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










Base de dados
Intervalo de ano de publicação
1.
J Neurosci Methods ; 207(2): 181-8, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22542732

RESUMO

Selective neuronal vulnerability is a common theme in both acute and chronic diseases affecting the nervous system. This phenomenon is particularly conspicuous after global cerebral ischemia wherein CA1 pyramidal neurons undergo delayed death while surrounding hippocampal regions are relatively spared. While injury in this model can be easily demonstrated using either histological or immunological stains, current methods used to assess the cellular injury present in these biological images lack the precision required to adequately compare treatment effects. To address this shortcoming, we devised a supervised work-flow that can be used to quantify ischemia-induced nuclear condensation using microscopic images. And while we demonstrate the utility of this technique using models of ischemic brain injury, the approach can be readily applied to other paradigms in which programmed cell death is a major component.


Assuntos
Isquemia Encefálica/patologia , Região CA1 Hipocampal/patologia , Núcleo Celular/patologia , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos
2.
Cell Death Differ ; 19(10): 1634-43, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22522596

RESUMO

The dual specificity phosphatase MAPK phosphatase-1 (MKP-1) feeds back on MAP kinase signaling to regulate metabolic, inflammatory and survival responses. MKP-1 is widely expressed in the central nervous system (CNS) and induced after ischemic stress, although its function in these contexts remains unclear. Here we report that MKP-1 activated several cell death factors, including BCL2 and adenovirus E1B 19 kDa interacting protein 3, and caspases 3 and 12 culminating in apoptotic cell death in vitro. MKP-1 also exerted inhibitory effects on the bZIP transcription factor CCAAT/enhancer-binding protein (C/EBPß), previously shown to have neuroprotective properties. These effects included reduced expression of the full-length C/EBPß variant and hypo-phosphorylation at the MEK-ERK1/2-sensitive Thr(188) site. Notably, enforced expression C/EBPß rescued cells from MKP-1-induced toxicity. Studies performed in knock-out mice indicate that the MKP-1 activity is required to exclude C/EBPß from the nucleus basally, and that MKP-1 antagonizes C/EBPß expression after global forebrain ischemia, particularly within the vulnerable CA1 sector of the hippocampus. Overall, MKP-1 appears to lower the cellular apoptotic threshold by inhibiting C/EBPß and enhancing both BH3 protein expression and cellular caspase activity. Thus, although manipulation of the MKP-1-C/EBPß axis could have therapeutic value in ischemic disorders, our observations using MKP-1 catalytic mutants suggest that approaches geared towards inhibiting MKP-1's phosphatase activity alone may be ineffective.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Fosfatase 1 de Especificidade Dupla/metabolismo , Animais , Apoptose , Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Caspase 12/metabolismo , Caspase 3/metabolismo , Hipóxia Celular , Linhagem Celular , Fosfatase 1 de Especificidade Dupla/deficiência , Fosfatase 1 de Especificidade Dupla/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mitocondriais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Ativação Transcricional
3.
J Gene Med ; 8(11): 1320-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16989006

RESUMO

Given their generous transgene capacity and inherent neurotropism, herpes simplex virus (HSV-1)-based viral vectors are promising tools for gene delivery to the central nervous system. Despite their widespread pre-clinical use, vector toxicity remains a concern with regard to the use of herpes vectors in humans. One potential source of toxicity stems from the tegument-associated virion host shutoff protein (vhs), which induces translational arrest in the host cell through non-specific mRNAse activity. In the current study we utilized a series of HSV-1 viruses containing a deletion in the U(L)41 open reading frame to investigate: (1) the requirement of intact vhs function in amplicon packaging and (2) whether vhs influences the post-transduction survival of dissociated cortical neurons. Our results demonstrate that while amplicon yield was reduced an order of magnitude, U(L)41 deletion was associated with reduced vector toxicity. Furthermore, partial reconstitution of vhs function using mRNAse-inactive point mutants improved amplicon titers without imparting the toxicity observed with wild-type controls. These findings offer a novel approach to improving the titer and toxicity profiles of HSV-based viral vectors.


Assuntos
Herpesvirus Humano 1/genética , Animais , Linhagem Celular , Sobrevivência Celular , Chlorocebus aethiops , Genes Virais , Herpesvirus Humano 1/fisiologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Células NIH 3T3 , Neurônios/citologia , Neurônios/virologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Ribonucleases/genética , Ribonucleases/metabolismo , Deleção de Sequência , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Montagem de Vírus
4.
Mol Med ; 7(8): 561-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11591892

RESUMO

BACKGROUND: Herpes simplex type I (HSV)-based vectors have been used experimentally for suicide gene therapy, immunomodulatory gene delivery, and direct oncolytic therapy. The current study utilizes the novel concept of regional delivery of an oncolytic virus in combination with or serving as the helper virus for packaging herpes-based amplicon vectors carrying a cytokine transgene, with the goal of identifying if this combination is more efficacious than either modality alone. MATERIALS AND METHODS: A replication competent oncolytic HSV (G207) and a replication incompetent HSV amplicon carrying the gene for the immunomodulatory cytokine IL-2 (HSV-IL2) were tested in murine syngeneic colorectal carcinoma and in rat hepatocellular carcinoma models. Liver tumors were treated with vascular delivery of (1) phosphate-buffered saline (PBS), (2) G207, (3) HSV-IL2, (4) G207 and HSV-IL2 mixed in combination (mG207/HSV- IL2), and (5) G207 as the helper virus for packaging the construct HSV-IL2 (pG207/HSV-IL2). RESULTS: Tumor burden was significantly reduced in all treatment groups in both rats and mice treated with high-dose G207, HSV-IL2, or both (p < 0.02). When a low dose of virus was used in mice, anti-tumor efficacy was improved by use of G207 and HSV-IL2 in combination or with HSV-IL2 packaged by G207 (p < 0.001). This improvement was abolished when CD4(+) and CD8(+) lymphocytes were depleted, implying that the enhanced anti-tumor response to low-dose combined therapy is immune mediated. CONCLUSIONS: Vascular regional delivery of oncolytic and amplicon HSV vectors can be used to induce improved anti-tumor efficacy by combining oncolytic and immunostimulatory strategies.


Assuntos
Terapia Genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Interleucina-2/genética , Neoplasias Hepáticas Experimentais/terapia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Sobrevivência Celular , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Interleucina-2/metabolismo , Neoplasias Hepáticas Experimentais/imunologia , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Distribuição Aleatória , Ratos , Ratos Endogâmicos BUF , Células Tumorais Cultivadas
5.
Mol Ther ; 4(3): 250-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11545616

RESUMO

There are two promising herpes viral-based anticancer strategies: one involves replication-defective viruses to transfer therapeutic transgenes, and the other involves replication-conditional oncolytic viruses, which selectively infect and destroy cancer cells directly. This study examines a novel dual herpesvirus preparation, which combines the immunostimulatory effects of amplicon-mediated IL2 expression with direct viral-induced oncolysis. The oncolytic virus G207 was used as the helper virus to package a herpes simplex virus (HSV)-amplicon vector carrying the gene IL2 (HSV-IL2), yielding a single preparation with two complementary modes of action. In vivo comparison was carried out in a syngeneic squamous cell carcinoma flank tumor model. We directly injected established tumors with HSV-IL2, G207, G207 mixed with HSV-IL2, or G207-packaged HSV-amplicon carrying the IL2 transgene (G207[IL2]). Significant inhibition of tumor growth was seen at 2 weeks in the G207[IL2]-treated tumors relative to controls (0.57+/-0.44 cm(3) versus 39.45+/-5.13 cm(3), P<0.00001), HSV-IL2 (20.97+/-4.60 cm(3)), and the G207 group (7.71+/-2.10 cm(3)). This unique use of a replication-conditional, oncolytic virus to package a replication-incompetent amplicon vector demonstrates impressive efficacy in vitro and in vivo, and avoids the theoretical concerns of recombination with reversion to wild type.


Assuntos
Imunoterapia , Interleucina-2/genética , Interleucina-2/imunologia , Neoplasias/genética , Neoplasias/terapia , Simplexvirus/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Morte Celular , Divisão Celular , Humanos , Interleucina-2/metabolismo , Interleucina-2/uso terapêutico , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Transfecção , Células Tumorais Cultivadas
6.
Blood ; 98(2): 287-95, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11435295

RESUMO

Herpes simplex virus (HSV)-based vectors have favorable biologic features for gene therapy of leukemia and lymphoma. These include high transduction efficiency, ability to infect postmitotic cells, and large packaging capacity. The usefulness of HSV amplicon vectors for the transduction of primary human B-cell chronic lymphocytic leukemia (CLL) was explored. Vectors were constructed encoding beta-galactosidase (LacZ), CD80 (B7.1), or CD154 (CD40L) and were packaged using either a standard helper virus (HSVlac, HSVB7.1, and HSVCD40L) or a helper virus-free method (hf-HSVlac, hf-HSVB7.1, and hf-HSVCD40L). Both helper-containing and helper-free vector stocks were studied for their ability to transduce CLL cells, up-regulate costimulatory molecules, stimulate allogeneic T-cell proliferation in a mixed lymphocyte tumor reaction, and generate autologous cytotoxic T lymphocytes (CTLs). Although helper-containing and helper-free amplicon stocks were equivalent in their ability to transduce CLL cells, a vigorous T-cell proliferative response was obtained using cells transduced with hf-HSVB7.1 but not with HSVB7.1. CLL cells transduced with either HSVCD40L or hf-HSVCD40L were compared for their ability to up-regulate resident B7.1 and to function as T-cell stimulators. Significantly enhanced B7.1 expression in response to CD40L was observed using hf-HSVCD40L but not with HSVCD40L. CLL cells transduced with hf-HSVCD40L were also more effective at stimulating T-cell proliferation than those transduced with HSVCD40L stocks and were successful in stimulating autologous CTL activity. It is concluded that HSV amplicons are efficient vectors for gene therapy of hematologic malignancies and that helper virus-free HSV amplicon preparations are better suited for immunotherapy.


Assuntos
Vetores Genéticos , Herpesvirus Humano 1/genética , Imunoterapia , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/terapia , Células Apresentadoras de Antígenos , Antígeno B7-1/genética , Ligante de CD40/genética , Citocinas/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Ativação Linfocitária , Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Transfecção , beta-Galactosidase/genética
7.
Gene Ther ; 8(2): 111-20, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11313780

RESUMO

Recently developed helper virus-free methods of herpes simplex virus (HSV) amplicon vector packaging provide stocks that are virtually devoid of the cytotoxic component normally associated with traditional helper virus-based packaging methods. These approaches involve cotransfection of amplicon plasmid DNA with either a five-cosmid set or a bacterial artificial chromosome (BAC) that contains the HSV genome without its cognate pac signals. Helper virus-free amplicon packaging produces low-titer stocks (<10(5) expressing particles/ml) that exhibit a high frequency of pseudotransduction. In an effort to enhance amplicon titers, we introduced in trans a genomic copy of the virion host shutoff (vhs) protein-encoding gene UL41 into both cosmid- and BAC-based packaging strategies. Cotransfection of this plasmid with the amplicon and packaging reagents results in a 10-fold higher amplicon titer, and stocks that do not exhibit the pseudotransduction phenomenon. To further enhance packaging efficiency, the HSV transcriptional activator VP16 was introduced into packaging cells 1 day before the packaging components. Pre-loading of packaging cells with VP16 led to an additional enhancement of amplicon titers, an effect that did not occur in the absence of vhs. Increased helper virus-free amplicon titers resulting from these modifications will make in vivo transduction experiments more feasible.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Proteínas Virais/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/genética , Cricetinae , Técnicas de Transferência de Genes , Vírus Auxiliares/genética , Camundongos , Reação em Cadeia da Polimerase/métodos , Ribonucleases , Proteínas Virais/genética , beta-Galactosidase/metabolismo
8.
J Mol Med (Berl) ; 78(3): 166-74, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10868479

RESUMO

Oncolytic viral therapy is a promising new method of cancer treatment. Peritoneal dissemination of cancer is a common and fatal clinical condition seen in many malignancies, with few effective therapies available. G207, a multimutated replication-competent herpes simplex virus type-1, effectively treats disseminated peritoneal cancer. This study evaluates viral proliferation and subsequent tumoricidal effects in vitro and in vivo after regional viral delivery. In vitro studies demonstrate that G207 efficiently kills five human gastric cancer cell lines, and that permissiveness to viral replication is correlated with cytotoxicity. In a murine xenograft model of human gastric carcinomatosis, peritoneal delivery of G207 effectively kills tumor and prolongs survival. Data from quantitative PCR characterizes peritoneal clearance of virus after intraperitoneal injection, and identifies G207 replication within tumor cells in vivo, similar to in vitro proliferation. Further analysis of various organs confirms that G207 does not replicate within normal tissue after peritoneal delivery. Wild-type KOS viral replication was also demonstrated in vivo, with significant toxicity secondary to dissemination and encephalitis. In vivo viral proliferation of G207 is restricted to tumor cells, is correlated with in vitro assays, and is an important mechanism of anticancer efficacy.


Assuntos
Herpesvirus Humano 1/genética , Neoplasias Peritoneais/terapia , Vírus/genética , Animais , Caquexia/virologia , Herpesvirus Humano 1/fisiologia , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Gástricas/terapia , Fatores de Tempo , Distribuição Tecidual , Resultado do Tratamento , Células Tumorais Cultivadas
9.
Mol Med ; 5(9): 606-16, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10551902

RESUMO

BACKGROUND: Costimulatory and cellular adhesion molecules are thought to be essential components of antigen presentation in the immune response to cancer. The current studies examine gene transfer utilizing herpes viral amplicon vectors (HSV) to direct surface expression of adhesion molecules, and specifically evaluate the potential of a tumor-expressing intercellular adhesion molecule-1 (ICAM-1) to elicit an anti-tumor response. MATERIALS AND METHODS: The human ICAM-1 (hICAM1) gene was inserted into an HSV amplicon vector and tested in a transplantable rat hepatocellular carcinoma and in a human colorectal cancer cell line. Cell surface ICAM-1 expression was assessed by flow cytometry. Lymphocyte binding to HSV-hICAM1-transduced cells was compared with that to cells transduced with HSV not carrying the ICAM gene. Tumorigenicity of HSV-hICAM1-transduced tumor cells were tested in syngeneic Buffalo rats. Additionally, immunization with irradiated (10,000 rads) HSV-hICAM1-transduced tumor cells was performed to determine its effect on tumor growth. RESULTS: A 20-min exposure of tumor cells at a multiplicity of infection (MOI) of 1 resulted in high-level cell surface expression of human ICAM in approximately 25% of tumor cells. Transduced rat or human tumor cells exhibited significantly enhanced binding of lymphocytes (p < 0.05). HSV-hICAM1-transduced cells elicited an increase in infiltration by CD4(+) lymphocytes in vivo and exhibited decreased tumorigenicity. Immunization with irradiated HSV-hICAM1-transduced cells protected against growth of subsequent injected parental tumor cells. CONCLUSIONS: HSV amplicon-mediated gene transfer is an efficient method for modifying the cell surface expression of adhesion molecules. Increased tumor expression of ICAM-1 represents a promising immune anti-cancer strategy.


Assuntos
Carcinoma Hepatocelular/imunologia , Imunidade Inata/genética , Molécula 1 de Adesão Intercelular/genética , Neoplasias Hepáticas/imunologia , Simplexvirus/genética , Animais , Testes de Carcinogenicidade , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Adesão Celular , Divisão Celular/genética , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica/métodos , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Ratos , Células Tumorais Cultivadas , Vacinas/administração & dosagem
10.
Exp Neurol ; 159(1): 65-72, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10486175

RESUMO

Brain ischemia is a cause of substantial morbidity and mortality during the later decades of life. In light of this, many studies have used in vitro and in vivo models of acute necrosis to test candidate therapeutic agents. More recently, the existence of a genetically programmed component of ischemic death has become widely accepted. We have used molecular genetic approaches to investigate the potential link between hypoxia-induced gene transcription and the delayed death of ischemic neurons. Hypoxia-induced gene expression is an evolutionarily conserved response comprising both transcriptional activation and posttranscriptional and posttranslational stabilization events. Members of the PER-ARNT-SIM (PAS) family of basic helix-loop-helix transcription factors have been shown to regulate hypoxic transcripts in nonneuronal cultured lines. However, evidence for ischemic activation of PAS proteins within the neuronal compartment or possible involvement in neuronal death is lacking. The tumor-suppressor protein p53 is a known transcriptional activator within the central nervous system that is clearly involved in the pathologic response to ischemia. This article will provide data that implicate the coordinate activities of p53 and the PAS protein HIF-1alpha in driving ischemia-induced delayed neuronal death. Background regarding mechanisms of ischemic neuronal death will also be provided with special attention paid to the role of de novo gene expression in promoting this pathologic sequence. The identification of the HIF-1alpha/p53-mediated signaling pathway in neurons highlights a novel target toward which anti-ischemic neuroprotective drug discovery can be applied.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas de Ligação a DNA/fisiologia , Hipóxia/metabolismo , Neurônios/citologia , Proteínas Nucleares/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Isquemia Encefálica/patologia , Morte Celular/fisiologia , Modelos Animais de Doenças , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Necrose , Neurônios/metabolismo , Fatores de Transcrição/fisiologia
11.
J Neurosci ; 19(16): 6818-24, 1999 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-10436039

RESUMO

Hypoxia-induced delayed neuronal death is known to require de novo gene expression; however, the molecular mediators that are involved remain undefined. The transcription factor hypoxia-inducible factor-1alpha (HIF-1alpha), in addition to promoting the expression of adaptive genes under conditions of hypoxia, has been implicated as being a necessary component in p53-mediated cell death in tumors. Using herpes amplicon-mediated gene transfer in cortical neuronal cultures, we demonstrate that delivery of a dominant-negative form of HIF-1alpha (HIFdn), capable of disrupting hypoxia-dependent transcription, reduces delayed neuronal death that follows hypoxic stress. In contrast, hypoxia-resistant p53-null primary cultures are not protected by HIFdn expression. These data indicate that, in hypoxic neurons, HIF-1alpha and p53 conspire to promote a pathological sequence resulting in cell death.


Assuntos
Morte Celular/fisiologia , Hipóxia Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Genes p53 , Neurônios/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , Genes Dominantes , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Camundongos Knockout , Fatores de Tempo , Transcrição Gênica , Células Tumorais Cultivadas
12.
Hum Gene Ther ; 10(10): 1599-606, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10428205

RESUMO

This study evaluates inhibition of human squamous cell carcinomas (SCCs) by a replication-competent multimutated herpes simplex virus type 1 (G207). Infectivity and cytotoxicity of the G207 virus were evaluated in vitro in seven human SCC cell lines. In vivo effects of the G207 virus on human tumor xenografts in an athymic rat model were then investigated by injecting established tumors with 1 x 10(7) virus particles and monitoring tumor growth. In addition, oral cavity tumors in immunocompetent hamster were infected with the G207 virus by selective intraarterial perfusion and the tumor response was monitored. In vitro studies demonstrated infection rates, measured 24 hr after exposure, exceeding 40% at an MOI of 2 in five of seven human SCC cell lines. Cytotoxic effects, as measured by percent cell death on day 5, exceeded 90% in five of seven SCC cell lines. In vivo inhibition of tumor growth in an athymic rat model was seen (p < 0.005) and in two of the cell lines a complete clinical response was seen in 12 of 14 tumors. In the hamster model, selective intraarterial perfusion with G207 virus showed selective infection of the tumor cells, with sparing of the adjacent normal mucosa, which leading to significant suppression of tumor growth (p < 0.005). The G207 virus displayed efficient and selective cytotoxicity and tumor growth inhibition against human SCC and may prove useful as a therapeutic agent for head and neck SCC.


Assuntos
Carcinoma de Células Escamosas/virologia , Neoplasias de Cabeça e Pescoço/virologia , Herpesvirus Humano 1/fisiologia , Animais , Ciclo Celular , Cricetinae , Galactosídeos , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/patogenicidade , Humanos , Indóis , Mucosa , Perfusão , Ratos , Ratos Nus , Coloração e Rotulagem/métodos , Células Tumorais Cultivadas
13.
FASEB J ; 13(11): 1325-34, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10428757

RESUMO

G207 is a multi-mutated, replication-competent type-1 herpes simplex virus designed to target, infect, and lyse neurological tumors. This study examines the feasibility of using G207 in the treatment of human colorectal cancer and defines the biological determinants of its antitumor efficacy. This virus was tested on five human colorectal cancer cell lines in vitro to determine efficacy of infection and tumor cell kill. These results were correlated to measures of tumor cell proliferation. In vivo testing was performed through direct injections of G207 into xenografts of human colorectal cancer tumors grown in flanks of athymic rats. To evaluate an alternate method of administration, hepatic portal vein infusion of G207 was performed in a syngeneic model of liver metastases in Buffalo rats. Among the five cell lines tested, infection rates ranged between 10% and 90%, which correlated directly with S-phase fraction (8.6%-36.6%) and was proportional to response to G207 therapy in vitro (1%-93%). Direct injection of G207 into nude rat flank tumors suppressed tumor growth significantly vs. control (0.58 +/- 0.60 cm(3) vs. 9.16 +/- 3.70 cm(3), P<0. 0001). In vivo tumor suppression correlated with in vitro effect. In the syngeneic liver tumor model, portal infusion resulted in significant reduction in number of liver nodules (13 +/- 10 nodules in G207-treated livers vs. 80 +/- 30 nodules in control livers, P<0.05). G207 infects and kills human colorectal cancer cells efficiently. In vitro cytotoxicity assay and tumor S-phase fraction can be used to predict response to treatment in vivo. This antineoplastic agent can be delivered effectively by both direct tumor injection and regional vascular infusion. G207 should be investigated further as therapy for colorectal cancer and liver metastases.


Assuntos
Neoplasias Colorretais/terapia , Herpesvirus Humano 1/fisiologia , Neoplasias Hepáticas/terapia , Animais , Morte Celular , Neoplasias Colorretais/patologia , Neoplasias Colorretais/virologia , Humanos , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/virologia , Mutação , Transplante de Neoplasias , Ratos , Ratos Nus , Replicação Viral
14.
Neuroreport ; 10(2): 337-44, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-10203332

RESUMO

The influence of NGF on the neuroanatomic substrate(s) subserving learning and memory was probed by somatic mosaic analysis. NGF XAT mice underwent unilateral hippocampal delivery of virus vector expressing cre recombinase to produce focal NGF gain of function mosaics. Activated mice expressing increased NGF, but not control mice transduced with the lacZ gene showed reorganization of the septohippocampal projection assessed by retrograde labeling with Fluorogold (FG). Mice mosaic for NGF function demonstrated in ipsilateral medial septum and diagonal band significant increases in FG-labeled cell bodies (94%), ChAT-labeled cells (55%), double-positive cells (190%) and increased somal size of double-positive cells (56%) than did non-activated mice. These results indicate that reorganization of the cholinergic septal input to a specific hippocampal region is promoted by gain of NGF function.


Assuntos
Fibras Colinérgicas/fisiologia , Hipocampo/fisiologia , Fatores de Crescimento Neural/fisiologia , Plasticidade Neuronal/fisiologia , Septo Pelúcido/fisiologia , Estilbamidinas , Animais , Tamanho Celular , Fibras Colinérgicas/patologia , Corantes Fluorescentes , Lobo Frontal/citologia , Lobo Frontal/fisiologia , Hipertrofia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mosaicismo/fisiopatologia , Neurônios/citologia , Neurônios/fisiologia , Neurônios Aferentes/patologia , Prosencéfalo/fisiologia , Valores de Referência , Septo Pelúcido/citologia , Transmissão Sináptica/fisiologia
15.
J Virol ; 73(4): 2832-40, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10074131

RESUMO

Lentivirus vectors based on human immunodeficiency virus (HIV) type 1 (HIV-1) constitute a recent development in the field of gene therapy. A key property of HIV-1-derived vectors is their ability to infect nondividing cells. Although high-titer HIV-1-derived vectors have been produced, concerns regarding safety still exist. Safety concerns arise mainly from the possibility of recombination between transfer and packaging vectors, which may give rise to replication-competent viruses with pathogenic potential. We describe a novel lentivirus vector which is based on HIV, simian immunodeficiency virus (SIV), and vesicular stomatitis virus (VSV) and which we refer to as HIV/SIVpack/G. In this system, an HIV-1-derived genome is encapsidated by SIVmac core particles. These core particles are pseudotyped with VSV glycoprotein G. Because the nucleotide homology between HIV-1 and SIVmac is low, the likelihood of recombination between vector elements should be reduced. In addition, the packaging construct (SIVpack) for this lentivirus system was derived from SIVmac1A11, a nonvirulent SIV strain. Thus, the potential for pathogenicity with this vector system is minimal. The transduction ability of HIV/SIVpack/G was demonstrated with immortalized human lymphocytes, human primary macrophages, human bone marrow-derived CD34(+) cells, and primary mouse neurons. To our knowledge, these experiments constitute the first demonstration that the HIV-1-derived genome can be packaged by an SIVmac capsid. We demonstrate that the lentivirus vector described here recapitulates the biological properties of HIV-1-derived vectors, although with increased potential for safety in humans.


Assuntos
Terapia Genética , Vetores Genéticos , HIV-1/genética , Lentivirus/genética , Vírus da Imunodeficiência Símia/genética , Animais , Humanos , Camundongos
16.
Cancer Immunol Immunother ; 47(5): 265-71, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10022470

RESUMO

In vivo cytokine gene transfer may greatly simplify autologous tumor vaccine production. Herpes simplex viral amplicon vectors (HSV) are efficient gene-transfer vehicles and may overcome many limitations of prior gene-transfer methods. The interleukin-2 (IL-2) and beta-galactosidase genes (lac) were inserted into an HSV amplicon vector and tested in a subcutaneous squamous cell carcinoma of lung origin to determine the efficiency of in vivo gene transfer and the utility of such a direct gene transfer approach in cancer therapy. Gene transfer and expression were assessed by histochemical staining and enzyme-linked immunosorbent assay (ELISA). Growth of injected tumors as well as non-injected tumors remote from the site of injection was assessed. Assessment of lymphocytic infiltrates into tumors was performed by immunohistochemistry. Survival was recorded. Direct in vivo injection of established tumors with a HSVi12 resulted in efficient gene transfer and production of IL-2 in the injected tumor but not at tumors remote from the sites of injection. There was a significant suppression of growth of the tumors injected with HSVi12 (P<0.01) when compared with tumors injected with HSV without i12. Of note, growth of tumors remote from sites of HSVi12 injection was also retarded and treatment was associated with a significant (P<0.05) improvement in survival. Direct intratumoral administration of HSV amplicon vectors can result in efficient transfer of cytokine genes and have antitumor efficacy. HSV vectors are therefore potentially useful agents in such in vivo gene-therapy strategies and simplify cytokine antitumor gene-therapy strategies.


Assuntos
Carcinoma de Células Escamosas/terapia , DNA Recombinante/uso terapêutico , Terapia Genética , Vetores Genéticos/uso terapêutico , Interleucina-2/genética , Neoplasias Pulmonares/terapia , Neoplasias Experimentais/terapia , Simplexvirus/genética , beta-Galactosidase/genética , Animais , Imuno-Histoquímica , Masculino , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas
17.
Blood ; 93(2): 643-54, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9885227

RESUMO

Herpes simplex virus amplicon vectors expressing RANTES (HSVrantes) and the T-cell costimulatory ligand B7.1 (HSVB7.1) were studied for their ability to elicit a tumor-specific T-cell response in a murine lymphoma model. HSVB7.1- and HSVrantes-transduced EL4 cells expressed high levels of B7.1 and RANTES as analyzed by flow cytometry and enzyme-linked immunosorbent assay, respectively. Inoculation of ex vivo HSVB7.1 transduced cells in syngeneic mice resulted in regression of both transduced cells and nontransduced cells inoculated contralaterally. Direct intratumoral injection of HSVB7.1 and/or HSVrantes alone or in combination into established EL4 tumors led to complete tumor regression in injected tumors as well as in nontransduced contralaterally implanted tumor, whereas control tumors or tumors injected with HSVlac expressing beta-galactosidase did not regress. Maximal protection was achieved with combined injection of HSVB7.1 and HSVrantes; mice showing tumor regression were resistant to rechallenge with parental EL4 cells, and tumor cell-specific cytolytic T-cell activity was observed in mice demonstrating regression. HSV amplicon-mediated delivery of immune effector molecules may represent a useful strategy for immunotherapy in the setting of pre-existing tumor.


Assuntos
Vetores Genéticos , Imunoterapia , Linfoma de Células T/terapia , Simplexvirus/genética , Linfócitos T/imunologia , Animais , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Expressão Gênica , Humanos , Linfoma de Células T/imunologia , Linfoma de Células T/patologia , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Linfócitos T Citotóxicos/imunologia , Transfecção , Células Tumorais Cultivadas
18.
Ann N Y Acad Sci ; 884: 345-60, 1999 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-10842605

RESUMO

The application of gene therapy to the inner ear is an emerging field of study. Most studies report the expression of marker genes (e.g., galactosidase) within the tissues of the cochlea. The first biologic response of an inner ear tissue (i.e., auditory neurons) to transduction by a gene therapy vector expressing a therapeutic gene (a herpes amplicon vector containing a BDNF gene) was observed in spiral explants obtained from early postnatal rat cochleae. This study was important because it demonstrated the feasibility of a gene augmentation approach to treat traumatized cochleae. Long-term expression of transduced or transfected genes in cochlear tissues have been obtained with adenovirus, adeno-associated virus, and herpes amplicon vectors. The herpes amplicon vector (i.e., HSVbdnflac) that evoked a biologic response in vitro has also been successfully used to support the survival of auditory neurons in vivo following loss of the auditory hair cells (i.e., loss of trophic factor). Gene therapy has been successfully applied to the cochlea of a laboratory animal, and future studies will define the types of vectors and therapeutic genes that will work best for the treatment of inner ear diseases in the clinic.


Assuntos
Surdez/terapia , Orelha Interna , Terapia Genética , Vetores Genéticos/uso terapêutico , Fatores de Crescimento Neural/uso terapêutico , Proteínas E1 de Adenovirus , Animais , Surdez/genética , Orelha Interna/efeitos dos fármacos , Previsões , Herpesvirus Humano 1 , Ratos , Gânglio Espiral da Cóclea/efeitos dos fármacos
19.
J Neurosci Methods ; 80(2): 137-47, 1998 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-9667386

RESUMO

To develop a reproducible gene transfer method for the murine CNS we evaluated delivery of various gene vehicles using mechanical or manual stereotaxic intracranial inoculation. A microprocessor controlled microsyringe pump (The World Precision Instruments/UltraMicroPump) programmable for volume, rate and syringe size and designed to dispense nanoliter and picoliter volumes was compared to a standard manual deliver method. Gene transfer efficiency of two viral vectors, two synthetic cationic lipid molecules, and naked DNA were evaluated in mice injected unilaterally in two brain regions. Animals received 1 microl over 10 min. of either HSVlac (1 x 10(5) b.f.u), AdLac (1 x 10(5) p.f.u), Tfx-10 or Tfx-20 (2.6 microg DNA in 2.0 microl Tfx; 1:1 charge ratio of DNA to liposome), or naked DNA (HSVlac plasmid, 10 microg/microl). After 4 days, animals from each group were perfused and tissue prepared for X-gal histochemical detection of beta-galactosidase expression. Blue cells were observed in the HSV, Adenovirus, and Tfx-20 groups only at the injection site in animals injected using the UMP. Animals injected manually exhibited fewer blue cells and positive cells were not restricted to the injection site. To quantify expression, tissue punches harvested from the injection sites as well as other brain regions were analyzed using a chemiluminescent reporter assay to detect beta-galactosidase (Galacto-Light). These data indicated increased activity in all animals injected with a lacZ containing vector via the UMP as compared to manual delivery: A 41% increase in the expression levels of beta-gal in HSVlac infected animals (p = 0.0029); a 29% increase in Adlac infected animals (p = 0.01); a 56% increase in Tfx-10 transduced animals (p = 0.04); a 24% increase in Tfx-20 transduced animals (p = 0.01); and a 69% increase in naked DNA gene transfer (p = 0.05). Total beta-galactosidase activity was greatest in HSVlac infected mice followed by Adlac > Tfx-20 > Tfx-10 = naked DNA.


Assuntos
Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Técnicas de Transferência de Genes/instrumentação , Microcomputadores , Adenoviridae/genética , Animais , DNA Viral/metabolismo , Genes Virais , Terapia Genética/instrumentação , Terapia Genética/métodos , Vetores Genéticos , Herpes Simples/genética , Herpes Simples/terapia , Lipossomos/metabolismo , Camundongos , Microinjeções , Reprodutibilidade dos Testes , Simplexvirus/genética , Técnicas Estereotáxicas/instrumentação , Proteínas Estruturais Virais/genética
20.
Am J Pathol ; 152(6): 1521-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626056

RESUMO

We investigated the transfer of the lacZ reporter gene into human trophoblastic cells using herpes simplex virus and adeno-associated virus vectors. We used an established choriocarcinoma cell line (BeWo cells) that can be induced to terminally differentiate after treatment with cyclic-AMP. Our results demonstrate that transduction of trophoblastic cells by the herpes simplex virus vector, HSV.CMVlac, and the adeno-associated virus vector, AAV.CMVlac, is affected by cellular differentiation. Treatment of BeWo cells with cyclic-AMP reduced transduction by HSV.CMVlac but increased transduction by the AAV vector. In contrast, when BeWo cells were transfected with herpes simplex virus and adeno-associated virus plasmids, lacZ expression was not affected by treatment with cyclic-AMP. Southern blot analysis demonstrated 2.75 times less herpes simplex virus DNA in cyclic-AMP treated BeWo cells, but 2.0 to 7.4 times more adeno-associated virus DNA in treated cells. We conclude that inefficient transduction of differentiated trophoblastic cells with HSV.CMVlac is because of diminished viral entry, whereas cellular differentiation is associated with increased entry of AAV.CMVlac. These observations suggest that adeno-associated virus vectors may be used to modify trophoblast function and study placental physiology. Additionally, trophoblast differentiation leads to alterations in the mechanisms of virus uptake that may affect maternal-to-fetus transmission.


Assuntos
Diferenciação Celular/fisiologia , Transfecção/efeitos dos fármacos , Trofoblastos/citologia , Trofoblastos/virologia , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Adenoviridae/genética , Southern Blotting , Diferenciação Celular/efeitos dos fármacos , Dependovirus/genética , Relação Dose-Resposta a Droga , Técnicas de Transferência de Genes , Vetores Genéticos , Herpesvirus Humano 1/genética , Humanos , Trofoblastos/efeitos dos fármacos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...