Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Intervalo de ano de publicação
2.
Gene Ther ; 22(9): 707-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25965393

RESUMO

Monocyte-derived conventional dendritic cells (ConvDCs) loaded with melanoma antigens showed modest responses in clinical trials. Efficacy studies were hampered by difficulties in ConvDC manufacturing and low potency. Overcoming these issues, we demonstrated higher potency of lentiviral vector (LV)-programmed DCs. Monocytes were directly induced to self-differentiate into DCs (SmartDC-TRP2) upon transduction with a tricistronic LV encoding for cytokines (granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4)) and a melanoma antigen (tyrosinase-related protein 2 (TRP2)). Here, SmartDC-TRP2 generated with monocytes from five advanced melanoma patients were tested in autologous DC:T cell stimulation assays, validating the activation of functional TRP2-specific cytotoxic T lymphocytes (CTLs) for all patients. We described methods compliant to good manufacturing practices (GMP) to produce LV and SmartDC-TRP2. Feasibility of monocyte transduction in a bag system and cryopreservation following a 24-h standard operating procedure were achieved. After thawing, 50% of the initial monocyte input was recovered and SmartDC-TRP2 self-differentiated in vitro, showing uniform expression of DC markers, detectable LV copies and a polyclonal LV integration pattern not biased to oncogenic loci. GMP-grade SmartDC-TRP2 expanded TRP2-specific autologous CTLs in vitro. These results demonstrated a simpler GMP-compliant method of manufacturing an effective individualized DC vaccine. Such DC vaccine, when in combination with checkpoint inhibition therapies, might provide higher specificity against melanoma.


Assuntos
Vacinas Anticâncer/uso terapêutico , Células Dendríticas/imunologia , Lentivirus/metabolismo , Melanoma/terapia , Proteínas de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Vetores Genéticos , Células HEK293 , Humanos , Imunoterapia/métodos , Lentivirus/genética , Melanoma/imunologia , Linfócitos T Citotóxicos/imunologia
3.
Cancer Gene Ther ; 20(3): 141-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23370333

RESUMO

We provide an overview of the latest developments in cancer gene therapy--from the bench to early-stage clinical trials. We describe the most recent work of worldwide teams including experienced scientists and clinicians, reflecting the recent emergence of gene therapy from the 'Valley of Death'. The treatment efficacy of clinical gene therapy has now been shown in a number of diseases including cancer and we are observing a renewed interest by big pharmaceutical and biotechnology companies most obviously demonstrated by Amgen's acquisition of Biovex for up to USD$1 billion. There is an opportunity to be cautiously hopeful regarding the future of gene therapy in the clinic and we review here some of the most recent progress in the field.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Animais , Biomarcadores Tumorais/genética , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Imunoterapia , Terapia de Alvo Molecular , Medicina de Precisão , Interferência de RNA
4.
Oncogene ; 32(25): 3028-38, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22847614

RESUMO

Activation of NRas signaling is frequently found in human myeloid leukemia and can be induced by activating mutations as well as by mutations in receptors or signaling molecules upstream of NRas. To study NRas-induced leukemogenesis, we retrovirally overexpressed wild-type NRas in a murine bone marrow transplantation (BMT) model in C57BL/6J mice. Overexpression of wild-type NRas caused myelomonocytic leukemias ∼3 months after BMT in the majority of mice. A subset of mice (30%) developed malignant histiocytosis similar to mice that received mutationally activated NRas(G12D)-expressing bone marrow. Aberrant Ras signaling was demonstrated in cells expressing mutationally active or wild-type NRas, as increased activation of Erk and Akt was observed in both models. However, more NRas(G12D) were found to be in the activated, GTP-bound state in comparison with wild-type NRas. Consistent with observations reported for primary human myelomonocytic leukemia cells, Stat5 activation was also detected in murine leukemic cells. Furthermore, clonal evolution was detected in NRas wild-type-induced leukemias, including expansion of clones containing activating vector insertions in known oncogenes, such as Evi1 and Prdm16. In vitro cooperation of NRas and Evi1 improved long-term expansion of primary murine bone marrow cells. Evi1-positive cells upregulated Bcl-2 and may, therefore, provide anti-apoptotic signals that collaborate with the NRas-induced proliferative effects. As activation of Evi1 has been shown to coincide with NRAS mutations in human acute myeloid leukemia, our murine model recapitulates crucial events in human leukemogenesis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Leucemia Mielomonocítica Aguda/metabolismo , Fatores de Transcrição/metabolismo , Proteínas ras/metabolismo , Animais , Apoptose , Transplante de Medula Óssea , Linhagem Celular Tumoral , Proliferação de Células , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Genes ras , Humanos , Leucemia Mielomonocítica Aguda/genética , Proteína do Locus do Complexo MDS1 e EVI1 , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proto-Oncogenes , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Proteínas ras/genética
5.
Gene Ther ; 18(8): 750-64, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21412283

RESUMO

Development of lentiviral vectors (LVs) in the field of immunotherapy and immune regeneration will strongly rely on biosafety of the gene transfer. We demonstrated previously the feasibility of ex vivo genetic programming of mouse bone marrow precursors with LVs encoding granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4), which induced autonomous differentiation of long-lived dendritic cells (DCs), referred to as self-differentiated myeloid-derived antigen-presenting-cells reactive against tumors (SMART-DCs). Here, LV biosafety was enhanced by using a DC-restricted and physiological promoter, the major histocompatibility complex (MHC) II promoter, and including co-expression of the herpes simplex virus-thymidine kinase (sr39HSV-TK) conditional suicide gene. Tricistronic vectors co-expressing sr39HSV-TK, GM-CSF and IL-4 transcriptionally regulated by the MHCII promoter or the ubiquitous cytomegalovirus (CMV) promoter were compared. Despite the different gene transfer effects, such as the kinetics, levels of transgene expression and persistency of integrated vector copies, both vectors induced highly viable SMART-DCs, which persisted for at least 70 days in vivo and could be ablated with the pro-drug Ganciclovir (GCV). SMART-DCs co-expressing the tyrosine-related protein 2 melanoma antigen administered subcutaneously generated antigen-specific, anti-melanoma protective and therapeutic responses in the mouse B16 melanoma model. GCV administration after immunotherapy did not abrogate DC vaccination efficacy. This demonstrates proof-of-principle of genetically programmed DCs that can be ablated pharmacologically.


Assuntos
Diferenciação Celular/genética , Células Dendríticas/imunologia , Vetores Genéticos , Lentivirus/genética , Melanoma Experimental/terapia , Animais , Movimento Celular , Sobrevivência Celular , Ganciclovir/farmacologia , Genes MHC da Classe II , Genes Transgênicos Suicidas , Interleucina-4 , Camundongos , Camundongos Endogâmicos C57BL , Simplexvirus/genética , Timidina Quinase/genética , Vacinação
6.
Transplant Proc ; 38(10): 3184-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17175217

RESUMO

Transplantation of many tissues requires histocompatibility matching of human leukocyte antigens (HLA) to prevent graft rejection, to reduce the level of immunosuppression needed to maintain graft survival, and to minimize the risk of graft-versus-host disease, particularly in the case of bone marrow transplantation. However, recent advances in fields of gene delivery and genetic regulation technologies have opened the possibility of engineering grafts that display reduced levels of HLA expression. Suppression of HLA expression could help to overcome the limitations imposed by extensive HLA polymorphisms that restrict the availability of suitable donors, necessitate the maintenance of large donor registries, and complicate the logistics of procuring and delivering matched tissues and organs to the recipient. Accordingly, we investigated whether knockdown of HLA by RNA interference (RNAi), a ubiquitous regulatory system that can efficiently and selectively inhibit the expression of specific gene products, would enable allogeneic cells to evade immune recognition. For efficient and stable delivery of short hairpin-type RNAi constructs (shRNA), we employed lentivirus-based gene transfer vectors, which provide a delivery system that can achieve integration into genomic DNA, thereby permanently modifying transduced graft cells. Our results show that lentivirus-mediated delivery of shRNA targeting pan-Class I and allele-specific HLA can achieve efficient and dose-dependent reduction in surface expression of HLA in human cells, associated with enhanced resistance to alloreactive T lymphocyte-mediated cytotoxicity, while avoiding MHC-non-restricted killing. We hypothesize that RNAi-induced silencing of HLA expression has the potential to create histocompatibility-enhanced, and, eventually, perhaps "universally" compatible cellular grafts.


Assuntos
Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Linfócitos T/imunologia , Sequência de Bases , Linhagem Celular , Citotoxicidade Imunológica , Primers do DNA , Inativação Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , HIV/imunologia , Humanos , Interferon gama/imunologia , Rim , Lentivirus , Interferência de RNA
7.
Leukemia ; 16(10): 1974-83, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12357348

RESUMO

Adult patients with acute leukemia have, in general, a poor prognosis, with long-term, disease-free survival achieved in only approximately one-third of cases. One of the proposed mechanisms for this poor overall response is the inability of the immune system to detect and eliminate residual malignant leukemia cells, which subsequently serve as a source of leukemic relapse. This review discusses the rationale of immunotherapy for acute leukemia and presents in vitro and in vivo model systems that were devised for pre-B acute lymphocytic leukemia (ALL) and acute myeloid leukemia (AML). New advances in the ex vivo manipulation of acute leukemia cells are presented, which attempt to modify these cells into functional antigen-presenting cells. These cells can then be used as autologous vaccines at the time of minimal residual disease after standard chemotherapy, to stimulate host immune responses against their own leukemia cells. The various approaches toward this aim include incubation of leukemia cells with cytokines or growth factors and gene manipulation of these cells. In particular, ex vivo culture of ALL cells with CD40 ligand, incubation of AML cells with granulocyte-macrophage colony-stimulating factor and interleukin-4 (GM-CSF/IL-4) and lentiviral transduction of ALL and AML cells for expression of immunomodulators (CD80 and GM-CSF) are current approaches under investigation for the development of autologous acute leukemia cell vaccines.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Técnicas de Transferência de Genes , Imunoterapia , Leucemia Mieloide/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Doença Aguda , Adulto , Vacinas Anticâncer/uso terapêutico , Humanos
8.
Leukemia ; 16(9): 1645-54, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12200676

RESUMO

Acute myeloid leukemia (AML) patients treated with available therapies achieve remission in approximately 60% of cases, but the long-term event-free survival is less than 30%. Use of immunotherapy during remission is a potential approach to increase survival. We propose to develop cell vaccines by genetic modification of AML cells with CD80, an essential T cell costimulator that is lacking in the majority of AML cases, and GM-CSF, to induce proliferation and activation of professional antigen-presenting cells. Here, we evaluated third generation self inactivating (SIN) lentiviral vectors, which have the potential advantage of improved safety. CD80 and GM-CSF expression by these vectors was higher than that reported with second generation vectors (Stripecke et al, Blood 2000; 96: 1317-1326). In some cases, endogenous GM-CSF expression by transduced AML cells induced phenotypic changes consistent with the maturation of leukemia blasts into antigen-presenting cells. Further, in all cases studied, GM-CSF expression was associated with higher proliferation and cell viability. Allogeneic and autologous mixed lymphocyte reactions performed with transduced irradiated AML cells expressing CD80 and/or GM-CSF demonstrated that expression of either transgene enhanced T cell activation. These pre-clinical data demonstrate the potential feasibility of third generation SIN vectors for use in AML immunotherapy.


Assuntos
Antígeno B7-1/genética , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Lentivirus/genética , Leucemia Mieloide/genética , Leucemia Mieloide/imunologia , Linfócitos T/imunologia , Doença Aguda , Adulto , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Diferenciação Celular , Divisão Celular , Citotoxicidade Imunológica/genética , Citometria de Fluxo , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Leucemia Mieloide/patologia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Linfócitos T/metabolismo , Transdução Genética , Células Tumorais Cultivadas
9.
Hum Gene Ther ; 12(4): 323-32, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11242525

RESUMO

The entry of retroviral vectors into cells requires two events: binding to a cell surface receptor and the subsequent fusion of viral and cellular membranes. The host range of a vector is therefore determined largely by the receptor specificity of the fusion protein contained in the outer viral envelope. Previous attempts to generate targeted retroviral vectors have included the addition of targeting ligands to the murine leukemia virus envelope protein (MuLV Env). Although such proteins frequently display modified cell-binding characteristics, the interaction with the targeted receptors fails to trigger virus-cell fusion. Here, we report the use of a binding-defective but fusion-competent hemagglutinin (HA) protein to complement the fusion defect in a chimeric MuLV Env targeted to the Flt-3 receptor. Retroviral vectors containing both proteins showed enhanced transduction of cells expressing Flt-3, which was abrogated by preincubating the target cells with soluble Flt-3 ligand. Furthermore, the fusion function of HA was absolutely required. These data demonstrate that it is possible to separate the binding and fusion events of retroviral entry, using two separate proteins, and suggest that varying the binding protein component in this scheme may allow a general strategy for targeting retroviral vectors.


Assuntos
Produtos do Gene env/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Leucemia Murina/genética , Fusão de Membrana , Receptores Virais/fisiologia , Retroviridae/genética , Proteínas do Envelope Viral/química , Células 3T3 , Animais , Sítios de Ligação/genética , Linhagem Celular , Marcação de Genes , Vetores Genéticos , Humanos , Camundongos , Mutação , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas Recombinantes de Fusão/genética , Tirosina Quinase 3 Semelhante a fms
10.
Blood ; 96(4): 1317-26, 2000 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10942373

RESUMO

Cell vaccines engineered to express immunomodulators have shown feasibility in eliminating leukemia in murine models. Vectors for efficient gene delivery to primary human leukemia cells are required to translate this approach to clinical trials. In this study, second-generation lentiviral vectors derived from human immunodeficiency virus 1 were evaluated, with the cytomegalovirus (CMV) promoter driving expression of granulocyte-macrophage-colony-stimulating factor (GM-CSF) and CD80 in separate vectors or in a bicistronic vector. The vectors were pseudotyped with vesicular stomatitis virus G glycoprotein and concentrated to high titers (10(8)-10(9) infective particles/mL). Human acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia cell lines transduced with the monocistronic pHR-CD80 vector or the bicistronic pHR-GM/CD vector became 75% to 95% CD80 positive (CD80(+)). More important, transduction of primary human ALL and AML blasts with high-titer lentiviral vectors was consistently successful (40%-95% CD80(+)). The average amount of GM-CSF secretion by the leukemia cell lines transduced with the pHR-GM-CSF monocistronic vector was 2182.9 pg/10(6) cells per 24 hours. Secretion was markedly lower with the bicistronic pHR-GM/CD vector (average, 225.7 pg/10(6) cells per 24 hours). Lower amounts of CMV-driven messenger RNA were detected with the bicistronic vector, which may account for its poor expression of GM-CSF. Primary ALL cells transduced to express CD80 stimulated T-cell proliferation in an autologous mixed lymphocyte reaction. This stimulation was specifically blocked with monoclonal antibodies reactive against CD80 or by recombinant cytotoxic T-lymphocyte antigen 4-immunoglobulin fusion protein. These results show the feasibility of efficiently transducing primary leukemia cells with lentiviral vectors to express immunomodulators to elicit antileukemic immune responses. (Blood. 2000;96:1317-1326)


Assuntos
Antígeno B7-1/genética , Terapia Genética , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Lentivirus , Leucemia Mieloide/genética , Leucemia Mieloide/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Doença Aguda , Antígeno B7-1/imunologia , Antígeno B7-1/uso terapêutico , Citotoxicidade Imunológica/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Transferência de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Humanos , Imunoterapia , Leucemia Mieloide/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Tumorais Cultivadas
11.
Hum Gene Ther ; 10(13): 2109-22, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10498243

RESUMO

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is a highly aggressive malignancy caused by the bcr-abl translocation oncogene. To explore alternative treatments for Ph+ ALL we tested gene-modified cell vaccines in the BALB/c-derived BM185 leukemia model. We compared the efficacy of BM185 cell vaccine expressing CD80 alone or in combination with IL-2 or GM-CSF. Mice injected with viable BM185 leukemia cells modified to express CD80 and GM-CSF (BM185/CD80+GM-CSF) showed the highest leukemia rejection rates. Cell vaccines consisting of irradiated BM185/CD80+GM-CSF cells administered subcutaneously stimulated a potent cytotoxic T lymphocyte (CTL) response against parental BM185. Histological examination of the vaccination site showed a large concentration of immune cells. Administration of the BM185/CD80+GM-CSF cell vaccine before intravenous challenge with parental cells caused strong inhibition of leukemia development. Vaccination after subcutaneous challenge with BM185 cells caused efficient elimination of leukemia promoting 40-60% long-term survival rates. The immunization efficacy of the BM185/CD80+ GM-CSF cell vaccine was directly correlated with the percentage of cells expressing the transgenes. In all, this preclinical study shows that leukemia cell vaccines coexpressing CD80 and GM-CSF can potentially be explored for immunotherapy in Ph+ ALL patients.


Assuntos
Antígeno B7-1/metabolismo , Vacinas Anticâncer/uso terapêutico , Terapia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Animais , Células Apresentadoras de Antígenos/imunologia , Vacinas Anticâncer/metabolismo , Vacinas Anticâncer/efeitos da radiação , Linhagem Celular , Citotoxicidade Imunológica , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica , Imunoterapia , Interleucina-2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Transplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Linfócitos T Citotóxicos/imunologia
12.
Gene Ther ; 6(7): 1305-12, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10455440

RESUMO

Green fluorescent protein (GFP) is a widely used intracellular reporter molecule to assess gene transfer and expression. A potential use for GFP is as a co-expressed marker, to select and enrich gene-modified cells by flow cytometry. Processed peptides derived from GFP and presented by the major histocompatibility complex on the cell surface could potentially induce T cell immune responses against GFP+ cells. Thus, clinical application of GFP is premature, since in vivo studies on its immunogenicity are lacking. Therefore, we investigated immune responses against EGFP (enhanced-GFP) in two transplantable murine models: the BALB/c (H-2d) BM185 pre-B leukemia and the C57BL/6 (H-2b) EL-4 T cell lymphoma. BM185 and EL-4 cell lines modified to express high levels of EGFP showed drastic reduction of disease development when transplanted into immunocompetent mice. BM185/ EGFP did lead to rapid development of disease in immunodeficient Nu/Nu mice. Mice surviving BM185/EGFP leukemia challenge developed high cytotoxic T lymphocyte (CTL) responses against EGFP-expressing cells. Furthermore, immune stimulation against BM185/EGFP cells could also be induced by immunization with EGFP+ transduced dendritic cells. The effects of the co-expression of EGFP and immunomodulators (CD80 plus GM-CSF) were also investigated as an irradiated leukemia vaccine. EGFP co-expression by the vaccine did not interfere with the development of CTLs against the parental leukemia or with the anti-leukemia response in vivo. These results indicate that the immune response against EGFP may interfere with its applicability in gene insertion/replacement strategies but could potentially be employed for leukemia cell vaccines.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Leucemia Experimental/imunologia , Proteínas Luminescentes/genética , Linfócitos T Citotóxicos/imunologia , Animais , Antígeno B7-1/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Proteínas de Fluorescência Verde , Leucemia Experimental/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinas de DNA
13.
Proc Natl Acad Sci U S A ; 96(6): 2988-93, 1999 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-10077624

RESUMO

We compared the efficiency of transduction by an HIV-1-based lentiviral vector to that by a Moloney murine leukemia virus (MLV) retroviral vector, using stringent in vitro assays of primitive, quiescent human hematopoietic progenitor cells. Each construct contained the enhanced green fluorescent protein (GFP) as a reporter gene. The lentiviral vector, but not the MLV vector, expressed GFP in nondivided CD34(+) cells (45.5% GFP+) and in CD34(+)CD38(-) cells in G0 (12.4% GFP+), 48 hr after transduction. However, GFP could also be detected short-term in CD34(+) cells transduced with a lentiviral vector that contained a mutated integrase gene. The level of stable transduction from integrated vector was determined after extended long-term bone marrow culture. Both MLV vectors and lentiviral vectors efficiently transduced cytokine-stimulated CD34(+) cells. The MLV vector did not transduce more primitive, quiescent CD34(+)CD38(-) cells (n = 8). In contrast, stable transduction of CD34(+)CD38(-) cells by the lentiviral vector was seen for over 15 weeks of extended long-term culture (9.2 +/- 5.2%, n = 7). GFP expression in clones from single CD34(+)CD38(-) cells confirmed efficient, stable lentiviral transduction in 29% of early and late-proliferating cells. In the absence of growth factors during transduction, only the lentiviral vector was able to transduce CD34(+) and CD34(+)CD38(-) cells (13.5 +/- 2.5%, n = 11 and 12.2 +/- 9.7%, n = 4, respectively). The lentiviral vector is clearly superior to the MLV vector for transduction of quiescent, primitive human hematopoietic progenitor cells and may provide therapeutically useful levels of gene transfer into human hematopoietic stem cells.


Assuntos
Antígenos CD , Terapia Genética , Vetores Genéticos , HIV-1 , Células-Tronco Hematopoéticas/fisiologia , Transdução Genética , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Antígenos CD34 , Antígenos de Diferenciação , Humanos , Lentivirus , Vírus da Leucemia Murina , Glicoproteínas de Membrana , NAD+ Nucleosidase
14.
Blood ; 92(10): 3537-45, 1998 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9808545

RESUMO

Autologous leukemia cells engineered to express immune-stimulating molecules may be used to elicit antileukemia immune responses. Gene delivery to human B-precursor acute lymphoblastic leukemia (ALL) cells was investigated using the enhanced green fluorescent protein (EGFP) as a reporter gene, measured by flow cytometry. Transfection of the Nalm-6 and Reh B-precursor ALL leukemia cell lines with an expression plasmid was investigated using lipofection, electroporation, and a polycationic compound. Only the liposomal compound Cellfectin showed significant gene transfer (3.9% to 12% for Nalm-6 cells and 3.1% to 5% for Reh cells). Transduction with gibbon-ape leukemia virus pseudotyped Moloney murine leukemia virus (MoMuLV)-based retrovirus vectors was investigated in various settings. Cocultivation of ALL cell lines with packaging cell lines showed the highest transduction efficiency for retroviral gene transfer (40.1% to 87.5% for Nalm-6 cells and 0.3% to 9% for Reh cells), followed by transduction with viral supernatant on the recombinant fibronectin fragment CH-296 (13% to 35.5% for Nalm-6 cells and 0.4% to 6% Reh cells), transduction on human bone marrow stroma monolayers (3.2% to 13.3% for Nalm-6 cells and 0% to 0.2% Reh cells), and in suspension with protamine sulfate (0.7% to 3.1% for Nalm-6 cells and 0% for Reh cells). Transduction of both Nalm-6 and Reh cells with human immunodeficiency virus-type 1 (HIV-1)-based lentiviral vectors pseudotyped with the vesicular stomatitis virus-G envelope produced the best gene transfer efficiency, transducing greater than 90% of both cell lines. Gene delivery into primary human B-precursor ALL cells from patients was then investigated using MoMuLV-based retrovirus vectors and HIV-1-based lentivirus vectors. Both vectors transduced the primary B-precursor ALL cells with high efficiencies. These studies may be applied for investigating gene delivery into primary human B-precursor ALL cells to be used for immunotherapy.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Resinas de Troca de Cátion/farmacologia , Separação Celular , Técnicas de Cocultura , DNA Recombinante/genética , Portadores de Fármacos/farmacologia , Eletroporação , Citometria de Fluxo , Genes Reporter , Terapia Genética , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , HIV-1/genética , Humanos , Vírus da Leucemia do Macaco Gibão/genética , Lipídeos/farmacologia , Lipossomos , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Vírus da Leucemia Murina de Moloney/genética , Fosfatidiletanolaminas/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Protaminas , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transfecção , Células Tumorais Cultivadas , Vírus da Estomatite Vesicular Indiana/genética
15.
Hum Gene Ther ; 9(14): 2049-62, 1998 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-9759932

RESUMO

We examined the potential of generating an immune response against Philadelphia chromosome-positive acute lymphoblastic leukemia. The immunostimulatory molecules chosen for this study were the cytokines IL-2 and GM-CSF and the costimulatory ligand CD80 (B7.1). We used a murine model based on a BALB/c pre-B cell line, BM185wt, in which leukemia is induced by the p185 BCR-ABL oncogenic product, which reproduces Philadelphia chromosome-positive ALL. BM185wt cells were transduced with retroviral vectors and the transduced clones expressing mIL-2, mGM-CSF, or mCD80 were used for challenge. Expression of the immunomodulators by BM185 cells was correlated with delay in leukemia development in immunocompetent mice, but not in immunodeficient mice, indicating an immune response against the modified leukemia cells. Expression of CD80 caused leukemia rejection in 50% of the cohort, which was associated with the CD4+ and CD8+ T cell-dependent development of anti-leukemia cytotoxic T lymphocytes. Furthermore, mice surviving the BM185/CD80 challenge or preimmunized with irradiated BM185/CD80 cells developed an immune response against subsequent challenge with the parental leukemia. These studies provide evidence that immunotherapeutic approaches can be developed for the treatment of ALL.


Assuntos
Antígeno B7-1/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interleucina-2/genética , Cromossomo Filadélfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Vacinas/imunologia , Adjuvantes Imunológicos/genética , Animais , Antígenos CD4/imunologia , Antígenos CD8/imunologia , Divisão Celular/imunologia , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/mortalidade , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Transdução Genética/genética , Células Tumorais Cultivadas
16.
RNA ; 3(9): 983-95, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9292498

RESUMO

The poly(A) tail plays an important role in translation initiation. We report the identification of a mechanism that operates in mammalian somatic cells, and couples mRNA poly(A) tail length with its translation state. The regulation of human ferritin L-chain mRNA by iron-responsive elements (IREs) and iron regulatory proteins (IRPs) is subject to this mechanism: translational repression imposed by IRP binding to the IRE of ferritin L-chain mRNA induces poly(A) tail shortening. For the accumulation of mRNAs with short poly(A) tails, IRP binding to an IRE per se is not sufficient, but must cause translational repression. Interestingly, puromycin and verrucarin (general translation inhibitors that dissociate mRNAs from ribosomes) mimick the negative effect of the specific translational repressor proteins on poly(A) tail length, whereas cycloheximide and anisomycin (general translation inhibitors that maintain the association between mRNAs and ribosomes) preserve long poly(A) tails. Thus, the ribosome association of the mRNA appears to represent the critical determinant. These findings identify a novel mechanism of regulated polyadenylation as a consequence of translational control. They reveal differences in poly(A) tail metabolism between polysomal and mRNP-associated mRNAs. A possible role of this mechanism in the maintenance of translational repression is discussed.


Assuntos
Poli A/metabolismo , Biossíntese de Proteínas , Proteínas Repressoras/metabolismo , Ribossomos/genética , Animais , Ferritinas/genética , Ferritinas/metabolismo , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Ferro/metabolismo , Cinética , Mamíferos , Poli A/genética , Capuzes de RNA , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Ribossomos/metabolismo
17.
Mol Cell Biol ; 17(7): 3915-23, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199326

RESUMO

To identify specific proteins interacting with guide RNAs (gRNAs) in mitochondrial ribonucleoprotein complexes from Leishmania tarentolae, fractionated and unfractionated mitochondrial extracts were subjected to UV cross-linking with added labeled gRNA and also with [alpha-32P]UTP-labeled endogenous RNA. An abundant 110-kDa protein (p110) localized in the T-V complex, which sediments in glycerol gradients at the leading edge of the 10S terminal uridylyltransferase peak, was found to interact with both types of labeled RNAs. The p110 protein was gel isolated and subjected to microsequence analysis, and the gene was cloned. The sequence revealed significant similarity with mitochondrial glutamate dehydrogenases. A polyclonal antiserum was raised against a recombinant fragment of the p110 gene and was used to demonstrate a stable and specific gRNA-binding activity by coimmunoprecipitation and competitive gel shift analyses. Complex formation was strongly inhibited by competition with poly(U) or by deletion or substitution of the gRNA 3' oligo(U) tail. Also, addition of a 3' oligo(U) tail to an unrelated transcript was sufficient for p110 binding. Both the gRNA-binding activity of the p110 protein and in vitro gRNA-independent and gRNA-dependent uridine insertion activities in the mitochondrial extract were inhibited by high concentrations of dinucleotides.


Assuntos
Glutamato Desidrogenase/genética , Leishmania/enzimologia , Mitocôndrias/fisiologia , RNA Guia de Cinetoplastídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Animais , Compartimento Celular , Clonagem Molecular , Genes de Protozoários , Leishmania/genética , Dados de Sequência Molecular , Peso Molecular , NADP/metabolismo , Poli U/metabolismo , Ribonucleoproteínas/metabolismo , Alinhamento de Sequência
18.
Mol Cell Biol ; 14(9): 5898-909, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8065323

RESUMO

We demonstrate that a bacteriophage protein and a spliceosomal protein can be converted into eukaryotic translational repressor proteins. mRNAs with binding sites for the bacteriophage MS2 coat protein or the spliceosomal human U1A protein were expressed in human HeLa cells and yeast. The presence of the appropriate binding protein resulted in specific, dose-dependent translational repression when the binding sites were located in the 5' untranslated region (UTR) of the reporter mRNAs. Neither mRNA export from the nucleus to the cytoplasm nor mRNA stability was demonstrably affected by the binding proteins. The data thus reveal a general mechanism for translational regulation: formation of mRNA-protein complexes in the 5' UTR controls translation initiation by steric blockage of a sensitive step in the initiation pathway. Moreover, the findings establish the basis for novel strategies to study RNA-protein interactions in vivo and to clone RNA-binding proteins.


Assuntos
Regulação Fúngica da Expressão Gênica , Regulação da Expressão Gênica , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Sequência de Bases , Transporte Biológico , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos/química , Proteínas Repressoras/genética , Saccharomyces cerevisiae
19.
Nucleic Acids Res ; 21(23): 5316-22, 1993 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-8265343

RESUMO

The regulation of the synthesis of ferritin and erythroid 5-aminolevulinate synthase in mammalian cells is mediated by the interaction of the iron regulatory factor (IRF) with a specific recognition site, the iron responsive element (IRE), in the 5' untranslated regions (UTRs) of the respective mRNAs. A new modular expression system was designed to allow reconstruction of this regulatory system in Saccharomyces cerevisiae. This comprised two components: a constitutively expressed reporter gene (luc; encoding luciferase) preceded by a 5' UTR including an IRE sequence, and an inducibly expressed cDNA encoding human IRF. Induction of the latter led to the in vivo synthesis of IRF, which in turn showed IRE-binding activity and also repressed translation of the luc mRNA bearing an IRE-containing 5' UTR. The upper stem-loop region of an IRE, with no further IRE-specific flanking sequences, sufficed for recognition and repression by IRF. Translational regulation of IRE-bearing mRNAs could also be demonstrated in cell-free yeast extracts. This work defines a minimal system for IRF/IRE translational regulation in yeast that requires no additional mammalian-specific components, thus providing direct proof that IRF functions as a translational repressor in vivo. It should be a useful tool as the basis for more detailed studies of eukaryotic translational regulation.


Assuntos
Regulação da Expressão Gênica , Biossíntese de Proteínas , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética , Sequência de Bases , Regulação Fúngica da Expressão Gênica , Humanos , Proteínas Reguladoras de Ferro , Dados de Sequência Molecular , RNA Mensageiro/genética , Proteínas Recombinantes , Saccharomyces cerevisiae/genética , Transcrição Gênica
20.
Cell ; 72(6): 881-92, 1993 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-8458082

RESUMO

Human, mouse, and Xenopus mRNAs encoding the U1 snRNP-specific U1A protein contain a conserved 47 nt region in their 3' untranslated regions (UTRs). In vitro studies show that human U1A protein binds to two sites within the conserved region that resemble, in part, the previously characterized U1A-binding site on U1 snRNA. Overexpression of human U1A protein in mouse cells results in down-regulation of endogenous mouse U1A mRNA accumulation. In vitro and in vivo experiments demonstrate that excess U1A protein specifically inhibits polyadenylation of pre-mRNAs that contain the conserved 3' UTR from human U1A mRNA. Thus, U1A protein regulates the production of its own mRNA via a mechanism that involves pre-mRNA binding and inhibition of polyadenylation.


Assuntos
Poli A/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA , Ribonucleoproteína Nuclear Pequena U1/genética , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Animais , Sequência de Bases , Regulação da Expressão Gênica , Humanos , Ligação de Hidrogênio , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligodesoxirribonucleotídeos/química , Ligação Proteica , Biossíntese de Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...