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










Base de datos
Intervalo de año de publicación
1.
J Gene Ther ; 3(1)2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30465046

RESUMEN

BACKGROUND: Recognition sequences for microRNAs (miRs) that are down-regulated in tumor cells have recently been used to render lytic viruses tumor-specific. Since different tumor types down-regulate different miRs, this strategy requires virus customization to the target tumor. We have explored a feature that is shared by many tumor types, the up-regulation of miR-21, as a means to generate an oncolytic herpes simplex virus (HSV) that is applicable to a broad range of cancers. METHODS: We assembled an expression construct for a dominant-negative (dn) form of the essential HSV replication factor UL9 and inserted tandem copies of the miR-21 recognition sequence (T21) in the 3' untranslated region. Bacterial Artificial Chromosome (BAC) recombineering was used to introduce the dnUL9 construct with or without T21 into the HSV genome. Virus was produced by transfection and replication was assessed in different tumor and control cell lines. RESULTS: Virus production was conditional on the presence of the T21 sequence. The dnUL9-T21 virus replicated efficiently in tumor cell lines, less efficiently in cells that contained reduced miR-21 activity, and not at all in the absence of miR-21. CONCLUSION: miR-21-sensitive expression of a dominant-negative inhibitor of HSV replication allows preferential destruction of tumor cells in vitro. This observation provides a basis for further development of a widely applicable oncolytic HSV.

2.
Gene Ther ; 25(1): 20-26, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29057994

RESUMEN

Enhanced afferent excitability is considered to be an important pathophysiological basis of interstitial cystitis/bladder pain syndrome (IC/BPS). In addition, transient receptor potential vanilloid-1 (TRPV1) receptors are known to be involved in afferent sensitization. Animals with hydrogen peroxide (HP)-induced cystitis have been used as a model exhibiting pathologic characteristics of chronic inflammatory condition of the bladder. This study investigated the effect of gene therapy with replication-defective herpes simplex virus (HSV) vectors encoding poreless TRPV1 (PL) or protein phosphatase 1 α (PP1α), a negative regulator of TRPV1, using a HP-induced rat model of cystitis. HSV vectors encoding green fluorescent protein, PL or PP1α were inoculated into the bladder wall of female rats. After 1 week, 1% HP or normal saline was administered into the bladder, and the evaluations were performed 2 weeks after viral inoculation. In HP-induced cystitis rats, gene delivery of PL or PP1α decreased pain behavior as well as a reduction in the intercontraction interval. Also, both treatments reduced nerve growth factor expression in the bladder mucosa, reduced bladder inflammation characterized by infiltration of inflammatory cells and increased bladder weight. Taken together, HSV-mediated gene therapy targeting TRPV1 receptors could be effective for the treatment of IC/BPS.


Asunto(s)
Cistitis/inducido químicamente , Cistitis/terapia , Terapia Genética/métodos , Vectores Genéticos , Peróxido de Hidrógeno/toxicidad , Proteína Fosfatasa 1/genética , Simplexvirus/genética , Canales Catiónicos TRPV/genética , Animales , Cistitis/enzimología , Cistitis/metabolismo , Virus Defectuosos/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Tamaño de los Órganos , Ratas , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología
3.
Gene Ther ; 24(5): 314-324, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28368370

RESUMEN

Morphine appears to be the most active metabolite of heroin; therefore, the effects of morphine are important in understanding the ramifications of heroin abuse. Opioid physical dependence (withdrawal response) may have very long-lasting effects on the motivation for reward, including the incubation of cue-induced drug-seeking behavior. However, the exact mechanisms of morphine withdrawal (MW) are not clear yet, and its treatment remains elusive. Periaqueductal gray (PAG) is one of the important sites in the pathogenesis of MW. Here, we used recombinant herpes simplex virus (HSV) vectors that encode the sod2 gene expressing manganese superoxide dismutase (MnSOD) to evaluate its therapeutic potential in MW. Microinjection of HSV vectors expressing MnSOD into the PAG reduced the MW syndrome. MnSOD vectors suppressed the upregulated mitochondrial superoxide, and endoplasmic reticulum stress markers (glucose-related protein 78 (GRP78) and activating transcription factor 6 alpha (ATF6α)) in the PAG induced by MW. Immunostaining showed that mitochondrial superoxide, GRP78 and ATF6α were colocalized with neuronal nuclei (a neuronal-specific marker), suggesting that they are located in the neurons in the PAG. These results suggest that overexpression of MnSOD by HSV vectors may relieve opioid dependence. This study may provide a novel therapeutic approach to morphine physical withdrawal response.


Asunto(s)
Terapia Genética , Morfina/efectos adversos , Sustancia Gris Periacueductal/metabolismo , Simplexvirus/genética , Síndrome de Abstinencia a Sustancias/terapia , Superóxido Dismutasa/genética , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Vectores Genéticos/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo
4.
Gene Ther ; 21(7): 694-702, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24830437

RESUMEN

Acute and chronic pain (post-herpetic neuralgia or PHN) are encountered in patients with herpes zoster that is caused by reactivation of varicella-zoster virus (VZV) from a state of neuronal latency. PHN is often refractory to current treatments, and additional strategies for pain relief are needed. Here we exploited a rat footpad model of PHN to show that herpes simplex virus (HSV) vector-mediated gene delivery of human preproenkephalin (vHPPE) effectively reduced chronic VZV-induced nocifensive indicators of pain. VZV inoculated at the footpad induced prolonged mechanical allodynia and thermal hyperalgesia that did not develop in controls or with ultraviolet light-inactivated VZV. Subsequent footpad administration of vHPPE relieved VZV-induced pain behaviors in a dose-dependent manner for extended periods, and prophylactic vector administration prevented VZV-induced pain from developing. Short-term pain relief following low-dose vHPPE administration could be effectively prolonged by vector re-administration. HPPE transcripts were increased three- to fivefold in ipsilateral ganglia, but not in the contralateral dorsal root ganglia. VZV hypersensitivity and its relief by vHPPE were not affected by peripheral delivery of opioid receptor agonist or antagonist, suggesting that the efficacy was mediated at the ganglion and/or spinal cord level. These results support further development of ganglionic expression of enkephalin as a novel treatment for the pain associated with Zoster.


Asunto(s)
Encefalinas/metabolismo , Ganglión/metabolismo , Vectores Genéticos/administración & dosificación , Neuralgia Posherpética/prevención & control , Neuralgia Posherpética/terapia , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Encefalinas/genética , Pie/virología , Terapia Genética , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Simplexvirus/genética , Médula Espinal/metabolismo
5.
Gene Ther ; 20(6): 589-96, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22996196

RESUMEN

The development of effective strategies for gene therapy has been hampered by difficulties verifying transgene delivery in vivo and quantifying gene expression non-invasively. Magnetic resonance imaging (MRI) offers high spatial resolution and three-dimensional views, without tissue depth limitations. The iron-storage protein ferritin is a prototype MRI gene reporter. Ferritin forms a paramagnetic ferrihydrite core that can be detected by MRI via its effect on the local magnetic field experienced by water protons. In an effort to better characterize the ferritin reporter for central nervous system applications, we expressed ferritin in the mouse brain in vivo using a neurotropic herpes simplex virus type 1 (HSV-1). We computed three-dimensional maps of MRI transverse relaxation rates in the mouse brain with ascending doses of ferritin-expressing HSV-1. We established that the transverse relaxation rates correlate significantly to the number of inoculated infectious particles. Our results are potentially useful for quantitatively assessing limitations of ferritin reporters for gene therapy applications.


Asunto(s)
Encéfalo/diagnóstico por imagen , Ferritinas/aislamiento & purificación , Terapia Genética , Herpesvirus Humano 1/aislamiento & purificación , Imagen por Resonancia Magnética/métodos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Ferritinas/química , Ferritinas/uso terapéutico , Expresión Génica , Regulación Viral de la Expresión Génica , Genes Reporteros , Herpesvirus Humano 1/genética , Ratones , Radiografía
6.
Gene Ther ; 20(2): 194-200, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22402319

RESUMEN

We investigated the effects of replication-defective herpes simplex virus (HSV) vector expression of interleukin-4 (IL-4) on bladder overactivity and nociception. HSV vector expressing murine interleukin-4 (S4IL4) or the control vector expressing ß-galactosidase (SHZ) were injected to the rat bladder wall. At 1 week after viral injection, in cystometry performed under urethane anesthesia, the S4IL4-treated group did not show the intercontraction intervals reduction during intravesical administration of 10 nM resiniferatoxin (RTx). At 2 weeks after viral injection, behavioral studies were performed on vector-injected animals in an awakened state. Freezing behavior induced by 3 µM RTx, administered for 1 min into the bladder, was significantly suppressed in the S4IL4 group compared with the SHZ group. Murine IL-4 levels examined by ELISA were significantly increased in bladder and bladder afferent dorsal root ganglia at 2 weeks after viral injection. The expression of IL-1ß and IL-2 and bladder inflammatory responses were significantly suppressed in the RTx-irritated bladder of S4IL4-injected rats. These results indicate that HSV vector-mediated interleukin-4 expression in the bladder and bladder afferent pathways reduces the inflammatory response, bladder overactivity and nociceptive behavior induced by bladder irritation in the rat model. Therefore, IL-4 gene therapy could be a new strategy for treating urinary frequency and/or bladder pain.


Asunto(s)
Terapia Genética , Interleucina-4/genética , Nocicepción , Simplexvirus/genética , Vejiga Urinaria Hiperactiva/terapia , Animales , Diterpenos/farmacología , Femenino , Reacción Cataléptica de Congelación , Ganglios Espinales/metabolismo , Expresión Génica , Vectores Genéticos , Inflamación/terapia , Interleucina-4/metabolismo , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Vejiga Urinaria Hiperactiva/fisiopatología
7.
Gene Ther ; 20(7): 761-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23254370

RESUMEN

Epstein-Barr virus (EBV)-associated B-cell lymphoproliferative disease (LPD) after hematopoietic stem cell or solid organ transplantation remains a life-threatening complication. Expression of the virus-encoded gene product, EBER, has been shown to prevent apoptosis via blockade of PKR activation. As PKR is a major cellular defense against Herpes simplex virus (HSV), and oncolytic HSV-1 (oHSV) mutants have shown promising antitumor efficacy in preclinical models, we sought to determine whether EBV-LPD cells are susceptible to infection by oHSVs. We tested three primary EBV-infected lymphocyte cell cultures from neuroblastoma (NB) patients as models of naturally acquired EBV-LPD. NB12 was the most susceptible, NB122R was intermediate and NB88R2 was essentially resistant. Despite EBER expression, PKR was activated by oHSV infection. Susceptibility to oHSV correlated with the expression of the HSV receptor, nectin-1. The resistance of NB88R2 was reversed by exogenous nectin-1 expression, whereas downregulation of nectin-1 on NB12 decreased viral entry. Xenografts derived from the EBV-LPDs exhibited only mild (NB12) or no (NB88R2) response to oHSV injection, compared with a NB cell line that showed a significant response. We conclude that EBV-LPDs are relatively resistant to oHSV virotherapy, in some cases, due to low virus receptor expression but also due to intact antiviral PKR signaling.


Asunto(s)
Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Trastornos Linfoproliferativos/genética , Virus Oncolíticos/genética , Apoptosis/genética , Moléculas de Adhesión Celular/metabolismo , ADN Viral/genética , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 4/inmunología , Humanos , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Nectinas , Viroterapia Oncolítica , Cultivo Primario de Células , Receptores Virales/genética
8.
Prilozi ; 31(2): 151-5, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21258284

RESUMEN

Imaging gene expression non-invasively and deep into opaque tissues has been a long-standing goal of molecular science. Optical gene reporters such as green fluorescent protein and luciferase have revolutionized cellular and molecular biology, however their in vivo application is limited, due to poor tissue penetration of visible light. The iron storage protein ferritin forms a paramagnetic ferrihydrite core that affects the relaxation rate of surrounding nuclear spins. Ferritin has recently emerged as an MRI gene reporter for molecular applications, however its detection with MRI still has relatively low sensitivity. In this work we present an improved ferritin chimera, genetically engineered to exhibit stronger paramagnetic properties.


Asunto(s)
Ferritinas/metabolismo , Genes Reporteros , Imagen Molecular/métodos , Ferritinas/genética , Ferritinas/aislamiento & purificación , Colorantes Fluorescentes , Proteínas Fluorescentes Verdes , Humanos , Aumento de la Imagen/métodos , Imagen por Resonancia Magnética/métodos , Microscopía Fluorescente
9.
Gene Ther ; 16(5): 660-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19225548

RESUMEN

We examined whether replication-defective herpes simplex virus (HSV) vectors encoding the 67 kDa form of the glutamic acid decarboxylase (GAD(67)) gene product, the gamma-aminobutyric acid (GABA) synthesis enzyme, can suppress detrusor overactivity (DO) in rats with spinal cord injury (SCI). One week after spinalization, HSV vectors expressing GAD and green fluorescent protein (GFP) (HSV-GAD) were injected into the bladder wall. Rats with SCI without HSV injection (HSV-untreated) and those injected with lacZ-encoding reporter gene HSV vectors (HSV-LacZ) were used as controls. Three weeks after viral injection, continuous cystometry was performed under awake conditions in all three groups. In the HSV-GAD group, the number and amplitude of non-voiding contractions (NVCs) were significantly decreased (40-45% and 38-40%, respectively) along with an increase in voiding efficiency, compared with HSV-untreated and HSV-LacZ groups, but micturition pressure was not different among the three groups. Intrathecal application of bicuculline partly reversed the decreased number and amplitude of NVCs, and decreased voiding efficiency in the HSV-GAD group. In the HSV-GAD group, GAD(67) mRNA and protein levels were significantly increased in the L6-S1 dorsal root ganglia (DRG) compared with the HSV-LacZ group, while 57% of DRG cells were GFP-positive, and these neurons showed increased GAD(67)-like immunoreactivity compared with the HSV-LacZ group. These results indicate that GAD gene therapy effectively suppresses DO after SCI predominantly through the activation of spinal GABA(A) receptors. Thus, HSV-based GAD gene transfer to bladder afferent pathways may represent a novel approach for treatment of neurogenic DO.


Asunto(s)
Terapia Genética/métodos , Glutamato Descarboxilasa/genética , Simplexvirus/genética , Traumatismos de la Médula Espinal/complicaciones , Vejiga Urinaria Hiperactiva/terapia , Animales , Estudios de Factibilidad , Femenino , Expresión Génica/genética , Vectores Genéticos , Glutamato Descarboxilasa/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Transgenes , Vejiga Urinaria/fisiopatología , Vejiga Urinaria Hiperactiva/etiología , Vejiga Urinaria Hiperactiva/fisiopatología
10.
Gene Ther ; 16(4): 558-69, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19242523

RESUMEN

Interstitial cystitis (IC)/painful bladder syndrome (PBS) is a painful debilitating chronic visceral pain disorder of unknown etiology that affects an estimated 1 million people in the United States alone. It is characterized by inflammation of the bladder that results in chronic pelvic pain associated with bladder symptoms of urinary frequency and urgency. Regardless of the etiology, IC/PBS involves either increased and/or abnormal activity in afferent nociceptive sensory neurons. Pain-related symptoms in patients with IC/PBS are often very difficult to treat. Both medical and surgical therapies have had limited clinical utility in this debilitating disease and numerous drug treatments, such as heparin, dimethylsulfoxide and amitriptyline, have proven to be palliative at best, and in some IC/PBS patients provide no relief whatsoever. Although opiate narcotics have been employed to help alleviate IC/PBS pain, this strategy is fraught with problems as systemic narcotic administration causes multiple unwanted side effects including mental status change and constipation. Moreover, chronic systemic narcotic use leads to dependency and need for dose escalation due to tolerance; therefore, new therapies are desperately needed to treat refractory IC/PBS. This has led our group to develop a gene therapy strategy that could potentially alleviate chronic pelvic pain using the herpes simplex virus-directed delivery of analgesic proteins to the bladder.


Asunto(s)
Cistitis Intersticial/terapia , Terapia Genética/métodos , Vectores Genéticos , Simplexvirus/genética , Cistitis Intersticial/fisiopatología , Técnicas de Transferencia de Gen , Humanos , Neuronas Aferentes/fisiología , Péptidos Opioides/fisiología , Vejiga Urinaria/inervación
11.
Gene Ther ; 14(18): 1344-52, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17611585

RESUMEN

Erectile dysfunction (ED) is frequently associated with injury to the cavernous nerve sustained during pelvic surgery. Functional recovery from cavernous nerve injury is generally incomplete and occurs over an extended time frame. We employed a therapeutic gene transfer approach with herpes simplex virus (HSV) vector expressing glial cell line-derived neurotrophic factor (GDNF). Rat cavernous nerve was injured bilaterally using a clamp and dry ice. For HSV-treated groups, 20 microl of purified vector stock was administered directly to and around the damaged nerve. Delivery of an HSV vector expressing both green fluorescent protein (GFP) and lacZ (HSV-LacZ) was used as a control. Intracavernous pressure along with systemic arterial pressure (ICP/AP) was measured 2 and 4 weeks after the nerve injury. Fluorogold (FG) was injected into the penile crus 7 days before killing to assess nerve survival. Approximately 60% of major pelvic ganglion (MPG) cells were GFP positive after viral administration. At 4 weeks after nerve injury, rats treated with HSV-GDNF exhibited significant recovery of ICP/AP compared with control vector or untreated groups. The HSV-GDNF group also yielded more FG-positive MPG cells than the control vector group. HSV vector-mediated delivery of GDNF presents a viable approach for the treatment of ED following cavernous nerve injury.


Asunto(s)
Disfunción Eréctil/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Simplexvirus/genética , Animales , Biomarcadores/análisis , Presión Sanguínea , Disfunción Eréctil/metabolismo , Ganglios Espinales/metabolismo , Ganglios Espinales/virología , Expresión Génica , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Inyecciones , Masculino , Modelos Animales , Óxido Nítrico Sintasa de Tipo I/análisis , Óxido Nítrico Sintasa de Tipo I/genética , Pene/lesiones , Pene/inervación , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factores de Tiempo
12.
Gene Ther ; 13(14): 1068-79, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16541122

RESUMEN

Accumulation of insoluble aggregates of amyloid-beta peptide (Abeta), a cleavage product of amyloid precursor protein (APP), is thought to be central to the pathogenesis of Alzheimer's disease (AD). Consequently, downregulation of APP, or enhanced clearance of Abeta, represent possible therapeutic strategies for AD. We generated replication-defective herpes simplex virus (HSV) vectors that inhibit Abeta accumulation, both in vitro and in vivo. In cell culture, HSV vectors expressing either (i) short hairpin RNA directed to the APP transcript (HSV-APP/shRNA), or (ii) neprilysin, an endopeptidase that degrades Abeta (HSV-neprilysin), substantially inhibited accumulation of Abeta. To determine whether these vectors showed similar activity in vivo, we developed a novel mouse model, in which overexpression of a mutant form of APP in the hippocampus, using a lentiviral vector (LV-APP(Sw)), resulted in rapid Abeta accumulation. Co-inoculation of LV-APP(Sw) with each of the HSV vectors showed that either HSV-APP/shRNA or HSV-neprilysin inhibited Abeta accumulation in this model, whereas an HSV control vector did not. These studies demonstrate the utility of HSV vectors for reducing Abeta accumulation in the brain, thus providing useful tools to clarify the role of Abeta in AD that may facilitate the development of novel therapies for this important disease.


Asunto(s)
Enfermedad de Alzheimer/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Neprilisina/genética , ARN Interferente Pequeño/genética , Simplexvirus/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Expresión Génica , Ingeniería Genética , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente
13.
Gene Ther ; 12(11): 891-901, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15908995

RESUMEN

Herpes simplex virus type 1 (HSV-1) is a neurotropic double-stranded DNA virus that causes cold sores, keratitis, and rarely encephalitis in humans. Nonpathogenic HSV-1 gene transfer vectors have been generated by elimination of viral functions necessary for replication. The life cycle of the native virus includes replication in epithelial cells at the site of initial inoculation followed by retrograde axonal transport to the nuclei of sensory neurons innervating the area of cutaneous primary infection. In this review, we summarize the current understanding of the molecular basis for HSV cell entry, nuclear transport of the genome, virion egress following replication, and retrograde and anterograde axonal transport in neurons. We discuss how each of these properties has been exploited or modified to allow the generation of gene transfer vectors with particular utility for neurological applications. Recent advances in engineering virus entry have provided proof of principle that vector targeting is possible. Furthermore, significant and potentially therapeutic modifications to the pathological responses to various noxious insults have been demonstrated in models of peripheral nerve disease. These applications exploit the natural axonal transport mechanism of HSV, allowing transgene expression in the cell nucleus within the inaccessible trigeminal ganglion or dorsal root ganglion, following the noninvasive procedure of subcutaneous vector inoculation. These findings demonstrate the importance of understanding basic virology in the design of vector systems and the powerful approach of exploiting favorable properties of the parent virus in the generation of gene transfer vectors.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/farmacocinética , Herpesvirus Humano 1/genética , Enfermedades del Sistema Nervioso/terapia , Transporte Activo de Núcleo Celular , Técnicas de Transferencia de Gen , Herpesvirus Humano 1/fisiología , Humanos , Neuronas/virología , Activación Viral
14.
Gene Ther ; 12(1): 48-58, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15470480

RESUMEN

The accessibility of adipose tissue and its ability to secrete various bioactive molecules suggest that adipose cells may be attractive targets for gene therapy applications. Here, we report the use of highly defective herpes simplex virus (HSV) vectors as suitable gene transfer agents for adipose cells in culture and fat tissue in animals. Using an in vitro model of human adipose differentiation, we first demonstrated that mature adipocytes and their precursor cells express the two principal HSV viral entry receptors HveA and HveC (nectin-1) and are efficiently transduced at a low multiplicity of infection by HSV-lacZ reporter gene and glial cell line-derived neurotrophic factor (GDNF) gene vectors. Extended expression of beta-galactosidase and secretion of GDNF occurred in transduced fat tissue explants from rabbits. In vivo gene transfer to rabbit subcutaneous adipose tissue resulted in local GDNF expression for at least 2 months. These experiments establish the efficient transduction of adipose cells by HSV vectors and suggest that fat tissue may represent a useful site for HSV-mediated gene delivery with potential for therapeutic applications.


Asunto(s)
Tejido Adiposo/metabolismo , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Factores de Crecimiento Nervioso/metabolismo , Simplexvirus/genética , Transducción Genética/métodos , Adipocitos/metabolismo , Animales , Células Cultivadas , Factor Neurotrófico Derivado de la Línea Celular Glial , Humanos , Conejos , Técnicas de Cultivo de Tejidos
15.
Adv Drug Deliv Rev ; 53(2): 155-70, 2001 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-11731025

RESUMEN

Herpes simplex virus (HSV) is an encapsulated DNA virus, with many favourable properties for use as a gene transfer vector. For gene therapy applications, it may be desirable to restrict transgene expression to pre-defined subsets of cells. One potential method for achieving targeted transgene expression using the HSV vector system might involve dictating the cell types to which the vector will transfer the therapeutic transgene of interest. HSV delivers its genetic payload to cells directly through the plasmalemma; the mechanisms are complex and involve multiple viral and cell surface determinants. We have investigated several ways in which each component of the cell entry cascade may be manipulated in order to restrict viral DNA and transgene delivery to particular cellular populations. Our results indicate that targeted transduction may be a viable approach to achieving our goal of targeted HSV-mediated transgene expression.


Asunto(s)
Marcación de Gen/métodos , Vectores Genéticos , Simplexvirus/genética , Animales , Humanos , Simplexvirus/ultraestructura
16.
Stem Cells ; 19(5): 358-77, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11553845

RESUMEN

Herpes simplex virus (HSV) is a neurotropic DNA virus. The viral genome is large (152 kb), and many genes are dispensable for viral function, allowing insertion of multiple or large transgene expression cassettes. The virus life cycle includes a latent phase, during which the viral genome remains as a stable episomal element within neuronal nuclei for the lifetime of the host, without disturbing normal function. We have exploited these features of HSV to construct a series of nonpathogenic gene therapy vectors that efficiently deliver therapeutic and experimental transgenes to neural and non-neural tissue. Importantly, transgene expression may be sustained long term; reporter gene expression has been demonstrated for over a year in the nervous system. This article discusses the generation of replication-defective HSV vectors and reviews recent studies investigating their use in several animal models of human disease. We have demonstrated correction or prevention of a number of important neurological phenotypes, including neurodegeneration, chronic pain, peripheral neuropathy, and malignancy. In addition, HSV-mediated transduction of non-neurological tissues allows their use as depot sites for synthesis of circulating and locally acting secreted proteins. New applications for this vector system include the genetic modification of stem cell populations; this may become an important means to direct cellular differentiation or deliver therapeutic genes systemically. Replication-defective HSV vectors are an effective and flexible vehicle for the delivery of transgenes to numerous tissues, with multiple applications.


Asunto(s)
Vectores Genéticos , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Transgenes , Genes Reporteros , Genoma Viral , Humanos , Modelos Biológicos , Mutación , Neoplasias/terapia , Enfermedades del Sistema Nervioso/terapia , Fenotipo , Factores de Tiempo
17.
J Urol ; 165(5): 1748-54, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11342969

RESUMEN

PURPOSE: Diabetic cystopathy resulting from sensory neuropathy may potentially be treated by direct gene therapy. It has been suggested that nerve growth factor (NGF) has an ameliorative effect in preventing the death in diabetes of afferent dorsal root ganglion neurons, which control bladder function. We investigated NGF gene transfer to the bladder and bladder afferent pathways for treating diabetic cystopathy. We used replication competent and replication defective herpes simplex virus type 1 (HSV-1) vectors that express a functionally active form of the beta-subunit of mouse NGF (beta-NGF) to examine the level and duration of therapeutic gene expression after administration of the vectors. MATERIALS AND METHODS: NGF expression during acute (3 days) and latent (21 days) infections was assessed by enzyme-linked immunosorbent assay (ELISA) and immunohistochemical testing after the injection of 1 x 106 to 1 x 108 pfu HSV-NGF expression vectors into the bladder wall of adult rats. RESULTS: HSV vectors with the strong human cytomegalovirus immediate early promoter used to drive beta-NGF gene expression exhibited increased NGF 3 days after infection in the bladder and L6 to S1 dorsal root ganglia, where bladder afferent neurons are located. ELISA analysis revealed that NGF in the bladder tissue and dorsal root ganglia was increased 7 to 9 and 2 to 4-fold, respectively, over the control vector. Increased NGF expression in L6 to S1 dorsal root ganglia neurons was also detected by immunohistochemical staining with antiNGF antibodies. Extended NGF expression was detected by ELISA 21 days after injection. Replication defective vectors containing HSV-1 latency promoter (LAP-2) driving NGF expressed NGF in the bladder and dorsal root ganglia 21 days after bladder injection. ELISA analysis confirmed an approximate 2 to 3-fold increase of NGF expression in the bladder and L6 to S1 dorsal root ganglia. CONCLUSIONS: The NGF gene may be transferred and expressed in the bladder and bladder afferent pathways using HSV vectors. To our knowledge our study represents the first demonstration of the effectiveness of gene therapy for altering neurotrophic expression in visceral sensory neurons. This technique of gene transfer may be useful for treating certain types of neurogenic bladder dysfunction, such as diabetic cystopathy, in which decreased NGF transport may be a causative factor.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Neuropatías Diabéticas/complicaciones , Terapia Genética , Vectores Genéticos , Herpesvirus Humano 1/genética , Factor de Crecimiento Nervioso/metabolismo , Neuronas Aferentes/metabolismo , Vejiga Urinaria Neurogénica/terapia , Vejiga Urinaria/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/metabolismo , Técnicas de Transferencia de Gen , Factor de Crecimiento Nervioso/genética , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria/inervación , Vejiga Urinaria Neurogénica/etiología , Vejiga Urinaria Neurogénica/metabolismo
19.
Gene Ther ; 8(7): 551-6, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11319622

RESUMEN

Endogenous opiate peptides acting pre- and post-synaptically in the dorsal horn of spinal cord inhibit transmission of nociceptive stimuli. We transfected neurons of the dorsal root ganglion in vivo by footpad inoculation with 30 microl (3 x 10(7) p.f.u.) of a replication-incompetent (ICP4-deleted) herpes simplex virus (HSV) vector with a cassette containing a portion of the human proenkephalin gene coding for 5 met- and 1 leu-enkephalin molecules under the control of the human cytomegalovirus immediate-early promoter (HCMV IEp) inserted in the HSV thymidine kinase (tk) locus. Vector-directed expression of enkephalin produced a significant antinociceptive effect measured by the formalin footpad test, that was most prominent in the delayed ("tonic") phase 20-70 min after the administration of formalin. The magnitude of the antinociceptive effect diminished over 4 weeks after transduction, but reinoculation of the vector reestablished the analgesic effect, without evidence for the development of tolerance. The antinociceptive effect was blocked completely by intrathecal naltrexone. These results suggest that gene therapy with an enkephalin-producing herpes-based vector may prove useful in the treatment of pain.


Asunto(s)
Analgesia/métodos , Encefalinas/genética , Ganglios Espinales/metabolismo , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Precursores de Proteínas/genética , Simplexvirus/genética , Animales , Encefalinas/metabolismo , Formaldehído , Humanos , Masculino , Naltrexona/farmacología , Dimensión del Dolor , Precursores de Proteínas/metabolismo , Ratas , Ratas Wistar , Transfección
20.
Mol Ther ; 3(1): 61-9, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11162312

RESUMEN

Sustained systemic dissemination of therapeutic proteins from peripheral sites is an attractive prospect for gene therapy applications. Replication-defective genomic herpes simplex virus type 1 (HSV-1) vectors were evaluated for their ability to express nerve growth factor (NGF) as a model gene product both locally and systemically. Intra-articular inoculation of NGF expression vectors in rabbits resulted in significant increases in joint lavage and blood plasma NGF that persisted for 1 year. A rhesus macaque injected intra-articularly displayed a comparable increase in plasma NGF for at least 6 months, at which time the serum NGF levels of this animal were sufficient to cause differentiation of PC12 cells in culture, but not to increase footpad epidermis innervation. Long-term reporter transgene expression was observed primarily in ligaments, a finding confirmed by direct inoculation of patellar ligament. Patellar ligament inoculation with a NGF vector resulted in elevated levels of circulating NGF similar to those observed following intra-articular vector delivery. These results represent the first demonstration of sustained systemic release of a transgene product using HSV vectors, raising the prospect of new applications for HSV-1 vectors in the treatment of systemic disease.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Herpesvirus Humano 1/genética , Factor de Crecimiento Nervioso/genética , Animales , Epidermis/inervación , Eliminación de Gen , Operón Lac , Ligamentos/metabolismo , Macaca mulatta , Factor de Crecimiento Nervioso/sangre , Células PC12 , Conejos , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...