Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Ther ; 19(3): 500-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21081904

RESUMO

Persistent pain remains a tremendous health problem due to both its prevalence and dearth of effective therapeutic interventions. To maximize pain relief while minimizing side effects, current gene therapy-based approaches have mostly exploited the expression of pain inhibitory products or interfered with pronociceptive ion channels. These methods do not enable control over the timing or duration of analgesia, nor titration to analgesic efficacy. Here, we describe a gene therapy strategy that potentially overcomes these limitations by providing exquisite control over therapy with efficacy in clinically relevant models of inflammatory pain. We utilize a herpes simplex viral (HSV) vector (vHGlyRα1) to express a ligand-regulated chloride ion channel, the glycine receptor (GlyR) in targeted sensory afferents; the subsequent exogenous addition of glycine provides the means for temporal and spatial control of afferent activity, and therefore pain. Use of an endogenous inhibitory receptor not normally present on sensory neurons both minimizes immunogenicity and maximizes therapeutic selectivity.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos , Manejo da Dor , Receptores de Glicina , Células Receptoras Sensoriais , Simplexvirus , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Ordem dos Genes , Terapia Genética , Vetores Genéticos/genética , Glicina/metabolismo , Glicina/farmacologia , Células HEK293 , Humanos , Ligantes , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Células Receptoras Sensoriais/metabolismo , Simplexvirus/genética
2.
Stem Cells ; 26(12): 3119-29, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18787207

RESUMO

The ability of embryonic stem cells to develop into multiple cell lineages provides a powerful resource for tissue repair and regeneration. Gene transfer offers a means to dissect the complex events in lineage determination but is limited by current delivery systems. We designed a high-efficiency replication-defective herpes simplex virus gene transfer vector (JDbetabeta) for robust and transient expression of the transcription factors Pax3 and MyoD, which are known to be involved in skeletal muscle differentiation. JDbetabeta-mediated expression of each gene in day 4 embryoid bodies (early-stage mesoderm) resulted in the induction of unique alterations in gene expression profiles, including the upregulation of known target genes relevant to muscle and neural crest development, whereas a control enhanced green fluorescent protein expression vector was relatively inert. This vector delivery system holds great promise for the use of gene transfer to analyze the impact of specific genes on both regulatory genetic events and commitment of stem cells to particular lineages.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteína MyoD/biossíntese , Fatores de Transcrição Box Pareados/biossíntese , Simplexvirus/metabolismo , Animais , Linhagem Celular Tumoral , Linhagem da Célula , Chlorocebus aethiops , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/metabolismo , Humanos , Músculo Esquelético/metabolismo , Fator de Transcrição PAX3 , Células Vero
3.
Biotechnol Bioeng ; 102(4): 1087-97, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18828174

RESUMO

Herpes simplex virus type 1 (HSV-1) represents an attractive vehicle for a variety of gene therapy applications. To render this virus safe for clinical use, its cytotoxic genes must be removed without losing its ability to express transgenes efficiently. Our vectors are deleted for the essential immediate early genes ICP4 and ICP27. These genes are controlled by unique promoters having enhancer elements responsive to a viral structural protein VP16. The expression of these genes occurs prior to the activation of all other lytic functions and is thus required to initiate and complete the virus replication cycle. For large scale manufacture of clinical grade vectors, efficient cell lines must be generated that express the essential viral gene products in trans during vector propagation. Here we describe methods for engineering HSV-1 production cell lines that improve vector growth by altering the kinetics of complementing gene expression. We examined the ability of Vero cells independently transduced with ICP4 and ICP27 under transcriptional control of their respective promoters to support the growth of a replication defective vector (JDTOZHE), deleted for ICP4, ICP27 and approximately 20 kb of internal elements that are not required for virus growth in Vero cells. Vector yield on this cell line was 3 logs lower than wild-type virus grown on Vero cells. To understand the mechanism underlying poor vector yield, we examined the expression of ICP4 and ICP27 during virus complementation. While ICP27 was expressed immediately on vector infection, the expression of ICP4 was considerably delayed by 8-10 h, suggesting that the ICP4 promoter was not adequately activated by VP16 delivered by the infectious vector particle. Use of the ICP0 promoter to express ICP4 from the cellular genome resulted in higher induction levels and faster kinetics of ICP4 expression and a 10-fold improvement in vector yield. This study suggests that vector complementation is highly dependent on the kinetics of complementing gene expression and can lead to large differences in vector yield.


Assuntos
Biotecnologia/métodos , Vetores Genéticos , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/genética , Replicação Viral , Animais , Chlorocebus aethiops , Deleção de Genes , Expressão Gênica , Regulação Viral da Expressão Gênica , Terapia Genética , Proteínas Imediatamente Precoces/genética , Células Vero
4.
Methods Mol Biol ; 433: 97-113, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18679619

RESUMO

Virus vectors have been employed as gene transfer vehicles for various pre-clinical and clinical gene therapy applications. Replication-competent herpes simplex virus (HSV) vectors that replicate specifically in actively dividing glial tumor cells have been used in Phase I-II human trials in patients with glioblastoma multiforme (GBM), a fatal form of brain cancer. Research during the last decade on the development of HSV vectors has resulted in the engineering of recombinant vectors that are totally replication defective, non-toxic, and capable of long-term transgene expression. This chapter describes methods for the construction of recombinant genomic HSV vectors based on the HSV-1 replication-defective vector backbones, steps in their purification, and their small-scale production for use in cell culture experiments as well as studies in animals.


Assuntos
Vetores Genéticos/biossíntese , Herpesvirus Humano 1/genética , Biologia Molecular/métodos , Animais , Células COS , Chlorocebus aethiops , DNA Viral/genética , Genoma Viral/genética , Herpesvirus Humano 1/fisiologia , Humanos , Transfecção , Vírion , Replicação Viral
5.
Methods Mol Biol ; 246: 257-99, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14970599

RESUMO

The human herpesviruses represent excellent candidate viruses for several types of gene vector applications. As a class, they are large DNA viruses with the potential to accommodate large or multiple transgene cassettes, and they have evolved to persist in a lifelong nonintegrated latent state without causing disease in the immune-competent host. Among the herpesviruses, herpes simplex virus type 1 (HSV-1) is an attractive vehicle because in natural infection, the virus establishes latency in neurons, a state in which viral genomes may persist for the life of the host as intranuclear episomal elements. The natural lifelong persistence of latent genomes in trigeminal ganglia (TG) without the development of sensory loss or histologic damage to the ganglion attests to the effectiveness of these natural latency mechanisms. Although the wild-type virus may be reactivated from latency under the influence of a variety of stresses, completely replication defective viruses can be constructed that retain the ability to establish persistent quiescent genomes in neurons, but that are unable to subsequently reactivate in the nervous system. These persistent genomes are devoid of lytic gene expression, but retain the ability to express latency-associated transcripts (LATs).


Assuntos
Técnicas de Transferência de Genes , Simplexvirus/genética , Animais , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Vetores Genéticos , Genoma Viral , Herpes Simples/fisiopatologia , Herpes Simples/virologia , Humanos , Simplexvirus/fisiologia , Latência Viral
6.
Methods Mol Biol ; 246: 339-52, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14970603

RESUMO

In contrast to traditional drugs that generally act by altering existing gene product function, gene therapy aims to target the root cause of the disease by altering the genetic makeup of the cell to treat the disease. Researchers have adapted several classes of viruses as gene-transfer vectors, taking advantage of natural viral mechanisms designed to efficiently and effectively deliver DNA to the host-cell nucleus. Among these, the human herpesviruses are excellent candidate vectors for a variety of applications. Herpes simplex virus type 1 (HSV-1) is a particularly attractive gene-transfer vehicle because natural infection in humans includes a latent state in which the viral genome persists in a nonintegrated form without causing disease in an immune-competent host. HSV-1 is a large DNA virus with a broad host range that can be engineered to accommodate multiple or large therapeutic transgenes (4). HSV vectors may be generally useful for gene transfer to a variety of tissues in which short-term or extended transgene expression of therapeutic transgenes achieve a therapeutic effect. We have used therapeutic vectors to successfully treat human disease models in animals, including cancer, Parkinson's disease, and nerve damage (5-10).


Assuntos
Vetores Genéticos , Simplexvirus/genética , Células-Tronco/virologia , Animais , Antígenos CD34/imunologia , Ensaio de Imunoadsorção Enzimática , Técnicas de Transferência de Genes , Humanos , Camundongos , Simplexvirus/fisiologia , Células-Tronco/imunologia , Transdução Genética , Replicação Viral
7.
Hum Gene Ther ; 24(2): 170-80, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23316929

RESUMO

We previously reported the effects of herpes simplex virus (HSV) vector-mediated enkephalin on bladder overactivity and pain. In this study, we evaluated the effects of vHPPE (E1G6-ENK), a newly engineered replication-deficient HSV vector encoding human preproenkephalin (hPPE). vHPPE or control vector was injected into the bladder wall of female rats 2 weeks prior to the following studies. A reverse-transcription PCR study showed high hPPE transgene levels in L6 dorsal root ganglia innervating the bladder in the vHPPE group. The number of freezing behaviors, which is a nociceptive reaction associated with bladder pain, was also significantly lower in the vHPPE group compared with the control group. The number of L6 spinal cord c-fos-positive cells and the urinary interleukin (IL)-1ß and IL-6 levels after resiniferatoxin (RTx) administration into the bladder of the vHPPE group were significantly lower compared with those of the control vector-injected group. In continuous cystometry, the vHPPE group showed a smaller reduction in intercontraction interval after RTx administration into the bladder. This antinociceptive effect was antagonized by naloxone hydrochloride. Thus, the HSV vector vHPPE encoding hPPE demonstrated physiological improvement in visceral pain induced by bladder irritation. Gene therapy may represent a potentially useful treatment modality for bladder hypersensitive disorders such as bladder pain syndrome/interstitial cystitis.


Assuntos
Encefalinas/metabolismo , Terapia Genética/métodos , Vetores Genéticos/metabolismo , Nociceptividade , Precursores de Proteínas/metabolismo , Simplexvirus/metabolismo , Bexiga Urinária Hiperativa/terapia , Analgésicos/antagonistas & inibidores , Analgésicos/farmacologia , Animais , Diterpenos/administração & dosagem , Encefalinas/genética , Feminino , Gânglios Espinais/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Interleucina-1beta/urina , Interleucina-6/urina , Naloxona/farmacologia , Precursores de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simplexvirus/genética , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/fisiopatologia , Bexiga Urinária Hiperativa/patologia , Cateterismo Urinário , Replicação Viral , Dor Visceral/terapia
8.
Cold Spring Harb Protoc ; 2011(5): pdb.prot5615, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21536761

RESUMO

INTRODUCTION: Engineering effective vectors has been crucial to the efficient delivery and expression of therapeutic gene products in vivo. Among these, HSV-1 represents an excellent candidate vector for delivery to the peripheral and central nervous systems. The natural biology of HSV-1 includes the establishment of a lifelong latent state in neurons in which the viral genome persists as an episomal molecule. Genomic HSV vectors can be produced that are completely replication-defective, nontoxic, and capable of long-term transgene expression. Herpes simplex virus (HSV) vectors are constructed by using a replication-deficient vector backbone (TOZ.1) for homologous recombination with a shuttle plasmid containing a cassette expressing the gene of interest inserted into the UL41 gene sequence. The TOZ.1 vector expresses a reporter gene (lacZ) in the UL41 locus, such that recombination of the transgenic cassette into the UL41 locus results in the loss of the reporter gene activity. The TOZ.1 vector also contains a unique PacI endonuclease site for digestion of parental viral DNA that substantially reduces the nonrecombinant background. Following homologous recombination of the shuttle plasmid into the PacI-digested TOZ.1 genome, the recombinants are identified as clear plaques. After three rounds of limiting dilution analysis, the structure of the recombinants can be confirmed by Southern blot or by polymerase chain reaction (PCR) analysis.


Assuntos
Vírus Defeituosos/genética , Vetores Genéticos , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/genética , Replicação Viral , Expressão Gênica , Engenharia Genética/métodos , Terapia Genética/métodos , Biologia Molecular/métodos , Plasmídeos
10.
Virology ; 375(1): 13-23, 2008 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-18321553

RESUMO

ICP0 is a multi-functional herpes simplex virus type 1 (HSV-1) immediate-early (IE) gene product that contributes to efficient virus growth and reactivation from latency. Here we show that HSV-1-induced cell-cycle arrest at the G2/M border requires ICP0 and Chk2 kinase and that ICP0 expression by transfection or infection induces ATM-dependent phosphorylation of Chk2 and Cdc25C. Infection of cells with a replication-defective mutant virus deleted for all the regulatory IE genes except ICP0 (TOZ22R) induced G2/M arrest whereas a mutant virus deleted in addition for ICP0 (QOZ22R) failed to do so. Chk2-deficient cells and cells expressing a kinase-deficient Chk2 did not undergo cell-cycle arrest in response to TOZ22R infection. Chk2 deficiency diminished the growth of wild-type HSV-1, but not the growth of an ICP0-deleted recombinant virus. Together, these results are consistent with the interpretation that ICP0 activates a DNA damage response pathway to arrest cells in G2/M phase and promote virus growth.


Assuntos
Ciclo Celular , Herpesvirus Humano 1/crescimento & desenvolvimento , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular , Quinase do Ponto de Checagem 2 , Deleção de Genes , Herpesvirus Humano 1/genética , Humanos , Proteínas Imediatamente Precoces/genética , Fosforilação , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Ubiquitina-Proteína Ligases/genética , Fosfatases cdc25/metabolismo
11.
J Virol ; 78(17): 8994-9006, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15308696

RESUMO

Herpes simplex virus type 1 (HSV-1) is a promising vector for gene therapy applications, particularly at peripheral nerves, the natural site of virus latency. Many gene vectors require large particle numbers for even early-phase clinical trials, emphasizing the need for high-yield, scalable manufacturing processes that result in virus preparations that are nearly free of cellular DNA and protein contaminants. HSV-1 is an enveloped virus that requires the development of gentle purification methods. Ideally, such methods should avoid centrifugation and may employ selective purification processes that rely on the recognition of a unique envelope surface chemistry. Here we describe a novel method that fulfills these criteria. An immobilized metal affinity chromatography (IMAC) method was developed for the selective purification of vectors engineered to display a high-affinity binding peptide. Feasibility studies involving various transition metal ions (Cu2+, Zn2+, Ni2+, and Co2+) showed that cobalt had the most desirable features, which include a low level of interaction with either the normal virus envelope or contaminating DNA and proteins. The introduction of a cobalt-specific recognition element into the virus envelope may provide a suitable target for cobalt-dependent purification. To test this possibility, we engineered a peptide with affinity for immobilized cobalt in frame in the heparan sulfate binding domain of HSV-1 glycoprotein B, which is known to be exposed on the surface of the virion particle and recombined into the viral genome. By optimizing the IMAC loading conditions and reducing cobalt ion leakage, we recovered 78% of the tagged HSV-1 recombinant virus, with a >96% reduction in contaminating proteins and DNA.


Assuntos
Cromatografia de Afinidade/métodos , Cobalto/metabolismo , Vetores Genéticos/isolamento & purificação , Herpesvirus Humano 1/isolamento & purificação , Adsorção , Sequência de Aminoácidos , Animais , Western Blotting , Cátions Bivalentes/metabolismo , Chlorocebus aethiops , DNA/análise , Engenharia Genética , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 1/fisiologia , Cinética , Dados de Sequência Molecular , Plasmídeos/genética , Proteínas/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Vero
12.
Mol Ther ; 8(4): 530-42, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14529825

RESUMO

Our laboratory has employed replication-defective herpes simplex virus type 1 gene transfer vectors for treatment of animal models of human malignant glioblastoma. The base vectors were defective for the immediate early (IE) genes ICP4, ICP27, and ICP22 but expressed the IE gene ICP0, which can arrest tumor cell division, and an IE thymidine kinase (alpha-tk) gene construct that mediates suicide gene therapy (SGT) in the presence of ganciclovir (GCV). Previously, we reported that SGT using ICP0/alpha-tk vectors in nude mouse models of glioblastoma was improved by coexpression of the gap-junction-forming protein connexin43 (Cx43) or human tumor necrosis factor alpha (TNF alpha). We also showed that further gains in therapeutic outcome could be achieved by combining TNF alpha-enhanced SGT with gamma-knife radiosurgery (GKR). To expand these observations, we have first repeated these studies in immunocompetent rats with brain tumors derived from implanted 9L gliosarcoma cells and second compared the most efficient vector from this study with a new recombinant vector, NUREL-C2, which expressed both TNF alpha and Cx43 along with ICP0 and alpha-tk. Results from the first part indicated that our ICP0/alpha-tk/TNF alpha vector in combination with GKR provides an effective therapy although this treatment was not statistically better than GKR combined with the ICP0/alpha-tk/Cx43 vector. Our observations in the second part suggested that NUREL-C2 may be more effective than the ICP0/alpha-tk/TNF alpha vector in combination treatments with GCV (P = 0.08) or GCV plus GKR (P = 0.10). GKR significantly enhanced the efficacy of NUREL-C2/GCV treatment (P = 0.02) as well as other virus/GCV treatments (P < or = 0.05). Conversely, the efficacy of GKR was significantly improved by both the ICP0/alpha-tk/TNF alpha vector and NUREL-C2 in combination with GCV (P = 0.02 and P < 0.01, respectively). Together these results indicate that NUREL-C2 may be an attractive candidate for Phase I gene-therapy safety studies in patients with recurrent malignant glioblastoma.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Terapia Genética , Vetores Genéticos , Gliossarcoma/tratamento farmacológico , Gliossarcoma/radioterapia , Simplexvirus , Animais , Neoplasias Encefálicas/imunologia , Conexina 43/genética , Conexina 43/metabolismo , Gliossarcoma/imunologia , Ratos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA