Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gene Ther ; 18(6): 579-83, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21248791

RESUMO

To determine the effects of manganese superoxide dismutase (MnSOD) plasmid liposome (PL) maternal radioprotection on fetal mice, timed pregnant female mice (E14 gestation) were irradiated to 3.0 Gy total body irradiation (TBI) dose, and the number, weight and growth and development over 6 months after birth of newborn mice was quantitated compared with irradiated controls. Maternal MnSOD-PL treatment at E13 improved pup survival at birth (5.4±0.9 per litter) compared with non-irradiated 3.0 Gy controls 4.9±1.1. There was no statistically significant difference in newborn abnormalities, male to female ratio in newborn litters, or other evidence of teratogenesis in surviving newborn mice from MnSOD-PL treated compared with irradiated controls. However, E14 3 Gy irradiated pups from gene therapy-treated mothers showed a significant increase in both growth and overall survival over 6 months after birth (P=0.0022). To determine if transgene product crossed the placenta pregnant E13 mice were injected intravenously with hemagglutinin-epitope-tagged MnSOD (100 µg plasmid in 100 µl liposomes), then after 24 h, fetal mice, placentas and maternal tissues were removed and tested by both immunohistochemistry and reverse transcriptase-PCR for transgene and product. There was no evidence of transgene or product in placenta or any fetal tissue while maternal liver was positive by both assays. The data provide evidence for fetal radioprotection by maternal MnSOD-PL gene therapy before irradiation, which is mediated by an indirect bystander effect and is associated with a significant improvement in both survival at birth and growth and development of newborn mice.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Terapia Genética/métodos , Lipossomos , Prenhez , Efeitos Tardios da Exposição Pré-Natal , Protetores contra Radiação/administração & dosagem , Superóxido Dismutase/genética , Irradiação Corporal Total/efeitos adversos , Animais , Feminino , Morte Fetal/prevenção & controle , Retardo do Crescimento Fetal/prevenção & controle , Feto/efeitos da radiação , Troca Materno-Fetal , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
2.
Gene Ther ; 17(9): 1181-90, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20827278

RESUMO

Duchenne muscular dystrophy is a fatal, genetic disorder in which dystrophin-deficient muscle progressively degenerates, for which dystrophin gene transfer could provide effective treatment. The host immune response to dystrophin, however, is an obstacle to therapeutic gene expression. Understanding the dystrophin-induced host immune response will facilitate the discovery of strategies to prolong expression of recombinant dystrophin in dystrophic muscle. Using whole-body irradiation of the dystrophic mdx mouse before gene transfer, we temporally removed the immune system; a 600 rad dose removed peripheral immune cells, which were restored by self-reconstitution, and a 900 rad dose removed central and peripheral immune cells, which were restored by adoptive transfer of bone marrow from a syngeneic, dystrophin-normal donor. The anti-dystrophin humoral response was delayed and dystrophin expression was partially preserved in irradiated, vector-treated mice. Nonirradiated, vector-treated control mice lost muscle dystrophin expression completely, had an earlier anti-dystrophin humoral response and demonstrated muscle fibers focally surrounded with T cells. We conclude that dystrophin gene transfer induced anti-dystrophin humoral immunity and cell-mediated responses that were significantly diminished and delayed by temporal removal of the host central or peripheral immune cells. Furthermore, manipulation of central immunity altered the pattern of regulatory T cells in muscle.


Assuntos
Distrofina/genética , Imunidade Humoral/efeitos da radiação , Distrofia Muscular de Duchenne/imunologia , Irradiação Corporal Total , Animais , DNA Complementar/administração & dosagem , Distrofina/imunologia , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Endogâmicos mdx , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos da radiação
3.
Gene Ther ; 15(5): 347-56, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18097469

RESUMO

We evaluated whether the improved esophageal radiation tolerance following Manganese Superoxide Dismutase (MnSOD)-Plasmid Liposomes was explained by improved engraftment of bone marrow-derived progenitors. C57BL/6NHsd female mice pretreated with intraesophageal MnSOD-PL were irradiated to 29 Gy to the esophagus and intravenously transplanted with marrow from male B6. 129S7-Gt (ROSA) 26S OR/J ROSA (Lac-Z+, G418-resistant) mice. After 14 days, esophagi were removed and side population and non-side population cells evaluated for donor multilineage (endothelin/vimentin/F480) positive esophageal cells. Serial intravenous transplantability was tested in second generation 29 Gy esophagus-irradiated mice. Esophagi from recipients receiving swallowed MnSOD-PL 24 h prior to irradiation demonstrated significantly increased esophageal repopulation with donor bone marrow-derived Lac-Z+, G418+, Y-probe+ multilineage cells (37.8+/-1.8>50 cell Lac-Z+ foci per esophagus) compared to irradiated controls (19.8+/-1.8) P<0.0001. Serial transfer to second-generation irradiated C57BL/6NHsd mice of intravenously injected SP or NSP first generation recipient esophagus cells was also significantly enhanced by MnSOD-PL intraesophageal pretreatment (74.4+/-3.6 SP-derived Lac-Z+ foci per esophagus, 48.6+/-5.4 NSP-derived) compared to irradiation controls (23.4+/-1.8 SP, 6.0+/-3.0 NSP), P<0.0001. Thus, intraesophageal MnSOD-PL administration enhances engraftment of marrow-derived progenitors.


Assuntos
Esôfago/lesões , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Transplante de Células-Tronco Hematopoéticas/métodos , Lesões Experimentais por Radiação/terapia , Superóxido Dismutase/genética , Administração Oral , Animais , Terapia Combinada , Esôfago/metabolismo , Esôfago/patologia , Feminino , Expressão Gênica , Vetores Genéticos/genética , Hibridização In Situ , Óperon Lac , Lipossomos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Superóxido Dismutase/metabolismo , Cicatrização , Cromossomo Y
4.
Cancer Res ; 56(5): 964-68, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8640786

RESUMO

Elevated interstitial fluid pressure (IFP) is a pathophysiological characteristic of most human and experimental tumors and may be responsible, in part, for the poor distribution of blood-borne therapeutic agents and low blood flow rate in tumors. Recent data in cervical carcinomas in patients suggest that fractionated radiation can lower tumor IFP and increase oxygen partial pressure (pO (2)) in some patients. The goals of this study were to find the minimum dose of radiation required to modulate IFP and pO(2) and to determine the time course of IFP changes due to radiation in a preclinical model. Xenografts of the LS174T human colon adenocarcinoma were grown in the right flank of nude (BALB/c) mice. IFP and pO(2) were measured before and 24 h after graded doses of irradiation. The mean +/- SD initial IFP in untreated tumors was 12.9 +/- 0.5 mm Hg (n=109), and the range was 3.0 to 40.3 mm Hg. The mean +/- SD and median initial pO(2) were 20.2 +/- 2.4 and 11.9 mm Hg, respectively (n=37). IFP and pO(2) were independent of tumor size. Fractionated radiation lowered IFP by 2.5 mm Hg when the total dose was 10 or 15 Gy (P<0.05), but IFP did not change in the controls or the 5-Gy radiation group (P>0.05). Irradiation increased the proportion of tumors at higher oxygen tensions when compared to control tumors. The IFP and tumor volumes were followed for up to 10 days after a single dose of 10, 20, or 30 Gy of irradiation. IFP decreased for all treatment groups. The decrease was most significant for the group receiving 30 Gy. On day five following irradiation, the IFP had decreased by 35%. The changes in IFP and pO(2) occurred before any macroscopic changes in tumor volume could be observed. The radiation-induced decrease in IFP could be, in part, responsible for the increased uptake of monoclonal antibodies following single or fractionated radiation that has been reported in the literature.


Assuntos
Adenocarcinoma/fisiopatologia , Neoplasias do Colo/fisiopatologia , Adenocarcinoma/patologia , Adenocarcinoma/radioterapia , Animais , Neoplasias do Colo/patologia , Neoplasias do Colo/radioterapia , Espaço Extracelular/efeitos da radiação , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Pressão Osmótica/efeitos da radiação , Consumo de Oxigênio/efeitos da radiação , Transplante Heterólogo
5.
Hum Gene Ther ; 9(9): 1381-6, 1998 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9650622

RESUMO

Toxicity to nontumor-derived tissue has proven to be a significant obstacle in achieving therapeutic levels of gamma irradiation in the treatment of cancer. The formation of reactive oxygen species (ROS) such as superoxide radicals (O2-) following irradiation is thought to be a major determinant of cellular damage. To this end, we describe the generation of two recombinant adenoviral vectors expressing the radical-scavenging enzymes MnSOD and CuZnSOD to test therapeutic strategies of radioprotection. Using a human lung epithelial cell line (IB-3), we have demonstrated that infections with both Ad.CMVMnSOD or Ad.CMVCuZnSOD significantly increase both the levels of SOD protein and enzymatic activity as compared to control cells. This increase in SOD expression reduced the level of apoptosis at 72 hr post-irradiation by 50% as compared to mock- or Ad.CMVLacZ-infected cells. Such studies provide the foundation for radioprotective gene therapies in the treatment of cancer.


Assuntos
Apoptose , Células Epiteliais/efeitos da radiação , Terapia Genética , Pulmão/citologia , Protetores contra Radiação , Superóxido Dismutase/genética , Adenoviridae/genética , Western Blotting , Linhagem Celular , Imunofluorescência , Vetores Genéticos , Humanos , Radiação Ionizante , Superóxido Dismutase/metabolismo , Transgenes
6.
Antioxid Redox Signal ; 3(3): 347-59, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11491649

RESUMO

Reactive oxygen species (ROS)-mediated damage to DNA is associated with induction of stress-activated protein kinases leading to secondary and tertiary effects on the nuclear matrix, cytoplasmic transport mechanisms, and altered mitochondrial and cell membranes. The cellular defenses against ROS damage are associated with up-regulation of gene products that can significantly alter cell biology, including antiapoptotic Bax family proteins and inflammatory proteins. Altered cell integrity can occur either directly or by indirect paracrine and juxtacrine interactions within tissues. Previous approaches toward therapeutic intervention against ROS damage have included administration of radical scavenger compounds, use of novel drugs that increase cellular production of constitutive antioxidants, or pharmacologic agents that modify the intracellular transport of antioxidants. Strategies to modify the cellular effects of ROS in hyperbaric oxygen injury to the lung, reperfusion injury to transplanted organs, and cancer have led to novel approaches of gene therapy in which the transgenes for antioxidant proteins can be expressed in specific tissues. Reducing tissue-damaging effects of ROS may have relevance to cancer patients by ameliorating normal tissue damage from ionizing irradiation therapy, photodynamic therapy, and cancer chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Antioxidantes/uso terapêutico , Terapia Genética , Neoplasias/terapia , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética , Animais , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Lewis/radioterapia , Carcinoma Pulmonar de Lewis/terapia , Linhagem Celular , Citocinas/farmacologia , Relação Dose-Resposta à Radiação , Transporte de Elétrons/efeitos da radiação , Sequestradores de Radicais Livres/uso terapêutico , Terapia Genética/métodos , Glutationa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Neoplasias/prevenção & controle , Oxirredução , Protetores contra Radiação/uso terapêutico , Transdução de Sinais , Superóxido Dismutase/fisiologia , Superóxido Dismutase/uso terapêutico , Células Tumorais Cultivadas
7.
Curr Gene Ther ; 3(3): 183-95, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12762478

RESUMO

Control of cancer by irradiation therapy alone or in conjunction with combination chemotherapy is often limited by organ specific toxicity. Ionizing irradiation toxicity is initiated by damage to normal tissue near the tumor target and within the transit volume of radiotherapy beams. Irradiation-induced cellular, tissue, and organ damage is mediated by acute effects, which can be dose limiting. A latent period follows recovery from the acute reaction, then chronic irradiation fibrosis (late effects) pose a second cause of organ failure. We have developed the technology for radioprotective gene therapy using the transgene for the antioxidant manganese superoxide dismutase, delivered to specific target organs (lung, esophagus, oral cavity, oropharynx, and bladder) using gene transfer vectors including plasmid/liposomes (PL) and adenovirus. Irradiation protection by MnSOD transgene overexpression at the cellular level has been demonstrated to be localized to the mitochondrial membrane. Using MnSOD transgene constructs lacking the mitochondrial localization leader sequence, and in other experiments attaching this localization signal to otherwise non-radioprotective cytoplasmic Cu/ZnSOD, mitochondrial localization has been demonstrated to be critical to protection. Organ specific injection of MnSOD-PL prior to irradiation demonstrates transgene expression for 48-72 hours, and an associated decrease in ionizing irradiation-induced expression of inflammatory cytokine mRNA and protein. Significant reduction of organ specific tissue injury has been demonstrated in several organ systems in rodent models. Application of MnSOD-PL gene therapy in the setting of fractionated chemo-radiotherapy is being tested in clinical trials for prevention of esophagitis during treatment of non-small cell carcinoma of the lung, and in prevention of mucositis during combination therapy of carcinomas of the head and neck. Encouraging results in pre-clinical models suggest that radioprotective gene therapy may facilitate dose escalation protocols to allow increases in the therapeutic ratio of cancer radiotherapy.


Assuntos
Terapia Genética , Neoplasias Induzidas por Radiação/terapia , Protetores contra Radiação/uso terapêutico , Superóxido Dismutase/genética , Animais , Camundongos , Camundongos Endogâmicos , Transgenes
8.
Int J Radiat Oncol Biol Phys ; 31(1): 171-8, 1995 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-7995749

RESUMO

PURPOSE: Microdosimetric expectations of Boron contents are extracted from a CRAY-Monte Carlo simulation of the nuclear reaction 10B(n, alpha)7Li as it occurs on a boronated membrane of a model cell and as the reaction fragments (alpha and Li) traverse into the cellular nucleus. METHODS AND MATERIALS: The present microdosimetry calculation is based upon the assumption that the therapeutic advantage of boron neutron capture therapy (BNCT), while depending upon the RBE and LET of the reaction particles, is equally dependent on the boron carrier preferential localization to tumor tissue, and the boron selectivity to cancerous cells and its specificity within subcellular compartments. In particular, boron fixes to cell membrane as it ought to, using monoclonal antibodies. The present Monte Carlo simulation computes stochastic expectations of alpha/Li energy depositions to the nucleus in a uniformly boronated membrane shell of a spherical cell. Differential energy gain was deduced from the stochastic energy depositions in events of neutron reactions with membrane boron compared against those with natural elements (O, H, N) in the cell. RESULTS: Microdosimetry data are presented in terms of specific energy (keV/micron3) and lineal energy (keV/micron) functions of the nucleus-to-cell volume ratios (NCVR). When folded with the geometric boron content and accounting for background reaction energies, the distributions yield effective energy gain to the cell nucleus per neutron capture event. Boron amount required to yield these energy gains are found to be of the order of picograms of boron per gram of cell mass. CONCLUSION: The boron content as inferred by the present Monte Carlo microdosimetry compares well with that deliverable by present pharmacokinetic means, but are orders of magnitude (mu-grams) less than those deduced previously from anthropomorphic macrodosimetry.


Assuntos
Terapia por Captura de Nêutron de Boro , Boro/química , Lítio/química , Partículas alfa , Membrana Celular/efeitos da radiação , Núcleo Celular/efeitos da radiação , Simulação por Computador , Método de Monte Carlo , Radioquímica , Dosagem Radioterapêutica
9.
Int J Radiat Oncol Biol Phys ; 43(1): 169-81, 1999 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-9989524

RESUMO

PURPOSE: A dose and volume limiting factor in radiation treatment of thoracic cancer is the development of fibrosis in normal lung. The goal of the present study was to determine whether expression prior to irradiation of a transgene for human manganese superoxide dismutase (MnSOD) or human copper/zinc superoxide dismutase (Cu/ZnSOD) protects against irradiation-induced lung damage in mice. METHODS AND MATERIALS: Athymic Nude (Nu/J) mice were intratracheally injected with 10(9) plaque-forming units (PFU) of a replication-incompetent mutant adenovirus construct containing the gene for either human MnSOD, human copper/zinc superoxide dismutase (Cu/ZnSOD) or LacZ. Four days later the mice were irradiated to the pulmonary cavity to doses of 850, 900, or 950 cGy. To demonstrate adenoviral infection, nested reverse transcriptase-polymerase chain reaction (RT-PCR) was carried out with primers specific for either human MnSOD or Cu/ZnSOD transgene on freshly explanted lung, trachea, or alveolar type II cells, and immunohistochemistry was used to measure LacZ expression. RNA was extracted on day 0, 1, 4, or 7 after 850 cGy of irradiation from lungs of mice that had previously received adenovirus or had no treatment. Slot blot analysis was performed to quantitate RNA expression for IL-1, tumor necrosis factor (TNF)-alpha, TGF-beta, MnSOD, or Cu/ZnSOD. Lung tissue was explanted and tested for biochemical activity of MnSOD or Cu/ZnSOD after adenovirus injection. Other mice were sacrificed 132 days after irradiation, lungs excised, frozen in OCT, (polyvinyl alcohol, polyethylene glycol mixture) sectioned, H&E stained, and evaluated for percent of the lung demonstrating organizing alveolitis. RESULTS: Mice injected intratracheally with adenovirus containing the gene for human MnSOD had significantly reduced chronic lung irradiation damage following 950 cGy, compared to control mice or mice injected with adenovirus containing the gene for human Cu/ZnSOD or LacZ. Immunohistochemistry for LacZ protein in adenovirus LacZ (Ad-LacZ)-injected mice demonstrated expression of LacZ in both the upper and lower airway. Nested RT-PCR showed lung expression of MnSOD and Cu/ZnSOD for at least 11 days following infection with each respective adenovirus construct. Nested RT-PCR using primers specific for human MnSOD demonstrated increased expression of the human MnSOD transgene in the trachea and alveolar type II cells 4 days after virus injection on the day of irradiation. At this time point, increased biochemical activity of MnSOD and Cu/ZnSOD respectively, was detected in lungs from these two adenovirus groups, compared to each other or to control or adenovirus LacZ mice. Slot blot analysis of RNA from lungs of mice in each group following 850 cGy irradiation demonstrated decreased expression of mRNA for interleukin-I (IL-1), TNF-alpha, and transforming growth factor-beta (TGF-beta) in the MnSOD adenovirus-injected mice, compared to irradiated control, LacZ, or Cu/ZnSOD adenovirus-injected, irradiated mice. Mice receiving adenovirus MnSOD showed decreased organizing alveolitis at 132 days in all three dose groups, compared to irradiated control or Ad-LacZ, or Ad-Cu/ZnSOD mice. CONCLUSIONS: Overexpression of MnSOD in the lungs of mice prior to irradiation prevents irradiation-induced acute and chronic damage quantitated as decreased levels of mRNA for IL-1, TNF-alpha, and TGF-beta in the days immediately following irradiation, and decrease in the percent of lung demonstrating fibrosis or organizing alveolitis at 132 days. These data provide a rational basis for development of gene therapy as a method of protection of the normal lung from acute and chronic sequelae of ionizing irradiation.


Assuntos
Adenoviridae/genética , Pneumonia em Organização Criptogênica/prevenção & controle , Terapia Genética/métodos , Pneumonite por Radiação/prevenção & controle , Superóxido Dismutase/genética , Transgenes , Animais , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Pulmão/metabolismo , Pulmão/efeitos da radiação , Camundongos , Camundongos Nus , RNA Mensageiro/metabolismo , Superóxido Dismutase/metabolismo
10.
Cancer Lett ; 63(1): 73-9, 1992 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-1555210

RESUMO

The rationale for melanoma specific antitumor agents containing phenolic amines is based in part upon the ability of the enzyme tyrosinase to oxidize these prodrugs to toxic intermediates. Two phenolic amine compounds, N-acetyl-4-S-cysteaminylphenol (N-Ac-4-S-CAP) and 4-S-cysteaminylphenol (4-S-CAP), demonstrated growth inhibitory activity with a variety of melanoma cell lines and were essentially non-toxic to non-melanoma cell lines. Theophylline, an inhibitor of cyclic nucleotide phosphodiesterase, increased in situ tyrosinase activity and enhanced the antimelanoma effects of 4-S-CAP and N-Ac-4-S-CAP in pigmented melanoma cell lines. Phenylthiourea, a specific inhibitor of tyrosinase activity, partially blocked the growth inhibitory activity of N-Ac-4-S-CAP in human pigmented melanoma cells. Buthionine sulfoximine, an inhibitor of the synthesis of the cellular antioxidant glutathione, potentiated the growth inhibitory activity of N-Ac-4-S-CAP in pigmented melanoma cells.


Assuntos
Antineoplásicos/metabolismo , Cisteamina/análogos & derivados , Melanoma/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Fenóis/metabolismo , Butionina Sulfoximina , Divisão Celular/efeitos dos fármacos , Cisteamina/metabolismo , Sinergismo Farmacológico , Humanos , Melanoma/patologia , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacologia , Teofilina/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
11.
Radiat Res ; 149(6): 570-8, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9611095

RESUMO

The molecular mechanisms underlying radiation-induced defects in the bone marrow which may contribute to the development of radiation-induced hematopoietic disorders such as aplastic anemia and leukemia are not known. Persistent changes in gene expression were examined after exposure of cells of a murine bone marrow stromal cell line to ionizing radiation. Analysis of mRNA transcript levels by differential display led to the identification of a band, C122, which increased in abundance 1 week after exposure. Northern blot hybridization verified these results and revealed a 12-fold increase in abundance of this message for up to 3 weeks after irradiation in vitro. DNA sequence analysis identified clone C122 as murine serum amyloid A 3 (Saa3), a member of the Saa family of acute-phase or inflammatory response genes. Saa message levels were then examined in vivo in the bone marrow of mice exposed to total-body irradiation. Semi-quantitative reverse transcription-polymerase chain reaction revealed a 15-20-fold increase in Saa3 message levels in the bone marrow of irradiated mice from 3 days to 2 weeks after exposure. Saa3 message levels continued to be 2-3-fold above control for up to 28 weeks in vivo. Two additional members of the murine Saa gene family, Saa1 and Saa2, were also detected in irradiated bone marrow. The expression of SAA1 and SAA2 was also detected in irradiated cells of human bone marrow stromal cell lines in vitro. These results suggest that SAA genes are involved in the radiation response in the bone marrow, but their role in the recovery of the marrow after irradiation or in the development of radiation-induced hematopoietic disorders remains to be established. Additionally, the persistent radiation-induced increase in expression suggests the potential utility of using SAA3 transcript levels as a molecular marker of past radiation exposure.


Assuntos
Células da Medula Óssea/efeitos da radiação , Regulação da Expressão Gênica , Proteína Amiloide A Sérica/genética , Animais , Sequência de Bases , Linhagem Celular , Feminino , Hematopoese/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos CBA , Dados de Sequência Molecular , RNA Mensageiro/análise , Células Estromais/efeitos da radiação
12.
Radiat Res ; 152(1): 29-40, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10381838

RESUMO

To investigate the effect of Trp53 (formerly known as p53) on stromal cells of the hematopoietic microenvironment, long-term bone marrow cultures were established from mice in which the Trp53 gene had been inactivated by homologous recombination (Trp53(-/-)) or their wild-type littermates (Trp53(+/+)). Long-term bone marrow cultures from Trp53(-/-) mice continued to produce nonadherent cells for 22 weeks, while Trp53(+/+) cultures ceased production after 15 weeks. There was a significant increase in the number of nonadherent cells produced in Trp53(-/-) long-term bone marrow cultures beginning at week 9 and continuing to week 22 (P < 0.02). The Trp53(-/-) cultures also showed significantly increased cobblestone island formation indicative of early hematopoietic stem cell-containing colonies beginning at week 10 (P < 0.01). Cobblestone islands persisted until weeks 15 and 22 in Trp53(+/+) and Trp53(-/-) cultures, respectively. Co-cultivation experiments in which Trp53(+/+) Sca1(+)lin- enriched hematopoietic stem cells were plated on Trp53(-/-) stromal cells showed increased cobblestone island formation compared to Trp53(-/-) Scal+lin- cells plated on Trp53(+/+) or Trp53(-/-) stromal cells. Radiation survival curves for clonal bone marrow stromal cells revealed a similar D0 for the Trp53(+/+) and Trp53(-/-) cell lines (1.62 +/- 0.16 and 1.49 +/- 0. 08 Gy, respectively; P = 0.408), and similar n (8.60 +/- 3.23 and 10.71 +/- 0.78, respectively) (P = 0.491). Cell cycle analysis demonstrated a G2/M-phase arrest that occurred 6 h after irradiation for both Trp53(+/+) and Trp53(-/-) stromal cell lines. After 10 Gy irradiation, there was no significant increase in the frequency of apoptosis detected in Trp53(+/+) compared to Trp53(-/-) marrow stromal cell lines. In the stromal cell lines, ICAM-1 was constitutively expressed on Trp53(+/+) but not Trp53(-/-) cells; however, a 24-h exposure to TNF-alpha induced detectable ICAM-1 on Trp53(-/-) cells and increased expression on Trp53(+/+) cells. To test the effect of Trp53 on the radiation biology of hematopoietic progenitor cells, the 32D cl 3 cell line was compared with a subclone in which expression of an E6 inserted transgene accelerates ubiquitin-dependent degradation of Trp53, thus preventing accumulation of Trp53 after genotoxic stress. The radiation survival curves were similar with no significant difference in the D0 or n, or in the percentage of cells undergoing apoptosis after 10 Gy irradiation between the two cell lines. Cells of the 32D-E6 cell line displayed a G2/M-phase arrest 6 h after 10 Gy, while cells of the parent line exhibited both a G2/M-phase arrest and a G1-phase arrest at 24 and 48 h. The results suggest a complex mechanism of action of Trp53 on the interactions between stromal and hematopoietic cells in long-term bone marrow cultures.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Ciclo Celular/efeitos da radiação , Genes p53 , Proteína Supressora de Tumor p53/fisiologia , Animais , Células da Medula Óssea/fisiologia , Adesão Celular , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Cruzamentos Genéticos , Feminino , Células-Tronco Hematopoéticas/citologia , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recombinação Genética , Células Estromais , Fatores de Tempo , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
13.
Radiat Res ; 154(4): 365-74, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11023599

RESUMO

The pulmonary ionizing radiation sensitivity of C57BL/6 Sod2(+/-) mice heterozygous for MnSOD deficiency was compared to that Sod2(+/+) control littermates. Embryo fibroblast cell lines from Sod2(-/-) (neonatal lethal) or Sod2(+/-) mice produced less biochemically active MnSOD and demonstrated a significantly greater in vitro radiosensitivity. No G(2)/M-phase cell cycle arrest after 5 Gy was observed in Sod2(-/-) cells compared to the Sod2(+/-) or Sod2(+/+) lines. Subclonal Sod2(-/-) or Sod2(+/-) embryo fibroblast lines expressing the human SOD2 transgene showed increased biochemical activity of MnSOD and radioresistance. Sod2(+/-) mice receiving 18 Gy whole-lung irradiation died sooner and had an increased percentage of lung with organizing alveolitis between 100 and 160 days compared to Sod2(+/+) wild-type littermates. Both Sod2(+/-) and Sod2(+/+) littermates injected intratracheally with human manganese superoxide dismutase-plasmid/liposome (SOD2-PL) complex 24 h prior to whole-lung irradiation showed decreased DNA strand breaks and improved survival with decreased organizing alveolitis. Thus underexpression of MnSOD in the lungs of heterozygous Sod2(+/-) knockout mice is associated with increased pulmonary radiation sensitivity and parallels increased radiation sensitivity of embryo fibroblast cell lines in vitro. The restoration of cellular radioresistance in vitro and in lungs in vivo by SOD2-PL transgene expression supports a potential role for SOD2-PL gene therapy in organ-specific radioprotection.


Assuntos
Terapia Genética , Pulmão/efeitos da radiação , Pneumonite por Radiação/terapia , Superóxido Dismutase/deficiência , Animais , Ciclo Celular/efeitos da radiação , Linhagem Celular , Ensaio Cometa , DNA/efeitos da radiação , Dano ao DNA , Relação Dose-Resposta à Radiação , Embrião de Mamíferos , Fibroblastos/enzimologia , Fibroblastos/efeitos da radiação , Humanos , Injeções , Lipossomos/administração & dosagem , Pulmão/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Pneumonite por Radiação/enzimologia , Pneumonite por Radiação/etiologia , Pneumonite por Radiação/genética , Tolerância a Radiação/genética , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Superóxido Dismutase/genética , Traqueia , Transgenes
14.
Radiat Res ; 155(1 Pt 1): 2-14, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11121210

RESUMO

Radiation of the esophagus of C3H/HeNsd mice with 35 or 37 Gy of 6 MV X rays induces significantly increased RNA transcription for interleukin 1 (Il1), tumor necrosis factor alpha (Tnf), interferon gamma inducing factor (Ifngr), and interferon gamma (Ifng). These elevations are associated with DNA damage that is detectable by a comet assay of explanted esophageal cells, apoptosis of the esophageal basal lining layer cells in situ, and micro-ulceration leading to dehydration and death. The histopathology and time sequence of events are comparable to the esophagitis in humans that is associated with chemoradiotherapy of non-small cell lung carcinoma (NSCLC). Intraesophageal injection of clinical-grade manganese superoxide dismutase-plasmid/liposome (SOD2-PL) 24 h prior to irradiation produced an increase in SOD2 biochemical activity in explanted esophagus. An equivalent therapeutic plasmid weight of 10 microgram ALP plasmid in the same 500 microliter of liposomes, correlated to around 52-60% of alkaline phosphatase-positive cells in the squamous layer of the esophagus at 24 h. Administration of SOD2-PL prior to irradiation mediated a significant decrease in induction of cytokine mRNA by radiation and decreased apoptosis of squamous lining cells, micro-ulceration, and esophagitis. Groups of mice receiving 35 or 37 Gy esophageal irradiation by a technique protecting the lungs and treating only the central mediastinal area were followed to assess the long-term effects of radiation. SOD2-PL-treated irradiated mice demonstrated a significant decrease in esophageal wall thickness at day 100 compared to irradiated controls. Mice with orthotopic thoracic tumors composed of 32D-v-abl cells that received intraesophageal SOD2-PL treatment showed transgenic mRNA in the esophagus at 24 h, but no detectable human SOD2 transgene mRNA in explanted tumors by nested RT-PCR. These data provide support for translation of this strategy of SOD2-PL gene therapy to studies leading to a clinical trial in fractionated irradiation to decrease the acute and chronic side effects of radiation-induced damage to the esophagus.


Assuntos
Citocinas/biossíntese , Estenose Esofágica/prevenção & controle , Esofagite/prevenção & controle , Terapia Genética/métodos , Lesões por Radiação/prevenção & controle , Proteção Radiológica/métodos , Superóxido Dismutase/genética , Animais , Apoptose/efeitos da radiação , Citocinas/genética , Estenose Esofágica/etnologia , Estenose Esofágica/metabolismo , Esofagite/etiologia , Esofagite/metabolismo , Feminino , Expressão Gênica , Humanos , Lipossomos , Masculino , Neoplasias do Mediastino/genética , Neoplasias do Mediastino/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Plasmídeos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Lesões por Radiação/etnologia , Lesões por Radiação/metabolismo , Tolerância a Radiação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/metabolismo , Transgenes
15.
Radiat Res ; 154(1): 73-86, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10856968

RESUMO

Reverse transcription-polymerase chain reaction and immunofluorescence analysis of D2XRII murine bone marrow stromal cells showed that gamma irradiation with doses of 2-50 Gy from (137)Cs stimulated expression of nitric oxide synthase 2 (Nos2, also known as iNos). The activation of Nos2 was accompanied by an increase in the fluorescence of 4,5-diaminofluorescein diacetate, a nitric oxide trap, and accumulation of 3-nitrotyrosine within cellular proteins in a dose-dependent manner. These effects were inhibited by actinomycin D and by N-[3-(aminomethyl)benzyl]acetamidine dihydrochloride, a specific inhibitor of Nos2. The induction of Nos2 expression and Nos2-dependent release of nitric oxide in D2XRII cells was observed within 24 h after irradiation and was similar in magnitude to that observed in cultures incubated with Il1b and Tnf. We conducted (1) confocal fluorescence imaging of 3-nitrotyrosine in bone marrow cells of irradiated C57BL/6J mice and (2) 3-nitrotyrosine fluorescence imaging of FDC-P1JL26 hematopoietic cells that were cocultured with previously irradiated D2XRII bone marrow stromal cells. Exposure to ionizing radiation increased the production of 3-nitrotyrosine in irradiated bone marrow cells in vivo and in nonirradiated FDC-P1JL26 cells cocultured with irradiated D2XRII cells for 1 or 4 h. We suggest that nitrative/oxidative stress to the transplanted multilineage hematopoietic cells due to exposure to nitric oxide released by host bone marrow stromal cells may contribute to the genotoxic events associated with malignant alterations in bone marrow tissue of transplant recipients who are prepared for engraftment by total-body irradiation.


Assuntos
Células da Medula Óssea/efeitos da radiação , Óxido Nítrico Sintase/metabolismo , Tirosina/análogos & derivados , Animais , Células da Medula Óssea/enzimologia , Comunicação Celular , Ativação Enzimática , Imunofluorescência , Células-Tronco Hematopoéticas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase Tipo II , Doses de Radiação , Radiação Ionizante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/enzimologia , Células Estromais/efeitos da radiação , Tirosina/metabolismo
16.
J Steroid Biochem Mol Biol ; 39(5A): 729-34, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1958509

RESUMO

Therapeutic strategies for human breast cancer using 125I-labeled steroid hormones are clinically attractive in light of the estrogen dependence of many human breast cancers and the favorable microdosimetry resulting from 125I decay. We determined the uptake specific estrogen receptor binding and radiotoxicity of 17 alpha-[125I]iodovinyl-11 beta-methoxyestradiol (125IVME2) in vitro using cultured MCF-7 human breast carcinoma cells. 125IVME2 rapidly enters MCF-7 cells and reaches a plateau in the presence of competing 10(-7) M 17 beta-estradiol. In the absence of competitor, uptake is substantially greater before reaching a plateau. Efflux of 125IVME2 from cells incubated in the absence of estradiol decreases to levels corresponding to specific binding. Under equilibrium conditions and in the absence of competitor, 125IVME2 binds to both specific and nonspecific sites but, in the presence of excess 17 beta-estradiol, the observed binding is nonspecific. 125IVME2 is cytotoxic to exponentially growing MCF-7 cells and produces a survival curve typical of those observed for [125I]iododeoxyuridine and 16 alpha-[125I]iodoestradiol.


Assuntos
Sobrevivência Celular/efeitos da radiação , Estradiol/análogos & derivados , Radioisótopos do Iodo , Ligação Competitiva , Transporte Biológico , Neoplasias da Mama , Linhagem Celular , Estradiol/metabolismo , Estradiol/farmacologia , Feminino , Humanos , Cinética , Receptores de Estrogênio/metabolismo
17.
Nucl Med Biol ; 20(6): 719-26, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8401372

RESUMO

We have developed a very efficient labeling technique for monoclonal antibodies with technetium-99m. Oxidation of sugar residues on the IgG class of antibodies leads to the generation of aldehyde groups which are further reacted with two newly developed hydrazide compounds. This methodology introduces sulfhydryl groups on the antibody through sugar residues which can be labeled with technetium-99m. We have studied the TAG-72 specific second generation antibody CC-49. The specific activity of the labeled antibody was high without loss of its immunoreactivity.


Assuntos
Anticorpos/química , Antígenos de Neoplasias/imunologia , Carboidratos/química , Glicoproteínas/imunologia , Marcação por Isótopo/métodos , Compostos de Organotecnécio/síntese química , Tecnécio/química , Adenocarcinoma/metabolismo , Animais , Anticorpos/imunologia , Especificidade de Anticorpos , Neoplasias do Colo/metabolismo , Humanos , Imunoglobulina G/química , Camundongos , Camundongos Nus , Transplante de Neoplasias , Compostos de Organotecnécio/imunologia , Compostos de Organotecnécio/farmacocinética , Distribuição Tecidual , Triazóis/química
18.
Nucl Med Biol ; 20(1): 1-6, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8461873

RESUMO

B72.3 monoclonal antibody has been successfully boronated using mercaptoundecahydro-closo-dodecaborate (boron cage compound). The reagent was incorporated by first reacting the lysine residues of the antibody with m-maleimidobenzoyl succinimide ester (MBS), followed by Michael addition to the maleimido group by the mercapto boron cage compound to form a physiologically stable thioether linkage. Boron content of the antibody was determined by atomic absorption spectroscopy. For biodistribution studies, boronated antibody was radioiodinated with idogen. 125I-labeled and boronated B72.3 monoclonal antibody demonstrated clear tumor localization when administered via tail vein injections to athymic nude mice bearing LS174-T tumor xenografts. Boronated antibody was calculated to deliver 10(6) boron atoms per tumor cell. Although this falls short of the specific boron content originally proposed as necessary for boron neutron capture therapy (BNCT), recent calculations suggest that far fewer atoms of 10B per tumor cell would be necessary to effect successful BNCT when the boron is targeted to the tumor cell membrane.


Assuntos
Anticorpos Monoclonais/biossíntese , Terapia por Captura de Nêutron de Boro , Adenocarcinoma/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Boroidretos , Boro/análise , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Espectrofotometria Atômica , Compostos de Sulfidrila , Células Tumorais Cultivadas
19.
Nucl Med Biol ; 21(2): 171-8, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9234280

RESUMO

A cleaning resin has been developed for the non-chromatographic purification of 99mTc-labeled monoclonal antibodies (MAbs). The resin used is a modified form of thiopropylsepharose 6B resin, in which its sulfhydryl groups have been tinylated with stannous chloride. The method requires only simple stirring of the radiolabeling reaction mixture with this tinylated resin and subsequent separation of it from the resin by filtration to obtain a 99mTc-labeled MAb of radiopharmaceutical purity. The method provides an alternative to chromatographic purification of the radiolabeled MAb (i.e. gel filtration or anion exchange chromatography) which has been used in other 99mTc-MAb preparations. For comparison studies, we labeled the B72.3 MAb with NeoRx's diamide dimercaptide chelate radiolabeling kit, split the reaction mixture into two equal portions and then purified one portion with anion exchange chromatography (NeoRx's chosen method) while the other portion was purified with our cleaning resin. Comparison of HPLC chromatograms, percent 99mTc-bound to MAb, biodistribution and scintigraphic results show that our cleaning resin methodology provides a 99mTc-labeled MAb of essentially equal purity and utility as does the established, chromatographic one.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Compostos Radiofarmacêuticos/isolamento & purificação , Resinas Vegetais , Sefarose/análogos & derivados , Tecnécio , Animais , Neoplasias do Colo/diagnóstico por imagem , Neoplasias do Colo/metabolismo , Humanos , Camundongos , Camundongos Nus , Estrutura Molecular , Cintilografia , Tecnécio/isolamento & purificação , Distribuição Tecidual/fisiologia , Transplante Heterólogo
20.
Anticancer Res ; 13(6A): 1953-6, 1993.
Artigo em Inglês | MEDLINE | ID: mdl-7507652

RESUMO

A model is described by which in vivo tumor-MOAB interactions may be investigated. The method is rapid and may aid in the selection of appropriate MOABS from a panel of MOABS for individualized patient treatment. Groups of athymic nude mice were injected intravenously with small cell lung cancer line SHP-77 cells which are trapped primarily in the lungs. Twenty-four hours post tumor cell inoculation, 186Rhenium tagged HNK1 MOAB (CD57) was injected intravenously. Controls which received no tumor cells were injected with unbound 186Re or radioactive MOAB. Biodistribution studies at 24 hours following MOAB injection showed significantly more radioactivity in lungs of mice inoculated with both SHP-77 cells and 186Re MOAB than did lungs of controls.


Assuntos
Carcinoma de Células Pequenas/radioterapia , Neoplasias Pulmonares/radioterapia , Radioimunoterapia , Rênio/uso terapêutico , Animais , Anticorpos Monoclonais/metabolismo , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos CD57 , Feminino , Humanos , Camundongos , Camundongos Nus , Radioisótopos/uso terapêutico , Rênio/farmacocinética , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA