Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Parasit Vectors ; 16(1): 168, 2023 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-37226181

RESUMO

BACKGROUND: Giardia duodenalis (referred to as Giardia) is a flagellated binucleate protozoan parasite, which causes one of the most common diarrheal diseases, giardiasis, worldwide. Giardia can be infected by Giardiavirus (GLV), a small endosymbiotic dsRNA virus belongs to the Totiviridae family. However, the regulation of GLV and a positive correlation between GLV and Giardia virulence is yet to be elucidated. METHODS: To identify potential regulators of GLV, we performed a yeast two-hybrid (Y2H) screen to search for interacting proteins of RdRp. GST pull-down, co-immunoprecipitation and bimolecular fluorescence complementation (BiFC) assay were used to verify the direct physical interaction between GLV RdRp and its new binding partner. In addition, their in vivo interaction and colocalization in Giardia trophozoites were examined by using Duolink proximal ligation assay (Duolink PLA). RESULTS: From Y2H screen, the Giardia chaperone protein, Giardia DnaJ (GdDnaJ), was identified as a new binding partner for GLV RdRp. The direct interaction between GdDnaJ and GLV RdRp was verified via GST pull-down, co-immunoprecipitation and BiFC. In addition, colocalization and in vivo interaction between GdDnaJ and RdRp in Giardia trophozoites were confirmed by Duolink PLA. Further analysis revealed that KNK437, the inhibitor of GdDnaJ, can significantly reduce the replication of GLVs and the proliferation of Giardia. CONCLUSION: Taken together, our results suggested a potential role of GdDnaJ in regulating Giardia proliferation and GLV replication through interaction with GLV RdRp.


Assuntos
Gastrópodes , Giardíase , Giardiavirus , Animais , Giardia/genética , Proliferação de Células , RNA Polimerase Dependente de RNA , Poliésteres
2.
Radiat Res ; 171(5): 588-95, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19580494

RESUMO

To determine whether increased mitochondrially localized catalase was radioprotective, a human catalase transgene was cloned into a small pSVZeo plasmid and localized to the mitochondria of 32D cl 3 cells by adding the mitochondrial localization sequence of MnSOD (mt-catalase). The cell lines 32D-Cat and 32D-mt-Cat had increased catalase biochemical activity as confirmed by Western blot analysis compared to the 32D cl 3 parent cells. The MnSOD-overexpressing 32D cl 3 cell line, 2C6, had decreased baseline catalase activity that was increased in 2C6-Cat and 2C6-mt-Cat subclonal cell lines. 32D-mt-Cat cells were more radioresistant than 32D-Cat cells, but both were radioresistant relative to 32D cl 3 cells. 2C6-mt-Cat cells but not 2C6-Cat cells were radioresistant compared to 2C6 cells. Intratracheal injection of the mt-catalase-plasmid liposome complex (mt-Cat-PL) but not the catalase-plasmid liposome complex (Cat-PL) increased the resistance of C57BL/6NHsd female mice to 20 Gy thoracic irradiation compared to MnSOD-plasmid liposomes. Thus mitochondrially targeted overexpression of the catalase transgene is radioprotective in vitro and in vivo.


Assuntos
Catalase/fisiologia , Mitocôndrias/enzimologia , Tolerância a Radiação , Animais , Catalase/genética , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Feminino , Glutationa/análise , Glutationa Peroxidase/análise , Humanos , Lipossomos , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos , Superóxido Dismutase/fisiologia , Transgenes
3.
In Vivo ; 20(6B): 791-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17203769

RESUMO

Radiation therapy of tumors of the head and neck region is compromised by dose limiting toxicity of normal tissues including the oral cavity and oropharyngeal mucosa. MnSOD-Plasmid Liposome (MnSOD-PL) intraoral gene therapy has been demonstrated to decrease normal tissue toxicity and also improve survival in mice with orthotopic SCC-VII squamous cell tumors on the floor of the mouth. Furthermore, intravenous administration of MnSOD-PL in mice with orthotopic tumors, or addition of MnSOD-PL to tumor cell lines in vitro produces a radiosensitizing effect attributable to differences in antioxidant pool responses of tumor cells compared to normal tissues following irradiation. To determine whether EGF receptor (EGFR) antagonists Iressa, or Cetuximab provided further improvement of radiation killing of squamous cell tumors, MnSOD-PL transfected or control SCCVII tumor cells were irradiated in vitro, and then the effect of EGFR receptor antagonists was tested. Cells transfected with MnSOD-PL were relatively radiosensitive D0 = 1.244 +/- 0.126 Gy compared to control D0 = 3.246 +/- 0.087 (p < 0.0001). Clonogenic radiation survival curves of SCCVII cells demonstrated radiosensitization by Iressa D0 = 2.770 +/- 0.134 Gy (p = 0.0264), but no significant radiosensitizing effect of Cetuximab D0 = 3.193 +/- 0.309 (p = 0.7338). The combination of MnSOD-PL plus Iressa further increased radiosensitivity of SCC-VII cells in vitro D0 = 0.785 +/- 0.01064 (p < 0.0001). The results suggest some synergy of the effectiveness of the EGFR antagonist Iressa on increasing the radiation killing of SCC-VII cells that supplements MnSOD-PL tumor radiosensitization.


Assuntos
Anticorpos Monoclonais/farmacologia , Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Superóxido Dismutase/genética , Animais , Anticorpos Monoclonais Humanizados , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Cetuximab , Relação Dose-Resposta à Radiação , Gefitinibe , Lipossomos , Camundongos , Transfecção , Transgenes/genética
4.
In Vivo ; 19(6): 965-74, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16277008

RESUMO

BACKGROUND: Intraesophageal manganese superoxide dismutase plasmid liposome (MnSOD-PL) gene therapy protects against irradiation damage. MATERIALS AND METHODS: To determine whether esophageal side population (SP) stem cells were protected, epitope-tagged (hemagglutinin) (HA) MnSOD-PL was administered to C57BL/6J mice 24 hours prior to 30 Gy esophageal irradiation. SP cells were isolated, and apoptosis and multi-lineage vimentin/endothelin/F4/80 (macrophage) colonies in vitro were quantitated. RESULTS: The number and percent of SP cells, apoptotic cells, or numbers of multi-lineage vimentin/endothelin/F4/80-positive in vitro colonies isolated from non-irradiated HA-MnSOD-PL-treated or 30 Gy-irradiated esophagus did not differ between groups. Irradiation in vitro significantly increased apoptosis in explanted non-SP cells from control (p = 0.021) compared to MnSOD-PL-treated mice. Irradiation-induced cell division was significantly increased in SP cells from control-irradiated mice (p = 0.001), but not MnSOD-PL-treated mice. Irradiation-induced apoptosis detected in vivo at 5 days was decreased by MnSOD-PL. CONCLUSION: MnSOD-PL gene therapy protects esophageal SP cells from irradiation in vitro and in vivo.


Assuntos
Esôfago/efeitos da radiação , Terapia Genética , Lesões Experimentais por Radiação/patologia , Proteção Radiológica , Células-Tronco/efeitos da radiação , Superóxido Dismutase/administração & dosagem , Animais , Anexina A5/metabolismo , Antígenos de Diferenciação/metabolismo , Apoptose/efeitos da radiação , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Divisão Celular/efeitos da radiação , Linhagem da Célula , Células Cultivadas , Endotelinas/metabolismo , Esôfago/enzimologia , Esôfago/patologia , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Lipossomos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Lesões Experimentais por Radiação/prevenção & controle , Radiação Ionizante , Células-Tronco/citologia , Superóxido Dismutase/genética , Vimentina/metabolismo
5.
In Vivo ; 19(6): 997-1004, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16277013

RESUMO

BACKGROUND: Ionizing irradiation-induced cellular and tissue damage is mediated in part by resultant radiochemical reactions and resultant oxidative stress. Irradiation-induced reactive oxygen and nitrogen species include: superoxide, nitric oxide, hydroxyl radical and hydrogen peroxide. The biochemical combination of superoxide and nitric oxide radicals forms peroxynitrite, a potent oxidant known to induce lipid peroxidation. MATERIALS AND METHODS: The antioxidant capacity and lipid peroxidation of the esophagus were determined following irradiation. RESULTS: In the present studies, measurements of total antioxidant capacity did not change in the esophagus of control irradiated or control plasmid pNGVL3-PL intraesophageally-injected mice. In contrast, manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) intraesophageally-treated mice showed a significant increase in antioxidant capacity persisting for seven days. Lipid peroxidative changes induced in the control irradiated mouse esophagus decreased over seven days after irradiation of C3H/HeNHsd mice exposed to 37 Gy in a single fraction. MnSOD-PL radioprotective gene therapy administered intraorally 24 hours prior to irradiation did not significantly reduce the kinetics of induction of total peroxidated lipids over the first seven days after irradiation but did decrease lipid peroxidation at days 14 and 21. CONCLUSION: These studies demonstrate the antioxidant function of MnSOD-PL gene therapy to the esophagus, which is detectable as a reduction in irradiation-induced lipid peroxidation.


Assuntos
Antioxidantes/administração & dosagem , Esôfago/efeitos da radiação , Terapia Genética , Peroxidação de Lipídeos/efeitos da radiação , Lesões Experimentais por Radiação/patologia , Superóxido Dismutase/administração & dosagem , Animais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Esôfago/enzimologia , Esôfago/patologia , Citometria de Fluxo , Corantes Fluorescentes , Lipossomos , Masculino , Malondialdeído/análise , Camundongos , Camundongos Endogâmicos C3H , Oxazinas , Plasmídeos , Lesões Experimentais por Radiação/prevenção & controle , Radiação Ionizante , Superóxido Dismutase/genética , Superóxido Dismutase/farmacocinética , Fatores de Tempo , Distribuição Tecidual , Transgenes
6.
Int J Pharm ; 270(1-2): 185-98, 2004 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-14726134

RESUMO

The objective of this work was to characterize a polymeric paste formulation of the anticancer drug paclitaxel that was injectable through a narrow gauge needle at room temperature and set to a solid implant in vivo for the intratumoral treatment of localized cancer. Pastes were manufactured from a triblock copolymer composed of poly(D,L-lactide-co-caprolactone)-block-polyethylene glycol-block-poly(D,L-lactide-co-caprolactone) (PLC-PEG-PLC) or triblock blended with a low molecular weight polymer methoxypolyethylene glycol (MePEG). Characterization of pastes was performed using differential scanning calorimetry (DSC), gel permeation chromatography (GPC) and drug release studies. Paste integrity in water was measured by determining the degree of fragmentation under initial agitation. MePEG was found to be miscible with the triblock polymer and paclitaxel dissolved in various blends of these polymers up to 15% drug loading. Pastes composed of 40:60 triblock:MePEG blends and 10% paclitaxel were found to inject through a 23-gauge needle and set to a solid pellet in phosphate-buffered saline at 37 degrees C. Such pellets released paclitaxel in a controlled manner over 7 weeks. Pastes composed of 40:60 triblock:MePEG blends containing 10% paclitaxel are proposed as suitable injectable formulations of the drug for intratumoral therapy.


Assuntos
Antineoplásicos/química , Paclitaxel/química , Poliésteres/química , Polietilenoglicóis/química , Antineoplásicos/administração & dosagem , Varredura Diferencial de Calorimetria , Química Farmacêutica , Cromatografia em Gel , Composição de Medicamentos , Estabilidade de Medicamentos , Óxido de Etileno , Injeções , Lactonas , Pomadas , Paclitaxel/administração & dosagem , Temperatura , Fatores de Tempo
7.
Radiat Res ; 182(1): 35-49, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24932534

RESUMO

The altered DNA damage response pathway in patients with Fanconi anemia (FA) may increase the toxicity of clinical radiotherapy. We quantitated oral cavity mucositis in irradiated Fanconi anemia Fancd2(-/-) mice, comparing this to Fancd2(+/-) and Fancd2(+/+) mice, and we measured distant bone marrow suppression and quantitated the effect of the intraoral radioprotector GS-nitroxide, JP4-039 in F15 emulsion. We found that FA mice were more susceptible to radiation injury and that protection from radiation injury by JP4-039/F15 was observed at all radiation doses. Adult 10-12-week-old mice, of FVB/N background Fancd2(-/-), Fancd2(+/-) and Fancd2(+/+) were head and neck irradiated with 24, 26, 28 or 30 Gy (large fraction sizes typical of stereotactic radiosurgery treatments) and subgroups received intraoral JP4-039 (0.4 mg/mouse in 100 µL F15 liposome emulsion) preirradiation. On day 2 or 5 postirradiation, mice were sacrificed, tongue tissue and femur marrow were excised for quantitation of radiation-induced stress response, inflammatory and antioxidant gene transcripts, histopathology and assay for femur marrow colony-forming hematopoietic progenitor cells. Fancd2(-/-) mice had a significantly higher percentage of oral mucosal ulceration at day 5 after 26 Gy irradiation (59.4 ± 8.2%) compared to control Fancd2(+/+) mice (21.7 ± 2.9%, P = 0.0063). After 24 Gy irradiation, Fancd2(-/-) mice had a higher oral cavity percentage of tongue ulceration compared to Fancd2(+/+) mice irradiated with higher doses of 26 Gy (P = 0.0123). Baseline and postirradiation oral cavity gene transcripts were altered in Fancd2(-/-) mice compared to Fancd2(+/+) controls. Fancd2(-/-) mice had decreased baseline femur marrow CFU-GM, BFUe and CFU-GEMM, which further decreased after 24 or 26 Gy head and neck irradiation. These changes were not seen in head- and neck-irradiated Fancd2(+/+) mice. In radiosensitive Fancd2(-/-) mice, biomarkers of both local oral cavity and distant marrow radiation toxicity were ameliorated by intraoral JP4-039/F15. We propose that Fancd2(-/-) mice are a valuable radiosensitive animal model system, which can be used to evaluate potential radioprotective agents.


Assuntos
Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/deficiência , Boca/efeitos da radiação , Mucosite/prevenção & controle , Óxidos de Nitrogênio/farmacologia , Lesões Experimentais por Radiação/prevenção & controle , Animais , Medula Óssea/imunologia , Contagem de Células , Linhagem Celular , Feminino , Fêmur/imunologia , Cabeça/efeitos da radiação , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos da radiação , Lipossomos , Masculino , Camundongos , Boca/efeitos dos fármacos , Pescoço/efeitos da radiação , Óxidos de Nitrogênio/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Protetores contra Radiação/administração & dosagem , Protetores contra Radiação/farmacologia
8.
PLoS One ; 8(6): e68965, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840861

RESUMO

As the major enamel matrix protein contributing to tooth development, amelogenin has been demonstrated to play a crucial role in tooth enamel formation. Previous studies have revealed amelogenin alternative splicing as a mechanism for amelogenin heterogeneous expression in mammals. While amelogenin and its splicing forms in mammalian vertebrates have been characterized, splicing variants of amelogenin gene still remains largely unknown in non-mammalian species. Here, using PCR and sequence analysis we discovered two novel amelogenin transcript variants in tooth organ extracts from a caudate amphibian, the salamander Plethodoncinereus. The one was shorter -S- (416 nucleotides including untranslated regions, 5 exons) and the other larger -L- (851 nt, 7 exons) than the previously published "normal" gene in this species -M- (812 nucleotides, 6 exons). This is the first report demonstrating the amelogenin alternative splicing in amphibian, revealing a unique exon 2b and two novel amelogenin gene transcripts in Plethodoncinereus.


Assuntos
Processamento Alternativo , Amelogenina/genética , Proteínas de Anfíbios/genética , Anfíbios/genética , Sequência de Aminoácidos , Anfíbios/crescimento & desenvolvimento , Animais , Sequência de Bases , Alinhamento de Sequência
9.
PLoS One ; 7(9): e45871, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029286

RESUMO

Amelogenin, the major enamel matrix protein in tooth development, has been demonstrated to play a significant role in tooth enamel formation. Previous studies have identified the alternative splicing of amelogenin in many mammalian vertebrates as one mechanism for amelogenin heterogeneous expression in teeth. While amelogenin and its splicing forms in mammalian vertebrates have been cloned and sequenced, the amelogenin gene, especially its splicing forms in non-mammalian species, remains largely unknown. To better understand the mechanism underlying amelogenin evolution, we previously cloned and characterized an amelogenin gene sequence from a squamate, the green iguana. In this study, we employed RT-PCR to amplify the amelogenin gene from the black spiny-tailed iguana Ctenosaura similis teeth, and discovered a novel splicing form of the amelogenin gene. The transcript of the newly identified iguana amelogenin gene (named C. Similis-T2L) is 873 nucleotides long encoding an expected polypeptide of 206 amino acids. The C. Similis-T2L contains a unique exon denominated exon X, which is located between exon 5 and exon 6. The C. Similis-T2L contains 7 exons including exon 1, 2, 3, 5, X, 6, and 7. Analysis of the secondary and tertiary structures of T2L amelogenin protein demonstrated that exon X has a dramatic effect on the amelogenin structures. This is the first report to provide definitive evidence for the amelogenin alternative splicing in non-mammalian vertebrates, revealing a unique exon X and the splicing form of the amelogenin gene transcript in Ctenosaura similis.


Assuntos
Amelogenina/genética , Iguanas/genética , Proteínas de Répteis/genética , Amelogenina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA/genética , Éxons , Expressão Gênica , Iguanas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Especificidade de Órgãos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Proteínas de Répteis/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA , Dente/metabolismo
10.
Hum Gene Ther ; 22(3): 336-42, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20873987

RESUMO

Manganese superoxide dismutase (MnSOD) is a genetically engineered therapeutic DNA/liposome containing the human MnSOD transgene. Preclinical studies in mouse models have demonstrated that the expression of the human MnSOD transgene confers protection of normal tissues from ionizing irradiation damage. This is a phase I study of MnSOD plasmid liposome (PL) in combination with standard chemoradiation in surgically unresectable stage III non-small-cell lung cancer. Chemotherapy (carboplatin and paclitaxel) was given weekly (for 7 weeks), concurrently with radiation. MnSOD PL was swallowed twice a week (total 14 doses), at three dose levels: 0.3, 3, and 30 mg. Dose escalation followed a standard phase I design. Esophagoscopy was done at baseline, day 4, and 6 weeks after radiation with biopsies of the squamous lining cells. DNA was extracted and analyzed by PCR for the detection of the MnSOD transgene DNA. Ten patients with AJCC stage IIIA (three) and IIIB (seven) completed the course of therapy. Five had squamous histology, two adenocarcinoma, one large cell, and two not specified. Patients were treated in three cohorts at three dose levels of MnSOD PL: 0.3 (three patients), 3 (three patients), and 30 mg (four patients). The median dose of radiation was 77.7 Gy (range 63-79.10 Gy). Overall response rate for the standard chemoradiation regimen was 70% (n = 10). There were no dose-limiting toxicities reported in all three dosing tiers. It is concluded that the oral administration of MnSOD PL is feasible and safe. The phase II recommended dose is 30 mg.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas , Terapia Genética , Lipossomos , Neoplasias Pulmonares , Plasmídeos , Superóxido Dismutase/metabolismo , Idoso , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carboplatina/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Terapia Combinada , Esôfago/efeitos dos fármacos , Esôfago/patologia , Feminino , Humanos , Lipossomos/administração & dosagem , Lipossomos/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/radioterapia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Paclitaxel/administração & dosagem , Plasmídeos/administração & dosagem , Plasmídeos/genética , Reação em Cadeia da Polimerase , Superóxido Dismutase/genética , Transgenes/genética , Resultado do Tratamento
11.
Radiat Res ; 174(3): 297-312, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20726721

RESUMO

The effect of deletion of the nitric oxide synthase 1 gene (NOS1(-/-)) on radiosensitivity was determined. In vitro, long-term cultures of bone marrow stromal cells derived from NOS1(-/-) were more radioresistant than cells from C57BL/6NHsd (wild-type), NOS2(-/-) or NOS3(-/-) mice. Mice from each strain received 20 Gy thoracic irradiation or 9.5 Gy total-body irradiation (TBI), and NOS1(-/-) mice were more sensitive to both. To determine the etiology of radiosensitivity, studies of histopathology, lower esophageal contractility, gastrointestinal transit, blood counts, electrolytes and inflammatory markers were performed; no significant differences between irradiated NOS1(-/-) and control mice were found. Video camera surveillance revealed the cause of death in NOS1(-/-) mice to be grand mal seizures; control mice died with fatigue and listlessness associated with low blood counts after TBI. NOS1(-/-) mice were not sensitive to brain-only irradiation. MnSOD-PL therapy delivered to the esophagus of wild-type and NOS1(-/-) mice resulted in equivalent biochemical levels in both; however, in NOS1(-/-) mice, MnSOD-PL significantly increased survival after both thoracic and total-body irradiation. The mechanism of radiosensitivity of NOS1(-/-) mice and its reversal by MnSOD-PL may be related to the developmental esophageal enteric neuronal innervation abnormalities described in these mice.


Assuntos
Esôfago/enzimologia , Lipossomos , Óxido Nítrico Sintase Tipo I/metabolismo , Plasmídeos , Superóxido Dismutase/genética , Animais , Células da Medula Óssea/efeitos da radiação , Estimulação Elétrica , Trânsito Gastrointestinal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/genética , Radiografia Torácica , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Hum Gene Ther ; 19(8): 820-6, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18699723

RESUMO

Manganese superoxide dismutase plasmid liposomes (MnSOD-PL) confer organ-specific in vivo ionizing irradiation protection. To prepare for potential intravenous clinical trials of systemic MnSOD-PL for radioprotection in humans, plasmid and bacterial sequences were removed and a new minicircle construct was tested. Minicircle MnSOD was purified and then cotransfected into 32D cl 3 murine interleukin-3-dependent hematopoietic progenitor cells along with another plasmid carrying the neo gene. Cells were selected in G418 (50 microg/ml) and cloned by limiting dilution. Biochemical analysis of minicircle MnSOD-transfected cells showed an MnSOD biochemical activity level of 5.8 +/- 0.5 U/mg compared with 2.7 +/- 0.1 U/mg for control 32D cl 3 cells (p = 0.0039). 32D-mc-MnSOD cells were as radioresistant as full-length MnSOD-PL transgene-expressing 2C6 cells, relative to 32D cl 3 parent cells, with an increased shoulder on the radiation survival curve (n = 4.8 +/- 0.2 and n = 4.6 +/- 0.2, respectively, compared with 1.5 +/- 0.5 for 32D cl 3 cells; p = 0.007). C57BL/6NHsd mice received intraoral mc-MnSOD-PL, mc-DsRed-PL control, full-length MnSOD-PL, or blank-PL and then were irradiated 24 hr later with 31 Gy to the esophagus. Mice receiving mc-MnSOD-PL showed increased survival compared with control mice or mice treated with mc-DsRed-PL (p = 0.0003 and 0.039, respectively), and comparable to full-length MnSOD-PL. Intravenous, systemic administration of mc-MnSOD-PL protected mice from total body irradiation (9.75 Gy). Therefore, minicircle DNA containing the human MnSOD transgene confers undiminished radioprotection in vitro and in vivo.


Assuntos
Terapia Genética , Lipossomos/administração & dosagem , Plasmídeos/administração & dosagem , Lesões Experimentais por Radiação/prevenção & controle , Protetores contra Radiação/uso terapêutico , Superóxido Dismutase/genética , Animais , Sequência de Bases , Linhagem Celular , Relação Dose-Resposta à Radiação , Esôfago/efeitos da radiação , Feminino , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Plasmídeos/genética , Tolerância a Radiação/genética , Radiação Ionizante , Transfecção , Transgenes , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA