Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Neurobiol Dis ; 68: 215-27, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24769160

RESUMO

G-protein coupled receptor 3 (GPR3), GPR6, and GPR12 belong to a family of constitutively active Gs-coupled receptors that activate 3'-5'-cyclic adenosine monophosphate (cAMP) and are highly expressed in the brain. Among these receptors, the endogenous expression of GPR3 in cerebellar granule neurons (CGNs) is increased following development. GPR3 is important for neurite outgrowth and neural maturation; however, the physiological functions of GPR3 remain to be fully elucidated. Here, we investigated the survival and antiapoptotic functions of GPR3 under normal and apoptosis-inducing culture conditions. Under normal culture conditions, CGNs from GPR3-knockout mice demonstrated lower survival than did CGNs from wild-type or GPR3-heterozygous mice. Cerebellar sections from GPR3-/- mice at P7, P14, and P21 revealed more caspase-3-positive neurons in the internal granular layer than in cerebellar sections from wild-type mice. Conversely, in a potassium-deprivation model of apoptosis, increased expression of these three receptors promoted neuronal survival. The antiapoptotic effect of GPR3 was also observed under hypoxic (1% O2/5% CO2) and reactive oxygen species (ROS)-induced apoptotic conditions. We further investigated the signaling pathways involved in the GPR3-mediated antiapoptotic effect. The addition of the PKA inhibitor KT5720, the MAP kinase inhibitor U0126, and the PI3 kinase inhibitor LY294002 abrogated the GPR3-mediated antiapoptotic effect in a potassium-deprivation model of apoptosis, whereas the PKC inhibitor Gö6976 did not affect the antiapoptotic function of GPR3. Furthermore, downregulation of endogenous GPR3 expression in CGNs resulted in a marked reduction in the basal levels of ERK and Akt phosphorylation under normal culture conditions. Finally, we used a transient middle cerebral artery occlusion (tMCAO) model in wild-type and GPR3-knockout mice to determine whether GPR3 expression modulates neuronal survival after brain ischemia. After tMCAO, GPR3-knockout mice exhibited a significantly larger infarct area than did wild-type mice. Collectively, these in vitro and in vivo results suggest that the developmental expression of constitutively active Gs-coupled GPR3 activates the ERK and Akt signaling pathways at the basal level, thereby protecting neurons from apoptosis that is induced by various stimuli.


Assuntos
Apoptose/genética , Cerebelo/citologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Fatores Etários , Animais , Cardiotônicos/farmacologia , Sobrevivência Celular/genética , Colforsina/farmacologia , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Isquemia/metabolismo , Isquemia/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
2.
Dev Biol ; 332(2): 309-24, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19500566

RESUMO

In the adult cerebellum, basket/stellate cells are scattered throughout the ML, but little is known about the process underlying the cell dispersion. To determine the allocation of stellate/basket cells within the ML, we examined their migration in the early postnatal mouse cerebellum. We found that after entering the ML, basket/stellate cells sequentially exhibit four distinct phases of migration. First, the cells migrated radially from the bottom to the top while exhibiting saltatory movement with a single leading process (Phase I). Second, the cells turned at the top and migrated tangentially in a rostro-caudal direction, with an occasional reversal of the direction of migration (Phase II). Third, the cells turned and migrated radially within the ML at a significantly reduced speed while repeatedly extending and withdrawing the leading processes (Phase III). Fourth, the cells turned at the middle and migrated tangentially at their slowest speed, while extending several dendrite-like processes after having completely withdrawn the leading process (Phase IV). Finally, the cells stopped and completed their migration. These results suggest that the dispersion of basket/stellate cells in the ML is controlled by the orchestrated activity of external guidance cues, cell-cell contact and intrinsic programs in a position- and time-dependent manner.


Assuntos
Movimento Celular/fisiologia , Cerebelo , Neurônios , Animais , Animais Recém-Nascidos , Células Cultivadas , Cerebelo/citologia , Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Humanos , Camundongos , Neurônios/citologia , Neurônios/fisiologia
3.
J Cell Biol ; 171(2): 255-65, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16247026

RESUMO

The arrest of meiotic prophase in mouse oocytes within antral follicles requires the G protein G(s) and an orphan member of the G protein-coupled receptor family, GPR3. To determine whether GPR3 activates G(s), the localization of Galpha(s) in follicle-enclosed oocytes from Gpr3(+/+) and Gpr3(-/-) mice was compared by using immunofluorescence and Galpha(s)GFP. GPR3 decreased the ratio of Galpha(s) in the oocyte plasma membrane versus the cytoplasm and also decreased the amount of Galpha(s) in the oocyte. Both of these properties indicate that GPR3 activates G(s). The follicle cells around the oocyte are also necessary to keep the oocyte in prophase, suggesting that they might activate GPR3. However, GPR3-dependent G(s) activity was similar in follicle-enclosed and follicle-free oocytes. Thus, the maintenance of prophase arrest depends on the constitutive activity of GPR3 in the oocyte, and the follicle cell signal acts by a means other than increasing GPR3 activity.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Meiose/fisiologia , Oócitos/metabolismo , Prófase/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/química , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Oócitos/citologia , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia
4.
Mol Ther ; 17(4): 707-15, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19223864

RESUMO

The herpes simplex virus (HSV) amplicon vector produces an initial host response that limits transgene expression. In this study, we hypothesized that restoration of the HSV gene infected cell protein (ICP0) into the amplicon could circumvent this host response and thus overcome silencing of encoded transgenes. To test this, we constructed an amplicon vector that encodes the ICP0 under control of its native promoter (ICP0+ amplicon). Expression of ICP0 was transient and, at a multiplicity of infection (MOI) of 1, did not significantly alter interferon (IFN)-based responses against the vector or cell kinetics/apoptosis of infected cells. Chromatin immunoprecipitation (ChIP) PCR analysis revealed that conventional amplicon DNA became associated with histone deacetylase 1 (HDAC1) immediately after infection, whereas ICP0+ amplicon DNA remained relatively unbound by HDAC1 for at least 72 hours after infection. Mice administered systemic ICP0+ amplicon exhibited significantly greater and more sustained transgene expression in their livers than did those receiving conventional amplicon, likely due to increased transcriptional or post-transcriptional activity rather than increased copy numbers of vector DNA. These findings indicate that restoration of ICP0 expression may be employed within HSV amplicon constructs to decrease transgene silencing in vitro and in vivo.


Assuntos
Proteínas Imediatamente Precoces/fisiologia , Simplexvirus/fisiologia , Transgenes , Ubiquitina-Proteína Ligases/fisiologia , Animais , Sequência de Bases , Imunoprecipitação da Cromatina , Primers do DNA , DNA Viral/genética , Histona Desacetilase 1 , Histona Desacetilases/metabolismo , Camundongos , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simplexvirus/genética
5.
J Cell Biol ; 167(5): 961-72, 2004 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-15569714

RESUMO

Neural activity induces the remodeling of pre- and postsynaptic membranes, which maintain their apposition through cell adhesion molecules. Among them, N-cadherin is redistributed, undergoes activity-dependent conformational changes, and is required for synaptic plasticity. Here, we show that depolarization induces the enlargement of the width of spine head, and that cadherin activity is essential for this synaptic rearrangement. Dendritic spines visualized with green fluorescent protein in hippocampal neurons showed an expansion by the activation of AMPA receptor, so that the synaptic apposition zone may be expanded. N-cadherin-venus fusion protein laterally dispersed along the expanding spine head. Overexpression of dominant-negative forms of N-cadherin resulted in the abrogation of the spine expansion. Inhibition of actin polymerization with cytochalasin D abolished the spine expansion. Together, our data suggest that cadherin-based adhesion machinery coupled with the actin-cytoskeleton is critical for the remodeling of synaptic apposition zone.


Assuntos
Caderinas/metabolismo , Espinhas Dendríticas/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal/fisiologia , Transmissão Sináptica/fisiologia , Actinas/antagonistas & inibidores , Actinas/biossíntese , Potenciais de Ação/fisiologia , Animais , Caderinas/genética , Células Cultivadas , Citocalasina D/farmacologia , Espinhas Dendríticas/ultraestrutura , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Microscopia Confocal , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Ratos , Receptores de AMPA/metabolismo , Proteínas Recombinantes de Fusão , Membranas Sinápticas/metabolismo
6.
Mol Ther ; 16(10): 1727-36, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18728642

RESUMO

The herpes simplex virus (HSV) amplicon is a plasmid-based, infectious gene delivery system that carries up to 150 kilobase (kb) of exogenous DNA. We previously characterized early host responses and stability of transgene expression in mice systemically injected with HSV amplicon vectors. Transgene expression was readily detected primarily in the liver but rapidly declined to undetectable levels within 2 weeks. Molecular analyses revealed induction of type I interferons (IFN) as the primary response, and early transcriptional silencing of the vector followed IFN's activation of signal transducers and activators of transcription 1 (STAT1). In this study, we investigate vector administration by stereotactic injection into the striatum. In the brain, induction of type I IFN was rather modest, and transgene expression lasted more than 1 year despite dose-dependent inflammation and infiltration of immune cells around injection sites. Further analyses revealed dose-dependent upregulation of immunosuppressive cytokines and molecular markers specific to regulatory T cells in the injected brain regions, which supported the immune-privileged properties of the brain parenchyma. Overall, our findings indicate that the spectrum of host responses can differ significantly depending on target organs and administrative routes, and that HSV amplicon vectors hold great potential for gene therapy of chronic neurological disorders.


Assuntos
Encéfalo/metabolismo , Expressão Gênica , Vetores Genéticos , Simplexvirus/genética , Transgenes , Animais , Sequência de Bases , Corpo Estriado/imunologia , Corpo Estriado/virologia , Primers do DNA , Camundongos , Camundongos Endogâmicos C57BL
7.
Mol Ther ; 16(9): 1546-55, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18648350

RESUMO

Replication-conditional (oncolytic) mutants of herpes simplex virus (HSV), are considered promising therapeutic alternatives for human malignancies, and chemotherapeutic adjuvants are increasingly sought to augment their efficacy. Histone deacetylase (HDAC) inhibitors are a new class of antineoplastic agents because of their potent activity in growth arrest, differentiation, and apoptotic death of cancer cells. The ability of the HDAC inhibitors to upregulate exogenous transgene expression and inhibit interferon (IFN) responses prompted our exploration of their use in improving the antitumor efficacy of oncolytic HSV. We discovered that the yield of viral progeny increased significantly when cultured glioma cells were treated with HDAC inhibitors before viral infection. Valproic acid (VPA), a commonly used antiepileptic agent with HDAC inhibitory activity, proved most effective when used to treat glioma cells before viral infection, but not concomitantly with viral infection. Pretreatment with VPA inhibited the induction of several IFN-responsive antiviral genes, augmented the transcriptional level of viral genes, and improved viral propagation, even in the presence of type I IFNs. Moreover, VPA pretreatment improved the propagation and therapeutic efficacy of oncolytic HSV in a human glioma xenograft model in vivo. These findings indicate that HDAC inhibitors can improve the efficacy of tumor virotherapies.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Glioma/tratamento farmacológico , Herpesvirus Humano 1/genética , Inibidores de Histona Desacetilases , Interferon beta/uso terapêutico , Vírus Oncolíticos , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virologia , Butiratos/uso terapêutico , Sinergismo Farmacológico , Quimioterapia Combinada , Terapia Genética , Vetores Genéticos/uso terapêutico , Glioma/genética , Glioma/virologia , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 1/metabolismo , Humanos , Interferon beta-1a , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/virologia , Camundongos , Camundongos Nus , Proteínas Recombinantes/uso terapêutico , Células Tumorais Cultivadas , Ácido Valproico/uso terapêutico , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Mol Ther ; 16(9): 1546-1555, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28189010

RESUMO

Replication-conditional (oncolytic) mutants of herpes simplex virus (HSV), are considered promising therapeutic alternatives for human malignancies, and chemotherapeutic adjuvants are increasingly sought to augment their efficacy. Histone deacetylase (HDAC) inhibitors are a new class of antineoplastic agents because of their potent activity in growth arrest, differentiation, and apoptotic death of cancer cells. The ability of the HDAC inhibitors to upregulate exogenous transgene expression and inhibit interferon (IFN) responses prompted our exploration of their use in improving the antitumor efficacy of oncolytic HSV. We discovered that the yield of viral progeny increased significantly when cultured glioma cells were treated with HDAC inhibitors before viral infection. Valproic acid (VPA), a commonly used antiepileptic agent with HDAC inhibitory activity, proved most effective when used to treat glioma cells before viral infection, but not concomitantly with viral infection. Pretreatment with VPA inhibited the induction of several IFN-responsive antiviral genes, augmented the transcriptional level of viral genes, and improved viral propagation, even in the presence of type I IFNs. Moreover, VPA pretreatment improved the propagation and therapeutic efficacy of oncolytic HSV in a human glioma xenograft model in vivo. These findings indicate that HDAC inhibitors can improve the efficacy of tumor virotherapies.

9.
Neuro Oncol ; 10(5): 690-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18715951

RESUMO

Therapies targeting glioma cells that diffusely infiltrate normal brain are highly sought after. Our aim was to identify novel approaches to this problem using glioma spheroid migration assays. Lithium, a currently approved drug for the treatment of bipolar illnesses, has not been previously examined in the context of glioma migration. We found that lithium treatment potently blocked glioma cell migration in spheroid, wound-healing, and brain slice assays. The effects observed were dose dependent and reversible, and worked using every glioma cell line tested. In addition, there was little effect on cell viability at lithium concentrations that inhibit migration, showing that this is a specific effect. Lithium treatment was associated with a marked change in cell morphology, with cells retracting the long extensions at their leading edge. Examination of known targets of lithium showed that inositol monophosphatase inhibition had no effect on glioma migration, whereas inhibition of glycogen synthase kinase-3 (GSK-3) did. This suggested that the effects of lithium on glioma cell migration could possibly be mediated through GSK-3. Specific pharmacologic GSK-3 inhibitors and siRNA knockdown of GSK-3alpha or GSK-3beta isoforms both reduced cell motility. These data outline previously unidentified pathways and inhibitors that may be useful for the development of novel anti-invasive therapeutics for the treatment of brain tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/enzimologia , Movimento Celular/efeitos dos fármacos , Glioma/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Cloreto de Lítio/farmacologia , Animais , Western Blotting , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Glioma/patologia , Quinase 3 da Glicogênio Sintase/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica/fisiopatologia , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos
10.
Clin Cancer Res ; 12(2): 643-52, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16428511

RESUMO

Conditionally replicating herpes simplex virus-1 (HSV-1) vectors are promising therapeutic agents for cancer. Insertion of therapeutic transgenes into the viral genome should confer desired anticancer functions in addition to oncolytic activities. Herein, using bacterial artificial chromosome and two recombinase-mediated recombinations, we simultaneously created four "armed" oncolytic HSV-1, designated vHsv-B7.1-Ig, vHsv-interleukin (IL)-12, vHsv-IL-18, and vHsv-null, which express murine soluble B7.1 (B7.1-Ig), murine IL-12, murine IL-18, and no transgene, respectively. These vHsv vectors possess deletions in the gamma34.5 genes and contain the green fluorescent protein gene as a histochemical marker and the immunostimulatory transgene inserted in the deleted ICP6 locus. The vHsv showed similar replicative capabilities in vitro. The in vivo efficacy was tested in A/J mice harboring s.c. tumors of syngeneic and poorly immunogenic Neuro2a neuroblastoma. The triple combination of vHsv-B7.1-Ig, vHsv-IL-12, and vHsv-IL-18 exhibited the highest efficacy among all single vHsv or combinations of two viruses. Combining 1 x 10(5) plaque-forming units each of the three armed viruses showed stronger antitumor activities than any single armed virus at 3 x 10(5) plaque-forming units in inoculated tumors as well as in noninoculated remote tumors. Studies using athymic mice indicated that this enhancement of antitumor efficacy was likely mediated by T-cell immune responses. The combined use of multiple oncolytic HSV-1 armed with different immunostimulatory genes may be a useful strategy for cancer therapy.


Assuntos
Antígeno B7-1 , Vetores Genéticos/uso terapêutico , Herpesvirus Humano 1/genética , Interleucina-12 , Interleucina-18 , Neuroblastoma/terapia , Terapia Viral Oncolítica , Animais , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Chlorocebus aethiops , Cromossomos Artificiais Bacterianos , Quimioterapia Combinada , Vetores Genéticos/genética , Humanos , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-12/metabolismo , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-18/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Nus , Neuroblastoma/genética , Neuroblastoma/imunologia , Recombinases/genética , Linfócitos T/imunologia , Transfecção , Transgenes/fisiologia , Células Tumorais Cultivadas , Células Vero
11.
Cancer Res ; 65(24): 11255-8, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16357128

RESUMO

The success of cancer virotherapy depends on its efficacy versus toxicity profile in human clinical trials. Progress towards clinical trials can be hampered by the relatively elevated doses of oncolytic viruses administered in animal models to achieve an anticancer effect and by the even higher doses required in humans to approximate an animal bioequivalent dose. Such elevated doses of injected viral proteins may also lead to undesirable toxicities and are also very difficult to produce in a biotechnological setting. We report that a relatively potent herpes simplex virus type 1 oncolytic virus (rQNestin34.5) produces 45% survivors at a dose of 3 x 10(4) plaque-forming units (pfu) in a 9-day-old mouse model of human glioma. Unlike our previous findings with less potent oncolytic viruses, though, the preadministration of cyclophosphamide did not enhance this survival or affect oncolytic virus tumor distribution and tumor volume. However, when oncolytic virus doses were reduced (3 x 10(3) and 3 x 10(2) pfu), cyclophosphamide significantly enhanced both animal survival and oncolytic virus tumor distribution and also reduced tumor volumes. These findings thus show that cyclophosphamide allows for dose reduction of doses of a relatively potent oncolytic virus, a finding with implications for the development of clinical trials.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Ciclofosfamida/uso terapêutico , Terapia Genética , Glioma/terapia , Herpesvirus Humano 1/fisiologia , Vírus Oncolíticos/genética , Animais , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Terapia Combinada , Vetores Genéticos/administração & dosagem , Glioma/patologia , Glioma/virologia , Humanos , Camundongos , Taxa de Sobrevida , Células Tumorais Cultivadas , Replicação Viral
12.
Cancer Res ; 65(7): 2832-9, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15805284

RESUMO

Oncolytic herpes simplex virus-1 (HSV-1) mutants possessing mutations in the ICP34.5 and ICP6 genes have proven safe through clinical trials. However, ICP34.5-null viruses may grow poorly in cells due to their inability to prevent host-cell shut-off of protein synthesis caused by hyperphosphorylation of eukaryotic initiation factor 2alpha. To increase tumor selectivity, glioma-selective expression of ICP34.5 in the context of oncolysis may be useful. Malignant gliomas remain an incurable disease. One molecular marker of malignant gliomas is expression of the intermediate filament nestin. Expression of nestin mRNA was confirmed in 6 of 6 human glioma lines and in 3 of 4 primary glioma cells. Normal human astrocytes were negative. A novel glioma-selective HSV-1 mutant (rQNestin34.5) was thus engineered by expressing ICP34.5 under control of a synthetic nestin promoter. Replication, cellular propagation, and cytotoxicity of rQNestin34.5 were significantly enhanced in cultured and primary human glioma cell lines compared with control virus. However, replication, cellular propagation, and cytotoxicity of rQNestin34.5 in normal human astrocytes remained quantitatively similar to that of control virus. In glioma cell lines infected with rQNestin34.5, the level of phospho-eukaryotic initiation factor 2alpha was lower than that of cells infected by control rHsvQ1, confirming selective ICP34.5 expression in glioma cells. In vivo, rQNestin34.5 showed significantly more potent inhibition of tumor growth compared with control virus. Treatment in the brain tumor model was instituted on animal's display of neurologic symptoms, which usually led to rapid demise. rQNestin34.5 treatment doubled the life span of these animals. These results show that rQNestin34.5 could be a potent agent for the treatment of malignant glioma.


Assuntos
Neoplasias Encefálicas/virologia , Glioma/virologia , Herpesvirus Humano 1/fisiologia , Proteínas de Filamentos Intermediários/genética , Proteínas do Tecido Nervoso/genética , Proteínas Virais/biossíntese , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Fator de Iniciação 2 em Eucariotos/metabolismo , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Immunoblotting , Proteínas de Filamentos Intermediários/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Proteínas do Tecido Nervoso/biossíntese , Nestina , Fosforilação , Regiões Promotoras Genéticas , Proteínas Virais/genética
13.
Cancer Res ; 65(15): 6850-7, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16061668

RESUMO

The treatment of malignant glioma is currently ineffective. Oncolytic viruses are being explored as a means to selectively lyse tumor cells in the brain. We have engineered a mutant herpes simplex virus type 1 with deletions in the viral UL39 and gamma(1)34.5 genes and an insertion of the two prodrug activating genes, CYP2B1 and secreted human intestinal carboxylesterase. Each of these can convert the inactive prodrugs, cyclophosphamide and irinotecan (CPT-11), into their active metabolites, respectively. This new oncolytic virus (MGH2) displays increased antitumor efficacy against human glioma cells both in vitro and in vivo when combined with cyclophosphamide and CPT-11. Importantly, cyclophosphamide, CPT-11, or the combination of cyclophosphamide and CPT-11 does not significantly affect oncolytic virus replication. Therefore, MGH2 provides effective multimodal therapy for gliomas in preclinical models when combined with these chemotherapy agents.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias Encefálicas/terapia , Carboxilesterase/genética , Citocromo P-450 CYP2B1/genética , Glioma/terapia , Herpesvirus Humano 1/fisiologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biotransformação , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virologia , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Carboxilesterase/biossíntese , Carboxilesterase/metabolismo , Linhagem Celular Tumoral , Ciclofosfamida/administração & dosagem , Citocromo P-450 CYP2B1/biossíntese , Citocromo P-450 CYP2B1/metabolismo , Terapia Genética/métodos , Glioma/genética , Glioma/virologia , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Irinotecano , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Replicação Viral
14.
Curr Gene Ther ; 6(3): 303-14, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16787182

RESUMO

The herpes simplex virus (HSV) amplicon vector is a versatile plasmid-based gene delivery vehicle with a large transgene capacity (up to 150 kb) and the ability to infect a broad range of cell types. The vector system was originally developed by Frenkel and her colleagues in 1980. Ever since, a great deal of effort by various investigators has been directed at minimizing the toxicity associated with the inevitable contamination by helper virus. In 1996, Fraefel and his colleagues successfully devised a cosmid-based packaging system that was free of contamination by helper virus (so-called helper virus-free packaging), which utilized as helper a set of 5 overlapping cosmid clones that covered the entire HSV genome, which lacked the DNA packaging/cleavage signals. With the helper virus-free system, broader applications of the vector became possible. Cloning of the entire HSV genome in bacteria artificial chromosome (BAC) plasmids enabled stable maintenance and propagation of the helper HSV genome in bacteria. It also allowed for the development of BAC-based helper virus-free packaging systems. In this article, we review various versions of DNA-based methods to prepare HSV amplicon vectors free of helper virus contamination. We also examine recent advances in vector design, including methods of vector construction, hybrid amplicon vectors, and the infectious BAC system. Future directions in improving packaging systems and vector designs are discussed.


Assuntos
DNA , Vetores Genéticos , Plasmídeos , Simplexvirus/genética , Cromossomos Artificiais Bacterianos
15.
J Virol Methods ; 123(1): 65-72, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15582700

RESUMO

Gene transfer into hepatocytes is highly desirable for the long-term goal of replacing deficient proteins and correcting metabolic disorders. Vectors based on herpes simplex virus type-1 (HSV-1) have been demonstrated to mediate efficient gene transfer into hepatocytes both in vitro and in vivo. Large transgene capacity and extrachromosomal persistence make HSV-1/EBV hybrid amplicon vectors an attractive candidate for hepatic gene replacement therapy. To assess liver-directed gene transfer, we constructed (i) a conventional HSV-1 amplicon vector encoding a secreted reporter protein (secreted alkaline phosphatase, SEAP) under the control of the HSV-1 immediate-early 4/5 promoter; (ii) a HSV-1 amplicon encoding SEAP under the control of the artificial CAG promoter (the chicken beta-actin promoter and cytomegalovirus (CMV) immediate-early enhancer); and (iii) a HSV-1/EBV hybrid amplicon, also encoding SEAP under the control of the CAG promoter. While all three vector constructs yielded high SEAP concentrations in vitro and in vivo, use of HSV-1/EBV hybrid amplicon vectors significantly prolonged the duration of gene expression. Using conventional amplicon vectors in cultured hepatocytes, SEAP was detected for two weeks, whereas SEAP was detected for at least six weeks when HSV-1/EBV amplicons were used. Intraparenchymal injection into the liver of SICD mice yielded high (up to 77 ng of SEAP per milliliter serum) and sustained (greater than three weeks) expression of SEAP. Serum transaminases (ALT/AST) were measured at different time points to monitor for hepatocellular damage. While initially elevated four times above baseline, the transaminase levels returned to normal after three to seven days. These results demonstrate the usefulness of HSV-1-based amplicons and SEAP for the evaluation of gene replacement strategies in the liver.


Assuntos
Fosfatase Alcalina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Hepatócitos/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 4/genética , Fosfatase Alcalina/metabolismo , Animais , Chlorocebus aethiops , Humanos , Camundongos , Camundongos SCID , Recombinação Genética , Transdução Genética , Transgenes , Células Vero
16.
Neoplasia ; 4(6): 523-30, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12407446

RESUMO

Magnetic resonance imaging (MRI) can provide high-resolution 3D maps of structural and functional information, yet its use of mapping in vivo gene expression has only recently been explored. A potential application for this technology is to noninvasively image transgene expression. The current study explores the latter using a nonregulatable internalizing engineered transferrin receptor (ETR) whose expression can be probed for with a superparamagnetic Tf-CLIO probe. Using an HSV-based amplicon vector system for transgene delivery, we demonstrate that: 1) ETR is a sensitive MR marker gene; 2) several transgenes can be efficiently expressed from a single amplicon; 3) expression of each transgene results in functional gene product; and 4) ETR gene expression correlates with expression of therapeutic genes when the latter are contained within the same amplicon. These data, taken together, suggest that MRI of ETR expression can serve as a surrogate for measuring therapeutic transgene expression.


Assuntos
Neoplasias Encefálicas/diagnóstico , Expressão Gênica , Glioma/diagnóstico , Imageamento por Ressonância Magnética , Receptores da Transferrina/genética , Transgenes/genética , Animais , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste , Sistema Enzimático do Citocromo P-450/metabolismo , Técnicas de Transferência de Genes , Genes erbB-1/fisiologia , Marcadores Genéticos , Vetores Genéticos , Glioma/genética , Glioma/patologia , Herpes Simples/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Técnicas Imunoenzimáticas , Ferro/farmacocinética , Camundongos , Sondas Moleculares , Óxidos/farmacocinética , Retroviridae , Transdução Genética , Células Tumorais Cultivadas
18.
PLoS One ; 4(6): e5922, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19526062

RESUMO

BACKGROUND: During postnatal murine and rodent cerebellar development, cerebellar granule precursors (CGP) gradually stop proliferating as they differentiate after migration to the internal granule layer (IGL). Molecular events that govern this program remain to be fully elucidated. GPR3 belongs to a family of Gs-linked receptors that activate cyclic AMP and are abundantly expressed in the adult brain. METHODOLOGY/PRINCIPAL FINDINGS: To investigate the role of this orphan receptor in CGP differentiation, we determined that exogenous GPR3 expression in rat cerebellar granule neurons partially antagonized the proliferative effect of Sonic hedgehog (Shh), while endogenous GPR3 inhibition by siRNA stimulated Shh-induced CGP proliferation. In addition, exogenous GPR3 expression in CGPs correlated with increased p27/kip expression, while GPR3 knock-down led to a decrease in p27/kip expression. In wild-type mice, GPR3 expression increased postnatally and its expression was concentrated in the internal granular layer (IGL). In GPR3 -/- mice, the IGL was widened with increased proliferation of CGPs, as measured by bromodeoxyuridine incorporation. Cell cycle kinetics of GPR3-transfected medulloblastoma cells revealed a G0/G1 block, consistent with cell cycle exit. CONCLUSIONS/SIGNIFICANCE: These results thus indicate that GPR3 is a novel antiproliferative mediator of CGPs in the postnatal development of murine cerebellum.


Assuntos
Cerebelo/citologia , Regulação da Expressão Gênica no Desenvolvimento , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/fisiologia , Animais , Encéfalo/metabolismo , Proliferação de Células , AMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Hedgehog/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
19.
PLoS One ; 4(1): e4235, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19156211

RESUMO

BACKGROUND: Although disease remission can frequently be achieved for patients with neuroblastoma, relapse is common. The cancer stem cell theory suggests that rare tumorigenic cells, resistant to conventional therapy, are responsible for relapse. If true for neuroblastoma, improved cure rates may only be achieved via identification and therapeutic targeting of the neuroblastoma tumor initiating cell. Based on cues from normal stem cells, evidence for tumor populating progenitor cells has been found in a variety of cancers. METHODOLOGY/PRINCIPAL FINDINGS: Four of eight human neuroblastoma cell lines formed tumorspheres in neural stem cell media, and all contained some cells that expressed neurogenic stem cell markers including CD133, ABCG2, and nestin. Three lines tested could be induced into multi-lineage differentiation. LA-N-5 spheres were further studied and showed a verapamil-sensitive side population, relative resistance to doxorubicin, and CD133+ cells showed increased sphere formation and tumorigenicity. Oncolytic viruses, engineered to be clinically safe by genetic mutation, are emerging as next generation anticancer therapeutics. Because oncolytic viruses circumvent typical drug-resistance mechanisms, they may represent an effective therapy for chemotherapy-resistant tumor initiating cells. A Nestin-targeted oncolytic herpes simplex virus efficiently replicated within and killed neuroblastoma tumor initiating cells preventing their ability to form tumors in athymic nude mice. CONCLUSIONS/SIGNIFICANCE: These results suggest that human neuroblastoma contains tumor initiating cells that may be effectively targeted by an oncolytic virus.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas/citologia , Neuroblastoma/metabolismo , Vírus Oncolíticos/metabolismo , Antígeno AC133 , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/biossíntese , Animais , Antígenos CD/biossíntese , Linhagem Celular Tumoral , Linhagem da Célula , Chlorocebus aethiops , Glicoproteínas/biossíntese , Humanos , Proteínas de Filamentos Intermediários/biossíntese , Proteínas de Neoplasias/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Nestina , Peptídeos , Células-Tronco/metabolismo , Transcrição Gênica , Células Vero
20.
Cancer Res ; 68(4): 1170-9, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281493

RESUMO

Malignant solid tumors remain a significant clinical challenge, necessitating innovative therapeutic approaches. Oncolytic viral therapy is a nonmutagenic, biological anticancer therapeutic shown to be effective against human cancer in early studies. Because matrix metalloproteinases (MMP) play important roles in the pathogenesis and progression of cancer, we sought to determine if "arming" an oncolytic herpes simplex virus (oHSV) with an MMP-antagonizing transgene would increase virus-mediated antitumor efficacy. We generated oHSVs that express human tissue inhibitor of metalloproteinases 3 (TIMP3) or firefly luciferase and designated them rQT3 and rQLuc, respectively. We evaluated the antitumor efficacy of these viruses against neuroblastoma and malignant peripheral nerve sheath tumor (MPNST) xenografts. Relative to rQLuc, rQT3-infected primary human MPNST and neuroblastoma cells exhibited equivalent virus replication but increased cytotoxicity and reduced MMP activity. In vivo, rQT3-treated tumors showed delayed tumor growth, increased peak levels of infectious virus, immature collagen extracellular matrix, and reduced tumor vascular density. Remarkably, rQT3 treatment reduced circulating endothelial progenitors, suggesting virus-mediated antivasculogenesis. We conclude that rQT3 enhanced antitumor efficacy through multiple mechanisms, including direct cytotoxicity, elevated virus titer, and reduced tumor neovascularization. These findings support the further development of combined TIMP-3 and oncolytic virotherapy for cancer.


Assuntos
Terapia Genética/métodos , Neoplasias de Bainha Neural/terapia , Neuroblastoma/terapia , Terapia Viral Oncolítica/métodos , Simplexvirus/fisiologia , Inibidor Tecidual de Metaloproteinase-3/genética , Animais , Chlorocebus aethiops , Terapia Combinada , Feminino , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/virologia , Humanos , Luciferases de Vaga-Lume/genética , Camundongos , Camundongos Nus , Neoplasias de Bainha Neural/sangue , Neoplasias de Bainha Neural/genética , Neoplasias de Bainha Neural/virologia , Neuroblastoma/sangue , Neuroblastoma/genética , Neuroblastoma/virologia , Simplexvirus/genética , Células Vero , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA