Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 10(23): 2252-2269, 2019 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-31040917

RESUMO

Immune checkpoint inhibitors (CPIs) are associated with a number of immune-related adverse events and low response rates. We provide preclinical evidence for use of a retroviral replicating vector (RRV) selective to cancer cells, to deliver CPI agents that may circumvent such issues and increase efficacy. An RRV, RRV-scFv-PDL1, encoding a secreted single chain variable fragment targeting PD-L1 can effectively compete with PD-1 for PD-L1 occupancy. Cell binding assays showed trans-binding activity on 100% of cells in culture when infection was limited to 5% RRV-scFv-PDL1 infected tumor cells. Further, the ability of scFv PD-L1 to rescue PD-1/PD-L1 mediated immune suppression was demonstrated in a co-culture system consisting of human-derived immune cells and further demonstrated in several syngeneic mouse models including an intracranial tumor model. These tumor models showed that tumors infected with RRV-scFv-PD-L1 conferred robust and durable immune-mediated anti-tumor activity comparable or superior to systemically administered anti-PD-1 or anti PD-L1 monoclonal antibodies. Importantly, the nominal level of scFv-PD-L1 detected in serum is ∼50-150 fold less than reported for systemically administered therapeutic antibodies targeting immune checkpoints. These results support the concept that RRV-scFv-PDL1 CPI strategy may provide an improved safety and efficacy profile compared to systemic monoclonal antibodies of currently approved therapies.

2.
Hum Gene Ther ; 29(4): 437-451, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29216761

RESUMO

Toca 511, a retroviral replicating vector (RRV), uses an internal ribosomal entry site (IRES) to express an optimized yeast cytosine deaminase (yCD2), which converts 5-fluorocytosine to 5-fluorouracil. This configuration is genetically stable in both preclinical mouse models and human clinical trials. However, the use of IRES (∼600 bp) restricts choices of therapeutic transgenes due to limits in RRV genome size. This study replaced IRES with 2A peptides derived from picornaviruses with or without a GSG linker. The data show that GSG-linked 2A (g2A) peptide resulted in higher polyprotein separation efficiency than non-GSG linked 2A peptide. The study also shows that RRV can tolerate insertion of two separate 2A peptides to allow expression of two transgenes without compromising the assembly and function of the virus in addition to insertion of a single 2A peptide to confirm genetic stability with yCD2, green fluorescent protein, and HSV-1 thymidine kinase. In a parallel comparison of the RRV-IRES-yCD2 and RRV-g2A-yCD2 configurations, the study shows the yCD2 protein expressed from RRV-g2A-yCD2 has higher activity, resulting in a higher survival benefit in an intracranial tumor mouse model. These data enable a wider range of potential product candidates that could be developed using the RRV platform.


Assuntos
Neoplasias Encefálicas/terapia , Terapia Genética , Vetores Genéticos , Sítios Internos de Entrada Ribossomal/genética , Animais , Neoplasias Encefálicas/genética , Citosina Desaminase/genética , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Humanos , Camundongos , Peptídeos/genética , Picornaviridae/genética , Replicação Viral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Curr Opin HIV AIDS ; 12(4): 383-389, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28509712

RESUMO

PURPOSE OF REVIEW: To discuss how aligning the collective power of scientists, regulators, drug companies, donors, implementers and advocates to achieve a single goal - accelerating access to simpler, safer, more robust and more affordable HIV treatment - can rapidly advance antiretroviral optimization efforts and enable scale-up. RECENT FINDINGS: Harmonization of traditionally sequential processes can address the delays commonly experienced in introducing new products to low-income and middle-income countries, by facilitating an 'end-to-end' approach that mitigates risk and encourages early planning for all aspects of product introduction. SUMMARY: Planning with the 'end-in-mind' can facilitate healthy markets, benefit the application of new technologies, and accelerate the development of improved products in parallel (versus traditionally sequential efforts).


Assuntos
Fármacos Anti-HIV/provisão & distribuição , Custos de Medicamentos , Infecções por HIV/tratamento farmacológico , Acessibilidade aos Serviços de Saúde , Fármacos Anti-HIV/economia , Países em Desenvolvimento , Infecções por HIV/economia , Humanos
4.
Mol Ther Nucleic Acids ; 6: 221-232, 2017 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-28325288

RESUMO

Tumor cells express a number of immunosuppressive molecules that can suppress anti-tumor immune responses. Efficient delivery of small interfering RNAs to treat a wide range of diseases including cancers remains a challenge. Retroviral replicating vectors (RRV) can be used to stably and selectively introduce genetic material into cancer cells. Here, we designed RRV to express shRNA (RRV-shPDL1) or microRNA30-derived shRNA (RRV-miRPDL1) using Pol II or Pol III promoters to downregulate PDL1 in human cancer cells. We also designed RRV expressing cytosine deaminase (yCD2) and miRPDL1 for potential combinatorial therapy. Among various configurations tested, we showed that RRV-miRPDL1 vectors with Pol II or Pol III promoter replicated efficiently and exhibited sustained downregulation of PDL1 protein expression by more than 75% in human cancer cell lines with high expression of PDL1. Immunologic effects of RRV-miRPDL1 were assessed by a trans-suppression lymphocyte assay. In vitro data showed downregulation of PDL1+ tumor cells restored activation of CD8+ T cells and bio-equivalency compared to anti-PDL1 antibody treatment. These results suggest RRV-miRPDL1 may be an alternative therapeutic approach to enhance anti-tumor immunity by overcoming PDL1-induced immune suppression from within cancer cells and this approach may also be applicable to other cancer targets.

5.
Hum Gene Ther Methods ; 27(2): 59-70, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26918465

RESUMO

We have developed retroviral replicating vectors (RRV) derived from Moloney murine gammaretrovirus with an amphotropic envelope and an encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES)-transgene cassette downstream of the env gene. During long-term (180 days) replication of the vector in animals, a bulge of 7 adenosine residues (A's) in the J-K bifurcation domain sometimes serially added A's. Therefore, vectors with 4-12 A's in the A bulge in the J-K bifurcation domain were generated, and the impact of the variants on transgene protein expression, vector stability, and IRES sequence upon multiple infection cycles was assessed in RRV encoding yeast-derived cytosine deaminase and green fluorescent protein in vitro. For transgene protein expression, after multiple infection cycles, RRV-IRES with 5-7 A's gave roughly comparable levels, 4 and 8 A's were within about 4-5-fold of the 6 A's, whereas 10 and 12 A's were marked lower. In terms of stability, after 10 infection cycles, expansion of A's appeared to be a more frequent event affecting transgene protein expression than viral genome deletions or rearrangement: 4 and 5 A's appeared completely stable; 6, 7, and particularly 8 A's showed some level of expansion in the A bulge; 10 and 12 A's underwent both expansion and transgene deletion. The strong relative translational activity of the 5 A's in the EMCV IRES has not been reported previously. The 5A RRV-IRES may have utility for preclinical and clinical applications where extended replication is required.


Assuntos
Vírus da Encefalomiocardite/genética , Vetores Genéticos , Sítios Internos de Entrada Ribossomal/genética , Vírus da Leucemia Murina de Moloney/genética , Transgenes/genética , Animais , Linhagem Celular Tumoral , Citosina Desaminase/genética , Feminino , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Biossíntese de Proteínas , RNA Viral/genética
6.
J Int AIDS Soc ; 17(3 Suppl 2): 19162, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25224619

RESUMO

INTRODUCTION: In planning for the introduction of vaginal microbicides and other new antiretroviral (ARV)-based prevention products for women, an in-depth understanding of potential end-users will be critically important to inform strategies to optimize uptake and long-term adherence. User-centred private sector companies have contributed to the successful launch of many different types of products, employing methods drawn from behavioural and social sciences to shape product designs, marketing messages and communication channels. Examples of how the private sector has adapted and applied these techniques to make decisions around product messaging and targeting may be instructive for adaptation to microbicide introduction. DISCUSSION: In preparing to introduce a product, user-centred private sector companies employ diverse methods to understand the target population and their lifestyles, values and motivations. ReD Associates' observational research on user behaviours in the packaged food and diabetes fields illustrates how 'tag along' or 'shadowing' techniques can identify sources of non-adherence. Another open-ended method is self-documentation, and IDEO's mammography research utilized this to uncover user motivations that extended beyond health. Mapping the user journey is a quantitative approach for outlining critical decision-making stages, and Monitor Inclusive Markets applied this framework to identify toilet design opportunities for the rural poor. Through an iterative process, these various techniques can generate hypotheses on user drop-off points, quantify where drop-off is highest and prioritize areas of further research to uncover usage barriers. Although research constraints exist, these types of user-centred techniques have helped create effective messaging, product positioning and packaging of health products as well as family planning information. These methods can be applied to microbicide acceptability testing outside of clinical trials to design microbicide marketing that enhances product usage. CONCLUSIONS: The introduction of microbicide products presents an ideal opportunity to draw on the insights from user-centred private sector companies' approaches, which can complement other methods that have been more commonly utilized in microbicide research to date. As microbicides move from clinical trials to real-world implementation, there will be more opportunities to combine a variety of approaches to understand end-users, which can lead to a more effective product launch and ultimately greater impact on preventing HIV infections.


Assuntos
Anti-Infecciosos/uso terapêutico , Comportamento/fisiologia , Transmissão de Doença Infecciosa/prevenção & controle , Infecções por HIV/prevenção & controle , Infecções por HIV/psicologia , Aceitação pelo Paciente de Cuidados de Saúde/psicologia , Setor Privado/estatística & dados numéricos , Administração Intravaginal , Feminino , Humanos
7.
J Virol ; 88(17): 10066-77, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24965455

RESUMO

UNLABELLED: We developed a Moloney mouse leukemia virus (MLV)-based retroviral replicating vector (RRV), Toca 511, which has displayed tumor specificity in resected brain tumor material and blood in clinical trials. Here, we investigated the interaction between Toca 511 and human host cells, and we show that RRVs do not induce type I interferon (IFN) responses in cultured human tumor cells or cultured human primary cells. However, exogenous type I IFN inhibited RRV replication in tumor cells and induced IFN-regulated genes, albeit at a lower level than in primary cells. Unexpectedly, RRVs did not induce IFN-α production upon incubation in vitro with human plasmacytoid dendritic cells (pDCs), whereas lentivirus vector and heat-treated RRVs did. Coincubation of RRVs with heat-treated RRVs or with lentivirus vector suppressed IFN-α production in pDCs, suggesting that native RRV has a dominant inhibitory effect on type I IFN induction. This effect is sensitive to trypsin treatment. In addition, heat treatment inactivated that activity but exposed an immune-stimulatory activity. The immune-stimulating component is sensitive to deglycosidases, trypsin, and phospholipase C treatment. Experiments with retroviral nonreplicating vectors and virus-like particles demonstrated that the immunosuppressive activity is not associated with the amphotropic envelope or the glyco-Gag protein. In summary, our data provide evidence that RRVs do not directly trigger type I IFN responses in IFN-responsive tumor cells. Moreover, RRVs appear to carry a heat-labile component that actively suppresses activation of cellular innate immune responses in pDCs. Inhibition of IFN induction by RRVs and the reduced response to IFN should facilitate tumor-specific infection in vivo. IMPORTANCE: RRVs have a convincing preference for replicating in tumor cells in animal models, and we observed similar preferences in the initial treatment of human glioblastoma patients. This study investigates the basis for the interaction between RRV and human host cells (tumor versus nontumor) in vitro. We found that RRVs do not trigger an IFN-α/ß response in tumor cells, but the cells are capable of responding to type I IFNs and of producing them when stimulated with known agonists. Surprisingly, the data show that RRVs can actively inhibit induction of cellular innate immunity and that this inhibitory activity is heat labile and trypsin sensitive and not attributable to the envelope protein. These data partially explain the observed in vivo tumor specificity.


Assuntos
Interferon Tipo I/metabolismo , Vírus da Leucemia Murina de Moloney/imunologia , Vírus da Leucemia Murina de Moloney/fisiologia , Neoplasias/imunologia , Replicação Viral , Células Cultivadas , Humanos , Vírus da Leucemia Murina de Moloney/genética
8.
Hum Gene Ther ; 25(8): 759-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24825189

RESUMO

We are developing a retroviral replicating vector (RRV) encoding cytosine deaminase as an anticancer agent for gliomas. Despite its demonstrated natural selectivity for tumors, and other safety features, such a virus could potentially cause off-target effects by productively infecting healthy tissues. Here, we investigated whether incorporation of a hematopoietic lineage-specific microRNA target sequence in RRV further restricts replication in hematopoietic lineage-derived human cells in vitro and in murine lymphoid tissues in vivo. One or four copies of a sequence perfectly complementary to the guide strand of microRNA 142-3p were inserted into the 3' untranslated region of the RRV genome expressing the transgene encoding green fluorescent protein (GFP). Viral spread and GFP expression of these vectors in hematopoietic lineage cells in vitro and in vivo were measured by qPCR, qRT-PCR, and flow cytometry. In hematopoietic lineage-derived human cell lines and primary human stimulated peripheral blood mononuclear cells, vectors carrying the 142-3pT sequence showed a remarkable decrease in GFP expression relative to the parental vector, and viral spread was not observed over time. In a syngeneic subcutaneous mouse tumor model, RRVs with and without the 142-3pT sequences spread equally well in tumor cells; were strongly repressed in blood, bone marrow, and spleen; and generated antiviral immune responses. In an immune-deficient mouse model, RRVs with 142-3pT sequences were strongly repressed in blood, bone marrow, and spleen compared with unmodified RRV. Tissue-specific microRNA-based selective attenuation of RRV replication can maintain antiviral immunity, and if needed, provide an additional safeguard to this delivery platform for gene therapy applications.


Assuntos
Células da Medula Óssea/virologia , Glioma/terapia , MicroRNAs/genética , Retroviridae/fisiologia , Replicação Viral , Animais , Células da Medula Óssea/fisiologia , Linhagem Celular Tumoral , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Leucócitos Mononucleares , Camundongos , Camundongos Nus , MicroRNAs/administração & dosagem , Transplante de Neoplasias , Especificidade de Órgãos , Transdução Genética
9.
Neuro Oncol ; 14(2): 145-59, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22070930

RESUMO

Patients with the most common and aggressive form of high-grade glioma, glioblastoma multiforme, have poor prognosis and few treatment options. In 2 immunocompetent mouse brain tumor models (CT26-BALB/c and Tu-2449-B6C3F1), we showed that a nonlytic retroviral replicating vector (Toca 511) stably delivers an optimized cytosine deaminase prodrug activating gene to the tumor lesion and leads to long-term survival after treatment with 5-fluorocytosine (5-FC). Survival benefit is dose dependent for both vector and 5-FC, and as few as 4 cycles of 5-FC dosing after Toca 511 therapy provides significant survival advantage. In the virally permissive CT26-BALB/c model, spread of Toca 511 to other tissues, particularly lymphoid tissues, is detectable by polymerase chain reaction (PCR) over a wide range of levels. In the Tu-2449-B6C3F1 model, Toca 511 PCR signal in nontumor tissues is much lower, spread is not always observed, and when observed, is mainly detected in lymphoid tissues at low levels. The difference in vector genome spread correlates with a more effective antiviral restriction element, APOBEC3, present in the B6C3F1 mice. Despite these differences, neither strain showed signs of treatment-related toxicity. These data support the concept that, in immunocompetent animals, a replicating retroviral vector carrying a prodrug activating gene (Toca 511) can spread through a tumor mass, leading to selective elimination of the tumor after prodrug administration, without local or systemic pathology. This concept is under investigation in an ongoing phase I/II clinical trial of Toca 511 in combination with 5-FC in patients with recurrent high-grade glioma (www.clinicaltrials.gov NCT01156584).


Assuntos
Neoplasias Encefálicas/terapia , Flucitosina/uso terapêutico , Fluoruracila/metabolismo , Vetores Genéticos , Glioma/terapia , Vírus da Leucemia Murina/genética , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/mortalidade , Terapia Combinada , Modelos Animais de Doenças , Feminino , Flucitosina/metabolismo , Flucitosina/farmacologia , Fluoruracila/farmacologia , Terapia Genética , Vetores Genéticos/administração & dosagem , Glioma/tratamento farmacológico , Glioma/genética , Glioma/mortalidade , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sobrevida , Células Tumorais Cultivadas
10.
PLoS One ; 5(10): e13478, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20976057

RESUMO

The combination of sucrose and starch in the presence of surface-adsorbed salivary α-amylase and bacterial glucosyltransferases increase the formation of a structurally and metabolically distinctive biofilm by Streptococcus mutans. This host-pathogen-diet interaction may modulate the formation of pathogenic biofilms related to dental caries disease. We conducted a comprehensive study to further investigate the influence of the dietary carbohydrates on S. mutans-transcriptome at distinct stages of biofilm development using whole genomic profiling with a new computational tool (MDV) for data mining. S. mutans UA159 biofilms were formed on amylase-active saliva coated hydroxyapatite discs in the presence of various concentrations of sucrose alone (ranging from 0.25 to 5% w/v) or in combination with starch (0.5 to 1% w/v). Overall, the presence of sucrose and starch (suc+st) influenced the dynamics of S. mutans transcriptome (vs. sucrose alone), which may be associated with gradual digestion of starch by surface-adsorbed amylase. At 21 h of biofilm formation, most of the differentially expressed genes were related to sugar metabolism, such as upregulation of genes involved in maltose/maltotriose uptake and glycogen synthesis. In addition, the groEL/groES chaperones were induced in the suc+st-biofilm, indicating that presence of starch hydrolysates may cause environmental stress. In contrast, at 30 h of biofilm development, multiple genes associated with sugar uptake/transport (e.g. maltose), two-component systems, fermentation/glycolysis and iron transport were differentially expressed in suc+st-biofilms (vs. sucrose-biofilms). Interestingly, lytT (bacteria autolysis) was upregulated, which was correlated with presence of extracellular DNA in the matrix of suc+st-biofilms. Specific genes related to carbohydrate uptake and glycogen metabolism were detected in suc+st-biofilms in more than one time point, indicating an association between presence of starch hydrolysates and intracellular polysaccharide storage. Our data show complex remodeling of S. mutans-transcriptome in response to changing environmental conditions in situ, which could modulate the dynamics of biofilm development and pathogenicity.


Assuntos
Biofilmes , Perfilação da Expressão Gênica , Amido/farmacologia , Streptococcus mutans/efeitos dos fármacos , Sacarose/farmacologia , Sequência de Bases , Primers do DNA , Genes Bacterianos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Streptococcus mutans/genética
11.
Biochem Biophys Res Commun ; 383(2): 167-71, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19341712

RESUMO

The gene for phosphatidylinositol-4-phosphate adaptor-2 (FAPP2) encodes a cytoplasmic lipid transferase with a plekstrin homology domain that has been implicated in vesicle maturation and transport from trans-Golgi to the plasma membrane. The introduction of ribozymes targeting the FAPP2 gene in colon carcinoma cells induced their apoptosis in the presence of Fas agonistic antibody. Furthermore, by quantitative PCR we showed that a siRNA specific to FAPP2, but not a randomized siRNA control, reduced FAPP2 gene expression in tumor cells. Transfection of FAPP2 siRNA into human tumor cells then incubated with FasL resulted in reduction of viable cell numbers. Also, FAPP2 siRNA transfected glioma and breast tumor cells showed significant increases in apoptosis upon incubation with soluble FasL, but the apoptosis did not necessarily correlate with increased Fas expression. These data demonstrate a previously unknown role for FAPP2 in conferring resistance to apoptosis and indicate that FAPP2 may be a target for cancer therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Apoptose , Proteína Ligante Fas/agonistas , Neoplasias/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Anticorpos/imunologia , Apoptose/genética , Linhagem Celular Tumoral , Regulação para Baixo , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , RNA Interferente Pequeno/genética
12.
Biochem Biophys Res Commun ; 382(3): 503-7, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19303400

RESUMO

A truncated form of 24kDa FGF-2 consisting of 86 NH(2)-terminal amino acids (ATE+31) inhibits cell migration in vitro and tumor development and angiogenesis in vivo. Focal adhesion kinase (FAK) is phosphorylated on tyrosine and serine sites after cell stimulation by growth factors. This study examined the effect of ATE+31 on FAK phosphorylation in human glioma cells. FAK and Pyk phosphorylation were evaluated at serines known to be involved with cell migration. We demonstrated that ATE+31 at 3 x 10(-11)M decreases phosphorylation levels of Tyr(407)-FAK and Ser(732)-FAK in the presence of platelet-derived growth factor (PDGF), that ATE+31 in the presence of PDGF alters the distribution of FAK and other phosphotyrosine proteins in the adhesion contacts, and that ATE+31 in the presence of PDGF has no effect on the activation of Pyk2. These data suggest that the inhibition of cell migration by ATE+31 occurs via Tyr(407)-FAK and Ser(732)-FAK.


Assuntos
Movimento Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Quinase 1 de Adesão Focal/metabolismo , Glioma/patologia , Neovascularização Patológica/patologia , Linhagem Celular Tumoral , Fator 2 de Crescimento de Fibroblastos/genética , Quinase 2 de Adesão Focal/metabolismo , Glioma/irrigação sanguínea , Humanos , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Serina/metabolismo , Tirosina/metabolismo
13.
Cancer Ther ; 6(B): 865-876, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19081762

RESUMO

The PATZ1 gene encodes a transcription factor that belongs to the BTB/POZ group of transcriptional regulators and has been implicated as a transcriptional repressor. We cloned cDNA from glioma cell lines and found they expressed transcript variant 2 of PATZ1. We designed a specific siRNA against PATZ1 and showed that this siRNA, but not a control randomized siRNA, reduced PATZ1 expression in glioma cells as determined by quantitative PCR. In a panel of human glioma cell lines incubated with proapoptotic FasL, those transfected with PATZ1 siRNA displayed reduced cell numbers by the MTT colorimetric assay, relative to those transfected with randomized siRNA. Further studies showed that in 10-08-MG, U-251MG, U-87MG, and T98G cells PATZ1 siRNA significantly increased apoptosis in response to incubation with soluble FasL, as shown by a morphologic acridine orange/ethidium bromide apoptotic assay. Using an apoptosis specific cDNA microarray we further demonstrated that down-regulation of PATZ1 by siRNA resulted in the upregulation of death receptor pro-apoptotic genes including caspase 8 and Death Receptor 5 (DR5) in U-373MG cells. Since DR5 is the receptor for TRAIL we tested whether PATZ1 downregulation also sensitized cells to TRAIL-induced apoptosis and found that PATZ1 siRNA, but not control siRNA, sensitized U-251MG and T98G glioma cells to TRAIL-induced apoptosis. Altogether, these data demonstrate a previously unknown role for the transcription factor PATZ1 in conferring resistance to apoptosis and indicate that modulation of PATZ1 expression may be a therapeutic strategy for gliomas.

14.
Proc Natl Acad Sci U S A ; 103(48): 18326-31, 2006 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-17110447

RESUMO

The TGF-beta signaling pathway is a key organizer of injury and immune responses, and recent studies suggest it fulfills critical roles in CNS function and maintenance. TGF-beta receptor activation results in phosphorylation of Smad proteins, which subsequently translocate to the nucleus to regulate gene transcription by binding to Smad binding elements (SBE). Using SBE-luciferase reporter mice, we recently discovered that the brain has the highest Smad baseline activity of any major organ in the mouse, and we now demonstrate that this signal is primarily localized to pyramidal neurons of the hippocampus. In vivo excitatory stimulation with kainic acid (KA) resulted in an increase in luciferase activity and phosphorylated Smad2 (Smad2P), and nuclear translocation of Smad2P in hippocampal CA3 neurons correlated significantly with luciferase activity. Although this activation was most prominent at 24 h after KA administration in neurons, Smad2P immunoreactivity gradually increased in astrocytes and microglial cells at 3 and 5 days, consistent with reactive gliosis. Bioluminescence measured over the skull in living mice peaked at 12-72 h and correlated with the extent of microglial activation and pathological markers of neurodegeneration 5 days after injury. Treatment with the glutamate receptor antagonist MK-801 strongly reduced bioluminescence and pathology. These results show that Smad2 signaling is a sensitive marker of neuronal activation and CNS injury that can be used to monitor KA-induced neuronal degeneration. This and related mouse models may provide valuable tools to study mechanisms and treatments for neurodegeneration.


Assuntos
Degeneração Neural/metabolismo , Degeneração Neural/patologia , Transdução de Sinais , Proteínas Smad/metabolismo , Animais , Biomarcadores , Células Cultivadas , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/lesões , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Maleato de Dizocilpina/farmacologia , Genes Reporter/genética , Ácido Caínico/farmacologia , Camundongos , Microscopia Confocal
15.
J Clin Invest ; 116(11): 3060-9, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17080199

RESUMO

Alzheimer's disease (AD) is characterized by progressive neurodegeneration and cerebral accumulation of the beta-amyloid peptide (Abeta), but it is unknown what makes neurons susceptible to degeneration. We report that the TGF-beta type II receptor (TbetaRII) is mainly expressed by neurons, and that TbetaRII levels are reduced in human AD brain and correlate with pathological hallmarks of the disease. Reducing neuronal TGF-beta signaling in mice resulted in age-dependent neurodegeneration and promoted Abeta accumulation and dendritic loss in a mouse model of AD. In cultured cells, reduced TGF-beta signaling caused neuronal degeneration and resulted in increased levels of secreted Abeta and beta-secretase-cleaved soluble amyloid precursor protein. These results show that reduced neuronal TGF-beta signaling increases age-dependent neurodegeneration and AD-like disease in vivo. Increasing neuronal TGF-beta signaling may thus reduce neurodegeneration and be beneficial in AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/fisiologia , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Dendritos/metabolismo , Dendritos/patologia , Gliose/metabolismo , Gliose/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/genética , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
16.
J Immunol ; 175(1): 547-54, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15972691

RESUMO

Smad2 and Smad3 (Smad2/3) proteins are key signaling molecules for TGF-beta and some related family members regulating the transcription of several hundred genes. TGF-beta have key roles in development, tissue homeostasis, and the pathogenesis of many human diseases, including cancer, fibrotic disorders, developmental defects, and neurodegeneration. To study the temporal and spatial patterns of Smad2/3-dependent signaling in normal and pathological conditions in the living organism, we engineered transgenic mice with a Smad-responsive luciferase reporter construct (SBE-luc mice). Using bioluminescent imaging, we assessed Smad2/3 signaling activity noninvasively in living mice. At baseline, this activity was highest in brain, intestine, heart, and skin, and correlated with biochemical measurements of reporter activity. Primary astrocytes cultured from SBE-luc mice showed specific activation of the reporter in response to Smad2/3-activating TGF-beta family members. Treatment of mice with the endotoxin LPS resulted in a fast and vigorous, but transient activation of the reporter in the intestine. Although the response was similarly rapid in brain, it remained increased, indicating important but different cellular responses to endotoxin challenge in these organs. Traumatic brain injury with a needle stab resulted in local activation of Smad2/3-dependent genes and a severalfold increase in bioluminescence in living mice. SBE-luc mice can therefore be used to study temporal, tissue-specific activation of Smad2/3-dependent signaling in living mice in normal or pathological conditions as well as for the identification of endogenous or synthetic modulators of this pathway.


Assuntos
Lesões Encefálicas/imunologia , Lesões Encefálicas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Lesões Encefálicas/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Genes Reporter , Humanos , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Lipopolissacarídeos/toxicidade , Luciferases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2 , Proteína Smad3 , Distribuição Tecidual , Transativadores/genética , Fator de Crescimento Transformador beta/genética
17.
Virus Res ; 83(1-2): 43-56, 2002 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-11864740

RESUMO

The hemagglutinin (HA) protein of influenza virus binds to terminal sialic acid residues present on cell surface glycoproteins and glycolipids. The specific amino acids involved in this interaction have been identified for a H3 subtype HA from the human non-pathogenic virus, A/Aichi/2/68, by both crystallographic and mutagenesis studies. We were interested to examine the receptor-binding pocket of a H7 subtype protein from the avian pathogenic virus A/FPV/Rostock/34. Accordingly, we made amino acid substitutions at six conserved residues (Y88, T126, H174, E181, L185, and G219), suggested by comparison with the receptor-binding pocket of the H3 protein, and analyzed the resulting proteins using pseudotyped retroviral vectors. The use of these vectors enabled us to quantitate both the ability of the mutant HA proteins to bind with receptor-expressing cells, and also to promote virus-cell fusion by measuring vector titer. Using this system, we identified a subset of mutants with impaired receptor-binding activity and a corresponding decrease in titer, but which retained the ability to induce syncytia in low pH cell-cell fusion assays. The most severely affected mutants contained more than one substitution, with the triple mutant Y88F/E181Q/G219K being the most defective. These observations highlight the importance of multiple contact points for the interaction between sialic acid and HA.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A/genética , Receptores Virais/metabolismo , Células 3T3 , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular Transformada , Membrana Celular/metabolismo , Expressão Gênica , Vetores Genéticos , Células Gigantes , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Humanos , Concentração de Íons de Hidrogênio , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Vírus da Influenza A/fisiologia , Fusão de Membrana , Camundongos , Dados de Sequência Molecular , Mutagênese , Estrutura Terciária de Proteína , Receptores Virais/química , Retroviridae , Homologia de Sequência de Aminoácidos , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...