Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Acta Obstet Gynecol Scand ; 100(9): 1557-1580, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33755191

RESUMO

INTRODUCTION: Immunosuppressant drugs are increasingly being used in the reproductive years. Theoretically, such medications could affect fetal health either through changes in the sperm DNA or through fetal exposure caused by a presence in the seminal fluid. This systematic overview summarizes existing literature on the spermatotoxic and genotoxic potentials of methotrexate (MTX), a drug widely used to treat rheumatic and dermatologic diseases, and mycophenolate mofetil (MMF), which alone or supplemented with ganciclovir (GCV) may be crucial for the survival of organ transplants. MATERIAL AND METHODS: The systematic overview was performed in accordance with the PRISMA guidelines: A systematic literature search of the MEDLINE and Embase databases was done using a combination of relevant terms to search for studies on spermatotoxic or genotoxic changes related to treatment with MTX, GCV or MMF. The search was restricted to English language literature, and to in vivo animal studies (mammalian species) and clinical human studies. RESULTS: A total of 102 studies were identified, hereof 25 human and 77 animal studies. For MTX, human studies of immunosuppressive dosages show transient effect on sperm quality parameters, which return to reference values within 3 months. No human studies have investigated the sperm DNA damaging effect of MTX, but in other organs the genotoxic effects of immunosuppressive doses of MTX are fluctuating. In animals, immunosuppressive and cytotoxic doses of MTX adversely affect sperm quality parameters and show widespread genotoxic damages in various organs. Cytotoxic doses transiently change the DNA material in all cell stages of spermatogenesis in rodents. For GCV and MMF, data are limited and the results are indeterminate, for which reason spermatotoxic and genotoxic potentials cannot be excluded. CONCLUSIONS: Data from human and animal studies indicate transient spermatotoxic and genotoxic potentials of immunosuppressive and cytotoxic doses of MTX. There are a limited number of studies investigating GCV and MMF.


Assuntos
Ganciclovir/toxicidade , Imunossupressores/toxicidade , Metotrexato/toxicidade , Ácido Micofenólico/toxicidade , Dano ao DNA/efeitos dos fármacos , Ganciclovir/farmacologia , Humanos , Imunossupressores/farmacologia , Masculino , Metotrexato/farmacologia , Ácido Micofenólico/farmacologia , Espermatozoides/efeitos dos fármacos
2.
Zhonghua Yan Ke Za Zhi ; 56(4): 279-285, 2020 Apr 11.
Artigo em Zh | MEDLINE | ID: mdl-32306620

RESUMO

Objective: To elevated the retinal toxicity of intravitreal ganciclovir in albino rabbit eyes. Methods: Experimental study. Twenty-four New Zealand albino rabbits (forty-eight eyes) were divided into four groups by random. Three groups were prepared for ganciclovir experiment, named A, B, C. Each group received intravitreal injection ganciclovir dose at 400 µg/0.05 ml, 2 mg/0.05 ml and 5 mg/0.05 ml respectively. The other group named D served as a control accepted intravitreal injection 0.9% normal saline 0.1 ml. Before and after 1, 2 and 4 weeks, flicker full field electroretina gram (ERG) was recorded. After 1, 2 and 4 weeks light and electron microscopic tests were recorded for further toxicity study. Results: There was significant difference in amplitude of maximal combined response a wave in one week(χ(2)=8.319, P=0.04), and pairwise comparison the 5 mg group (140.50 µV) was significantly lower than the control group (165.00 µV) (χ(2)=-2.830, P=0.028). Maximal combined response b wave in four weeks(χ(2)=-10.626, P=0.014), and pairwise comparison the 5 mg group (261.50 µV) was significantly lower than the control group (398.00 µV) (χ(2)=-2.973, P=0.018). 30 Hz flicker response in one, two and four weeks(χ(2)=17.589, 8.225, 8.997, P=0.001, 0.042, 0.02), and pairwise comparison the 5 mg group (71.3µV, 106.00µV, 63.60µV) was significantly lower than the control group (118.50µV, 129.00µV, 116.50µV) (χ(2)=-4.142, -2.826, -2.713, P=0.000, 0.028, 0.040). There was no histologic retinal toxicity evidence of group 400 µg and control group observed by light microscopy in any stage of the study. Histologic changes of group 2 mg four week later, group 5 mg two and four week later include inner nuclear layer loose arranged, nuclear of ganglia were widened and outer plexiform layer stained less in four week later. By electron microscopic observation, the ultrastructure of retina changed to different degrees and became worse in each experimental group with significant mitochondrial swelling and hydropic changes were seen in the inner segments of photoreceptors, loosely arranged and disordered in the outer segment of photoreceptors four weeks later. Conclusions: The retinal function and morphology were normal in group 400 µg. Group 2 mg and 5 mg had retinal toxicity, and 5 mg was more severe. Therefore, the clinical application of ganciclovir in the treatment of acute retinal necrosis (ARN) should select the minimum effective dose to avoid the occurrence of retinal toxicity. (Chin J Ophthalmol, 2020, 56:279-285).


Assuntos
Ganciclovir/toxicidade , Retina/efeitos dos fármacos , Animais , Injeções Intravítreas , Coelhos , Distribuição Aleatória , Retina/patologia , Retina/ultraestrutura , Testes de Toxicidade
3.
Gastroenterology ; 153(4): 1068-1081.e7, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28711628

RESUMO

BACKGROUND & AIMS: When the glial fibrillary acidic protein (GFAP) promoter is used to express cellular toxins that eliminate glia in mice, intestinal epithelial permeability and proliferation increase; this led to the concept that glia are required for maintenance of the gastrointestinal epithelium. Many enteric glia, however, particularly in the mucosa, do not express GFAP. In contrast, virtually all enteric glia express proteolipid protein 1 (PLP1). We investigated whether elimination of PLP1-expressing cells compromises epithelial maintenance or gastrointestinal motility. METHODS: We generated mice that express tamoxifen-inducible Cre recombinase under control of the Plp1 promoter and carry the diptheria toxin subunit A (DTA) transgene in the Rosa26 locus (Plp1CreER;Rosa26DTA mice). In these mice, PLP1-expressing glia are selectively eliminated without affecting neighboring cells. We measured epithelial barrier function and gastrointestinal motility in these mice and littermate controls, and analyzed epithelial cell proliferation and ultrastructure from their intestinal tissues. To compare our findings with those from previous studies, we also eliminated glia with ganciclovir in GfapHSV-TK mice. RESULTS: Expression of DTA in PLP1-expressing cells selectively eliminated enteric glia from the small and large intestines, but caused no defects in epithelial proliferation, barrier integrity, or ultrastructure. In contrast, administration of ganciclovir to GfapHSV-TK mice eliminated fewer glia but caused considerable non-glial toxicity and epithelial cell death. Elimination of PLP1-expressing cells did not reduce survival of neurons in the intestine, but altered gastrointestinal motility in female, but not male, mice. CONCLUSIONS: Using the Plp1 promoter to selectively eliminate glia in mice, we found that enteric glia are not required for maintenance of the intestinal epithelium, but are required for regulation of intestinal motility in females. Previous observations supporting the concept that maintenance of the intestinal epithelium requires enteric glia can be attributed to non-glial toxicity in GfapHSV-TK mice and epithelial-cell expression of GFAP. Contrary to widespread notions, enteric glia are therefore not required for epithelial homeostasis. However, they regulate intestinal motility in a sex-dependent manner.


Assuntos
Sistema Nervoso Entérico/fisiologia , Motilidade Gastrointestinal , Mucosa Intestinal/fisiologia , Intestinos/inervação , Neuroglia/fisiologia , Animais , Proliferação de Células , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/ultraestrutura , Feminino , Ganciclovir/toxicidade , Genótipo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Homeostase , Integrases/genética , Integrases/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestrutura , Intestinos/efeitos dos fármacos , Intestinos/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fenótipo , Regiões Promotoras Genéticas , RNA não Traduzido/genética , Fatores Sexuais , Fatores de Tempo
4.
Arch Toxicol ; 90(8): 1975-81, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27224990

RESUMO

Some nucleoside analogues are used to treat herpes simplex and other viral infections. They are known to impair spermatogenesis, but published data are scarce. We studied the effects of four nucleosides on SerW3 cells, a rat Sertoli cell line. Cells were cultured for 3 days in DMEM supplemented with four different concentrations of each drug. Aciclovir and ganciclovir were added at concentrations of 0.3, 1, 3 and 10 mg/l medium; penciclovir and its prodrug famciclovir were used at higher concentrations (3, 10, 30, 100 mg/l medium). After a culture period of 3 days, we analysed the expression of connexin43, N-cadherin and the cytoskeleton protein vimentin by Western blot. Aciclovir caused a clear-cut effect at the highest concentration tested (10 mg/l), which is less than the peak plasma concentration achieved in patients during intravenous therapy with the drug. Connexin43, vimentin and N-cadherin content decreased to 49.8 ± 17, 44.0 ± 4 and 75.4 ± 1.5 % of the control values, respectively (n = 3; mean ± SD). Similar effects were observed with the prodrug ganciclovir (43.2 ± 10.8; 54.1 ± 11.9; 84.4 ± 10.8 % of controls). Penciclovir caused less pronounced effects at 10 mg/l medium (82.1 ± 20.6; 90.0 ± 12.0; 76.5 ± 17.7 % of controls). Only a slight effect was observed with famciclovir. Even at a 10-fold concentration (100 mg/l), just moderate changes were induced. In summary, we observed clear-cut effects with aciclovir and ganciclovir on Sertoli cells in vitro at therapeutically relevant concentrations and identified connexin43 as the most sensitive marker.


Assuntos
2-Aminopurina/análogos & derivados , Aciclovir/toxicidade , Antivirais/toxicidade , Células de Sertoli/efeitos dos fármacos , 2-Aminopurina/toxicidade , Aciclovir/análogos & derivados , Animais , Biomarcadores/metabolismo , Western Blotting , Caderinas/genética , Técnicas de Cultura de Células , Linhagem Celular , Conexina 43/genética , Relação Dose-Resposta a Droga , Famciclovir , Ganciclovir/toxicidade , Guanina , Masculino , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/genética , Ratos , Células de Sertoli/metabolismo , Vimentina/genética
5.
J Hepatol ; 60(2): 298-305, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24060854

RESUMO

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) that express glial fibrillary acidic protein (GFAP) are located between the sinusoidal endothelial cells and hepatocytes. HSCs are activated during liver injury and cause hepatic fibrosis by producing excessive extracellular matrix. HSCs also produce many growth factors, chemokines and cytokines, and thus may play an important role in acute liver injury. However, this function has not been clarified due to unavailability of a model, in which HSCs are depleted from the normal liver. METHODS: We treated mice expressing HSV-thymidine kinase under the GFAP promoter (GFAP-Tg) with 3 consecutive (3 days apart) CCl4 (0.16 µl/g; ip) injections to stimulate HSCs to enter the cell cycle and proliferate. This was followed by 10-day ganciclovir (40 µg/g/day; ip) treatment, which is expected to eliminate actively proliferating HSCs. Mice were then subjected to hepatic ischemia/reperfusion (I/R) or endotoxin treatment. RESULTS: CCl4/ganciclovir treatment caused depletion of the majority of HSCs (about 64-72%), while the liver recovered from the initial CCl4-induced injury (confirmed by histology, serum ALT and neutrophil infiltration). The magnitude of hepatic injury due to I/R or endotoxemia (determined by histopathology and serum ALT) was lower in HSC-depleted mice. Their hepatic expression of TNF-α, neutrophil chemoattractant CXCL1 and endothelin-A receptor also was significantly lower than the control mice. CONCLUSIONS: HSCs play an important role both in I/R- and endotoxin-induced acute hepatocyte injury, with TNF-α and endothelin-1 as important mediators of these effects.


Assuntos
Células Estreladas do Fígado/patologia , Células Estreladas do Fígado/fisiologia , Fígado/lesões , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Animais , Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Quimiocina CXCL1/genética , Modelos Animais de Doenças , Endotelina-1/genética , Ganciclovir/toxicidade , Expressão Gênica , Proteína Glial Fibrilar Ácida , Células Estreladas do Fígado/efeitos dos fármacos , Interleucina-6/genética , Lipopolissacarídeos/toxicidade , Fígado/patologia , Fígado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Traumatismo por Reperfusão/genética , Timidina Quinase/genética , Fator de Necrose Tumoral alfa/genética
7.
Nat Med ; 6(3): 320-6, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10700235

RESUMO

To provide long-term therapy in patients with severe toxin-induced hepatic parenchymal damage, donor hepatocytes would need to replicate and replace a large portion of the damaged parenchyma. Using a mouse model developed to reproduce this type of hepatic injury, we found that hepatocyte transplantation only slightly improved survival after transplantation despite the fact that many non-survivors showed moderate liver repopulation by donor cells. Perhaps accounting for this outcome, donor parenchyma in non-survivors did not have typical lobular organization. These results indicate that the re-creation of functional parenchyma by transplanted hepatocytes requires time, during which donor cells proliferate and then establish normal parenchymal architecture.


Assuntos
Transplante de Células , Ganciclovir/toxicidade , Neoplasias Hepáticas Experimentais/patologia , Neoplasias Hepáticas Experimentais/terapia , Fígado/citologia , Fosfatase Alcalina/genética , Animais , Intervalo Livre de Doença , Elementos Facilitadores Genéticos , Terapia Genética , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas Experimentais/induzido quimicamente , Metalotioneína/genética , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/biossíntese , Albumina Sérica/genética , Timidina Quinase/genética
8.
Yakugaku Zasshi ; 138(1): 107-110, 2018.
Artigo em Japonês | MEDLINE | ID: mdl-29311455

RESUMO

The concentration of ganciclovir eye drops for hospital preparation was changed from 0.5% to 2.0% at the Nagasaki University Hospital from March 2015. We investigated the incidence of side effects in 12 patients using 2.0% ganciclovir eye drops and evaluated the cytotoxicity of 2.0% ganciclovir eye drops using cultured rabbit corneal cells in vitro. As a side effect of 2.0% ganciclovir eye drops, three patients exhibited an early feeling of transient stimulation. The 2.0% ganciclovir eye drops did not demonstrate cell cytotoxicity for cultured corneal cells after 5 min, but did after 10 min. These findings suggested that the 2.0% ganciclovir eye drops can be used without corneal epithelium disorder in clinical settings.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Ganciclovir/efeitos adversos , Animais , Células Cultivadas , Córnea/citologia , Córnea/efeitos dos fármacos , Testes Imunológicos de Citotoxicidade , Relação Dose-Resposta a Droga , Ganciclovir/toxicidade , Hospitais Universitários , Humanos , Incidência , Japão/epidemiologia , Soluções Oftálmicas , Coelhos , Fatores de Tempo
9.
Toxicol Sci ; 162(2): 488-498, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29228316

RESUMO

Ganciclovir (GCV) has been implicated in the development of testicular alterations. Exposure on gestational day (GD) 10 in rats induced permanent effects, including focal reduction or absence of germ cells (Sertoli cell-only tubules). Because the timing of exposure can be critical for testicular effects, we exposed rat dams to 300 mg/kg GCV (3 100 mg/kg subcutaneous injections) on GD10, 14 and 19, when germ cells have high rates of migration, proliferation and are mitotically quiescent, respectively. Males exposed to GCV in utero on GD10 and 14 were evaluated for androgenization markers, serum and fecal androgens, and testicular histomorphometry at adulthood. Double-labeling immunofluorescence for DAZL and Ki67 were used to assess gonocytes number and the proliferative activity of germ and somatic cells in fetal testes on GD15 and 20, ie, 24 h after GCV exposure. Adult rats exposed on GD14 showed delayed puberty onset, despite normal androgen levels. Also, there was a 50% reduction in testicular weight and about 30% of seminiferous tubules lacking germ cells. Effects on GD10 animals were less pronounced. In the fetal testis, the number of gonocytes was reduced by 50% in rats exposed on GD14, but normal in GD19 fetuses. GCV also reduced Sertoli cell proliferation immunolabeling in GD19 fetuses and Sertoli cell number in adults. In conclusion, GCV toxicity on germ cells seems to be linked to their proliferation rate and GD14 is a critical window in rats, when GCV exposure causes an acute massive loss of germ cells that persists until adulthood.


Assuntos
Antivirais/administração & dosagem , Ganciclovir/administração & dosagem , Organogênese/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Maturidade Sexual/efeitos dos fármacos , Testículo/efeitos dos fármacos , Animais , Antivirais/toxicidade , Proliferação de Células/efeitos dos fármacos , Feminino , Ganciclovir/toxicidade , Células Germinativas/efeitos dos fármacos , Células Germinativas/patologia , Idade Gestacional , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Wistar , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Fatores de Tempo
10.
Methods Mol Biol ; 1506: 179-192, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27830553

RESUMO

Hepatocyte transplantation is the best approach to maintain and propagate differentiated hepatocytes from different species. Host liver has to be adapted for transplanted hepatocytes productive engraftment and proliferation being required a chronic liver injury to eliminate host hepatocytes and provide a proliferative advantage to the transplanted hepatocytes. Most valuable mouse models for xenograft hepatocyte transplantation are based on genetically modified animals to cause a chronic liver damage and to limit host hepatocyte regeneration potential. We present a methodology that generates a chronic liver damage and can be applied to any host mouse strain and animal species based on the inoculation of a recombinant adenovirus to express herpes simplex thymidine kinase in host hepatocytes sensitizing them to ganciclovir treatment. This causes a prolonged liver damage that allows hepatocyte transplantation and generation of regenerative nodules in recipient mouse liver integrated by transplanted cells and host sinusoidal. Obtained chimeric animals maintain functional chimeric nodules for several weeks, ready to be used in any study.


Assuntos
Adenoviridae/genética , Transplante de Células/métodos , Hepatócitos/transplante , Regeneração Hepática/efeitos dos fármacos , Fígado/fisiologia , Condicionamento Pré-Transplante/métodos , Animais , Separação Celular/métodos , Transplante de Células/efeitos adversos , Transplante de Células/instrumentação , Doença Hepática Crônica Induzida por Substâncias e Drogas , Modelos Animais de Doenças , Ganciclovir/toxicidade , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Simplexvirus/genética , Timidina Quinase/genética , Transdução Genética/métodos , Quimeras de Transplante/fisiologia , Quimeras de Transplante/cirurgia , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos , Proteínas não Estruturais Virais/genética
11.
Environ Toxicol Chem ; 36(8): 2205-2216, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28198039

RESUMO

Potential environmental risks of the old antiviral pharmaceuticals ganciclovir (GCV) and valganciclovir (VGCV) were reassessed based on new environmental fate and chronic ecotoxicity tests and on actual use data for Europe. Valganciclovir is hydrolyzed to GCV by intestinal and hepatic esterases, and hence the new environmental tests only refer to GCV. A sorption study showed that GCV will not sorb significantly, excluding the soil as a relevant environmental compartment. Despite earlier data suggesting nondegradability, a new water/sediment fate test showed GCV to be primarily and ultimately degraded and to be nonpersistent. The chronic ecotoxicity tests with algae and daphnids resulted in no inhibition at the highest tested concentrations, whereas a fish partial life cycle test, selected in view of mammalian mutagenicity and reprotoxicity data, showed effects on growth of the young fish, but not on gametogenesis, fertilization, embryogenesis, or teratogenicity. Predicted environmental concentrations were derived based on actual per capita use data for European countries for 2004 to 2014, and the highest was selected for the risk assessment. A comparison of predicted environmental concentrations with predicted no-effect concentrations shows no significant risk for wastewater treatment, surface waters, groundwater, or sediment. In addition, potential risks to (semi)aquatic top predators or to human consumers of water and fish are exceedingly low. Environ Toxicol Chem 2017;36:2205-2216. © 2017 The Author. Environmental Toxicology and Chemistry Published by Wiley Periodicals, Inc. on behalf of SETAC.


Assuntos
Antivirais/análise , Exposição Ambiental/análise , Ganciclovir/análogos & derivados , Ganciclovir/análise , Poluentes Químicos da Água/análise , Animais , Antivirais/toxicidade , Clorófitas/efeitos dos fármacos , Daphnia/efeitos dos fármacos , Europa (Continente) , Peixes/metabolismo , Água Doce/química , Ganciclovir/toxicidade , Sedimentos Geológicos/química , Humanos , Valor Preditivo dos Testes , Reprodução/efeitos dos fármacos , Medição de Risco , Solo/química , Valganciclovir , Poluentes Químicos da Água/toxicidade
12.
Cancer Gene Ther ; 13(2): 150-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16096650

RESUMO

Adenoviral vectors are widely used in cancer gene therapy. After systemic administration however, the majority of the virus homes to the liver and the expressed transgene may cause hepatotoxicity. To restrict transgene expression to tumor cells, tumor- or tissue-specific promoters are utilized. The tumor antigen epithelial glycoprotein-2 (EGP-2), also known as Ep-CAM, is expressed in many cancers from different epithelial origins. In this study, the EGP-2 promoter was shown to restrict the expression of luciferase and thymidine kinase in an adenoviral context in different cell lines. In vivo, the EGP-2 promoter mediated efficient expression of luciferase in tumors but showed a 3-log lower activity in liver tissue when compared with the cytomegalovirus (CMV) promoter. Similarly, the EGP-2 promoter mediated specific cell killing after ganciclovir treatment in EGP-2-positive cells. Moreover, in vivo, this treatment regiment did not cause any rise in the liver enzymes aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT), demonstrating absence of liver toxicity. In contrast, CMV-mediated expression of thymidine kinase in combination with ganciclovir treatment resulted in high ASAT and ALAT values. This study demonstrates the value of the EGP-2 promoter to restrict transgene expression to a broad range of tumor types, thereby preventing liver toxicity.


Assuntos
Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Regulação Neoplásica da Expressão Gênica , Terapia Genética/métodos , Neoplasias/terapia , Regiões Promotoras Genéticas/genética , Adenoviridae , Alanina Transaminase/metabolismo , Animais , Aspartato Aminotransferases/metabolismo , Linhagem Celular Tumoral , Primers do DNA , Molécula de Adesão da Célula Epitelial , Ganciclovir/toxicidade , Vetores Genéticos/genética , Humanos , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timidina Quinase/metabolismo , Timidina Quinase/toxicidade , Testes de Toxicidade , Transgenes/genética
13.
Nucleic Acids Res ; 32(20): e161, 2004 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-15561996

RESUMO

Genetic ablation experiments are used to resolve problems regarding cell lineages and the in vivo function of certain groups of cells. We describe a two-component conditional ablation technology using a mouse carrying an X-linked puDeltatk transgene, which is only activated in cells expressing Cre. Ablation of the Cre-expressing cells can be temporally regulated by the time of ganciclovir (GCV) administration. This strategy was demonstrated using a Col2Cre transgenic line. Differentiating chondrocytes in bigenic animals could be ablated at different developmental stages resulting in disorganized growth plates and dwarfism. Macrocephaly, macroglossia and umbilical hernia were also observed in ablated 18.5 dpc embryos. Crosses between the puDeltatk selector transgenic line and existing cre lines will facilitate numerous temporally regulated tissue-specific ablation experiments.


Assuntos
Genes Transgênicos Suicidas , Integrases/genética , Camundongos Transgênicos/genética , Timidina Quinase/genética , Proteínas Virais/genética , Acetiltransferases/genética , Animais , Desenvolvimento Ósseo , Diferenciação Celular , Proliferação de Células , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Embrião de Mamíferos/citologia , Ganciclovir/toxicidade , Integrases/metabolismo , Camundongos , Camundongos Transgênicos/embriologia , Camundongos Transgênicos/crescimento & desenvolvimento , Proteínas Virais/metabolismo
14.
Cancer Res ; 57(9): 1699-703, 1997 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9135011

RESUMO

Although considerable attention has been directed in the field of gene therapy toward elucidating the mechanism by which a transduced cell could kill a bystander cell, little is known about how bystander cells may affect transduced cells. We hypothesized that bystander cells, particularly if they were capable of gap junctional communication, could protect cells transduced with the herpes simplex virus thymidine kinase (HSV-TK) from ganciclovir (GCV)-induced cytotoxicity. To test this hypothesis, we used a rat hepatocyte cell line (WB) that can carry out efficient gap junctional communication, a WB clone transduced with HSV-TK (WB-TK), and a communication-incompetent subclone of WB cells (aB1). We cocultured WB-TK cells with either WB or aB1 cells, treated them with GCV, and then plated the cells into selective media that permitted us to quantify independently the surviving fraction of WB-TK cells or bystander cells. We found that WB bystander cells conferred up to a 1000-fold protection on WB-TK cells treated with GCV. aB1 cells conferred detectable, but significantly less, protection. These findings demonstrate that herpes simplex virus thymidine kinase-transduced cells can be significantly protected by bystander cells, particularly those that can carry out gap junctional communication. Whether this "Good Samaritan" effect improves the overall efficacy of gene therapy, by prolonging the survival of the source of toxic metabolites, or decreases effectiveness by increasing the survival of transduced cells will need to be determined in vivo.


Assuntos
Ganciclovir/toxicidade , Timidina Quinase/administração & dosagem , Animais , Comunicação Celular , Células Cultivadas , Junções Comunicantes/fisiologia , Terapia Genética/métodos , Ratos , Transdução Genética
15.
Cancer Res ; 53(21): 5274-83, 1993 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8221662

RESUMO

Tumor cells expressing the herpes simplex virus thymidine kinase (HSV-TK) gene are sensitive to the drug ganciclovir (GCV). We demonstrate here that HSV-TK-positive cells exposed to GCV were lethal to HSV-TK-negative cells as a result of a "bystander effect." HSV-TK-negative cells were killed in vitro when the population of cultured cells contained only 10% HSV-TK-positive cells. The mechanism of this "bystander effect" on HSV-TK-negative cells appeared to be related to the process of apoptotic cell death when HSV-TK-positive cells were exposed to GCV. Flow cytometric and electron microscopic analyses suggested that apoptotic vesicles generated from the dying gene-modified cells were phagocytized by nearby, unmodified tumor cells. Prevention of apoptotic vesicle transfer prevented the bystander effect. The toxic effect of HSV-TK-positive cells on HSV-TK-negative cells was reproduced in an in vivo model. A mixed population of tumor cells consisting of HSV-TK-positive and HSV-TK-negative cells was inoculated s.c. into mice. Regression of the tumor mass occurred when the inoculum consisted of as few as 10% HSV-TK-expressing tumor cells. The bystander effect was also demonstrated in i.p. tumor studies. Initial experiments demonstrated that prolonged survival (> 70 days) occurred when a mixture containing 50% HSV-TK-positive and 50% HSV-TK-negative cells was injected i.p. followed by GCV treatment. Further, survival was prolonged for mice with a preexisting HSV-TK-negative i.p. tumor burden by injecting HSV-TK-positive cells and GCV. These results suggest that genetic modification of tumor cells may be useful for developing cancer therapies.


Assuntos
Apoptose , Ganciclovir/toxicidade , Vírus do Sarcoma Murino de Kirsten , Infecções por Retroviridae/patologia , Sarcoma Experimental/patologia , Timidina Quinase/genética , Infecções Tumorais por Vírus/patologia , Animais , Linhagem Celular Transformada , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Infecções por Retroviridae/genética , Sarcoma Experimental/genética , Sarcoma Experimental/ultraestrutura , Simplexvirus/enzimologia , Simplexvirus/genética , Infecções Tumorais por Vírus/genética
16.
Cancer Res ; 59(3): 669-75, 1999 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9973216

RESUMO

The ability of herpes simplex virus type 1 thymidine kinase (HSV-TK)-expressing cells incubated with ganciclovir (GCV) to induce cytotoxicity in neighboring HSV-TK-negative (bystander) cells has been well documented. Although it has been suggested that this bystander cell killing occurs through the transfer of phosphorylated GCV, there is little direct proof that bystander cells can accumulate GCV nucleotides. We have studied the ability of U251 human glioblastoma cells expressing HSV-TK (U251tk cells) to induce cytotoxicity in neighboring U251 bystander cells that lack the viral kinase (U251beta gal cells) and evaluated whether this bystander cell killing is mediated by GCV nucleotides. The cytotoxicity studies demonstrated that the ratio of HSV-TK-expressing cells:bystander cells was important in determining the sensitivity of both cell types to GCV. U251tk cells cocultured with an equal number of U251beta gal cells (a 50:50 ratio) exhibited a sensitivity to GCV similar to that observed in the absence of bystander cells, with >99.8% cell kill at 1 microm GCV. However, in cultures with 10% U251tk cells and 90% bystander cells (a 10:90 ratio), 1 microM GCV decreased the survival of U251tk cells by only 54%. Strong bystander cell killing was observed at both ratios. In a 50:50 coculture of U251tk and U251beta gal cells, the survival of bystander cells was decreased by >99.5% with 3 microM GCV, whereas 30 microM GCV was required to effect a similar decrease in bystander cell survival when 90% of the culture consisted of U251beta gal cells. To determine whether this bystander cell killing may be mediated by GCV nucleotides, we developed a technique to separate the two cell populations after coculture. A U251 bystander cell line was developed from the parental cell line by transfection with the cDNA coding for green fluorescent protein (U251gfp cells), which permitted the separation of U251gfp cells from nonfluorescing U251tk cells by flow cytometry with cell sorting. With this technique, bystander cells were isolated in a viable state with >97% purity within 1 h after harvest, permitting analysis of the nucleotide pools for the presence of phosphorylated GCV. The results demonstrated that significant levels of the triphosphate of GCV (GCVTP) accumulated in bystander cells within 4 h of coculture, and this accumulation was dependent upon the percentage of HSV-TK-expressing cells as well as the concentration of GCV and the length of incubation. The proportion of GCVTP in bystander cells was consistently 50-80% of that in HSV-TK-expressing cells in the 50:50 or 10:90 cocultures, suggesting a facile transfer of phosphorylated GCV. However, the actual amount of GCVTP was as much as 8-fold lower in both the U251tk and U251beta gal cells cocultured at a ratio of 10:90 compared to those cocultured at a ratio of 50:50, which is consistent with the lesser effect on cell survival. When U251tk and U251gfp cells were cultured with 1-beta-D-arabinofuranosylthymine (araT), the 5'-triphosphate of araT accumulated in the bystander cells, demonstrating that the transfer of phosphorylated compounds between these cell types is not restricted to GCV nucleotides. However, the proportion of araT-5'-triphosphate in bystander cells compared to that in HSV-TK-expressing cells was lower than that for GCVTP, and the amount was not sufficient to decrease survival in the bystander population.


Assuntos
Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/toxicidade , Ganciclovir/análogos & derivados , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Herpesvirus Humano 1/enzimologia , Timidina Quinase/metabolismo , Arabinonucleotídeos/metabolismo , Técnicas de Cocultura , Ganciclovir/farmacocinética , Ganciclovir/toxicidade , Glioblastoma/enzimologia , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/biossíntese , Timidina Quinase/biossíntese
17.
Cancer Res ; 54(8): 2055-9, 1994 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8174104

RESUMO

Transfer of the herpes simplex virus thymidine kinase (HSVtk) gene into tumor cells using retroviral vectors followed by administration of ganciclovir provides a potential strategy for the treatment of malignancy. Because of the limitations of using retroviral vectors for clinical application, the feasibility of using a recombinant adenovirus containing HSVtk was examined. Cell lines derived from human malignant mesotheliomas and non-small cell lung cancers infected with a recombinant adenovirus containing HSVtk showed strong expression of HSVtk protein as determined by immunohistochemical staining. Infection with a recombinant adenovirus containing HSVtk rendered cells sensitive to doses of ganciclovir that were 2-3 logs lower than uninfected cells or those infected with a control virus. A strong "bystander effect" was noted in mesothelioma lines; there was no diminution in the efficacy of ganciclovir treatment until the ratio of infected:uninfected cells fell below 1:10. This study thus demonstrates in vitro efficacy of an adenovirus-transduced HSVtk drug sensitization gene therapy system in thoracic malignancies. Recombinant adenovirus transfer of the HSVtk gene followed by ganciclovir may have promise as an in situ treatment for tumors.


Assuntos
Divisão Celular/efeitos dos fármacos , Ganciclovir/toxicidade , Simplexvirus/enzimologia , Timidina Quinase/genética , Transfecção/métodos , Adenoviridae , Carcinoma Pulmonar de Células não Pequenas , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Vetores Genéticos , Humanos , Neoplasias Pulmonares , Mesotelioma , Recombinação Genética , Simplexvirus/genética , Neoplasias Torácicas , Timidina Quinase/biossíntese , Células Tumorais Cultivadas
18.
Cancer Res ; 61(20): 7399-403, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11606369

RESUMO

The efficacy of suicide herpes simplex virus-1 thymidine kinase (HSVtk)/ganciclovir (GCV) gene therapy is often limited by intrinsic resistance of tumor cells. Here we show that repair of GCV incorporated in DNA is a factor involved in GCV resistance. A protective role of DNA repair in GCV-induced cell killing is supported by the following findings: (a) GCV-exposed Chinese hamster ovary-HSVtk cells exhibited both reduced repair of GCV and cloning efficiency in the presence of a specific polymerase beta (beta-pol) inhibitor, prunasin; (b) DNA beta-pol-deficient mouse fibroblasts were more sensitive to the cytotoxic, apoptosis-inducing, and genotoxic (DNA breakage and chromosomal aberration-inducing) effects of GCV as compared with wild-type and beta-pol-complemented cell lines; (c) methoxyamine, an inhibitor of beta-pol-dependent short-patch base excision repair, sensitized wild-type and complemented beta-pol cells to GCV, whereas it had no effect on the sensitivity of beta-pol-null cells to GCV. Because methoxyamine-mediated sensitization of beta-pol wild-type and beta-pol-complemented cells to GCV did not reach the level of null cells, we suggest that both beta-pol-dependent short- and long-patch base excision repair are involved in protection of cells to GCV. Some implications for HSVtk/GCV gene therapy are being discussed.


Assuntos
Dano ao DNA , DNA Polimerase beta/metabolismo , Ganciclovir/toxicidade , Animais , Células CHO , Cricetinae , DNA/metabolismo , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/deficiência , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Ganciclovir/metabolismo , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Hidroxilaminas/farmacologia , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transfecção
19.
Cancer Res ; 61(7): 3016-21, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306481

RESUMO

We previously reported that the retroviral vector expressing the herpes simplex virus-thymidine kinase gene under the control of 0.3-kb human alpha-fetoprotein (AFP) gene promoter (AF0.3) provided the cytotoxicity to ganciclovir (GCV) in high-AFP-producing human hepatoma cells but not in low-AFP-producing cells. Therefore, specific enhancement of AFP promoter activity is likely to be required to induce enough cytotoxicity in low-AFP-producing hepatoma cells. In this study, we constructed a hybrid promoter, [HRE]AF, in which a 0.4-kb fragment of human vascular endothelial growth factor 5'-flanking sequences containing hypoxia-responsive element (HRE) was fused to AF0.3 promoter. By means of the reporter gene transfection assay, hypoxia-inducible transcriptions that were mediated by [HRE]AF promoter were detected in low- and non-AFP-producing human hepatoma cells, but not in nonhepatoma cells. When the herpes simplex virus-thymidine kinase gene controlled by [HRE]AF promoter was transduced into hepatoma and nonhepatoma cells by a retroviral vector, the exposure to 1% O2 induced GCV cytotoxicity specifically in the hepatoma cells. Moreover, in nude mice bearing solid tumor xenografts, only the tumors consisting of the virus-infected hepatoma cells gradually disappeared by GCV administration. These results indicate that the hypoxia-inducible enhancer of the human vascular endothelial growth factor gene, which is directly linked to human AFP promoter, involves selective and enhanced tumoricidal activity in gene therapy for hepatocellular carcinoma.


Assuntos
Carcinoma Hepatocelular/terapia , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Neoplasias Hepáticas/terapia , Regiões Promotoras Genéticas , alfa-Fetoproteínas/genética , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Hipóxia Celular/genética , Fatores de Crescimento Endotelial/genética , Ganciclovir/toxicidade , Vetores Genéticos/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Linfocinas/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Retroviridae/genética , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Timidina Quinase/metabolismo , Ativação Transcricional , Transdução Genética , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , alfa-Fetoproteínas/biossíntese
20.
Cancer Res ; 56(18): 4087-95, 1996 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8797571

RESUMO

Noninvasive imaging of herpes simplex virus type 1 thymidine kinase (HSV1-tk) gene expression is possible with a clinical gamma camera and by single-photon emission tomography (SPECT) using 131I-labeled 2'-fluoro-2'-deoxy-1-beta-D-arabinofuranosyl-5-iodo-uracil (FIAU). Studies were performed in rats bearing s.c. tumors. Tumors were produced by injection of wild-type RG2 glioma or W256 mammary carcinoma cells into one flank and RG2TK+ glioma or W256TK+ mammary carcinoma cells (that had been transduced in vitro with the HSV1-tk gene) into the opposite flank. In some animals, HSV1-tk gene transduction of the pre-established wild-type tumors was accomplished in vivo by direct intratumoral injection of retroviral vector-producer cells. Imaging studies were performed 2 weeks after tumor transduction to allow time for production and spread of the retroviruses through the tumor and for sufficient growth and increase in size of the tumors to facilitate imaging. The gamma camera and SPECT images revealed highly specific localization of [131I]FIAU-derived radioactivity to areas of HSV1-tk gene expression at 24, 36, and 48 h after i.v. administration of 1.6-2.8 mCi of [131I]FIAU. Comparative analysis of quantitative autoradiographic images obtained from the same tumors confirmed that the high levels of [131I]FIAU-derived radioactivity (> 1% dose) were localized to areas of HSV1-tk gene expression demonstrated by immunohistochemical staining for HSV1-tk protein. In contrast, significantly lower levels of [131I]FIAU-derived radioactivity (< 0.01%) were observed in the surrounding nontransduced tumor tissue, contralateral wild-type tumors, and other tissues that showed no immunohistochemical staining for the HSV1-tk protein. The magnitude of FIAU accumulation in RG2TK+, W256TK+, and wild-type tumors corresponded to the in vitro ganciclovir sensitivity of the cell lines used to produce these tumors, which indicates that the magnitude of FIAU accumulation reflects the level of HSV1-tk gene expression. We suggest that "clinically relevant" levels of HSV1-tk gene expression in transfected tissue can be imaged with [131I]FIAU and a gamma camera or SPECT, and that a significant improvement in imaging sensitivity and resolution is expected with [124I]FIAU and PET.


Assuntos
Antivirais/toxicidade , Ganciclovir/toxicidade , Técnicas de Transferência de Genes , Terapia Genética , Glioma/diagnóstico por imagem , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Timidina Quinase/biossíntese , Animais , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/síntese química , Arabinofuranosiluracila/metabolismo , Arabinofuranosiluracila/farmacocinética , Radioisótopos de Carbono , Linhagem Celular , Câmaras gama , Glioma/enzimologia , Glioma/patologia , Glioma/terapia , Imuno-Histoquímica/métodos , Radioisótopos do Iodo , Imageamento por Ressonância Magnética/métodos , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/terapia , Ratos , Ratos Nus , Sensibilidade e Especificidade , Simplexvirus , Timidina Quinase/análise , Timidina Quinase/genética , Tomografia Computadorizada de Emissão de Fóton Único/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA