais/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
PLoS One ; 7(5): e37498, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22624040

RESUMO

Infection with Kaposi's sarcoma associated herpesvirus (KSHV) has been linked to the development of primary effusion lymphoma (PEL), a rare lymphoproliferative disorder that is characterized by loss of expression of most B cell markers and effusions in the body cavities. This unique clinical presentation of PEL has been attributed to their distinctive plasmablastic gene expression profile that shows overexpression of genes involved in inflammation, adhesion and invasion. KSHV-encoded latent protein vFLIP K13 has been previously shown to promote the survival and proliferation of PEL cells. In this study, we employed gene array analysis to characterize the effect of K13 on global gene expression in PEL-derived BCBL1 cells, which express negligible K13 endogenously. We demonstrate that K13 upregulates the expression of a number of NF-κB responsive genes involved in cytokine signaling, cell death, adhesion, inflammation and immune response, including two NF-κB subunits involved in the alternate NF-κB pathway, RELB and NFKB2. In contrast, CD19, a B cell marker, was one of the genes downregulated by K13. A comparison with K13-induced genes in human vascular endothelial cells revealed that although there was a considerable overlap among the genes induced by K13 in the two cell types, chemokines genes were preferentially induced in HUVEC with few exceptions, such as RANTES/CCL5, which was induced in both cell types. Functional studies confirmed that K13 activated the RANTES/CCL5 promoter through the NF-κB pathway. Taken collectively, our results suggest that K13 may contribute to the unique gene expression profile, immunophenotype and clinical presentation that are characteristics of KSHV-associated PEL.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/virologia , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Humanos , Luciferases , Linfoma de Efusão Primária/etiologia , Linfoma de Efusão Primária/genética , Análise em Microsséries , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Virais/genética
20.
PLoS One ; 7(5): e36601, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22590573

RESUMO

BACKGROUND: Kaposi's sarcoma associated herpesvirus encoded viral FLICE inhibitory protein (vFLIP) K13 activates the NF-κB pathway by binding to the NEMO/IKKγ subunit of the IκB kinase (IKK) complex. However, it has remained enigmatic how K13-NEMO interaction results in the activation of the IKK complex. Recent studies have implicated TRAF6, TAK1 and linear ubiquitin chains assembled by a linear ubiquitin chain assembly complex (LUBAC) consisting of HOIL-1, HOIP and SHARPIN in IKK activation by proinflammatory cytokines. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that K13-induced NF-κB DNA binding and transcriptional activities are not impaired in cells derived from mice with targeted disruption of TRAF6, TAK1 and HOIL-1 genes and in cells derived from mice with chronic proliferative dermatitis (cpdm), which have mutation in the Sharpin gene (Sharpin(cpdm/cpdm)). Furthermore, reconstitution of NEMO-deficient murine embryonic fibroblast cells with NEMO mutants that are incapable of binding to linear ubiquitin chains supported K13-induced NF-κB activity. K13-induced NF-κB activity was not blocked by CYLD, a deubiquitylating enzyme that can cleave linear and Lys63-linked ubiquitin chains. On the other hand, NEMO was required for interaction of K13 with IKK1/IKKα and IKK2/IKKß, which resulted in their activation by "T Loop" phosphorylation. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that K13 activates the NF-κB pathway by binding to NEMO which results in the recruitment of IKK1/IKKα and IKK2/IKKß and their subsequent activation by phosphorylation. Thus, K13 activates NF-κB via a mechanism distinct from that utilized by inflammatory cytokines. These results have important implications for the development of therapeutic agents targeting K13-induced NF-κB for the treatment of KSHV-associated malignancies.


Assuntos
Herpesvirus Humano 8/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Sarcoma de Kaposi/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Virais/metabolismo , Animais , Doença Crônica , Dermatite/genética , Dermatite/metabolismo , Dermatite/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Herpesvirus Humano 8/genética , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Jurkat , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , NF-kappa B/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/patologia , Fator 6 Associado a Receptor de TNF/genética , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa