Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Ther ; 20(8): 1622-32, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22801446

RESUMO

Severe and fatal viral infections remain common after hematopoietic stem cell transplantation. Adoptive transfer of cytotoxic T lymphocytes (CTLs) specific for Epstein-Barr virus (EBV), cytomegalovirus (CMV), and adenoviral antigens can treat infections that are impervious to conventional therapies, but broader implementation and extension to additional viruses is limited by competition between virus-derived antigens and time-consuming and laborious manufacturing procedures. We now describe a system that rapidly generates a single preparation of polyclonal (CD4(+) and CD8(+)) CTLs that is consistently specific for 15 immunodominant and subdominant antigens derived from 7 viruses (EBV, CMV, Adenovirus (Adv), BK, human herpes virus (HHV)-6, respiratory syncytial virus (RSV), and Influenza) that commonly cause post-transplant morbidity and mortality. CTLs can be rapidly produced (10 days) by a single stimulation of donor peripheral blood mononuclear cells (PBMCs) with a peptide mixture spanning the target antigens in the presence of the potent prosurvival cytokines interleukin-4 (IL4) and IL7. This approach reduces the impact of antigenic competition with a consequent increase in the antigenic repertoire and frequency of virus-specific T cells. Our approach can be readily introduced into clinical practice and should be a cost-effective alternative to common antiviral prophylactic agents for allogeneic hematopoietic stem cell transplant (HSCT) recipients.


Assuntos
Linfócitos T Citotóxicos/imunologia , Viroses/terapia , Antígenos Virais/imunologia , Células Cultivadas , Citomegalovirus/imunologia , Citometria de Fluxo , Herpesvirus Humano 4/imunologia , Humanos , Imunoterapia Adotiva , Viroses/imunologia
2.
Mol Ther ; 19(12): 2239-48, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21878902

RESUMO

Despite activity as single agent cancer therapies, Rapamycin (rapa) and its rapalogs may have their greatest effects when combined with other therapeutic modalities. In addition to direct antitumor activity, rapalogs reverse multiple tumor-intrinsic immune evasion mechanisms. These should facilitate tumor-specific T cell activity, but since rapa directly inhibits effector T cells, this potential immune enhancement is lost. We hypothesized that if T cells were rendered resistant to rapa they could capitalize on its downregulation of tumor immune evasion. We therefore modified T cells with a rapa-resistant mutant of mTor, mTorRR, and directed them to B lymphomas by coexpressing a chimeric antigen receptor (CAR) for CD19 (CAR.CD19-28ζ). T cells expressing transgenic mTorRR from a piggyBac transposon maintain mTor signaling, proliferate in the presence of rapa and retain their cytotoxic function and ability to secrete interferon-γ (IFNγ) after stimulation, effector functions that were inhibited by rapa in control T cells. In combination, rapa and rapa-resistant-CAR.CD19-28ζ-expressing T cells produced greater antitumor activity against Burkitt's lymphoma and pre-B ALL cell lines in vitro than CAR.CD19-28ζ T cells or rapa alone. In conclusion, the combination of rapa and rapa-resistant, CAR.CD19-28ζ-expressing T cells may provide a novel therapy for the treatment of B cell malignancies and other cancers.


Assuntos
Linfoma de Burkitt/prevenção & controle , Resistencia a Medicamentos Antineoplásicos , Imunossupressores/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/prevenção & controle , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Antígenos CD19/genética , Antígenos CD19/metabolismo , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Western Blotting , Linfoma de Burkitt/genética , Linfoma de Burkitt/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Terapia Combinada , Feminino , Citometria de Fluxo , Humanos , Interferon gama/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/imunologia , Receptores de Antígenos/genética , Receptores de Antígenos/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Serina-Treonina Quinases TOR/metabolismo
3.
Mol Ther ; 19(12): 2133-43, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21772253

RESUMO

Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) can be modified to function as heterologous tumor directed effector cells that survive longer in vivo than tumor directed T cells without virus specificity, due to chronic stimulation by viral antigens expressed during persistent infection in seropositive individuals. We evaluated the nonviral piggyBac (PB) transposon system as a platform for modifying EBV-CTLs to express a functional human epidermal growth factor receptor 2-specific chimeric antigen receptor (HER2-CAR) thereby directing virus-specific, gene modified CTLs towards HER2-positive cancer cells. Peripheral blood mononuclear cells (PBMCs) were nucleofected with transposons encoding a HER2-CAR and a truncated CD19 molecule for selection followed by specific activation and expansion of EBV-CTLs. HER2-CAR was expressed in ~40% of T cells after CD19 selection with retention of immunophenotype, polyclonality, and function. HER2-CAR-modified EBV-CTLs (HER2-CTLs) killed HER2-positive brain tumor cell lines in vitro, exhibited transient and reversible increases in HER2-CAR expression following antigen-specific stimulation, and stably expressed HER2-CAR beyond 120 days. Adoptive transfer of PB-modified HER2-CTLs resulted in tumor regression in a murine xenograft model. Our results demonstrate that PB can be used to redirect virus-specific CTLs to tumor targets, which should prolong tumor-specific T cell survival in vivo producing more efficacious immunotherapy.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Imunoterapia , Receptor ErbB-2/genética , Receptores de Antígenos/genética , Proteínas Recombinantes de Fusão/genética , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno , Antígenos CD19/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/terapia , Citometria de Fluxo , Herpesvirus Humano 4/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptor ErbB-2/imunologia , Receptores de Antígenos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Transdução Genética , Células Tumorais Cultivadas
4.
Blood ; 114(23): 4784-91, 2009 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19759356

RESUMO

Adoptive transfer of autologous Epstein-Barr virus-specific cytotoxic T lymphocytes (EBV-CTLs) to solid organ transplant (SOT) recipients has been shown safe and effective for the treatment of EBV-associated posttransplantation lymphoproliferative disorders (PTLDs). SOT recipients, however, require the continuous administration of immunosuppressive drugs to prevent graft rejection, and these agents may significantly limit the long-term persistence of transferred EBV-CTLs, precluding their use as prophylaxis. Tacrolimus (FK506) is one of the most widely used immunosuppressive agents in SOT recipients, and its immunosuppressive effects are largely dependent on its interaction with the 12-kDa FK506-binding protein (FKBP12). We have knocked down the expression of FKBP12 in EBV-CTLs using a specific small interfering RNA (siRNA) stably expressed from a retroviral vector and found that FKBP12-silenced EBV-CTLs are FK506 resistant. These cells continue to expand in the presence of the drug without measurable impairment of their antigen specificity or cytotoxic activity. We confirmed their FK506 resistance and anti-PTLD activity in vivo using a xenogenic mouse model, suggesting that the proposed strategy may be of value to enhance EBV-specific immune surveillance in patients at high risk of PTLD after transplantation.


Assuntos
Técnicas de Silenciamento de Genes , Vetores Genéticos/farmacologia , Herpesvirus Humano 4/imunologia , Imunossupressores/uso terapêutico , RNA Interferente Pequeno/fisiologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Proteína 1A de Ligação a Tacrolimo/antagonistas & inibidores , Tacrolimo/farmacologia , Transferência Adotiva , Animais , Antígenos Virais/imunologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/imunologia , Células Cultivadas/transplante , Citotoxicidade Imunológica , Resistência a Medicamentos/genética , Humanos , Linfoma não Hodgkin/terapia , Linfoma não Hodgkin/virologia , Camundongos , Camundongos SCID , Processamento de Proteína Pós-Traducional , RNA Mensageiro/antagonistas & inibidores , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Retroviridae/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/transplante , Proteína 1A de Ligação a Tacrolimo/efeitos dos fármacos , Proteína 1A de Ligação a Tacrolimo/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Ther ; 18(11): 2006-17, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20842106

RESUMO

Adoptive transfer of antigen-specific cytotoxic T lymphocytes has shown promise for the therapy of cancer. However, tumor-specific T cells are susceptible to diverse inhibitory signals from the tumor microenvironment. The Akt/protein kinase B plays a central role in T-cell proliferation, function, and survival and we hypothesized that expression of constitutively active Akt (caAkt) in T cells could provide resistance to many of these tumor-associated inhibitory mechanisms. caAkt expression in activated human T cells increased proliferation and cytokine production, a likely result of their sustained expression of nuclear factor-κB (NF-κB) and provided resistance to apoptosis by upregulating antiapoptotic molecules. caAkt expressing T cells (caAkt-T-cells) were also relatively resistant to suppression by and conversion into regulatory T cells (Tregs). These characteristics provided a survival advantage to T cells cocultured with tumor cells in vitro; CD3/28-stimulated T cells expressing a chimeric antigen receptor (CAR) specific for disialoganglioside (GD2) that redirected their activity to the immunosuppressive, GD2-expressing neuroblastoma cell line, LAN-1, resisted tumor-induced apoptosis when co-expressing transgenic caAkt. In conclusion, caAkt-transduced T cells showed resistance to several evasion strategies employed by tumors and may therefore enhance the antitumor activity of adoptively transferred T lymphocytes.


Assuntos
Apoptose , NF-kappa B/metabolismo , Neuroblastoma/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linfócitos T/metabolismo , Western Blotting , Proliferação de Células , Citometria de Fluxo , Gangliosídeos/metabolismo , Humanos , Ativação Linfocitária , NF-kappa B/genética , Neuroblastoma/imunologia , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transdução Genética , Fator de Crescimento Transformador beta/farmacologia , Carga Tumoral/imunologia , Células Tumorais Cultivadas
6.
Mol Cell Biol ; 27(8): 2910-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17296724

RESUMO

As a key mediator of type I interferon (IFN) (IFN-alpha/beta) responses, IFN regulatory factor 7 (IRF7) is essential to host immune defenses. Activation of IRF7 generally requires virus-induced C-terminal phosphorylation, which leads to its nuclear accumulation and activation of target genes. Here we use the Epstein-Barr virus (EBV) oncoprotein LMP1, which activates IRF7, to identify factors involved in IRF7 activation. We demonstrate for the first time that RIP activates IRF7 and that RIP and IRF7 interact under physiological conditions in EBV-positive Burkitt's lymphoma cells. We provide evidence that both RIP and IRF7 are ubiquitinated in these cells and that IRF7 preferentially interacts with ubiquitinated RIP. RIP is required for full activation of IRF7 by LMP1, with LMP1 stimulating the ubiquitination of RIP and its interaction with IRF7. Moreover, LMP1 stimulates RIP-dependent K63-linked ubiquitination of IRF7, which regulates protein function rather than proteasomal degradation of proteins. We suggest that RIP may serve as a general activator of IRF7, responding to and transmitting the signals from various stimuli, and that ubiquitination may be a general mechanism for enhancing the activity of IRF7.


Assuntos
Fator Regulador 7 de Interferon/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Ubiquitina/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Genes Dominantes , Humanos , Fator Regulador 7 de Interferon/genética , Células Jurkat , Camundongos , Modelos Biológicos , Ligação Proteica , Fator 6 Associado a Receptor de TNF/metabolismo
7.
Mol Cell Biol ; 22(10): 3460-73, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11971977

RESUMO

Although both RAG-1 and RAG-2 are required for all steps of V(D)J recombination, little is known about the specific contribution of either protein to these steps. RAG-1 contains three acidic active-site amino acids that are thought to coordinate catalytic metal ions. To search for additional catalytic amino acids and to better define the functional anatomy of RAG-1, we mutated all 86 conserved basic amino acids to alanine and evaluated the mutant proteins for DNA binding, nicking, hairpin formation, and joining. We found several amino acids outside of the canonical nonamer-binding domain that are critical for DNA binding, several step arrest mutants with defects in nicking or hairpin formation, and four RAG-1 mutants defective specifically for joining. Analysis of coding joints formed by some of these mutants revealed excessive deletions, frequent use of short sequence homologies, and unusually long palindromic junctional inserts, known as P nucleotides, that result from aberrant hairpin opening. These features characterize junctions found in scid mice, which are deficient for the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), suggesting that the RAG proteins and DNA-PKcs perform overlapping functions in coding joint formation. Interestingly, the amino acids that are altered in 12 of our mutants are also mutated in human inherited immunodeficiency syndromes. Our analysis of these mutants provides insights into the molecular mechanisms underlying these disorders.


Assuntos
Aminoácidos/metabolismo , DNA Nucleotidiltransferases/genética , Proteínas de Homeodomínio/genética , Recombinação Genética/genética , Alanina/metabolismo , Sequência de Aminoácidos , Animais , DNA/metabolismo , DNA Nucleotidiltransferases/química , DNA Nucleotidiltransferases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Genes RAG-1 , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Conformação de Ácido Nucleico , Oligonucleotídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transposases/química , Transposases/genética , Transposases/metabolismo , VDJ Recombinases
8.
Immunotargets Ther ; 4: 55-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-27471712

RESUMO

Non-Hodgkin's lymphoma (NHL) represents a heterogeneous group of malignancies with high diversity in terms of biology, clinical responses, and prognosis. Standard therapy regimens produce a 5-year relative survival rate of only 69%, with the critical need to increase the treatment-success rate of this patient population presenting at diagnosis with a median age of 66 years and many comorbidities. The evidence that an impaired immune system favors the development of NHL has opened the stage for new therapeutics, and specifically for the adoptive transfer of ex vivo-expanded antigen-specific T-cells. In this review, we discuss how T-cells specific for viral-associated antigens, nonviral-associated antigens expressed by the tumor, T-cells redirected through the expression of chimeric antigen receptors, and transgenic T-cell receptors against tumor cells have been developed and used in clinical trials for the treatment of patients with NHLs.

9.
J Immunother Cancer ; 3: 5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25734008

RESUMO

BACKGROUND: Virus-specific T-cells (VSTs) proliferate exponentially after adoptive transfer into hematopoietic stem cell transplant (HSCT) recipients, eliminate virus infections, then persist and provide long-term protection from viral disease. If VSTs behaved similarly when modified with tumor-specific chimeric antigen receptors (CARs), they should have potent anti-tumor activity. This theory was evaluated by Cruz et al. in a previous clinical trial with CD19.CAR-modified VSTs, but there was little apparent expansion of these cells in patients. In that study, VSTs were gene-modified on day 19 of culture and we hypothesized that by this time, sufficient T-cell differentiation may have occurred to limit the subsequent proliferative capacity of the transduced T-cells. To facilitate the clinical testing of this hypothesis in a project supported by the NHLBI-PACT mechanism, we developed and optimized a good manufacturing practices (GMP) compliant method for the early transduction of VSTs directed to Epstein-Barr virus (EBV), Adenovirus (AdV) and cytomegalovirus (CMV) using a CAR directed to the tumor-associated antigen disialoganglioside (GD2). RESULTS: Ad-CMVpp65-transduced EBV-LCLs effectively stimulated VSTs directed to all three viruses (triVSTs). Transduction efficiency on day three was increased in the presence of cytokines and high-speed centrifugation of retroviral supernatant onto retronectin-coated plates, so that under optimal conditions up to 88% of tetramer-positive VSTs expressed the GD2.CAR. The average transduction efficiency of early-and late transduced VSTs was 55 ± 4% and 22 ± 5% respectively, and early-transduced VSTs maintained higher frequencies of T cells with central memory or intermediate memory phenotypes. Early-transduced VSTs also had higher proliferative capacity and produced higher levels of TH1 cytokines IL-2, TNF-α, IFN-γ, MIP-1α, MIP-1ß and other cytokines in vitro. CONCLUSIONS: We developed a rapid and GMP compliant method for the early transduction of multivirus-specific T-cells that allowed stable expression of high levels of a tumor directed CAR. Since a proportion of early-transduced CAR-VSTs had a central memory phenotype, they should expand and persist in vivo, simultaneously protecting against infection and targeting residual malignancy. This manufacturing strategy is currently under clinical investigation in patients receiving allogeneic HSCT for relapsed neuroblastoma and B-cell malignancies (NCT01460901 using a GD2.CAR and NCT00840853 using a CD19.CAR).

10.
J Vis Exp ; (69): e4235, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23149543

RESUMO

The piggyBac transposon system is naturally active, originally derived from the cabbage looper moth. This non-viral system is plasmid based, most commonly utilizing two plasmids with one expressing the piggyBac transposase enzyme and a transposon plasmid harboring the gene(s) of interest between inverted repeat elements which are required for gene transfer activity. PiggyBac mediates gene transfer through a "cut and paste" mechanism whereby the transposase integrates the transposon segment into the genome of the target cell(s) of interest. PiggyBac has demonstrated efficient gene delivery activity in a wide variety of insect, mammalian, and human cells6 including primary human T cells. Recently, a hyperactive piggyBac transposase was generated improving gene transfer efficiency. Human T lymphocytes are of clinical interest for adoptive immunotherapy of cancer. Of note, the first clinical trial involving transposon modification of human T cells using the Sleeping beauty transposon system has been approved. We have previously evaluated the utility of piggyBac as a non-viral methodology for genetic modification of human T cells. We found piggyBac to be efficient in genetic modification of human T cells with a reporter gene and a non-immunogenic inducible suicide gene. Analysis of genomic integration sites revealed a lack of preference for integration into or near known proto-oncogenes. We used piggyBac to gene-modify cytotoxic T lymphocytes to carry a chimeric antigen receptor directed against the tumor antigen HER2, and found that gene-modified T cells mediated targeted killing of HER2-positive tumor cells in vitro and in vivo in an orthotopic mouse model. We have also used piggyBac to generate human T cells resistant to rapamycin, which should be useful in cancer therapies where rapamycin is utilized. Herein, we describe a method for using piggyBac to genetically modify primary human T cells. This includes isolation of peripheral blood mononuclear cells (PBMCs) from human blood followed by culture, gene modification, and activation of T cells. For the purpose of this report, T cells were modified with a reporter gene (eGFP) for analysis and quantification of gene expression by flow cytometry. PiggyBac can be used to modify human T cells with a variety of genes of interest. Although we have used piggyBac to direct T cells to tumor antigens, we have also used piggyBac to add an inducible safety switch in order to eliminate gene modified cells if needed. The large cargo capacity of piggyBac has also enabled gene transfer of a large rapamycin resistant mTOR molecule (15 kb). Therefore, we present a non-viral methodology for stable gene-modification of primary human T cells for a wide variety of purposes.


Assuntos
Elementos de DNA Transponíveis , Técnicas de Transferência de Genes , Linfócitos T Citotóxicos/fisiologia , Anticorpos/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Humanos , Ativação Linfocitária , Plasmídeos/genética , Linfócitos T Citotóxicos/imunologia , Transposases/genética
11.
Hum Gene Ther ; 23(3): 311-20, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21992617

RESUMO

We characterized a recently developed hyperactive piggyBac (pB) transposase enzyme [containing seven mutations (7pB)] for gene transfer in human cells in vitro and to somatic cells in mice in vivo. Despite a protein level expression similar to that of native pB, 7pB significantly increased the gene transfer efficiency of a neomycin resistance cassette transposon in both HEK293 and HeLa cultured human cells. Native pB and SB100X, the most active transposase of the Sleeping Beauty transposon system, exhibited similar transposition efficiency in cultured human cell lines. When delivered to primary human T cells ex vivo, 7pB increased gene delivery two- to threefold compared with piggyBac and SB100X. The activity of hyperactive 7pB transposase was not affected by the addition of a 24-kDa N-terminal tag, whereas SB100X manifested a 50% reduction in transposition. Hyperactive 7pB was compared with native pB and SB100X in vivo in mice using hydrodynamic tail-vein injection of a limiting dose of transposase DNA combined with luciferase reporter transposons. We followed transgene expression for up to 6 months and observed approximately 10-fold greater long-term gene expression in mice injected with a codon-optimized version of 7pB compared with mice injected with native pB or SB100X. We conclude that hyperactive piggyBac elements can increase gene transfer in human cells and in vivo and should enable improved gene delivery using the piggyBac transposon system in a variety of cell and gene-therapy applications.


Assuntos
Transposases/genética , Animais , Elementos de DNA Transponíveis , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Células HEK293 , Células HeLa , Humanos , Camundongos , Transposases/metabolismo
12.
Discov Med ; 9(47): 297-303, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20423673

RESUMO

Adoptive transfer of T cells can enhance immune-mediated elimination of tumor cells and provides a specific, non-toxic cancer therapy. This approach has been effective in treating some hematologic and solid malignancies. In addition, the ability to genetically modify T cells to enhance their activity and persistence as well as overcome tumor immune evasion mechanisms has the potential to increase the success of these therapies in a wide range of tumors. In this review we discuss methods for gene transfer and specific modifications that have been made to T cells.


Assuntos
Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Técnicas de Transferência de Genes , Engenharia Genética , Humanos , Evasão da Resposta Imune/imunologia
13.
J Immunother ; 32(8): 826-36, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19752751

RESUMO

Optimal implementation of adoptive T-cell therapy for cancer will likely require multiple and maintained genetic modifications of the infused T cells and their progeny so that they home to tumor sites and recognize tumor cells, overcome tumor immune evasion strategies, and remain safe. Retroviral vectors readily transduce T cells and integrate into the host cell genome, but have a limited capacity for multigene insertion and cotransduction and are prohibitively expensive to produce at clinical grade. Genetic modification of T cells using transposons as integrating plasmids is an attractive alternative because of the increased simplicity and cost of production. Of available transposons, piggyBac has the higher transposase activity and larger cargo capacity, and we now evaluate piggyBac for potential adoptive therapies with primary T cells. PiggyBac transposons mediated stable gene expression in approximately 20% of primary T cells without selection. Treatment and maintenance of T cells with interleukin-15 increased stable transgene expression up to approximately 40% and expression was sustained through multiple logs of expansion for over 9 weeks in culture. We demonstrate simultaneous integration of 2 independent transposons in 20% of T cells, a frequency that could be increased to over 85% by selection of a transgenic surface marker (truncated CD19). PiggyBac could also deliver transposons of up to 13 kb with 10,000-fold expansion of transduced T cells in culture and finally we demonstrate delivery of a functional suicide gene (iCasp9). PiggyBac transposons may thus be used to express the multiple integrated transgenes that will likely be necessary for the broader success of T-cell therapy.


Assuntos
Caspase 9/metabolismo , Elementos de DNA Transponíveis/imunologia , Vetores Genéticos , Imunoterapia Adotiva , Linfócitos T/metabolismo , Apoptose , Vacinas Anticâncer , Caspase 9/genética , Caspase 9/imunologia , Células Cultivadas , Engenharia Genética , Humanos , Interleucina-15/metabolismo , Linfócitos T/citologia , Transgenes/genética
14.
J Biol Chem ; 280(13): 12262-70, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15664995

RESUMO

Genes containing an interferon (IFN)-stimulated response element (ISRE) can be divided into two groups according to their inducibility by IFN and virus infection: one induced only by IFN and the other induced by both IFN and virus infection. Although it is now clear that IFN regulatory factor 7 (IRF7) is a multifunctional gene essential for induction of type I IFNs, regulation of the IRF7 promoter (IRF7p) is poorly understood. The IRF7 gene includes two IFN responsive elements, an IRF-binding element (IRFE) in the promoter region and an ISRE in the first intron, and is induced by the IFN-triggered Jak-STAT pathway by binding of the IFN-stimulated gene factor 3 (ISGF3) complex to the ISRE. In this study, we demonstrate that IRF3 and IRF7, which with the coactivators CREB-binding protein and P300 form the virus-activated factor (VAF) complex upon Sendai virus infection, bind to the IRF7 ISRE and IRFE and can directly activate IRF7 transcription. Promoter reporter assays show that both the ISRE and IRFE are responsive to activation by IRF7 and IRF3. In cells transiently expressing IRF7 or/and IRF3, the VAF level and binding of VAF are clearly increased after Sendai virus infection. Studies with Jak1 kinase inactive 293 cells that were stably transfected with a Jak1 kinase dead dominant negative construct, and the mutant cell lines SAN (IFNalpha-/beta-), U2A (IRF9-), U4A (Jak1-), and DKO (IRF1-/IRF2-) show that the IRF7 transcription activated directly by VAF is distinct from and independent of the IFN signaling pathway. Thus, IRF7 transcription is autoregulated by binding of the IRF7-containing VAF to its own ISRE and IRFE. The results show two distinct mechanisms for the activation of the IRF7 promoter, by IFN and by virus infection. A regulatory network between type I IFNs and IRF7 is proposed. The distinct pathways may reflect special roles for an efficient antiviral response at different stages of virus infection.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Interferons/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Células HeLa , Humanos , Imunoprecipitação , Fator Regulador 3 de Interferon , Fator Regulador 7 de Interferon , Fator Gênico 3 Estimulado por Interferon , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Íntrons , Janus Quinase 1 , Modelos Biológicos , Modelos Genéticos , Mutação , Proteínas Nucleares/metabolismo , Plasmídeos/metabolismo , Proteínas Tirosina Quinases/metabolismo , RNA Mensageiro/metabolismo , Elementos de Resposta , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
15.
J Virol ; 79(18): 11671-6, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140744

RESUMO

We have reported evidence for a positive regulatory circuit between interferon regulatory factor 7 (IRF7) and the Epstein-Barr virus (EBV) oncoprotein 1 (LMP1) (S. Ning, A. M. Hahn, and J. S. Pagano, J. Virol. 77:9359-9368, 2003). To explore a possible braking mechanism for this circuit, several type II EBV-infected cell lines that express different levels of LMP1 and IRF7 proteins and therefore are convenient for studying modulation of expression of LMP1 were analyzed. Endogenous levels of IRF7 and LMP1 were directly correlated. Transient expression of an IRF7 dominant-negative mutant decreased LMP1 levels. Endogenous IRF5 and IRF7 proteins were shown to physically associate in EBV-positive cells. Transient expression of IRF5 decreased activation of the LMP1 promoter by IRF7 in a dose-dependent manner. Finally, transfection of either an IRF5 dominant-negative construct or IRF5 small interfering RNA in these cells resulted in increases in endogenous levels of LMP1. These results indicate that IRF5 can downregulate IRF7's induction of expression of LMP1 most likely by interacting with IRF7 and provide a means of modulating a regulatory circuit between IRF7 and LMP1.


Assuntos
Proteínas de Ligação a DNA/genética , Herpesvirus Humano 4/genética , Fatores de Transcrição/genética , Proteínas da Matriz Viral/genética , Antígenos Virais/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/imunologia , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/virologia , Expressão Gênica , Genes Reguladores , Genes Virais , Herpesvirus Humano 4/imunologia , Humanos , Fator Regulador 7 de Interferon , Fatores Reguladores de Interferon , Modelos Biológicos , Mutação , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Fatores de Transcrição/imunologia , Proteínas da Matriz Viral/imunologia
16.
J Virol ; 79(15): 10040-52, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16014964

RESUMO

Virus infection stimulates potent antiviral responses; specifically, Epstein-Barr virus (EBV) infection induces and activates interferon regulatory factor 7 (IRF-7), which is essential for production of alpha/beta interferons (IFN-alpha/beta) and upregulates expression of Tap-2. Here we present evidence that during cytolytic viral replication the immediate-early EBV protein BZLF-1 counteracts effects of IRF-7 that are central to host antiviral responses. We initiated these studies by examining IRF-7 protein expression in vivo in lesions of hairy leukoplakia (HLP) in which there is abundant EBV replication but the expected inflammatory infiltrate is absent. This absence might predict that factors involved in the antiviral response are absent or inactive. First, we detected significant levels of IRF-7 in the nucleus, as well as in the cytoplasm, of cells in HLP lesions. IRF-7 activity in cell lines during cytolytic viral replication was examined by assay of the IRF-7-responsive promoters, IFN-alpha4, IFN-beta, and Tap-2, as well as of an IFN-stimulated response element (ISRE)-containing reporter construct. These reporter constructs showed consistent reduction of activity during lytic replication. Both endogenous and transiently expressed IRF-7 and EBV BZLF-1 proteins physically associate in cell culture, although BZLF-1 had no effect on the nuclear localization of IRF-7. However, IRF-7-dependent activity of the IFN-alpha4, IFN-beta, and Tap-2 promoters, as well as an ISRE promoter construct, was inhibited by BZLF-1. This inhibition occurred in the absence of other EBV proteins and was independent of IFN signaling. Expression of BZLF-1 also inhibited activation of IRF-7 by double-stranded RNA, as well as the activity of a constitutively active mutant form of IRF-7. Negative regulation of IRF-7 by BZLF-1 required the activation domain but not the DNA-binding domain of BZLF-1. Thus, EBV may subvert cellular antiviral responses and immune detection by blocking the activation of IFN-alpha4, IFN-beta, and Tap-2 by IRF-7 through the medium of BZLF-1 as a negative regulator.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Herpesvirus Humano 4/fisiologia , Transativadores/genética , Transcrição Gênica , Proteínas Virais/genética , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Biópsia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulação para Baixo , Fator Regulador 7 de Interferon , Interferon-alfa/genética , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/metabolismo , Leucoplasia Pilosa/metabolismo , Leucoplasia Pilosa/patologia , Leucoplasia Pilosa/virologia , Regiões Promotoras Genéticas , Pele/metabolismo , Pele/patologia , Pele/virologia , Transativadores/metabolismo , Proteínas Virais/metabolismo , Replicação Viral
17.
J Virol ; 77(17): 9359-68, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12915551

RESUMO

We have shown previously that interferon regulatory factor 7 (IRF7), a multifunctional protein intimately involved in latent Epstein-Barr virus (EBV) infection, is induced as well as activated by EBV latent membrane protein 1 (LMP1), the principal EBV oncoprotein. Since the LMP1 promoter (LMP1p) contains an interferon-stimulated response element (ISRE), we hypothesized that IRF7 might be able to regulate LMP1 expression and thus participate in a regulatory circuit between these two genes. In this study, IRF7 was shown first to activate LMP1p in transient transfection assays. Compared with EBV nuclear antigen 2 (EBNA2), the most potent viral transactivator of LMP1p, IRF7 has a lesser effect (approximately 10% that of EBNA2) on induction of LMP1p. Study with IRF7 deletion mutants showed that IRF7 functional domains have similar effects on both the beta interferon (IFN-beta) and LMP1 promoters in BJAB and 293 cells, and study with IRF7 phosphomimetic mutants showed that IRF7 phosphorylation may be involved in the activation of these two promoters. Further, the ISRE in LMP1p responds to IRF7 induction and IRF7 binds to this element. In the EBV-positive cell line P3HR1, which lacks the complete EBNA2 and EBV-encoded leader protein genes and hence expresses low-level LMP1, IRF7 alone can notably increase the endogenous LMP1 mRNA and protein levels. These results indicate that LMP1 is regulated by this host cell gene in addition to the viral factor, EBNA2, and may help to explain how LMP1 is expressed in type II latency in the absence of EBNA2. Moreover, IRF7 can regulate a viral gene in addition to a host cellular gene such as the IFN-beta gene. Together with the previous data that LMP1 can induce IRF7 expression and facilitate IRF7 phosphorylation and nuclear translocation, these results suggest a positive regulatory circuit between IRF7 and LMP1.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/fisiologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , DNA Viral/genética , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Antígenos Nucleares do Vírus Epstein-Barr/genética , Antígenos Nucleares do Vírus Epstein-Barr/fisiologia , Regulação Viral da Expressão Gênica , Humanos , Fator Regulador 7 de Interferon , Modelos Biológicos , Mutagênese Sítio-Dirigida , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção , Regulação para Cima , Proteínas Virais
18.
Mol Cell ; 9(4): 871-8, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11983177

RESUMO

V(D)J recombination generates two types of products: coding joints, which constitute the rearranged variable regions of antigen receptor genes, and signal joints, which often form on immunologically irrelevant, excised circular molecules that are lost during cell division. It has been widely believed that signal joints simply convert reactive broken DNA ends into safe, inert products. Yet two curious in vivo observations made us question this assumption: signal ends are far more abundant than coding ends, and signal joints form only after RAG expression is downregulated. In fact, we find that signal joints are not at all inert; they are cleaved quite efficiently in vivo and in vitro by a nick-nick mechanism and form an excellent substrate for RAG-mediated transposition in vitro, possibly explaining how genomic stability in lymphocytes may be compromised.


Assuntos
DNA Nucleotidiltransferases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Recombinação Genética/genética , Substituição de Aminoácidos , Animais , Células CHO/enzimologia , Cricetinae , Cricetulus , DNA Circular/metabolismo , DNA Recombinante/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Endonucleases/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Camundongos , Modelos Genéticos , Plasmídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequências Reguladoras de Ácido Nucleico , Especificidade por Substrato , Transfecção , VDJ Recombinases
19.
Immunity ; 19(1): 21-31, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12871636

RESUMO

V(H) replacement has been proposed as one way to modify unwanted antibody specificities, but analysis of this mechanism has been limited without a dynamic cellular model. We describe a human cell line that spontaneously undergoes serial V(H) gene replacement mediated by cryptic recombination signal sequences (cRSS) located near the 3' end of V(H) genes. Recombination-activating gene products, RAG-1 and RAG-2, bind and cleave the cRSS to generate DNA deletion circles during the V(H) replacement process. A V(H) replacement contribution to normal repertoire development is revealed by the identification of V(H) replacement "footprints" in IgH sequences and double-stranded DNA breaks at V(H) cRSS sites in immature B cells. Surprisingly, the residual 3' sequences of replaced V(H) genes contribute charged amino acids to the CDR3 region, a hallmark of autoreactive antibodies.


Assuntos
Linfócitos B/imunologia , Genes de Imunoglobulinas , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Sequência de Bases , Células Cultivadas , Regiões Determinantes de Complementaridade , Dano ao DNA , Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio/fisiologia , Humanos , Dados de Sequência Molecular , Proteínas Nucleares , Reação em Cadeia da Polimerase , Recombinação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA