Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Ann Neurol ; 70(2): 207-12, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21796661

RESUMO

OBJECTIVE: Preclinical evidence indicates that gene transfer to the dorsal root ganglion using replication-defective herpes simplex virus (HSV)-based vectors can reduce pain-related behavior in animal models of pain. This clinical trial was carried out to assess the safety and explore the potential efficacy of this approach in humans. METHODS: We conducted a multicenter, dose-escalation, phase I clinical trial of NP2, a replication-defective HSV-based vector expressing human preproenkephalin (PENK) in subjects with intractable focal pain caused by cancer. NP2 was injected intradermally into the dermatome(s) corresponding to the radicular distribution of pain. The primary outcome was safety. As secondary measures, efficacy of pain relief was assessed using a numeric rating scale (NRS), the Short Form McGill Pain Questionnaire (SF-MPQ), and concurrent opiate usage. RESULTS: Ten subjects with moderate to severe intractable pain despite treatment with >200mg/day of morphine (or equivalent) were enrolled into the study. Treatment was well tolerated with no study agent-related serious adverse events observed at any point in the study. Subjects receiving the low dose of NP2 reported no substantive change in pain. Subjects in the middle- and high-dose cohorts reported pain relief as assessed by NRS and SF-MPQ. INTERPRETATION: Treatment of intractable pain with NP2 was well tolerated. There were no placebo controls in this relatively small study, but the dose-responsive analgesic effects suggest that NP2 may be effective in reducing pain and warrants further clinical investigation.


Assuntos
Encefalinas/genética , Encefalinas/uso terapêutico , Terapia Genética/métodos , Manejo da Dor , Precursores de Proteínas/genética , Precursores de Proteínas/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Analgésicos Opioides/uso terapêutico , Relação Dose-Resposta a Droga , Encefalinas/metabolismo , Feminino , Vetores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade , Morfina/uso terapêutico , Estudos Multicêntricos como Assunto , Neoplasias/fisiopatologia , Medição da Dor , Precursores de Proteínas/metabolismo , Inquéritos e Questionários
2.
J Virol ; 84(14): 7360-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20463073

RESUMO

Viral vector-based gene expression libraries from normal or diseased tissues offer opportunities to interrogate cellular functions that influence or participate directly in specific biological processes. Here we report the creation and characterization of a herpes simplex virus (HSV)-based expression library consisting of cDNAs derived from PC12 pheochromocytoma cells. A replication-defective HSV vector backbone was engineered to contain both a bacterial artificial chromosome (BAC) and the Invitrogen in vitro Gateway recombination system, creating DBAC-GW. A cDNA library was produced and transferred into the DBAC-GW genome by in vitro recombination and selection in bacteria to produce DBAC-L. DBAC-L contained at least 15,000 unique cDNAs, as shown by DNA array analysis of PCR-amplified cDNA inserts, representing a wide range of cancer- and neuron-related cellular functions. Transfection of the recombinant DBAC-L DNA into complementing animal cells produced more than 1 million DBAC-L virus particles representing the library genes. By microarray analysis of vector-infected cells, we observed that individual members of this vector population expressed unique PC12 cDNA-derived mRNA, demonstrating the power of this system to transfer and express a variety of gene activities. We discuss the potential utility of this and similarly derived expression libraries for genome-wide approaches to identify cellular functions that participate in complex host-pathogen interactions or processes related to disease and to cell growth and development.


Assuntos
Expressão Gênica , Biblioteca Gênica , Vetores Genéticos , Simplexvirus/genética , Animais , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Células PC12 , Ratos
3.
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
4.
Pain Med ; 10(7): 1325-30, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19818042

RESUMO

The first human trial of gene therapy for chronic pain, a phase 1 study of a nonreplicating herpes simplex virus (HSV)-based vector engineered to express preproenkephalin in patients with intractable pain from cancer, began enrolling subjects in December 2008. In this article, we describe the rationale underlying this potential approach to treatment of pain, the preclinical animal data in support of this approach, the design of the study, and studies with additional HSV-based vectors that may be used to develop treatment for other types of pain.


Assuntos
Ensaios Clínicos como Assunto , Terapia Genética/métodos , Terapia Genética/tendências , Manejo da Dor , Dor/genética , Doença Crônica , Humanos , Resultado do Tratamento
5.
Eur J Neurosci ; 28(7): 1241-54, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18973552

RESUMO

Phosphorylation of the vanilloid receptor (TRPV1) by protein kinase C epsilon (PKCepsilon) plays an important role in the development of chronic pain. Here, we employ a highly defective herpes simplex virus vector (vHDNP) that expresses dominant negative PKCepsilon (DNPKCepsilon) as a strategy to demonstrate that PKCepsilon is essential for: (i) maintenance of basal phosphorylation and normal TRPV1 responses to capsaicin (CAPS), a TRPV1 agonist and (ii) enhancement of TRPV1 responses by phorbol esters. Phorbol esters induced translocation of endogenous PKCepsilon to the plasma membrane and thereby enhanced CAPS currents. These results were extended to an in-vivo pain model in which vHDNP delivery to dorsal root ganglion neurons caused analgesia in CAPS-treated, acutely inflamed rat hind paws. These findings support the conclusion that in addition to receptor sensitization, PKCepsilon is essential for normal TRPV1 responses in vitro and in vivo.


Assuntos
Gânglios Espinais/metabolismo , Nociceptores/metabolismo , Proteína Quinase C-épsilon/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Capsaicina/farmacologia , Carcinógenos/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Modelos Animais de Doenças , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Vetores Genéticos/genética , Humanos , Nociceptores/efeitos dos fármacos , Dor/induzido quimicamente , Dor/metabolismo , Dor/fisiopatologia , Técnicas de Patch-Clamp , Ésteres de Forbol/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/genética , Ratos , Células Receptoras Sensoriais/efeitos dos fármacos , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/agonistas
6.
Mol Biol Cell ; 16(8): 3727-39, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15930127

RESUMO

Volatile anesthetics including isoflurane affect all cells examined, but their mechanisms of action remain unknown. To investigate the cellular basis of anesthetic action, we are studying Saccharomyces cerevisiae mutants altered in their response to anesthetics. The zzz3-1 mutation renders yeast isoflurane resistant and is an allele of GCN3. Gcn3p functions in the evolutionarily conserved general amino acid control (GCN) pathway that regulates protein synthesis and gene expression in response to nutrient availability through phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF2alpha). Hyperphosphorylation of eIF2alpha inhibits translation initiation during amino acid starvation. Isoflurane rapidly (in <15 min) inhibits yeast cell division and amino acid uptake. Unexpectedly, phosphorylation of eIF2alpha decreased dramatically upon initial exposure although hyperphosphorylation occurred later. Translation initiation was inhibited by isoflurane even when eIF2alpha phosphorylation decreased and this inhibition was GCN-independent. Maintenance of inhibition required GCN-dependent hyperphosphorylation of eIF2alpha. Thus, two nutrient-sensitive stages displaying unique features promote isoflurane-induced inhibition of translation initiation. The rapid phase is GCN-independent and apparently has not been recognized previously. The maintenance phase is GCN-dependent and requires inhibition of general translation imparted by enhanced eIF2alpha phosphorylation. Surprisingly, as shown here, the transcription activator Gcn4p does not affect anesthetic response.


Assuntos
Fator de Iniciação 2B em Eucariotos/metabolismo , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Isoflurano/farmacologia , Iniciação Traducional da Cadeia Peptídica/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica , Clonagem Molecular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Farmacorresistência Fúngica , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 2B em Eucariotos/genética , Mutação/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1 , Proteínas de Saccharomyces cerevisiae/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
J Neurosurg ; : 1-11, 2018 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-30544352

RESUMO

OBJECTIVEMalignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas that harbor a high potential for metastasis and have a devastating prognosis. Combination chemoradiation aids in tumor control and decreases tumor recurrence but causes deleterious side effects and does not extend long-term survival. An effective treatment with limited toxicity and enhanced efficacy is critical for patients suffering from MPNSTs.METHODSThe authors recently identified that interleukin-13 receptor alpha 2 (IL-13Rα2) is overexpressed on MPNSTs and could serve as a precision-based target for delivery of chemotherapeutic agents. In the work reported here, a recombinant fusion molecule consisting of a mutant human IL-13 targeting moiety and a point mutant variant of Pseudomonas exotoxin A (IL-13.E13 K-PE4E) was utilized to treat MPNST in vitro in cell culture and in an in vivo murine model.RESULTSIL-13.E13 K-PE4E had a potent cytotoxic effect on MPNST cells in vitro. Furthermore, intratumoral administration of IL-13.E13 K-PE4E to orthotopically implanted MPNSTs decreased tumor burden 6-fold and 11-fold in late-stage and early-stage MPNST models, respectively. IL-13.E13 K-PE4E treatment also increased survival by 23 days in the early-stage MPNST model.CONCLUSIONSThe current MPNST treatment paradigm consists of 3 prongs: surgery, chemotherapy, and radiation, none of which, either singly or in combination, are curative or extend survival to a clinically meaningful degree. The results presented here provide the possibility of intratumoral therapy with a potent and highly tumor-specific cytotoxin as a fourth treatment prong with the potential to yield improved outcomes in patients with MPNSTs.

8.
Hum Gene Ther ; 17(10): 1006-18, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16989604

RESUMO

The use of nonviral delivery systems results in transient gene expression, in part because of the low efficiency of DNA integration. Previously, vectors based on transposon systems such as Sleeping Beauty have been shown to be able to increase stable transfection efficiencies in cell culture and in animal models. Himar1, a reconstructed active transposon belonging to the Tc1/mariner superfamily, also has been used as a vector for stable gene delivery, but the rate of transposition after transfection is low. In this paper, we evaluate the potential of the hyperactive Himar1 transposase C9, in combination with the Himar1 inverted repeat transposon, as a gene delivery vector. The C9 transposase is a hyperactive mutant of Himar1 with two amino acid substitutions, Q131R and E137K, that result in an increase in activity relative to wild type. Here we demonstrate that cotransfection of the C9 transposase with a Himar1-based vector increases the frequency of stable gene expression in human cells in a transposase concentration-dependent manner. In addition, we establish that C9 transposase mediates integration of the transgene in mammalian cells at a frequency similar to that of Sleeping Beauty under some of the conditions tested. Last, we show significantly higher levels of reporter gene expression in vivo in mouse liver and in synovium of rabbit knee joints after injection of the transposon plasmid expressing the transgene and the C9 transposase. These data suggest that vectors based on the Himar1 transposable element, in conjunction with the hyperactive mutant transposase C9, may be suitable vectors for gene therapy applications.


Assuntos
Elementos de DNA Transponíveis , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Transposases/genética , Animais , Sequência de Bases , Técnicas de Cultura de Células , Células HeLa , Humanos , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Articulação do Joelho/metabolismo , Fígado/metabolismo , Camundongos , Dados de Sequência Molecular , Coelhos , Transfecção , Transposases/metabolismo
9.
Cancer Gene Ther ; 12(5): 487-96, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15692608

RESUMO

To improve the effectiveness of herpes simplex virus (HSV) thymidine kinase/ganciclovir (HSV-tk/GCV) suicide gene therapy, the replication-defective HSV vector TOIkappaB expressing both HSV-TK and a mutant form of the NF-kappaB inhibitor IkappaBalpha (IkappaBalphaM) was developed. TOIkappaB was constructed by recombining the IkappaBalphaM gene into the U(L)41 locus of a replication-defective lacZ expression vector, TOZ.1. Expression of IkappaBalphaM was confirmed by Western blotting, and the ability of the mutant protein to inhibit NF-kappaB nuclear translocation was examined by electrophoretic mobility shift assay. In human glioblastoma U-87MG cells, the p50/p50 dimer of NF-kappaB was already translocated to the nucleus without receptor-dependent signaling by TNF-alpha. Following infection with TOIkappaB, nuclear translocation of NF-kappaB in U-87MG cells was significantly inhibited and caspase-3 activity increased compared with TOZ.1-infected cells. The cytotoxicity of TOIkappaB for U-87MG cells was investigated by colorimetric MTT assay. At an MOI of 3, TOIkappaB infection killed 85% of the cells compared to 20% killed by TOZ.1 infection. In the presence of GCV, these numbers increased to 95-100% for TOIkappaB and 80-85% for TOZ.1. TOIkappaB neurotoxicity measured on cultured murine neurons was relatively low and similar to that of TOZ.1. The survival of nude mice implanted into the brain with U-87MG tumor cells was markedly prolonged by intratumoral TOIkappaB injection and GCV administration. Survival of TOIkappaB+GCV group was significantly longer (P<.02, Wilcoxon test) than for the control groups (TOZ.1 or TOIkappaB only, PBS or PBS+GCV). These results suggest that IkappaBalphaM expression may be a safe enhancement of replication-defective HSV-based suicide gene therapy in vitro and in vivo.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Glioblastoma/terapia , Herpesvirus Humano 1/genética , Proteínas I-kappa B/genética , Timidina Quinase/genética , Animais , Apoptose , Encéfalo/citologia , Caspase 3 , Caspases/metabolismo , Terapia Combinada , Feminino , Genes Transgênicos Suicidas , Vetores Genéticos , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Camundongos , Mutação , Inibidor de NF-kappaB alfa , Plasmídeos/genética , Timidina Quinase/metabolismo , Células Tumorais Cultivadas
10.
Genetics ; 161(2): 563-74, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12072454

RESUMO

Volatile anesthetics affect all cells and tissues tested, but their mechanisms and sites of action remain unknown. To gain insight into the cellular activities of anesthetics, we have isolated genes that, when overexpressed, render Saccharomyces cerevisiae resistant to the volatile anesthetic isoflurane. One of these genes, WAK3/TAT1, encodes a permease that transports amino acids including leucine and tryptophan, for which our wild-type strain is auxotrophic. This suggests that availability of amino acids may play a key role in anesthetic response. Multiple lines of evidence support this proposal: (i) Deletion or overexpression of permeases that transport leucine and/or tryptophan alters anesthetic response; (ii) prototrophic strains are anesthetic resistant; (iii) altered concentrations of leucine and tryptophan in the medium affect anesthetic response; and (iv) uptake of leucine and tryptophan is inhibited during anesthetic exposure. Not all amino acids are critical for this response since we find that overexpression of the lysine permease does not affect anesthetic sensitivity. These findings are consistent with models in which anesthetics have a physiologically important effect on availability of specific amino acids by altering function of their permeases. In addition, we show that there is a relationship between nutrient availability and ubiquitin metabolism in this response.


Assuntos
Sistemas de Transporte de Aminoácidos/fisiologia , Anestésicos Inalatórios/farmacologia , Isoflurano/farmacologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Farmacorresistência Fúngica/genética , Leucina/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Análise de Sequência de DNA , Triptofano/metabolismo , Ubiquitina/metabolismo
11.
J Neurotrauma ; 19(1): 61-8, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11852979

RESUMO

Proximal spinal nerve injury results in the death of motor neurons in ventral horn. We have previously demonstrated this cell death can be prevented by HSV-mediated transfer of the gene coding for the antiapoptotic peptide Bcl-2 7 days prior to injury, but that expression of Bcl-2 does not preserve ChAT expression in the lesioned cells. In the current study, we examined two related issues: whether Bcl-2 delivered by HSV-mediated gene transfer 30 min after injury could similarly protect motor neurons from cell death, and whether the additional HSV-mediated expression of the glial cell derived neurotrophic factor (GDNF) could improve the result. At 30 min after avulsion of the L4, L5, and L6 spinal nerves, replication defective genomic HSV-based vectors coding for Bcl-2, GDNF, a reporter transgene (lacZ), or the Bcl-2 and GDNF vectors together were injected into spinal cord. Transduction of motor neurons with either the Bcl-2-expressing vector or the GDNF-expressing vector resulted in a substantial increase in the number of surviving motor neurons, and coinjection of the two vectors together resulted in cell survival that was similar to the result obtained with either vector alone. Neither the Bcl-2-expressing vector nor the GDNF-expressing vector delivered alone protected choline acetyltransferase (ChAT) expression in lesioned neurons. However, simultaneous injection of the Bcl-2- and the GDNF-expressing vectors together resulted in a substantial increase in the number of ChAT in cells in the lesioned ventral horn. Together, these findings suggest an approach to improving cell survival and regeneration following proximal root injury.


Assuntos
Terapia Genética , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Radiculopatia/terapia , Raízes Nervosas Espinhais/lesões , Animais , Morte Celular , Sobrevivência Celular , Colina O-Acetiltransferase/genética , Regulação Viral da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Masculino , Neurônios Motores , Radiculopatia/patologia , Ratos , Ratos Sprague-Dawley , Simplexvirus/genética , Raízes Nervosas Espinhais/patologia , Transgenes
12.
J Gastrointest Surg ; 7(2): 229-35; discussion 235-6, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12600447

RESUMO

Small bowel toxicity represents a major dose-limiting side effect of radiation treatment for many malignancies. We examined the effects of overexpressing human manganese superoxide dismutase (MnSOD) in the small intestine in mice to prevent radiation enteritis. Mice were treated with the human MnSOD gene delivered enterally using a nontoxic, replication-defective herpes simplex virus (HSV)-1-based vector. HSV vectors containing the human MnSOD transgene and green fluorescent protein (GFP) transgene, or GFP transgene alone, were constructed and injected intraluminally into a 2cm length of small intestine of C3H/HeNsd mice. Total body irradiation of 15 Gy was delivered to mice inoculated 24 hours earlier with either HSV-MnSOD (10(3) to 10(8) plaque-forming units), control HSV-GFP, or no vector. At 24 or 72 hours after irradiation, mice were killed and villi areas were measured from appropriate segments of the small intestine. Control irradiated mice showed a decreased villi area of 82% by day 3 after irradiation, whereas treatment of mice with HSV-MnSOD 10(8) plaque-forming units led to only a 16% decrease in villi area (P < 0.001) before radiation. Similar findings were seen on day 3 and were associated with a significant (P < 0.001) preservation of enteric protein content in HSV-MnSOD-treated mice. A dose-dependent effect of MnSOD in preventing radiation-induced small bowel injury was evident. These data demonstrate that overexpression of human MnSOD via a replication-defective herpes viral vector is an efficacious method of protecting the small intestine from ionizing radiation damage.


Assuntos
Terapia Genética/métodos , Intestino Delgado/patologia , Intestino Delgado/efeitos da radiação , Lesões por Radiação/prevenção & controle , Superóxido Dismutase/genética , Análise de Variância , Animais , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Feminino , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Camundongos , Camundongos Endogâmicos C3H , Probabilidade , Doses de Radiação , Lesões Experimentais por Radiação , Valores de Referência , Sensibilidade e Especificidade , Superóxido Dismutase/farmacologia , Transgenes
13.
Methods Mol Biol ; 246: 309-22, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14970601

RESUMO

Gene transfer to the nervous system is an attractive option to treat a wide variety of neurological insults. The expression of trophic factor and/or antiapoptotic genes may be beneficial in halting the slow neurodegeneration in such conditions as Parkinson's disease (4,5), the rapid neuronal cell death following trauma to the brain or spinal cord (6,7), or in treating peripheral neuropathies associated with diabetes or use of chemotherapeutic agents (8,9). Introduction of dominant-negative mutant genes or antisense RNA to treat diseases such as Huntington's disease, or transfer of genes to replace lost or mutated endogenous proteins to treat disorders such as lysosomal storage diseases, may prove useful. In addition, gene transfer to overexpress endogenous antinociceptive proteins has great potential in pain management. The problem faced by all of these applications is finding a suitable methodology that will facilitate the transfer of exogenous genes to the appropriate nerve cells; virusbased vectors have proven quite efficient in transferring genes to many different cell types.


Assuntos
Sistema Nervoso Central/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Simplexvirus/genética , Animais , Neurônios/virologia , Células PC12 , Ratos , Simplexvirus/isolamento & purificação
14.
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
15.
Methods Mol Biol ; 246: 323-37, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14970602

RESUMO

Glial tumors occur as intraaxial masses in the brain and are uniformly fatal due to lack of effective therapy. Resection combined with radiation and chemotherapy fails to eradicate malignant cells infiltrating into normal brain, and recurrence at the original site is ultimately fatal. Gene transfer offers the potential to enhance tumor cell killing while sparing surrounding normal brain. Several approaches have been developed to deliver genes to tumor cells in order to kill these cells. The first strategy involves the use of viral vectors that are replication-competent, but depend on attributes unique to the tumor cell to support viral growth. Both replication-competent adenovirus and herpes simplex virus (HSV) vectors have been employed in pre-clinical studies and most recently in human clinical trials. For this purpose, HSV vectors have been engineered that replicate in dividing cells, such as tumor cells, but not in normal neurons. The use of conditional replication competent viruses could allow for their spread in tumor tissue while minimizing damage to normal brain, thus increasing the specificity and effectiveness. Such mutants include those lacking the viral thymidine kinase (tk) gene (4-7), ribonucleotide reductase gene (8,9), a protein kinase gene, or a gene (gamma34.5) required for growth specifically in neurons (11-13).


Assuntos
Neoplasias Encefálicas/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Glioma/terapia , Simplexvirus/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/cirurgia , Terapia Combinada , Glioma/genética , Glioma/cirurgia , Injeções Intralesionais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Radiocirurgia , Ratos , Ratos Endogâmicos F344 , Timidina Quinase/genética , Fator de Necrose Tumoral alfa/genética
16.
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
17.
Biotechnol Prog ; 18(3): 476-82, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12052062

RESUMO

Herpes simplex virus type-1 (HSV-1) is a neurotrophic human pathogen that establishes life-long latency in the nervous system. Our laboratory has extensively engineered this virus to retain the ability to persist in neurons without expression of lytic genes or disease phenotype. Highly defective, replication-incompetent HSV mutants are thus potentially ideal for transfer of therapeutic transgenes to human nerves where long-term therapy of nervous system disease may be provided. A prerequisite for using recombinant HSV vectors for therapeutic gene delivery to humans is the development of methods for large-scale manufacture of HSV vectors. Here we report studies to identify infection parameters that result in high-yield production of immediate early gene deletion mutant HSV vectors in complementing cells that supply the deleted essential viral functions in trans. Virus yield was correlated with various culture media conditions that included pH, glucose metabolism, and serum levels. The results demonstrated that systematic media exchange to remove lactate derived from high-level glucose consumption, maintenance of tissue culture pH at 6.8, and the use of 5% fetal bovine serum gave the highest yield of infectious virus. The data indicate that these are important parameters to consider for high-yield, large-scale virus production.


Assuntos
Vírus Defeituosos/genética , Vetores Genéticos , Herpesvirus Humano 1/genética , Animais , Chlorocebus aethiops , Vírus Defeituosos/crescimento & desenvolvimento , Herpesvirus Humano 1/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Células Vero , Ensaio de Placa Viral
18.
J Pain Res ; 7: 71-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24470772

RESUMO

Chronic pain represents a major medical burden not only in terms of suffering but also in terms of economic costs. Traditional medical approaches have so far proven insufficient in treating chronic pain and new approaches are necessary. Gene therapy with herpes simplex virus (HSV)-based vectors offers the ability to directly target specific regions of the neuraxis involved in pain transmission including the primary afferent nociceptor. This opens up new targets to interact with that are either not available to traditional systemic drugs or cannot be adequately acted upon without substantial adverse off-target effects. Having access to the entire neuron, which HSV-based vector gene therapy enables, expands treatment options beyond merely treating symptoms and allows for altering the basic biology of the nerve. In this paper, we discuss several HSV-based gene therapy vectors that our group and others have used to target specific neuronal functions involved in the processing of nociception in order to develop new therapies for the treatment of chronic pain.

19.
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
20.
Neurosci Lett ; 527(2): 85-9, 2012 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-22565023

RESUMO

Gene transfer to target delivery of neurotrophic factors to the primary sensory afferent for treatment of polyneuropathy, or of inhibitory neurotransmitters for relief of chronic pain, offers the possibility of a highly selective targeted release of bioactive molecules within the nervous system. Preclinical studies with non-replicating herpes simplex virus (HSV)-based vectors injected into the skin to transduce neurons in the dorsal root ganglion have demonstrated efficacy in reducing-pain related behaviors in animal models of inflammatory pain, neuropathic pain, and pain caused by cancer, and in preventing progression of sensory neuropathy caused by toxins, chemotherapeutic drugs or resulting from diabetes. Successful completion of the first phase 1 clinical trial of HSV-mediated gene transfer in patients with intractable pain from cancer has set the stage for further clinical trials of this approach.


Assuntos
Terapia Genética , Sistema Nervoso Periférico/metabolismo , Animais , Dor Crônica/genética , Dor Crônica/metabolismo , Dor Crônica/terapia , Ensaios Clínicos como Assunto , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/terapia , Encefalinas/genética , Encefalinas/metabolismo , Vetores Genéticos , Herpesviridae/genética , Humanos , Neoplasias/complicações , Neoplasias/fisiopatologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Neurotransmissores/genética , Neurotransmissores/metabolismo , Dor Intratável/etiologia , Dor Intratável/genética , Dor Intratável/metabolismo , Dor Intratável/terapia , Polineuropatias/genética , Polineuropatias/metabolismo , Polineuropatias/terapia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa