Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 310
Filtrar
1.
Osteoporos Int ; 29(5): 1147-1154, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29464277

RESUMO

We investigated changes in health-related quality of life (HRQoL) due to hip fracture in Mexican adults aged ≥ 50 years during the first year post-fracture. Mean accumulated loss was 0.27 quality-adjusted life years (QALYs). HRQoL before fracture was the main contributor to explain the loss of QALYs. INTRODUCTION: We aimed to estimate the health-related quality of life (HRQoL) loss over 1 year in patients sustaining a hip fracture in Mexico. METHODS: Individuals aged ≥ 50 years old with diagnosis of a low-energy-induced hip fracture enrolled in the International Costs and Utilities Related to Osteoporotic Fractures Study (ICUROS) composed the study population. After a recall of their own pre-fracture status, HRQoL was prospectively collected in three phases over 12 months of follow-up using EQ-5D-3L. The UK preference weight set was applied to calculate the utility values. The accumulated quality-adjusted life years (QALYs) loss in the first year post-fracture was estimated using the trapezoid method. Multivariate regression analysis allowed identifying determinants of QALYs loss. RESULTS: One hundred ninety-three patients (mean ± SD age 77.2 ± 9.9 years; 80% women; 15.5% with prior fracture in the last 5 years; 78% in low-income category) were evaluated. Mean (95% CI) utility value before fracture was 0.64 (0.59-0.68). It dropped to 0.01 (0.01-0.02) immediately after fracture and then improved to 0.46 (0.42-0.51) and 0.60 (0.55-0.64) at 4 and 12 months post-fracture, respectively. Disregarding fracture-related mortality, accumulated QALYs loss over the first year was 0.27 (0.24-0.30) QALYs. Mobility, self-care, and usual activities were the most affected domains throughout the whole year. HRQoL before fracture was the main contributor to explain the loss of QALYs. CONCLUSIONS: Hip fractures reduce dramatically the HRQoL, with the loss sustained at least over the first year post-fracture in Mexico. The utility values derived from this study can be used in future economic evaluations.


Assuntos
Fraturas do Quadril/reabilitação , Fraturas por Osteoporose/reabilitação , Qualidade de Vida , Idoso , Idoso de 80 Anos ou mais , Efeitos Psicossociais da Doença , Feminino , Seguimentos , Humanos , Masculino , México , Pessoa de Meia-Idade , Estudos Prospectivos , Psicometria , Anos de Vida Ajustados por Qualidade de Vida , Autocuidado , Fatores Socioeconômicos
2.
Gene Ther ; 20(7): 733-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23171918

RESUMO

Specific and efficient gene delivery to the lung has been hampered by liver sequestration of adenovirus serotype 5 (Ad5) vectors. The complexity of Ad5 liver tropism has largely been unraveled, permitting improved efficacy of Ad5 gene delivery. However, Kupffer cell (KC) scavenging and elimination of Ad5 still represent major obstacles to lung gene delivery strategies. KC uptake substantially reduces bioavailability of Ad5 for target tissues and compensatory dose escalation leads to acute hepatotoxicity and a potent innate immune response. Here, we report a novel lung-targeting strategy through redirection of Ad5 binding to the concentrated leukocyte pool within the pulmonary microvasculature. We demonstrate that this leukocyte-binding approach retargets Ad5 specifically to lung endothelial cells and prevents KC uptake and hepatocyte transduction, resulting in 165,000-fold enhanced lung targeting, compared with Ad5. In addition, myeloid cell-specific binding is preserved in single-cell lung suspensions and only Ad.MBP-coated myeloid cells achieved efficient endothelial cell transduction ex vivo. These findings demonstrate that KC sequestration of Ad5 can be prevented through more efficient uptake of virions in target tissues and suggest that endothelial transduction is achieved by leukocyte-mediated 'hand-off' of Ad.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Terapia Genética , Células Mieloides/citologia , Tropismo Viral , Células Endoteliais/citologia , Células Endoteliais/virologia , Vetores Genéticos , Hepatócitos/citologia , Hepatócitos/virologia , Humanos , Células de Kupffer/citologia , Células de Kupffer/virologia , Fígado/citologia , Fígado/virologia , Pulmão/citologia , Pulmão/virologia , Células Mieloides/virologia , Transdução Genética
3.
Hum Reprod ; 28(9): 2398-406, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23820419

RESUMO

STUDY QUESTION: Is targeted adenovirus vector, Ad-SSTR-RGD-TK (Adenovirus -human somatostatin receptor subtype 2- arginine, glycine and aspartate-thymidine kinase), given in combination with ganciclovir (GCV) against immortalized human leiomyoma cells (HuLM) a potential therapy for uterine fibroids? SUMMARY ANSWER: Ad-SSTR-RGD-TK/GCV, a targeted adenovirus, effectively reduces cell growth in HuLM cells and to a significantly greater extent than in human uterine smooth muscle cells (UtSM). WHAT IS KNOWN ALREADY: Uterine fibroids (leiomyomas), a major cause of morbidity and the most common indication for hysterectomy in premenopausal women, are well-defined tumors, making gene therapy a suitable and potentially effective non-surgical approach for treatment. Transduction of uterine fibroid cells with adenoviral vectors such as Ad-TK/GCV (herpes simplex virus thymidine kinase gene) decreases cell proliferation. STUDY DESIGN, SIZE, DURATION: An in vitro cell culture method was set up to compare and test the efficacy of a modified adenovirus vector with different multiplicities of infection in two human immortalized cell lines for 5 days. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immortalized human leiomyoma cells and human uterine smooth muscle cells were infected with different multiplicities of infection (MOI) (5-100 plaque-forming units (pfu)/cell) of a modified Ad-SSTR-RGD-TK vector and subsequently treated with GCV. For comparison, HuLM and UtSM cells were transfected with Ad-TK/GCV and Ad-LacZ/GCV. Cell proliferation was measured using the CyQuant assay in both cell types. Additionally, western blotting was used to assess the expression of proteins responsible for regulating proliferation and apoptosis in the cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transduction of HuLM cells with Ad-SSTR-RGD-TK/GCV at 5, 10, 50 and 100 pfu/cell decreased cell proliferation by 28, 33, 45, and 84%, respectively (P < 0.05) compared with untransfected cells, whereas cell proliferation in UtSM cells transfected with the same four MOIs of Ad-SSTR-RGD-TK/GCV compared with that of untransfected cells was decreased only by 8, 23, 25, and 28%, respectively (P < 0.01). Western blot analysis showed that, in comparison with the untargeted vector Ad-TK, Ad-SSTR-RGD-TK/GCV more effectively reduced expression of proteins that regulate the cell cycle (Cyclin D1) and proliferation (PCNA, Proliferating Cell Nuclear Antigen), and it induced expression of the apoptotic protein BAX, in HuLM cells. LIMITATIONS, REASONS FOR CAUTION: Results from this study need to be replicated in an appropriate animal model before testing this adenoviral vector in a human trial. WIDER IMPLICATIONS OF THE FINDINGS: Effective targeting of gene therapy to leiomyoma cells enhances its potential as a non-invasive treatment of uterine fibroids.


Assuntos
DNA Recombinante/metabolismo , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/metabolismo , Leiomioma/metabolismo , Miométrio/metabolismo , Transdução Genética , Neoplasias Uterinas/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Adenoviridae/patogenicidade , Apoptose , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , DNA Recombinante/efeitos adversos , DNA Recombinante/uso terapêutico , Feminino , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/efeitos adversos , Vetores Genéticos/uso terapêutico , Humanos , Leiomioma/terapia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Uterinas/terapia
4.
Gynecol Obstet Invest ; 76(2): 119-24, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23920223

RESUMO

Postoperative abdominal/pelvic peritoneal adhesions are a major source of morbidity (bowel obstruction, infertility, ectopic gestation as well as chronic pelvic pain) in women. In this study, we screened various transduction and transcription modifications of adenovirus (Ad) to identify those that support maximal Ad-mediated gene delivery to human adhesion fibroblasts, which in turn would enhance the efficacy of this novel treatment/preventative strategy for postoperative adhesions. We transduced primary cultures of human peritoneal adhesion fibroblasts with fiber-modified Ad vectors Ad5-RGD-luc, Ad5-Sigma-luc, Ad5/3-luc and Ad5-CAV2-luc as well as transcriptional targeting viruses Ad5-survivin-luc, Ad5-heparanase-luc, Ad5-mesothelin (MSLN)-CRAd-luc and Ad5-secretory leukoprotease inhibitor (SLPI)-luc, and compared their activity to wild-type Ad5-luc. At 48 h, luciferase activity was measured and normalized to the total protein content in the cells. Among the fiber-modified Ad vectors, Ad5-Sigma-luc and among the transcriptional targeting modified Ad vectors, Ad5-MSLN-CRAd-luc showed significantly increased expression levels of luciferase activity at 5, 10 and 50 plaque forming units/cell in adhesion fibroblast cells compared with wild-type Ad5-luc (p < 0.05). Specific modifications of Ad improve their gene delivery efficiency towards human peritoneal adhesion fibroblasts. Developing a safe localized method to prevent/treat postoperative adhesion formation would have a major impact on women health.


Assuntos
Adenoviridae/genética , Fibroblastos , Terapia Genética , Aderências Teciduais/terapia , Transdução Genética , Células Cultivadas , Fibroblastos/enzimologia , Expressão Gênica , Vetores Genéticos , Humanos , Luciferases/genética , Luciferases/metabolismo , Mesotelina , Aderências Teciduais/prevenção & controle , Transcrição Gênica
5.
Nat Med ; 7(12): 1339-46, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11726975

RESUMO

Dendritic-cell (DC) trafficking and function in tumors is poorly characterized, with studies confined to myeloid DCs (DC1s). Tumors inhibit DC1 migration and function, likely hindering specific immunity. The role of plasmacytoid DCs (DC2s) in tumor immunity is unknown. We show here that malignant human ovarian epithelial tumor cells express very high levels of stromal-derived factor-1, which induces DC2 precursor (preDC2) chemotaxis and adhesion/transmigration, upregulates preDC2 very late antigen (VLA)-5, and protects preDC2s from tumor macrophage interleukin-10-induced apoptosis, all through CXC chemokine receptor-4. The VLA-5 ligand vascular-cell adhesion molecule-1 mediated preDC2 adhesion/transmigration. Tumor preDC2s induced significant T-cell interleukin-10 unrelated to preDC2 differentiation or activation state, and this contributed to poor T-cell activation. Myeloid precursor DCs (preDC1s) were not detected. Tumors may weaken immunity by attracting preDC2s and protecting them from the harsh microenvironment, and by altering preDC1 distribution.


Assuntos
Carcinoma/imunologia , Quimiocinas CXC/farmacologia , Células Dendríticas/efeitos dos fármacos , Neoplasias Ovarianas/imunologia , Células-Tronco/efeitos dos fármacos , Apoptose , Carcinoma/irrigação sanguínea , Quimiocina CXCL12 , Quimiotaxia de Leucócito , Células Dendríticas/citologia , Feminino , Humanos , Interleucina-10/farmacologia , Ativação Linfocitária , Neoplasias Ovarianas/irrigação sanguínea , Receptores de Fibronectina/biossíntese , Células-Tronco/citologia , Linfócitos T/imunologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
6.
Hum Reprod ; 25(8): 2068-83, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20573677

RESUMO

BACKGROUND: Novel therapeutic approaches for endometriosis based on molecular strategies may prove to be useful. Conditionally replicative adenoviruses (CRAds) are designed to exploit key differences between target and normal cells. The wild-type adenovirus (Adwt) promoter can be replaced by tissue-specific promoters, allowing viral replication only in target cells. Viral infectivity can be enhanced by altering Ad tropism via fiber modification. We investigated whether CRAds can be used to target endometriosis and determined the most efficient transcriptional- and transductional-targeting strategy. METHODS: An in vitro study was carried out using human endometriotic cell lines, 11Z (epithelial) and 22B (stromal), normal human ovarian surface epithelial cell line (NOSE006) and primary human endometriosis cells. A total of 9 promoters and 12 Ad tropism modifications were screened by means of a luciferase reporter assay. From this screening data, three CRAds (CRAd-S-pK7, CRAd-S-RGD, CRAd-S-F5/3sigma1, all incorporating the survivin promoter but with different fiber modifications) were selected to perform experiments using Adwt and a replication-deficient virus as controls. CRAds were constructed using a plasmid recombination system. Viral-binding capacity, rates of entry and DNA replication were evaluated by quantitative real-time PCR of viral genome copy. Cell-killing effects were determined by crystal violet staining and a cell viability assay for different concentrations of viral particles per cell. RESULTS: Comparison of promoters demonstrated that the survivin promoter exhibited the highest induction in both endometriotic cell lines. Among the fiber-modified viruses, the polylysine modification (pK7) showed the best infection enhancement. CRAd-S-pK7 was validated as the optimal CRAd to target endometriosis in terms of binding ability, entry kinetics, DNA replication and cell-killing effect. CRAd-S-pK7 also exhibited a high level of DNA replication in primary endometriosis cells. CONCLUSIONS: CRAd-S-pK7 has the best infection and cell-killing effect in the context of endometriosis. It could prove to be a useful novel method to target refractory cases of endometriosis.


Assuntos
Adenoviridae/genética , Endometriose/terapia , Replicação Viral , Linhagem Celular , Endometriose/genética , Feminino , Humanos , Regiões Promotoras Genéticas , Ligação Viral , Replicação Viral/genética
7.
Gene Ther ; 15(10): 716-29, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18369326

RESUMO

Use of cells as therapeutic carriers has increased in the past few years and has developed as a distinct concept and delivery method. Cell-based vehicles are particularly attractive for delivery of biotherapeutic agents that are difficult to synthesize, have reduced half-lives, limited tissue penetrance or are rapidly inactivated upon direct in vivo introduction. Initial studies using cell-based approaches served to identify some of the key factors for the success of this type of therapeutic delivery. These factors include the efficiency of cell loading with a therapeutic payload, the means of cell loading and the nature of therapeutics that cells can carry. However, one important aspect of cell-based delivery yet to be fully investigated is the process of actual delivery of the cell payload in vivo. In this regard, the potential ability of cell carriers to provide site-specific or targeted delivery of therapeutics deserves special attention. The present review focuses on a variety of targeting approaches that may be utilized to improve cell-based therapeutic delivery strategies. The different aspects of targeting that can be applied to cell vehicles will be discussed, including physical methods for directing cell distribution, intrinsic cell-mediated homing mechanisms and the feasibility of engineering cells with novel targeting mechanisms. Development of cell targeting strategies will further advance cell vehicle applications, broaden the applicability of this delivery approach and potentiate therapeutic outcomes.


Assuntos
Transplante de Células/métodos , Terapia Genética/métodos , Neoplasias/terapia , Movimento Celular , Proliferação de Células , Humanos , Neoplasias/patologia
8.
Gene Ther ; 15(4): 298-308, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18046426

RESUMO

CD40 ligation has been shown to promote antigen-presenting functions of dendritic cells, which express CD40 receptor. Here we reported significantly altered biodistribution and immune responses with the use of CD40-targeted adenovirus. Compared with unmodified adenovirus 5, the CD40-targeted adenovirus following intravenous administration (i.v.) resulted in increased transgene expressions in the lung and thymus, which normally do not take up significant amounts of adenovirus. Intradermal injection saw modified adenovirus being mainly processed in local draining lymph nodes and skin. Following intranasal administration (i.n.), neither unmodified nor targeted viruses were found to be in the liver or spleen, which predominantly took up the virus following i.v. administration. However, inadvertent infection of the brain was found with unmodified adenoviruses, with the second highest gene expression among 14 tissues examined. Importantly, such undesirable effects were largely ablated with the use of targeted vector. Moreover, the targeted adenovirus elicited more sustained antigen-specific cellular immune responses (up to 17-fold) at later time points (30 days post boosting), but also significantly hampered humoral responses irrespective of administration routes. Additional data suggest the skewed immune responses induced by the targeted adenoviruses were not due to the identity of the transgene but more likely a combination of overall transgene load and CD40 stimulation.


Assuntos
Adenoviridae/genética , Antígenos CD40/genética , Células Dendríticas/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Distribuição Tecidual , Transgenes
9.
Hum Reprod ; 23(3): 514-24, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18184643

RESUMO

BACKGROUND: To circumvent the paucity of the primary adenovirus (Ad5) receptor and the non-specific Ad5 tropism in the context of uterine leiomyoma cells, Ad5 modification strategies would be beneficial. METHODS: We screened several modified adenoviruses to identify the most efficient and selective virus toward human leiomyoma cells to be used as candidate for delivering therapeutic genes. We propagated: wild-type Ad5-luc, fiber-modified viruses: ad5 RGD-luc, Ad5-Sigma-luc, Ad5/3-luc and Ad5-CAV2-luc, as well as transcriptional targeted viruses: ad5 survivin-luc, Ad5-heparanase-luc, Ad5-MSLN-CRAD-luc and Ad5-SLPI-luc, on 293 cells and purified them by double CsCL density centrifugation. Then we transfected primary cultures of human leiomyoma cells derived from fibroids of four different patients, telomerase-immortalized human leiomyoma cell line (huLM), telomerase-immortalized normal human myometrial cell line (HM9) and immortalized normal human liver cells (THLE3) with the viruses at 5, 10 and 50 plaque-forming units (PFU)/cell. After 48 h, luciferase activities were measured and normalized to the total cellular protein content. RESULTS: Ad5-RGD-luc and Ad5-CAV2-luc, Ad5-SLPI-luc and Ad5-MSLN-CRAD-luc at 5, 10 and 50 pfu/cell showed significantly higher expression levels of luciferase activity in both primary and immortalized human leiomyoma cells when compared with Ad5-Luc. Additionally, these modified viruses demonstrated selectivity toward leiomyoma cells, compared with myometrial cells and exhibited lower liver cell transduction, compared with Ad5-luc, at the same dose levels. CONCLUSIONS: Ad5-CAV2-luc, Ad5-RGD-luc, Ad5-SLPI-luc and Ad5-MSLN-CRAD-luc are promising delivery vehicles in the context of leiomyoma gene therapy.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Leiomioma/terapia , Leiomioma/virologia , Feminino , Humanos , Fígado/citologia , Mesotelina , Miométrio/citologia , Miométrio/virologia , Receptores Virais/genética
11.
J Clin Invest ; 83(4): 1144-52, 1989 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2539391

RESUMO

alpha 1-Antitrypsin (alpha 1AT) deficiency is a hereditary disorder associated with reduced serum alpha 1AT levels and the development of pulmonary emphysema. An alpha 1AT gene is defined as "Null" when no alpha 1AT in serum is attributed to that alpha 1AT gene. Although all alpha 1AT Null genes have identical phenotypic consequences (i.e. no detectable alpha 1AT in the serum), different genotypic mechanisms can cause the Null state. This study defines the molecular basis for the alpha 1AT gene Nullmattawa, identified and cloned from genomic DNA of an individual with the Null-Null phenotype and emphysema resulting from the heterozygous inheritance of the Nullmattawa and Nullbellingham genes. Sequencing of exons Ic-V and all exon-intron junctions of the Nullmattawa gene demonstrated it was identical to the common normal M1(Val213) alpha 1AT gene except for the insertion of a single nucleotide within the coding region of exon V, causing a 3' frameshift with generation of a premature stop signal. Family analysis using oligonucleotide probes specific for the Nullmattawa sequence demonstrated the gene was inherited in an autosomal fashion. Examination of blood monocytes demonstrated that a normal-sized, 1.8-kb alpha 1AT mRNA transcript is associated with the Nullmattawa gene and in vitro translation of mRNA with the Nullmattawa mutation showed it translated at a normal rate but produced a truncated alpha 1AT protein. Additionally, retroviral transfer of the alpha 1AT Nullmattawa cDNA to murine fibroblasts demonstrated no detectable intracellular or secreted alpha 1AT, despite the presence of alpha 1AT Nullmattawa mRNA transcripts. These findings are consistent with the concept that the molecular pathophysiology of Nullmattawa is likely manifested at a posttranslational level. The identification of the Nullmattawa gene supports the concept that Null alpha 1AT alleles represent a heterogenous group in which very different mechanisms cause the identical phenotypic state.


Assuntos
Alelos , Elementos de DNA Transponíveis , Mutação , Enfisema Pulmonar/genética , Deficiência de alfa 1-Antitripsina , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Linhagem , Biossíntese de Proteínas , Enfisema Pulmonar/metabolismo , RNA Mensageiro/análise , Transcrição Gênica , Transfecção , alfa 1-Antitripsina/biossíntese , alfa 1-Antitripsina/genética
12.
J Clin Invest ; 103(4): 543-53, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10021463

RESUMO

To maintain the integrity of the vascular barrier, endothelial cells (EC) are resistant to cell death. The molecular basis of this resistance may be explained by the function of antiapoptotic genes such as bcl family members. Overexpression of Bcl-2 or Bcl-XL protects EC from tumor necrosis factor (TNF)-mediated apoptosis. In addition, Bcl-2 or Bcl-XL inhibits activation of NF-kappaB and thus upregulation of proinflammatory genes. Bcl-2-mediated inhibition of NF-kappaB in EC occurs upstream of IkappaBalpha degradation without affecting p65-mediated transactivation. Overexpression of bcl genes in EC does not affect other transcription factors. Using deletion mutants of Bcl-2, the NF-kappaB inhibitory function of Bcl-2 was mapped to bcl homology domains BH2 and BH4, whereas all BH domains were required for the antiapoptotic function. These data suggest that Bcl-2 and Bcl-XL belong to a cytoprotective response that counteracts proapoptotic and proinflammatory insults and restores the physiological anti-inflammatory phenotype to the EC. By inhibiting NF-kappaB without sensitizing the cells (as with IkappaBalpha) to TNF-mediated apoptosis, Bcl-2 and Bcl-XL are prime candidates for genetic engineering of EC in pathological conditions where EC loss and unfettered activation are undesirable.


Assuntos
Apoptose , Proteínas I-kappa B , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Animais , Linhagem Celular Transformada , Células Cultivadas , Cicloeximida/farmacologia , Proteínas de Ligação a DNA/metabolismo , Endotélio Vascular/citologia , Humanos , Camundongos , Inibidor de NF-kappaB alfa , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição RelA , Ativação Transcricional , Fator de Necrose Tumoral alfa/farmacologia , Proteína bcl-X
13.
J Clin Invest ; 105(6): 813-21, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10727450

RESUMO

We assessed the effect of modified antigen presenting cells (APCs) expressing high levels of Fas ligand (APC-FasL) on post-viral chronic inflammatory disease. FasL-deficient B6-gld/gld mice infected with murine cytomegalovirus (MCMV) cleared the virus from their lungs, kidneys, and livers within 2 weeks of infection. However, inflammation persisted in these organs for more than 8 weeks, with a chronically increased T-cell response to MCMV-infected APCs and production of autoantibodies. Administration of APC-AdFasL at 4 weeks suppressed this inflammation and diminished the T-cell response and autoantibody production. APC-AdFasL that had been transfected with ultraviolet-irradiated MCMV were more effective than uninfected APC-AdFasL in ameliorating the chronic inflammation. APC-AdFasL migrated preferentially to the spleen, where they triggered apoptosis of lymphocytes in the marginal zone of the spleen. These results confirm that Fas-mediated apoptosis is not required for clearance of virus, but is required for down-modulation of the virally induced chronic inflammatory response. This organwide effect of APC-AdFasL appears to be mediated by elimination of activated T lymphocytes in the spleen before their emigration to the target organs.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Apoptose , Infecções por Citomegalovirus/imunologia , Glicoproteínas de Membrana/fisiologia , Adenoviridae/genética , Animais , Células Apresentadoras de Antígenos/transplante , Autoanticorpos/biossíntese , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/terapia , Proteína Ligante Fas , Feminino , Vetores Genéticos/genética , Hepatite Viral Animal/etiologia , Hepatite Viral Animal/imunologia , Hepatite Viral Animal/patologia , Inflamação , Rim/patologia , Fígado/patologia , Pulmão/patologia , Doenças Pulmonares Intersticiais/etiologia , Doenças Pulmonares Intersticiais/imunologia , Doenças Pulmonares Intersticiais/patologia , Macrófagos/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos MRL lpr , Camundongos Mutantes , Nefrite/etiologia , Nefrite/imunologia , Nefrite/patologia , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/patologia , Subpopulações de Linfócitos T/imunologia , Transfecção , Receptor fas/fisiologia
14.
J Clin Invest ; 96(6): 2980-9, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8675670

RESUMO

Specific killing of erbB-2-overexpressing tumor cells can be achieved using expression of an intracellular antibody directed against the erbB-2 oncoprotein. We have developed a strategy using a recombinant adenovirus encoding an anti-erbB-2 single chain antibody to achieve targeted tumor cell killing in vivo and can show significantly prolonged survival of animals carrying a human ovarian carcinoma tumor burden within their peritoneal cavities. This strategy of gene therapy for ovarian carcinoma offers the potential to achieve highly specific, targeted killing of human tumor cells and thus establishes the rationale to undertake human clinical trials on this basis.


Assuntos
Anticorpos/imunologia , Neoplasias Ovarianas/patologia , Receptor ErbB-2/imunologia , Adenoviridae , Animais , Anticorpos/uso terapêutico , Formação de Anticorpos , Feminino , Terapia Genética , Vetores Genéticos , Células HeLa , Humanos , Imunoterapia , Camundongos , Camundongos Nus , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/terapia , Cavidade Peritoneal , Receptor ErbB-2/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/uso terapêutico , Transfecção , Transplante Heterólogo
15.
Cancer Gene Ther ; 14(1): 105-16, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17024232

RESUMO

Treatment of advanced lung cancer is one of the major challenges in current medicine because of the high morbidity and mortality of the disease. Advanced stage lung cancer is refractory to conventional therapies and has an extremely poor prognosis. Thus, new therapeutic approaches are needed. Lung tumor formation depends on angiogenesis in which the vascular endothelial growth factor (VEGF) produced by cancer cells plays a pivotal role. Neutralizing VEGF with a soluble VEGF receptor suppresses tumor growth; however, the anticancer effect with this therapy is weakened after the intratumoral vascular network is completed. In this study, we turned the expression of VEGF by tumors to therapeutic advantage using a conditionally replication-competent adenovirus (CRAd) in which the expression of E1 is controlled by the human VEGF promoter. This virus achieved good levels of viral replication in lung cancer cells and induced a substantial anticancer effect in vitro and in vivo. As a further enhancement, the cancer cell killing effect was improved with tropism modification of the virus to express the knob domain of Ad3, which improved infectivity for cancer cells. These VEGF promoter-based CRAds also showed a significant cell killing effect for various types of cancer lines other than lung cancer. Conversely, the VEGF promoter has low activity in normal tissues, and the CRAd caused no damage to normal bronchial epithelial cells. Since tumor-associated angiogenesis via VEGF signalling is common in many types of cancers, these CRAds may be applicable to a wide range of tumors. We concluded that VEGF promoter-based CRAds have the potential to be an effective strategy for cancer treatment.


Assuntos
Adenoviridae/genética , Terapia Genética , Neoplasias/terapia , Regiões Promotoras Genéticas , Fator A de Crescimento do Endotélio Vascular/genética , Replicação Viral , Adenoviridae/fisiologia , Sequência de Bases , Primers do DNA , Humanos , Transgenes
16.
Cancer Gene Ther ; 14(4): 421-30, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17235353

RESUMO

Oncolytic viruses represent a novel cancer treatment strategy. Despite their promising preclinical data, however, corresponding clinical trials have disappointed. To aid preclinical analyses, we hypothesized that three-dimensional tumor cell clusters or spheroids might provide an assay system superior to conventional monolayer cell cultures. Spheroids show viral infection, replication and oncolytic patterns distinct from conventional monolayer assays. Therefore, viral tumor penetration and oncolysis measurements may be improved with such three-dimensional models. Also, preclinical analyses of oncolytic viruses frequently measure mitochondrial activity, but more accurate measures of oncolysis might involve quantitation of intracellular protein release. Therefore, we measured luciferase released from luciferase-expressing spheroids and found unique patterns that maintained consistency with various viruses and doses. The relative variations between viruses and doses may represent temporal differences in oncolysis dynamics. Analysis of five recombinant replicative adenoviruses with promise for clinical application showed that Ad5/3-Delta24 produced the most luciferase release 1 week after infection and achieved the earliest and highest peak luciferase release level. Ad5/3-Delta24 also effected the earliest subtotal spheroid cell death. These findings closely parallel monolayer oncolysis assays with these agents. Therefore, the luciferase-expressing tumor spheroid assay represents a promising three-dimensional model for preclinical analysis of replicative oncolytic agents.


Assuntos
Adenoviridae/fisiologia , Bioensaio , Luciferases/análise , Vírus Oncolíticos/fisiologia , Replicação Viral , Adenoviridae/genética , Humanos , Luciferases/genética , Vírus Oncolíticos/genética , Esferoides Celulares/virologia , Células Tumorais Cultivadas
17.
Bone ; 40(4): 797-812, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17189720

RESUMO

This review begins with an introduction to the malignant bone tumor, osteosarcoma [OS] and then moves to a discussion of the commonly used vectors for gene transfer. We first briefly highlight non-viral vectors including polymeric and liposomal delivery systems but concentrate predominantly on the 5 leading viral vectors used in cancer gene therapy, specifically retroviruses, adeno-associated viruses, herpes viruses and lentiviruses with the most detailed analysis reserved for adenoviruses. The 3 main strategies for gene therapy in osteosarcoma are next summarized. As part of this review, the several prodrug-converting enzymes utilized in OS suicide gene therapy are examined. The text then turns to a discussion of adenovirus-mediated gene transfer and the need for tumor targeting via transductional or transcriptional approaches. Because of practical problems with use of replication-incompetent viruses in achieving complete tumor kill in vivo, virotherapy utilizing replication competent viruses has come to the fore. This topic is, thus, next reviewed which allows for a natural transition to a discussion of armed therapeutic viruses many of which are conditionally replicating adenoviruses carrying transgenes with established anti-tumor efficacy. We recognize that several other issues have arisen which hamper progress in the field of cancer gene therapy. We, therefore, review viral-induced toxicity in the host and vector delivery issues which have been found to potentially influence safety. We end with a brief perspective including commenting on animal models used in examining delivery strategies for osteosarcoma gene therapy. The challenges remaining are touched upon most especially the need to deal with pulmonary metastatic disease from OS.


Assuntos
Neoplasias Ósseas/terapia , Terapia Genética/métodos , Osteossarcoma/terapia , Genes Transgênicos Suicidas , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Mutação , Terapia Viral Oncolítica , Segurança , Vírus/genética
18.
Mol Cell Biol ; 10(1): 47-56, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1967187

RESUMO

The Mmineral springs alpha 1-antitrypsin (alpha 1AT) allele, causing alpha 1AT deficiency and emphysema, is unique among the alpha 1AT-deficiency alleles in that it was observed in a black family, whereas most mutations causing alpha 1AT deficiency are confined to Caucasian populations of European descent. Immobilized pH gradient analysis of serum demonstrated that alpha 1AT Mmineral springs migrated cathodal to the normal M2 allele. Evaluation of Mmineral springs alpha 1AT as an inhibitor of neutrophil elastase, its natural substrate, demonstrated markedly lower than normal function. Characterization of the alpha 1AT Mmineral springs gene demonstrated that it differed from the common normal M1(Ala213) allele by a single-base substitution causing the amino acid substitution Gly-67 (GGG)----Glu-67 (GAG). Capitalizing on the fact that this mutation creates a polymorphism for the restriction endonuclease AvaII, family analysis demonstrated that the Mmineral springs alpha 1AT allele was transmitted in an autosomal-codominant fashion. Evaluation of genomic DNA showed that the index case was homozygous for the alpha 1AT Mmineral springs allele. Cytoplasmic blot analysis of blood monocytes of the Mmineral springs homozygote demonstrated levels of alpha 1AT mRNA transcripts comparable to those in cells of a normal M1 (Val213) homozygote control. Evaluation of in vitro translation of Mmineral springs alpha 1AT mRNA transcripts demonstrated a normal capacity to direct the translation of alpha 1AT. Evaluation of secretion of alpha 1AT by the blood monocytes by pulse-chase labeling with [35S]methionine, however, demonstrated less secretion by the Mmineral springs cells than normal cells. To characterize the posttranslational events causing the alpha 1AT-secretory defect associated with the alpha 1AT Mmineral springs gene, retroviral gene transfer was used to establish polyclonal populations of murine fibroblasts containing either a normal human M1 alpha 1AT cDNA or an Mmineral springs alpha 1AT cDNA and expressing comparable levels of human alpha 1AT mRNA transcripts. Pulse-chase labeling of these cells with [35S]methionine demonstrated less secretion of human alpha 1AT from the Mmineral springs cells than from the M1 cells, and evaluation of cell lysates also demonstrated lower amounts of intracellular human alpha 1AT in the Mmineral springs cells than in the normal M1 control cells. Thus, the Gly-67 --> Glu mutation that characterizes Mmineral springs causes reduced alpha 1AT secretion on the basis of aberrant posttranslational alpha 1AT biosynthesis by a mechanism distinct from that associated with the alpha 1AT Z allele, whereby intracellular aggregation of the mutant protein is etiologic of the alpha 1AT-secretory defect. Furthermore, for the alpha 1AT protein that does reach the circulation, this mutation markedly affects the ability of the molecule to inhibit neutrophil elastase; i.e., the alpha 1AT Mmineral springs allele predisposes to emphysema on the basis of serum apha 1AT deficiency coupled with alpha AT dysfunction.


Assuntos
Enfisema/genética , Deficiência de alfa 1-Antitripsina , Alelos , Sequência de Bases , Clonagem Molecular , Regulação da Expressão Gênica , Humanos , Ponto Isoelétrico , Dados de Sequência Molecular , Monócitos/enzimologia , Linhagem , Polimorfismo de Fragmento de Restrição , Biossíntese de Proteínas , RNA Mensageiro/genética , Transcrição Gênica , alfa 1-Antitripsina/genética
19.
Nat Biotechnol ; 18(7): 723-7, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10888838

RESUMO

Rapid advances are being made in the engineering of replication-competent viruses to treat cancer. Adenovirus is a mildly pathogenic human virus that propagates prolifically in epithelial cells, the origin of most human cancers. While virologists have revealed many details about its molecular interactions with the cell, applied scientists have developed powerful technologies to genetically modify or regulate every viral protein. In tandem, the limited success of nonreplicative adenoviral vectors in cancer gene therapy has brought the old concept of adenovirus oncolysis back into the spotlight. Major efforts have been directed toward achieving selective replication by the deletion of viral functions dispensable in tumor cells or by the regulation of viral genes with tumor-specific promoters. However, the predicted replication selectivity has not been realized because of incomplete knowledge of the complex virus-cell interactions and the leakiness of cellular promoters in the viral genome. Capsid modifications are being developed to achieve tumor targeting and enhance infectivity. Cellular and viral functions that confer greater oncolytic potency are also being elucidated. Ultimately, the interplay of the virus with the immune system will likely dictate the success of this approach as a cancer therapy.


Assuntos
Adenoviridae/genética , Adenoviridae/fisiologia , Terapia Genética/métodos , Neoplasias/terapia , Animais , Vetores Genéticos , Humanos , Modelos Biológicos , Neoplasias/imunologia
20.
Nat Biotechnol ; 14(11): 1574-8, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9634824

RESUMO

The utility of adenoviral vectors for gene therapy is currently limited due, in part, to the widespread distribution of the cellular receptor for the adenovirus fiber that precludes the targeting of specific cell types. In order to develop a targeted adenovirus, it is therefore necessary both to ablate endogenous viral tropism and to introduce novel tropism. We hypothesized that these two goals could be achieved by employing a neutralizing anti-fiber antibody, or antibody fragment, chemically conjugated to a cell-specific ligand. To test this concept, we chose to target the folate receptor, which is overexpressed on the surface of a variety of malignant cells. Therefore, we conjugated folate to the neutralizing Fab fragment of an anti-fiber monoclonal antibody. This Fab-folate conjugate was complexed with an adenoviral vector carrying the luciferase reporter gene and was shown to redirect adenoviral infection of target cells via the folate receptor at a high efficiency. Furthermore, when complexed with an adenoviral vector carrying the gene for herpes simplex virus thymidine kinase, the Fab-folate conjugate mediated the specific killing of cells that overexpress the folate receptor. This work thus represents the first demonstration of the retargeting of a recombinant adenoviral vector via a non-adenoviral cellular receptor.


Assuntos
Adenoviridae/genética , Proteínas de Transporte/metabolismo , Vetores Genéticos , Imunoconjugados/metabolismo , Fragmentos Fab das Imunoglobulinas/metabolismo , Receptores de Superfície Celular/metabolismo , Adenoviridae/enzimologia , Animais , Linhagem Celular , Receptores de Folato com Âncoras de GPI , Regulação Viral da Expressão Gênica/genética , Técnicas de Transferência de Genes , Genes Reporter , Terapia Genética , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Luciferases/genética , Peptídeos/imunologia , Proteínas de Protozoários , Simplexvirus/enzimologia , Timidina Quinase/genética , Tropismo/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa