Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Genet Mol Res ; 15(2)2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27173256

RESUMO

The aim of the current study was to examine matrix metalloproteinase-2 (MMP-2) and tissue inhibitor of metalloproteinase-2 (TIMP-2) expression in patients with cervical disc herniation (CDH). A total of 127 specimens from CDH patients undergoing posterior spinal surgery were obtained for the case group, which was divided into three subgroups: lateral protrusion (N = 102), median protrusion (N = 18), and paramedian protrusion (N = 7). Another 55 specimens from subjects who had cervical spine trauma and underwent spinal canal decompression were obtained for the control group. Routine hematoxylin and eosin staining was performed for pathological diagnosis. Immunohistochemical (IHC) analysis was used to determine MMP-2 and TIMP-2 expression. Under light microscopy, MMP-2-positive cells presented brown-yellow or dark brown staining in the cell membrane or cytoplasm. MMP-2 expression in the case group was significantly higher than that in controls (P < 0.05). Furthermore, MMP-2 expression in the lateral and median protrusion groups was significantly higher compared to that in the paramedian protrusion group (both P < 0.05), while there was no apparent difference in MMP-2 expression between the lateral and median protrusion groups (P > 0.05). IHC results showed that TIMP-2 expression in cases was significantly lower than that in controls (P < 0.05). Spearman correlation analysis indicated that MMP- 2 was negatively correlated with TIMP-2 expression (r = -0.418, P < 0.001). In conclusion, MMP-2 expression increased, whereas TIMP- 2 expression decreased in CDH patients, suggesting that MMP-2 and TIMP-2 expression may contribute to CDH development.


Assuntos
Deslocamento do Disco Intervertebral/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Adulto , Estudos de Casos e Controles , Vértebras Cervicais/metabolismo , Vértebras Cervicais/patologia , Feminino , Humanos , Deslocamento do Disco Intervertebral/genética , Deslocamento do Disco Intervertebral/patologia , Masculino , Metaloproteinase 2 da Matriz/genética , Pessoa de Meia-Idade , Inibidor Tecidual de Metaloproteinase-2/genética
2.
Orthop Traumatol Surg Res ; 98(5): 576-82, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22906427

RESUMO

PURPOSE: To evaluate the feasibility and reliability of free vascularized fibular graft with skin island flap for reconstruction of large diaphyseal bone defect. METHOD: The clinical results of vascularized fibular graft and experiences related to the importance and reliability of a monitoring island flap for the reconstruction of various long-bone defects were reviewed in 87 patients. RESULTS: Bony reconstruction was achieved in 82 of the 87 patients. Arterial thrombosis of anastomosed vessel in two patients and venous congestion of monitoring flap in nine patients occurred in the early postoperative periods. All of them were managed by immediate thrombectomy and reanastomosis, alternatively the thrombotic veins were replaced by new veins to anastomose with the superficial veins in five patients. Partial flap necrosis was noted in six patients, but additional surgical intervention was not required. The vascularized fibula survived and bony fusion was achieved in all patients. Postoperative stress fractures of the fibula graft occurred in 19 (21.8%) patients (once in seven patients, twice in five patients, three or more times in seven) as the mechanical stress to the graft increased. Included fracture on the tibia in 12 patients, humerus in one and femur in six. Treatments included casting in 11 patients, percutaneous pinning in one case, and adjustment of external fixator in seven patients. Bony union was finally achieved an average of 9.6 months after fracture. CONCLUSIONS: Correct alignment between the recipient bone and the external fixator is a prerequisite to preventing graft fracture. Vascularized fibula transfer is a valuable procedure for long-bone defects, and a skin island-monitoring flap is a simple, extremely useful, and reliable method for assessing the vascular status of vascularized fibula. LEVEL OF EVIDENCE: Level IV. Retrospective study.


Assuntos
Transplante Ósseo/métodos , Diáfises/cirurgia , Fíbula/transplante , Procedimentos de Cirurgia Plástica/métodos , Transplante de Pele/métodos , Retalhos Cirúrgicos/irrigação sanguínea , Fraturas da Tíbia/cirurgia , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Seguimentos , Sobrevivência de Enxerto , Humanos , Masculino , Pessoa de Meia-Idade , Período Pós-Operatório , Reprodutibilidade dos Testes , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
3.
Cytokine Growth Factor Rev ; 18(5-6): 389-94, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17692556

RESUMO

Intravesical administration of interferon alpha-2b protein (IFN) has been successfully used in the treatment of patients with superficial bladder tumors. Local dosing of IFN minimizes well-known systemic side effects of the drug, but exposure to bladder tumors is limited by the duration of instillation and transient concentrations achieved in the urothelium. Intravesical delivery of the gene encoding interferon results in an alternative strategy for IFN-based therapy of the disease, enabling sustained exposure of IFN protein that results from production by tumor and non-tumor cells in the urothelium. Efficient gene delivery and expression of IFN has been achieved using a recombinant adenovirus gene delivery system (rAd-IFN) in conjunction with the novel small molecule excipient Syn3. Studies with rAd-IFN/Syn3 in animal models result in urine concentrations of IFN that persisted for weeks and correlated with potent anti-tumor effects. The objective of this review is to communicate the rationale and preclinical findings that support ongoing clinical investigation of intravesical rAd-IFN/Syn3 in superficial bladder cancer.


Assuntos
Ácidos Cólicos/administração & dosagem , Dissacarídeos/administração & dosagem , Excipientes/administração & dosagem , Interferon-alfa/administração & dosagem , Interferon-alfa/genética , Neoplasias da Bexiga Urinária/terapia , Adenoviridae/genética , Administração Intravesical , Animais , Ácidos Cólicos/uso terapêutico , Dissacarídeos/uso terapêutico , Excipientes/uso terapêutico , Terapia Genética , Humanos , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Proteínas Recombinantes , Neoplasias da Bexiga Urinária/metabolismo , Urotélio/metabolismo
4.
Exp Eye Res ; 77(3): 355-65, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12907168

RESUMO

Previous studies suggest that local gene therapy with rAd-p21(WAF1/Cip-1) [. Arch. Ophthalmol. 120, (2002) 941-949] may provide an effective adjunctive anti-proliferative treatment to prevent glaucoma surgery failure. To further investigate rAd-p21 in this indication, we have characterized several parameters of local gene delivery to conjunctiva including, vector delivery and transgene expression in target tissue, inflammatory response, biodistribution to non-target tissues, and immune response. Quantitative PCR and RT-PCR assays were employed to evaluate rAd-p21 dissemination and gene transfer following a single subconjunctival injection. In target tissue, significant levels of rAd-p21 DNA were found in 6/6 animals 1 and 4 days after injection. rAd-p21 DNA and RNA could be detected in the un-injected contralateral eye but at levels that were 10000-100000 lower than in the injected eye. Expression of human p21 transgene in conjunctival fibroblasts was confirmed by immunohistochemistry. Biodistribution of rAd-p21 following subconjunctival injection was substantially limited to ocular tissue. In 1/6 rabbits, rAd-p21 DNA was found in whole blood, liver, and spleen at levels that were barely detectable. All non-target organs were negative on day 4. In contrast, in a rabbit injected intravenously as a positive control, all blood samples and tissues samples were positive. rAd-p21 delivery to conjunctiva followed by filtration surgery caused an early acute inflammatory response, which by day 14 was indistinguishable from placebo-treated eyes. Neutralizing anti-adenovirus antibodies were detected following administration of rAd-p21 to conjunctiva, however, vector delivery and transgene expression were unaffected in a subsequent administration to the contralateral eye in the same animal. These results show that local delivery to conjunctiva may be a suitable delivery mode for ocular gene therapy.


Assuntos
Adenoviridae/genética , Túnica Conjuntiva/fisiologia , Técnicas de Transferência de Genes , Adenoviridae/imunologia , Animais , Anticorpos Antivirais/imunologia , DNA Viral/análise , Expressão Gênica/genética , Vetores Genéticos/genética , Glaucoma/cirurgia , Humanos , Imuno-Histoquímica/métodos , Inflamação/imunologia , Injeções , Fígado/metabolismo , Reação em Cadeia da Polimerase/métodos , RNA Viral/análise , Coelhos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Baço/metabolismo , Fatores de Tempo , Transgenes/genética
5.
Cancer Gene Ther ; 8(10): 788-95, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11687902

RESUMO

A recombinant adenovirus expressing human interferon alpha2b driven by the cytomegalovirus promoter, IACB, was shown to produce and secrete biologically active protein in vitro and in vivo. Intravenous administration of IACB in Buffalo rats resulted in circulating levels of biologically active human interferon at 70,000 international units/mL for up to 15 days. Distribution of interferon protein after IACB administration was different from that seen with the subcutaneous delivery of interferon protein. Higher levels of interferon protein were observed in liver and spleen after IACB delivery compared to protein delivery. The antitumor efficacy of IACB, as measured by suppression of tumor growth, was tested in athymic nude mice bearing established human tumor xenografts from different types of human cancer. Subcutaneous tumors most responsive to the intratumoral administration of IACB ranked as U87MG (glioblastoma) and K562 (chronic myelogenous leukemia), followed by Hep 3B (hepatocellular carcinoma) and LN229 cells (glioblastoma). Intravenous administration of IACB in animals bearing U87MG or Hep 3B xenografts was also effective in suppressing tumor growth, although to a lesser extent than the intratumoral administration. IACB was also tested in a metastatic model in beige/SCID mice generated with H69 (small cell lung carcinoma) cells and was found to prolong survival in tumor-bearing animals. This suggested that interferon gene delivery can be effective in suppressing tumor growth in a wide variety of cells.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos , Interferon-alfa/genética , Neoplasias Experimentais/terapia , Animais , Western Blotting , Divisão Celular , Citomegalovirus/genética , Primers do DNA/química , Feminino , Humanos , Interferon alfa-2 , Interferon-alfa/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Neoplasias Experimentais/mortalidade , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Transplante Heterólogo , Células Tumorais Cultivadas
6.
J Virol ; 75(18): 8733-41, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11507218

RESUMO

In vitro experiments have demonstrated intercellular trafficking of the VP22 tegument protein of herpes simplex virus type 1 from infected cells to neighboring cells, which internalize VP22 and transport it to the nucleus. VP22 also can mediate intercellular transport of fusion proteins, providing a strategy for increasing the distribution of therapeutic proteins in gene therapy. Intercellular trafficking of the p53 tumor suppressor protein was demonstrated in vitro using a plasmid expressing full-length p53 fused in-frame to full-length VP22. The p53-VP22 chimeric protein induced apoptosis both in transfected tumor cells and in neighboring cells, resulting in a widespread cytotoxic effect. To evaluate the anti-tumor activity of p53-VP22 in vivo, we constructed recombinant adenoviruses expressing either wild-type p53 (FTCB) or a p53-VP22 fusion protein (FVCB) and compared their effects in p53-resistant tumor cells. In vitro, treatment of tumor cells with FVCB resulted in enhanced p53-specific apoptosis compared to treatment with equivalent doses of FTCB. However, in normal cells there was no difference in the dose-related cytotoxicity of FVCB compared to that of FTCB. In vivo, treatment of established tumors with FVCB was more effective than equivalent doses of FTCB. The dose-response curve to FVCB was flatter than that to FTCB; maximal antitumor responses could be achieved using FVCB at doses 1 log lower than those obtained with FTCB. Increased antitumor efficacy was correlated with increased distribution of p53 protein in FVCB-treated tumors. This study is the first demonstration that VP22 can enhance the in vivo distribution of therapeutic proteins and improve efficacy in gene therapy.


Assuntos
Adenovírus Humanos , Vetores Genéticos , Herpesvirus Humano 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Estruturais Virais/metabolismo , Animais , Apoptose , Células COS , Caspase 9 , Caspases/metabolismo , Chlorocebus aethiops , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ativação Enzimática , Expressão Gênica , Humanos , Neoplasias Experimentais/fisiopatologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteínas Estruturais Virais/genética
7.
Mol Ther ; 3(5 Pt 1): 768-78, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11356081

RESUMO

Recombinant adenoviral (rAd) vectors are capable of mediating high-efficiency gene transfer in vivo. Under conditions requiring systemic administration, however, the use of rAd vectors can be problematic due to the presence of circulating anti-adenovirus antibodies developed either through natural infection or during the course of treatment. We developed a passive immunization model in SCID/Beige mice to assess the effect of human and mouse anti-adenovirus antibodies on systemic administration of a rAd vector expressing beta-galactosidase (rAd-betagal). In this model, the in vitro neutralizing activity of human or mouse antibodies used for passive immunization correlated well with inhibition of transduction of the liver following i.v. administration of rAd-betagal. Depletion of antibodies to individual adenovirus structural proteins (hexon, penton, fiber) by affinity chromatography demonstrated that antibodies to each of the three virion components contributed to neutralization of infectivity in vitro and to inhibition of transduction in vivo. Depletion of antibodies against all three structural proteins from human or mouse immune serum prior to passive immunization restored in vivo transduction activity to levels comparable to those obtained with nonimmune serum. Our data suggest that depletion of both murine and human anti-adenoviral antibodies can restore transduction in vivo during systemic rAd gene therapy in hosts previously exposed to adenovirus.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Transdução Genética , Adenoviridae/imunologia , Animais , Anticorpos/metabolismo , Proteínas de Fluorescência Verde , Humanos , Imunização Passiva , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Biossíntese de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta-Galactosidase/metabolismo
8.
Cancer Gene Ther ; 8(12): 936-47, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781656

RESUMO

Co-incubation of a replication-deficient, recombinant adenovirus carrying the wild-type p53 gene (rAd-p53) and hematopoietic stem cell (HSC) products from patients with breast cancer can significantly reduce tumor cell contamination. Whereas this approach provides a powerful tumor cell purging strategy, potential detrimental effects on the HSC population have not been investigated. The ability of human HSC to reconstitute hematopoiesis in severe combined immunodeficient (SCID) mice and to undergo secondary transplantation provides the only nonclinical measure of self-renewing, stem cell function. The objective of this study was to investigate whether co-incubation with rAd-p53 compromised the SCID repopulating activity (SRA) of HSC. Granulocyte colony-stimulating factor-mobilized human CD34+ cells were co-cultured with rAd-p53 at our targeted clinical dose, and the ability of these cells to establish multilineage hematopoiesis in sublethally irradiated, nonobese diabetic (NOD)-SCID mice was investigated. The persistence of human cells in the mice was investigated by flow cytometry, granulocyte-macrophage colony-forming unit assay, and polymerase chain reaction of human Alu sequences. Further, limiting dilution analysis provided a quantitative comparison between the SRA of CD34+ cells co-incubated with rAd-p53 and control CD34+ cells (no rAd-p53 co-incubation). We conclude that co-incubation with rAd-p53 has little effect on the SRA of HSC.


Assuntos
Purging da Medula Óssea , Genes p53 , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Adenoviridae , Animais , Purging da Medula Óssea/métodos , Feminino , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hematopoese , Mobilização de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
9.
Cancer Gene Ther ; 7(11): 1469-80, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11129289

RESUMO

SCH58500 (ACN53) is a replication-deficient, type 5 adenovirus (Ad) expressing human wild-type p53 tumor suppressor. It is currently undergoing clinical trials as a cancer therapeutic. Many SCH58500 clinical trials incorporate an arm comparing traditional chemotherapy against chemotherapy combined with SCH58500. Paclitaxel was chosen for combination therapy in the preclinical study reported here due to its extensive use as a first-line therapy in ovarian cancer, its synergy with SCH58500 in preclinical cancer models, and its activation of p53-independent apoptosis, which might result in a "lowered threshold" for tumor cell death. SCID mice bearing human tumor xenografts were dosed with intratumoral vehicle, control Ad vector, or SCH58500, with or without paclitaxel. Real-time quantitative reverse transcriptase polymerase chain reaction assays were developed and validated to quantitate expression of p53, the p53 downstream effector gene p21, and the apoptosis-related genes, bax, bcl-2, and survivin. Protein expression was confirmed using immunohistochemical assays for p53 and p21. Only tumors injected with SCH58500 had detectable levels of exogenous p53 DNA and mRNA. After SCH58500 treatment, 3-11-fold elevations of p21 expression were observed in tumor xenografts containing nonfunctional p53 (MDA-MB-468, MDA-MB-231, MIAPaCa2, DU-145, and SK-OV-3), but no change in p21 mRNA in wild-type p53 PA-1 tumors. Immunohistochemical assays confirmed induction of p21 protein in MDAMB-468 and SK-OV-3 cells, but not in PA-1 cells. Ad vector alone or paclitaxel alone had no effect on p21 mRNA levels in most tumors. However, paclitaxel suppressed p21 expression induced by SCH58500 4-fold in DU-145 and SK-OV-3 tumors. Paclitaxel also affected expression of the housekeeping gene gapdh. There was no consistent pattern to the changes in bax, bcl-2, or survivin after SCH58500 treatment with or without paclitaxel between tumor types, although there were consistent responses within individual tumor lines. The mRNA ratios for bax/bcl-2 and bax/survivin were also not informative across tumor types. Of the genes examined, only p21 gave a predictable response 24 hours after p53 gene therapy and therefore, p21 expression may be useful for confirming SCH58500 activity in human tumor biopsies.


Assuntos
Genes p53/genética , Terapia Genética/métodos , Proteínas Associadas aos Microtúbulos , Paclitaxel/uso terapêutico , Proteína Supressora de Tumor p53/biossíntese , Adenoviridae/genética , Animais , Apoptose , Terapia Combinada , Feminino , Humanos , Imuno-Histoquímica , Proteínas Inibidoras de Apoptose , Camundongos , Camundongos SCID , Proteínas de Neoplasias , Transplante de Neoplasias , Neoplasias Ovarianas/terapia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina , Células Tumorais Cultivadas , Proteína X Associada a bcl-2 , Proteínas rho de Ligação ao GTP/biossíntese
10.
Urology ; 53(5): 1049-53, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10223504

RESUMO

OBJECTIVES: Replication deficient adenoviral vectors (rAds) are used as gene delivery systems that can efficiently transduce a variety of tissues and may be appropriate vectors to develop gene therapeutics for many urologic applications. However, the bladder epithelium has been shown to be highly resistant to transgene expression after intracystic administration. A potential explanation for this low gene transfer efficiency may be the protective structure of the urothelium. Since this protective barrier can be disrupted by organic solvents, we assessed whether ethanol co-administration can enhance adenovirus-mediated transgene expression. METHODS: Normal and bladder tumor-bearing rats received a single intracystic administration of rAd encoding beta-galactosidase (rAd-beta gal) or p53 (rAd-p53). rAd was administered in a saline solution or in solutions with increasing concentrations of ethanol. Transgene expression was evaluated in the bladder tissues. RESULTS: A dramatic increase in urothelial beta-galactosidase transgene expression was achieved by rAd-beta gal administered in a 22% ethanol solution. Transgene expression was enhanced in normal urothelium and in superficial bladder tumors. p53 transgene expression was similarly enhanced. CONCLUSIONS: Co-administration of 22% ethanol enhanced local rAd-mediated transgene expression in the normal and neoplastic bladder epithelium in rodents. Improvement of rAd-mediated transgene expression is progress toward local gene delivery to the urothelium and may enable local gene therapy for superficial bladder cancer or other bladder diseases.


Assuntos
Adenoviridae , Etanol/farmacologia , Técnicas de Transferência de Genes , Solventes/farmacologia , Bexiga Urinária/efeitos dos fármacos , Animais , Feminino , Ratos , Ratos Sprague-Dawley , Urotélio/efeitos dos fármacos
11.
Clin Nephrol ; 50(3): 188-93, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9776424

RESUMO

A patient with the acquired immunodeficiency syndrome (AIDS) and sickle cell anemia presented to the University of Wisconsin Hospital on two separate occasions with pneumocystis carinii pneumonia (PCP). On both occasions he was treated with high-dose intravenous trimethoprim/sulfamethoxazole (TMP/SMX). Several days into each treatment course he developed hyperkalemia and systemic acidosis consistent with hyperkalemic renal tubular acidosis (RTA). The abnormalities resolved in the first instance with the addition of amphotericin B while continuing TMP/SMX, and in the second upon discontinuation of the TMP/SMX. While an increasing number of cases with TMP/SMX-induced hyperkalemia have been reported, hyperkalemic RTA is an uncommon complication of TMP/SMX therapy, occurring in patients with predisposing factors for acidosis such as aldosterone defects, medullary dysfunction and renal insufficiency.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/tratamento farmacológico , Acidose Tubular Renal/induzido quimicamente , Anti-Infecciosos/efeitos adversos , HIV-1 , Hiperpotassemia/induzido quimicamente , Pneumonia por Pneumocystis/tratamento farmacológico , Combinação Trimetoprima e Sulfametoxazol/efeitos adversos , Infecções Oportunistas Relacionadas com a AIDS/complicações , Acidose Tubular Renal/diagnóstico , Adulto , Anemia Falciforme/complicações , Quimioterapia Combinada , Humanos , Hiperpotassemia/diagnóstico , Masculino , Pneumonia por Pneumocystis/complicações , Recidiva
12.
Cancer Gene Ther ; 5(4): 207-14, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9694072

RESUMO

As a prelude to considering retinoblastoma (RB) gene therapy for cancer, a series of human tumor cell lines with either full-length, mutated, or undetectable RB protein were treated with recombinant adenovirus encoding RB (ACNRB). Both RB protein expression and the cytotoxic and antiproliferative effects of ACNRB treatment were evaluated. While the transgene expression of a reporter virus encoding the beta-galactosidase enzyme (rAd-beta-gal) varied among cell lines, the reintroduction and expression of the RB gene resulted in a pronounced inhibition of cellular proliferation in RB-altered cell lines. An antiproliferative response was observed with control adenovirus treatment in some cell lines. ACNRB treatment did not cause detectable cytotoxicity in either RB+ or RB-altered cells. Dose-dependent cytostasis was observed in RB- cell lines. In vivo tumor suppression was observed in a breast xenograft model subsequent to the treatment of established tumors with ACNRB. These data support a role for RB gene therapy of tumors with RB mutations and provide a basis for the further evaluation of ACNRB gene therapy of human cancer.


Assuntos
Adenoviridae/genética , Antineoplásicos/farmacologia , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Retinoblastoma/genética , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fase G1/efeitos dos fármacos , Fase G1/genética , Vetores Genéticos/genética , Humanos , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Camundongos , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Retinoblastoma/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
13.
Clin Cancer Res ; 4(7): 1649-59, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9676839

RESUMO

p53 tumor suppressor gene therapy has been proposed for cancers characterized by inactivation of p53 function, and successful therapy will require efficient strategies for gene delivery. To maximize transgene expression in tumors, a clinical strategy has been proposed to treat neoplasms in the liver via hepatic artery administration of a recombinant adenovirus encoding wild-type p53 (rAd-p53). We have developed a syngeneic rat model using a p53mut hepatocellular carcinoma cell line (McA-RH7777) that results in multifocal liver tumor nodules to provide experimental support for this strategy. Treatment of McA-RH7777 cells with rAd-p53 in vitro resulted in efficient transgene expression, growth suppression, and apoptosis. Intrahepatic artery dosing with rAd-p53 or an adenovirus encoding beta-galactosidase (rAd-betagal) increased transgene expression in tumor tissue and decreased systemic exposure when compared with i.v. dosing. Daily hepatic artery dosing of rAd-p53 suppressed tumor growth when compared with untreated rats or animals treated with rAd-betagal. These data demonstrate the potential for arterial gene delivery to tumors using recombinant adenoviruses, and support continued investigation of rAd-p53 gene therapy for liver malignancies.


Assuntos
Adenoviridae , Carcinoma Hepatocelular/terapia , Genes p53 , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Neoplasias Hepáticas Experimentais/terapia , Proteínas de Neoplasias/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Carcinoma Hepatocelular/metabolismo , Vírus Defeituosos , Feminino , Expressão Gênica , Artéria Hepática , Humanos , Infusões Intra-Arteriais , Infusões Intravenosas , Neoplasias Hepáticas Experimentais/metabolismo , Proteínas de Neoplasias/genética , Ratos , Transgenes , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
14.
Blood ; 89(7): 2259-67, 1997 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9116267

RESUMO

Transfer of "anti-HIV-1 genes" into hematopoietic stem cells of human immunodeficiency virus-1 (HIV-1)-infected individuals may be a potent therapeutic approach to render mature cells arising from transduced stem cells resistant to the destructive events associated with HIV-1 infection. To determine the feasibility of gene therapy for acquired immunodeficiency syndrome in individuals already infected with HIV-1, granulocyte colony-stimulating factor mobilized peripheral blood CD34+ cells were isolated from HIV-1-infected individuals and transduced with retroviral vectors containing three different anti-HIV-1-genes: the Rev binding domain of the Rev Responsive Element (RRE decoy) (L-RRE-neo), a double hammerhead ribozyme vector targeted to cleave the tat and rev transcripts (L-TR/TAT-neo), and the trans-dominant mutant of rev (M10) (L-M10-SN). As a control, a vector mediating only neomycin resistance (LN) was used. After 3 days of transduction on allogeneic stroma in the presence of stem cell factor, interleukin-6 (IL-6), and IL-3, the cultures were G418-selected, and then challenged with HIV-1(JR-FL) and a primary HIV-1 isolate. Compared with the control cultures, the L-RRE-neo-, L-TR/TAT-neo-, and L-M10-SN-transduced cultures displayed up to 1,000-fold inhibition of HIV-1 replication after challenge with HIV-1(JR-FL) and the primary HIV-1 isolate. Growth of the hematopoietic cells in long-term bone marrow culture was not perturbed by the presence of any of the anti-HIV-1 genes. This study shows that anti-HIV-1 genes can be introduced into CD34+ cells from individuals already infected with HIV-1, and strongly inhibit HIV-1 replication in primary monocytes derived from the CD34+ progenitors.


Assuntos
Genes rev , Terapia Genética , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos/farmacologia , HIV-1/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/virologia , RNA Catalítico/farmacologia , Sequências Reguladoras de Ácido Nucleico , Retroviridae/genética , Replicação Viral , Sítios de Ligação , Diferenciação Celular , Células Cultivadas , Produtos do Gene rev/metabolismo , Produtos do Gene tat/metabolismo , Infecções por HIV/sangue , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Monócitos/virologia , RNA Catalítico/genética , Proteínas Recombinantes de Fusão/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana , Produtos do Gene tat do Vírus da Imunodeficiência Humana
15.
J Formos Med Assoc ; 96(3): 157-71, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9080753

RESUMO

While the relative incidence of serious nephrotoxicities in the population consuming nonsteroidal anti-inflammatory drugs (NSAIDs) is very low, the frequency of adverse events in patients at risk has considerably increased due to the rising popularity of the use of the drugs in recent years. Under normal conditions, NSAIDs have relatively little effect on the kidney because of low renal production of prostaglandins. However, in the presence of renal hypoperfusion in which local synthesis of vasodilator prostaglandins is increased to protect the glomerular hemodynamics and to maintain appropriate renal tubular transport of fluid and electrolytes, inhibition of prostaglandin synthesis by NSAIDs can lead to vasoconstrictive acute renal failure as well as fluid and electrolyte disorders such as sodium retention and resistance to diuretics, hyponatremia and hyperkalemia. Conditions that increase the risk for NSAID-induced nephrotoxicities include volume depletion from diuretics and other causes, edematous states such as congestive heart failure and cirrhosis of the liver, old age and underlying renal disease, especially in the presence of renal functional impairment. In addition, renal parenchymal diseases may develop in susceptible patients taking NSAIDs. These include acute tubulointerstitial nephritis, frequently associated with nephrotic syndrome, and chronic progressive renal disease, with or without renal papillary necrosis. Rare cases of vasculitis and glomerulonephritis have also been reported. Finally, NSAIDs may aggravate hypertension by interacting with antihypertensive drugs, especially with diuretics and beta-blockers. Withdrawal of NSAIDs in patients at risk can frequently reverse or improve the nephrotoxicities. It is recommended that physicians be aware of the clinical settings that increase the risk for NSAID-induced nephrotoxicities and take preventive or therapeutic measures accordingly.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Rim/efeitos dos fármacos , Animais , Anti-Hipertensivos/efeitos adversos , Interações Medicamentosas , Hemodinâmica/efeitos dos fármacos , Humanos , Rim/fisiologia , Prostaglandinas/fisiologia
16.
Mod Pathol ; 9(3): 339-47, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8685238

RESUMO

Inactivation of the retinoblastoma susceptibility (RB) gene plays a role in the pathogenesis of a variety of human malignancies. Recently, it has become feasible to study RB expression in archival tissues, and it is expected that immunohistochemical studies on routinely processed tumors will further elucidate the biologic and clinical significance of RB mutations. Our study was designed to address two issues that are critical for the interpretation of such studies, i.e., whether mutant RB protein (pRB) can reliably be distinguished from normal pRB and whether there are significant differences in the performance characteristics of various anti-RB antibodies. We studied cell blocks of 26 mutant RB cell lines (11 lines without any RB expression, nine lines expressing truncated mRNA/pRB, six lines carrying missense mutations) with five different anti-RB monoclonal antibodies, using a recently described procedure that includes an antigen retrieval step. The specific staining pattern for pRB was nuclear. Cytoplasmic staining was found to be nonspecific and could be strong. Some truncated and all full-length mutant pRBs localized to the nucleus, creating positive nuclear staining that might be indistinguishable from the staining pattern of cells carrying wild-type RB. The five antibodies tested showed significant differences in sensitivity, specificity, and background reactivity. Our data suggest that a significant subset of mutant pRB has preserved nuclear translocation capacity, that not all anti-RB antibodies are equally suitable for immunohistochemical analysis of RB expression, and that any such analysis is bound to include a certain, albeit probably small, number of positive stains, despite the absence of functional pRB.


Assuntos
Mutação , Neoplasias/genética , Neoplasias/patologia , Proteína do Retinoblastoma/química , Proteína do Retinoblastoma/genética , Humanos , Imuno-Histoquímica , Neoplasias/química , Inclusão em Parafina , RNA Mensageiro/análise , Células Tumorais Cultivadas
17.
J Cell Biochem ; 57(4): 641-6, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7615648

RESUMO

A simple scheme was developed to study the mRNA stability of the proliferating cell nuclear antigen (PCNA) gene during cellular transition from the G1/S boundary to a quiescent state. By this scheme, CHO.K1 cells were grown to about 80% confluence and then serum-starved for 40 h for synchronization in a quiescent state. The quiescent cells were serum-stimulated for a period of time (between 8 h and 12 h) and then grown in serum-free medium until being harvested for further analyses. The cellular PCNA mRNA level was analyzed by Northern blotting. As compared with that in cells which were continuously incubated in serum-containing medium, the decline of the mRNA level, after reaching the peak, in these serum-deprived cells was virtually devoid of mRNA synthesis. Thus, this mRNA decay was taken for the measurement of mRNA stability. The advantage of the scheme is that, unlike the treatment of transcription inhibitors, it does not prevent the cells from completing the rest of the cell cycle before returning to the resting state, and so the mRNA stability observed is cell cycle dependent. In contrast with the previous report that the stability of PCNA mRNA in quiescent cells is less by severalfold than that in S phase cells, our study shows that the mRNA stability of PCNA remained constant during the cellular transition from G1/S boundary to quiescent state.


Assuntos
Ciclo Celular , Antígeno Nuclear de Célula em Proliferação/genética , RNA Mensageiro/biossíntese , Animais , Northern Blotting , Células CHO , Cricetinae , Meios de Cultura Livres de Soro , Técnicas de Cultura/métodos , Dactinomicina/farmacologia , Fase G1/genética , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , Fase S/genética
18.
In Vitro Cell Dev Biol Anim ; 31(3): 207-14, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7757303

RESUMO

We have established and characterized a new glioblastoma cell line, termed GT9, from a biopsy sample of a female adult patient with glioblastoma multiforme. The line has now undergone over 60 passages and has been successfully cultured after cryopreservation. Immunofluorescence analyses with a panel of monoclonal antibodies were positive for glial fibrillary acidic protein and vimentin, and negative for neurofilament, galactocerebroside, and fibronectin, a pattern typical of glial cells. Based on a tetraploid, the composite karyotype of GT9 cells included the loss of chromosome 10, gain of chromosome 7, and the presence of double minute chromosomes, three of the most common karyotypic abnormalities in glioblastoma. Sequence analysis of p53 cDNA revealed a homozygous double mutation at codon 249 (commonly mutated in aflatoxin-associated hepatocellular carcinoma) and codon 250. Moreover, there was a complete absence of wild-type p53. However, unlike the majority of human glioblastomas previously described, the expression of platelet-derived growth factor-B (PDGF-B), a potent mitogenic autocrine factor, was low in GT9 cells. The expression and phosphorylation of c-Jun and Jun-B, downstream mediators of the PDGF pathway, were also low. Thus, deregulation of the PDGF pathway does not appear to be involved in the pathogenesis of the GT9 glioblastoma. Conversely, Jun-D, a negative regulator of cell growth, was also low. In addition, Phosphorylated Egr-1, a recently reported suppressor of PDGF-B/v-sis-transformed cells, was also low, suggesting that the lack of activation of the PDGF pathway was not due to these suppressive mechanisms.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Genes p53 , Glioblastoma/genética , Proteínas Imediatamente Precoces , Mutação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína Supressora de Tumor p53/genética , Sequência de Aminoácidos , Anticorpos Monoclonais , Aberrações Cromossômicas , Cromossomos Humanos Par 10 , Cromossomos Humanos Par 7 , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce , Feminino , Imunofluorescência , Proteína Glial Fibrilar Ácida/análise , Glioblastoma/patologia , Humanos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas , Vimentina/análise
19.
J Am Soc Nephrol ; 5(9): 1647-52, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7780052

RESUMO

The binge drinking of alcohol combined with the ingestion of a nonsteroidal anti-inflammatory drug (NSAID) is a recently described cause of reversible acute renal failure. The pathogenetic mechanisms leading to acute tubular necrosis in this setting include the initial compromise in renal perfusion due to alcohol-induced extracellular volume contraction and the superimposed renal hemodynamic alterations induced by the NSAID that interfere with the renal autoregulation. Although alcohol may also cause rhabdomyolysis leading to acute tubular necrosis, this is usually not apparent in these cases. Previously, only three such cases have been reported but the incidence is likely to be higher in view of the prevalence of alcohol and NSAID use. Herein is presented another patient in whom the features of flank pain and acute renal failure in association with binge drinking and NSAID ingestion constitute a characteristic syndrome.


Assuntos
Dor Abdominal/induzido quimicamente , Injúria Renal Aguda/induzido quimicamente , Consumo de Bebidas Alcoólicas , Anti-Inflamatórios não Esteroides/efeitos adversos , Injúria Renal Aguda/terapia , Adulto , Feminino , Humanos , Necrose Tubular Aguda/induzido quimicamente , Necrose Tubular Aguda/terapia
20.
Hum Gene Ther ; 5(9): 1079-88, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7833367

RESUMO

We have constructed recombinant human adenoviruses that express wild-type human p53 under the control of either the Ad 2 major late promoter (MLP) or the human cytomegalovirus (CMV) immediate early gene promoter. Each construct replaces the Ad 5 E1a and E1b coding sequences necessary for viral replication with the p53 cDNA and MLP or CMV promoter. These p53/Ad recombinants are able to express p53 protein in a dose-dependent manner in infected human cancer cells. Tumor suppressor activity of the expressed p53 protein was assayed by several methods. [3H]Thymidine incorporation assays showed that the recombinant adenoviruses were capable of inhibiting DNA synthesis in a p53-specific, dose-dependent fashion. Ex vivo treatment of Saos-2 tumor cells, followed by injection of the treated cells into nude mice, led to complete tumor suppression using the MLP/p53 recombinant. Following a single injection of CMV/p53 recombinant adenovirus into the peritumoral space surrounding an in vivo established tumor derived from a human small cell lung carcinoma cell line (NIH-H69), we were able to detect p53 mRNA in the tumors at 2 and 7 days post-injection. Continued treatment of established H69 tumors with MLP/p53 recombinant led to reduced tumor growth and increased survival time compared to control treated animals. These results indicate that recombinant adenoviruses expressing wild-type p53 may be useful vectors for gene therapy of human cancer.


Assuntos
Adenovírus Humanos/genética , Carcinoma de Células Pequenas/terapia , Vírus Defeituosos/genética , Genes p53 , Terapia Genética , Vetores Genéticos , Neoplasias/terapia , Animais , Sequência de Bases , Carcinoma de Células Pequenas/patologia , Citomegalovirus/genética , Replicação do DNA , DNA Complementar/genética , DNA Recombinante/genética , DNA Viral/genética , Feminino , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Neoplasias/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...