Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Transplant Proc ; 41(1): 319-22, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19249545

RESUMEN

Cell-mediated immunity, especially of human CD8+ cytotoxic T lymphocytes (CTLs) is believed to have an important role in the long-term survival of pig islet xenografts. Protection against human CD8+ CTL cytotoxicity may reduce the direct damage to pig islets and enable long-term xenograft survival in pig-to-human islet xenotransplantation. We have previously reported that c-FLIP(S/L) genes, which are potent inhibitors of death receptor-mediated proapoptotic signals through binding competition with caspase-8 for recruitment to the Fas-associated via death domain (FADD), markedly suppress human CD8+ CTL-mediated xenocytotoxicity. In addition, the cytoprotective effects of c-FLIP(L) seem to be significantly stronger than those of c-FLIP(S). Accordingly, in the present study, expression of c-FLIP(L) was induced in intact pig islets by adenoviral transduction. Consequently, the cytoprotective capacity of the transgene in pig islets was examined in in vitro and in vivo exposure to human CD8+ CTLs. Cells from untransduced islets or mock islets were sensitive to CD8+ CTL-mediated lysis (59.3% +/- 15.9% and 64.0% +/- 8.9% cytotoxicity, respectively). In contrast, cells from pig islets transduced with the c-FLIP(L) gene were markedly protected from lysis (30.5% +/- 3.5%). Furthermore, prolonged xenograft survival was elicited from pig islets transduced with this molecule as assessed using an islet transplant model using the rat kidney capsule. Thus, these data indicate that intact pig islets can be transduced to express c-FLIP(L) with adenovirus. Pig islets expressing c-FLIP(L) are significantly resistant to human CTL killing and further exhibit beneficial effects to prolong xenograft survival.


Asunto(s)
Adenoviridae/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Regulación de la Expresión Génica , Islotes Pancreáticos/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Supervivencia Celular , Humanos , Inmunohistoquímica , Islotes Pancreáticos/citología , Trasplante de Islotes Pancreáticos/inmunología , Ratas , Ratas Endogámicas Lew , Porcinos , Transfección , Trasplante Heterólogo/inmunología
2.
Transplant Proc ; 41(1): 334-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19249550

RESUMEN

Islet transplantation can provide insulin independence in patients with type 1 diabetes mellitus. However, islet allograft recipients exhibit a gradual decline in insulin independence, and only 10% do not require insulin at 5 years. This decline may reflect drug toxicity to islet beta cells. Rapamycin, a central immunosuppressant in islet transplantation, is a mammalian target of rampamycin inhibitor that induces autophagy. The relative contributions of autophagy in transplanted islets are poorly understood. Therefore, in the present study we sought to evaluate the effects of rapamycin on islet beta cells. Rapamycin treatment of islets resulted in accumulation of membrane-bound light chain 3 (LC3-II) protein, an early marker of autophagy. In addition, rapamycin treatment of isolated islets elicited not only reduction of viability but also downregulation of in vitro potency. To further examine the occurrence of autophagy in rapamycin-treated islets, we used GFP (green fluorescent protein)-LC3 transgenic mice that express a fluorescent autophagosome marker. The GFP-LC3 signals were markedly increased in rapamycin treated islets compared with control islets. In addition, to show improvement by blockade of autophagic signaling, islets were treated with rapamycin in the presence of 3-methyladenine, which inhibits autophagy. Thereafter, both islet viability and islet potency were dramatically improved. The number of GFP-LC3 dots clearly increased after 3-MA treatment. Thus, rapamycin treatment of islets induces autophagy in vitro. This phenomenon may contribute to the progressive graft dysfunction of transplanted islets. Therapeutically targeting this novel signaling may yield significant benefits for long-term islet survival.


Asunto(s)
Autofagia/efectos de los fármacos , Trasplante de Islotes Pancreáticos/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Sirolimus/farmacología , Animales , Autofagia/fisiología , Genes Reporteros , Glucosa/farmacología , Cadenas Ligeras de Inmunoglobulina/genética , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal , Transfección
3.
Transplant Proc ; 41(1): 391-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19249564

RESUMEN

Human CD8(+) cytotoxic T lymphocyte (CTL)-mediated cytotoxicity, which participates in xenograft rejection, is mediated mainly by the Fas/FasL apoptotic pathway. We previously developed methods to inhibit human CTL xenocytotoxicity by extracellular remodeling using overexpression of membrane-bound human FasL on pig xenograft cells, and by intracellular blockade of death receptor-mediated apoptotic signals, such as the Fas/FasL pathway using the pig c-FLIP(L) molecule. To investigate the cooperative effects of both membrane-bound FasL and pig c-FLIP(L), we cotransfected both genes into pig endothelial cells (PEC). The double remodeling with these molecules effectively prevented CD8(+) CTL killing. Although double transfectants and single high transfectants of either membrane-bound FasL or c-FLIP(L) gene displayed similar inhibition of CTL cytotoxicity, the expression levels of these 2 molecules in double transfectants were almost half the expression levels of single transfectants. Furthermore, to show in vivo prolongation of xenograft survival, we transplanted PEC transfectants under the rat kidney capsule. Prolonged survival was displayed by PEC double transfectant xenografts whereas those from either parental PEC or MOCK (vehicle control) were completely rejected by day 5 posttransplantation. These data suggested that intracellular and extracellular remodeling by coexpression of membrane-bound FasL and pig c-FLIP(L) in xenograft cells may prevent an innate cellular response to xenografts. The gene compatibility of these molecules to generate transgenic pigs may be sufficient to create a window of opportunity to facilitate long-term xenograft survival.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Ligando Fas/genética , Linfocitos T Citotóxicos/inmunología , Trasplante Heterólogo/inmunología , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular/inmunología , Citotoxicidad Inmunológica , ADN Complementario/genética , Humanos , Inmunohistoquímica , Plásmidos/genética , Porcinos , Linfocitos T Citotóxicos/citología , Transfección
4.
Am J Transplant ; 8(2): 288-97, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18211505

RESUMEN

Overcoming cell-mediated immunity, especially of human CD8(+) CTLs, is important for the success of xenotransplantation. Our group has previously reported that the cytotoxicity of human CD8(+) CTLs against pig endothelial cells (PEC) is highly detrimental and mediated in major part by the Fas/FasL apoptotic pathway. Cellular FLICE inhibitory protein (c-FLIP) was originally identified as an inhibitor of death-receptor signaling through binding competition with caspase-8 for recruitment to Fas-associated via death domain (FADD). Two major c-FLIP variants result from alternative mRNA splicing: a short, 26-KDa protein (c-FLIP(S)) and a long, 55-KDa form (c-FLIP(L)). The cytoprotective effects of c-FLIP(S/L) in xenograft cells remain controversial. This study demonstrates that the overexpression of c-FLIP(S/L) genes markedly suppress human CD8(+) CTL-mediated xenocytotoxicity and, in addition, the cytoprotective effects of c-FLIP(L) appear to be significantly stronger than those of c-FLIP(S). Furthermore, to prove the prolonged effects of xenograft survival, PEC transfectants with c-FLIP(S/L) genes were transplanted under rat kidney capsules. Prolonged survival was elicited from FLIP(S/L) transfectants, whereas parental PEC was completely rejected through day 5, posttransplant. Thus, intracellular remodeling with the overexpression of c-FLIP(S/L) in xenograft cells may avoid innate cellular attacks against xenografts and facilitate long-term xenograft survival.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Transfusión de Linfocitos , Linfocitos T Citotóxicos/inmunología , Trasplante Heterólogo/inmunología , Animales , Caspasas/metabolismo , Línea Celular , Citotoxicidad Inmunológica , Fragmentación del ADN , Endotelio Vascular , Vectores Genéticos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos
5.
Transplant Proc ; 40(2): 559-63, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18374129

RESUMEN

Although the use of organs from alpha1,3-galactosyltransferase gene knockout pigs may prolong xenograft survival, resulting in overcoming antibody-mediated hyperacute rejection, pig xenografts will be destroyed directly by cell-mediated immunity, such as NK cells, macrophages, and CD8+ cytotoxic T lymphocytes (CTLs). Therefore, conquering cell-mediated immunity, especially of human CD8+ CTLs, is of particular importance to the success of long-term xenograft survival. We have previously reported that the cytotoxicity of human CD8+ CTLs is strong against pig endothelial cells (PEC) and mediated in major part by the Fas/FasL apoptotic pathway. Cellular FLICE inhibitory protein (c-FLIP) was originally identified as a potent inhibitor of death-receptor signaling through binding competition with caspase-8 for recruitment to Fas-associated via death domain (FADD). Two major c-FLIP variants result from alternative mRNA splicing: a short, 26-kDa protein (c-FLIP S) and a long, 55-kDa form (c-FLIP L). The present study demonstrated that overexpression of c-FLIP S/L genes in PEC markedly suppressed human CD8+ CTL-mediated xenocytotoxicity; moreover, the cytoprotective effects of c-FLIP L appeared to be significantly stronger than those of c-FLIP S. Furthermore, to prove the in vivo prolongation effects of xenograft survival, we transplanted PEC transfectants with c-FLIP(S/L) genes under the rat kidney capsule. Prolonged survival was displayed by xenografts of FLIP S/L PEC transfectants, whereas xenografts of parental PEC were completely rejected by day 5 posttransplantation. Thus, intracellular blocking of death receptor-mediated apoptotic signals by overexpression of c-FLIP S/L in xenograft cells may prevent innate cellular attacks against xenografts opening the window of opportunity for long-term xenograft survival.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T Citotóxicos/inmunología , Trasplante Heterólogo/inmunología , Animales , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Línea Celular , Supervivencia Celular/inmunología , Fragmentación del ADN , Endotelio Vascular , Expresión Génica , Humanos , Plásmidos , Porcinos
6.
Transplant Proc ; 40(2): 477-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18374107

RESUMEN

The success of pancreatic islet transplantation is limited because of the severe shortage of allogeneic pancreas donors. Accordingly, pig islets are considered to be an attractive, promising alternative. However, cell-mediated immunity, especially CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity, remains a formidable barrier to prevent long-term islet survival in xenograft recipients. Therefore, it is particularly important to explore methods to specifically prevent cell-mediated immunity against pig islets. Our group previously demonstrated that the overexpression of either membrane-bound human FasL or human decoy Fas antigen in pig endothelial cells prevented CTL xenocytotoxicity. In this study, we assessed the cytoprotective effects of adenoviral-mediated overexpression of either membrane-bound human FasL or human decoy Fas antigen in pig islets to inhibit CTL xenocytotoxicity. The CTL-mediated killing of pig islets infected with an adenoviral vector carrying either membrane-bound human FasL or human decoy Fas was significantly reduced compares with that of control pig islets transfected with adenoviral vector encoding enhanced green fluorescent protein (EGFP). Moreover, we transfected pig islets with these molecules to confirm their cytoprotective effects in in vivo studies. The significant long-term survival of pig islets expressing these molecules was elicited through days 3 to 5 posttransplantation. Thus, these results demonstrated that the remodeling of either death receptor or death ligand on pig islets by adenoviral gene transfer prevented innate cellular immunity against xeno-islet grafts facilitating long-term xenograft survival.


Asunto(s)
Adenoviridae/genética , Proteína Ligando Fas/genética , Supervivencia de Injerto/fisiología , Trasplante de Islotes Pancreáticos/fisiología , Trasplante Heterólogo/fisiología , Animales , Línea Celular , Clonación Molecular , Endotelio Vascular , Regulación de la Expresión Génica , Vectores Genéticos , Humanos , Ratas , Porcinos , Receptor fas/genética
7.
Transplant Proc ; 38(10): 3286-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17175251

RESUMEN

Pig islets are considered to be most suitable source of islets for xenotransplantation into patients with type 1 diabetes mellitus. However, cellular rejection, especially CD8+ CTL-mediated cytotoxicity, remains a formidable barrier preventing long-term xenograft survival. Our previous study demonstrated that human CD8+ CTLs were highly detrimental to xenograft cells and that this strong cytotoxicity of human CTLs was mediated mainly by the Fas/FasL apoptotic pathway. Furthermore, we exploited novel methods for inhibiting human CD8+ CTL-mediated xenocytotoxicity with overexpression of membrane-bound human FasL and human decoy Fas antigen in xenografted cells. In the present study, we assessed the cytoprotective effects of these novel inhibitory molecules overexpressed by an adenoviral-mediated system in pig islets. Isolated pig islets were transfected with adenovirus vector encoding either human decoy Fas or membrane-bound human FasL genes. Thirty percent to 60% of transfected pig islets expressed these molecules producing 60% to 88% suppression of CTL killing compared with parental pig islets. These data indicated that pig islet grafts isolated from transgenic pigs with either membrane-bound human FasL or human decoy Fas antigen genes may control the innate cellular response to xenografts, and creating a window of opportunity to facilitate xenograft survival.


Asunto(s)
Adenoviridae/genética , Citotoxicidad Inmunológica , Proteína Ligando Fas/genética , Islotes Pancreáticos/inmunología , Transfusión de Linfocitos , Linfocitos T Citotóxicos/trasplante , Receptor fas/genética , Animales , Humanos , Porcinos , Transfección , Trasplante Heterólogo
8.
Transplant Proc ; 38(10): 3315-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17175259

RESUMEN

Human CD8+ cytotoxic T lymphocyte (CTL)-mediated cytotoxicity in xenograft recipients is an important obstacle for successful xenotransplantation of pig organs into humans. In our previous study, we demonstrated that xenocytotoxicity of human CD8+ CTL detrimental to pig endothelial cells (PEC) is mediated mainly by the Fas/FasL apoptotic pathway. Furthermore, we developed new methods to prevent this CTL killing by extracellular remodeling using overexpression of human decoy Fas antigen and membrane-bound human FasL on pig xenograft cells. The cellular FLICE-inhibitory protein (c-FLIP), a caspase-8 inhibitor that lacks the cysteine domain, is a negative regulator of death receptor-mediated apoptosis. c-FLIP proteins exist as long (c-FLIP(L)) and short (c-FLIPs) splice variants, both capable of protecting cells from death receptor-mediated apoptosis. In this report, we have demonstrated that both pig c-FLIPs and pig c-FLIP(L) significantly inhibit human CD8+ CTL-mediated xenocytotoxicity toward stably transfected PEC, although the expression level of pig Fas antigen on cell surface was not changed. These data suggested that intracellular remodeling with overexpression of pig c-FLIP in xenograft cells may decrease the innate cellular responses against xenografts, facilitating long-term xenograft survival.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Citotoxicidad Inmunológica , Endotelio Vascular/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Línea Celular , Cartilla de ADN , ADN Complementario , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/inmunología , Supervivencia de Injerto , Humanos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Transfección , Trasplante Heterólogo/inmunología
9.
Mutat Res ; 414(1-3): 9-14, 1998 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-9630478

RESUMEN

The in vivo genotoxicity of five heterocyclic amines-Trp-P-2 (13 mg/kg), IQ (13 mg/kg), MeIQ (13 mg/kg), MeIQx (13 mg/kg), and PhIP (40 mg/kg)-in the mucosa of gastrointestinal and urinary tract organs (stomach, duodenum, jejunum, ileum, colon, and bladder) was studied by the alkaline single cell gel electrophoresis (SCG) (Comet) assay. Male CD-1 mice were sacrificed 1, 3, and 8 h after intraperitoneal injection. All the heterocyclic amines studied yielded statistically significant DNA damage in the colon but not the small intestine (duodenum, jejunum, and ileum) or urinary bladder. In this study, five heterocyclic amines were injected intraperitoneally to avoid the consequences of ingestion. Thus, the extensive damage to colon DNA was concluded to be due, at least in part, to a systemic effect.


Asunto(s)
Aminas/toxicidad , Colon/ultraestructura , Mucosa Gástrica/ultraestructura , Mucosa Intestinal/ultraestructura , Pruebas de Mutagenicidad/métodos , Mutágenos/toxicidad , Animales , Carbolinas/toxicidad , Electroforesis en Gel de Agar , Imidazoles/toxicidad , Masculino , Ratones , Quinolinas/toxicidad , Vejiga Urinaria/ultraestructura
10.
Mutat Res ; 395(1): 57-73, 1997 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-9465914

RESUMEN

We used a modification of the alkaline single cell gel electrophoresis (SCG) (Comet) assay to test the in vivo genotoxicity of 6 heterocyclic amines, Trp-P-1 (25 mg/kg), Trp-P-2 (13 mg/kg), IQ (13 mg/kg), MeIQ (13 mg/kg), MeIQx (13 mg/kg) and PhIP (40 mg/kg), in mouse liver, lung, kidney, brain, spleen, bone marrow and stomach mucosa. Mice were sacrificed 1, 3, and 24 h after intraperitoneal injection. Trp-P-2, IQ, MeIQ, and MeIQx yielded statistically significant DNA damage in the stomach, liver, kidney, lung and brain; Trp-P-1 in the stomach, liver and lung; and PhIP in the liver, kidney and brain. None of the heterocyclic amines induced DNA damage in the spleen and bone marrow. Our results suggest that the alkaline SCG assay applied to multiple organs is a good way to detect organ-specific genotoxicity of heterocyclic amines in mammals.


Asunto(s)
Aminas/toxicidad , Daño del ADN/efectos de los fármacos , Electroforesis/métodos , Mutágenos/toxicidad , Animales , Encéfalo/efectos de los fármacos , Carbolinas/toxicidad , Imidazoles/toxicidad , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Quinolinas/toxicidad , Quinoxalinas/toxicidad , Estómago/efectos de los fármacos
11.
Mutat Res ; 395(2-3): 189-98, 1997 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-9465930

RESUMEN

In Japan, ortho-phenylphenol (OPP), biphenyl (BP), and thiabendazole (2-(4'-thiazolyl)benzimidazole, TBZ) are commonly used as a postharvest treatment to preserve imported citrus fruits during transport and storage. We used a modification of the alkaline single cell gel electrophoresis (SCG) (Comet) assay to test the in vivo genotoxicity of those agents in mouse stomach, liver, kidney, bladder, lung, brain, and bone marrow. CD-1 male mice were sacrificed 3, 8, and 24 h after oral administration of the test compounds. OPP (2000 mg/kg) induced DNA damage in the stomach, liver, kidney, bladder, and lung, BP (2000 mg/kg) and TBZ (200 mg/kg) induced DNA damage in all the organs studied. For OPP, increased DNA damage peaked at 3-8 h and tended to decrease at 24 h. For BP, on the contrary, increased DNA migration peaked at 24 h. That delay may have been due to the fact that OPP is metabolized by cytochrome 450 and prostaglandin H synthase to phenylbenzoquinone (PBQ), a DNA binding metabolite, and BP is metabolized to PBQ via OPP and m-phenylphenol. The positive response to TBZ, an aneugen, supports the in vivo DNA-damaging action of TBZ.


Asunto(s)
Compuestos de Bifenilo/toxicidad , Electroforesis/métodos , Pruebas de Mutagenicidad/métodos , Tiabendazol/toxicidad , Animales , Médula Ósea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Daño del ADN , Fungicidas Industriales/toxicidad , Hígado/efectos de los fármacos , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos , Estómago/efectos de los fármacos , Factores de Tiempo , Distribución Tisular
12.
Mutat Res ; 419(1-3): 13-20, 1998 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-9804871

RESUMEN

The micronucleus test is widely used to assess in vivo clastogenicity because of its convenience, but it is not appropriate for some carcinogenic chemical classes. Halogenated compounds, for example, are inconsistent micronucleus inducers. We assessed the genotoxicity of 7 haloalkanes and haloalkenes carcinogenic to rodents in 7 mouse organs-stomach, liver, kidney, bladder, lung, brain, and bone marrow-using the alkaline single cell gel electrophoresis (SCG) assay. The carcinogens we studied were 1, 2-dibromo-3-chloropropane (DBCP), 1,3-dichloropropene (mixture of cis and trans) (DCP), 1,2-dibromoethane (EDB), 1,2-dichloroethane (EDC), vinyl bromide, dichloromethane, and carbon tetrachloride; only DBCP induces micronuclei in mouse bone marrow. Except for carbon tetrachloride, halocompounds studied are mutagenic to Salmonella typhimurium. Mice were sacrificed 3 or 24 h after carcinogen administration. DCP and EDC induced DNA damage in all of the organs studied. Vinyl bromide yielded DNA damage in all of the organs except for bone marrow. DBCP induced DNA damage in the stomach, liver, kidney, lung, and bone marrow; EDB in the stomach, liver, kidney, bladder, and lung; and dichloromethane in the liver and lung. Since no deaths, morbidity, clinical signs, organ pathology, or microscopic signs of necrosis were observed, the DNA damage was not attributable to cytotoxicity. On the other hand, the positive response in the liver induced by carbon tetrachloride, which was accompanied by necrosis, was considered to be a false positive response. We suggest that the alkaline SCG assay can be used in multiple organs to detect in vivo genotoxicity that is not expressed in bone marrow cells in mice given non-necrogenic doses of halocompounds.


Asunto(s)
Alcanos/toxicidad , Alquenos/toxicidad , Carcinógenos/toxicidad , Mutágenos/toxicidad , Compuestos Alílicos/toxicidad , Animales , Tetracloruro de Carbono/toxicidad , Electroforesis/métodos , Dicloruros de Etileno/toxicidad , Halógenos/toxicidad , Hidrocarburos Bromados/toxicidad , Hidrocarburos Clorados , Masculino , Ratones , Pruebas de Micronúcleos , Propano/análogos & derivados , Propano/toxicidad , Compuestos de Vinilo/toxicidad
13.
Mutat Res ; 415(1-2): 1-12, 1998 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-9711257

RESUMEN

We used a modification of the alkaline single cell gel electrophoresis (SCG) (Comet) assay to test the in vivo genotoxicity of four hydrazine derivatives--1,2-dimethylhydrazine (SDMH), 1,1-dimethylhydrazine (UDMH), hydrazine (HZ), and procarbazine (PCZ)--in mouse liver, lung, kidney, brain, and bone marrow, and in the mucosa of stomach, colon, and bladder. Mice were sacrificed 3 and 24 h after intra-peritoneal (i.p.) and oral (p.o.) administration. SDMH at 20 mg/kg i.p. yielded statistically significant DNA damage in all tested organs except for lung. In the gastrointestinal tract, SDMH was genotoxic in the stomach and the colon after i.p. treatment but only in the colon after 20 and 30 mg/kg p.o. treatment. UDMH at 50 mg/kg i.p. yielded DNA damage in the liver and lung at 3 h. PCZ at 200 mg/kg i.p. caused DNA damage in the liver, kidney, lung, brain, and bone marrow. UDMH and PCZ were positive in the stomach and colon p.o. but not by i.p. treatment. HZ at 100 mg/kg yielded DNA damage in the stomach, liver, and lung when given i.p. and in the brain when p.o. Thus, the administration route is important when evaluating organ-specific genotoxicity in multiple organs.


Asunto(s)
1,2-Dimetilhidrazina/toxicidad , Carcinógenos/toxicidad , Colon/efectos de los fármacos , Dimetilhidrazinas/toxicidad , Hidrazinas/toxicidad , Procarbazina/toxicidad , 1,2-Dimetilhidrazina/administración & dosificación , Administración Oral , Animales , Carcinógenos/administración & dosificación , Daño del ADN , Dimetilhidrazinas/administración & dosificación , Hidrazinas/administración & dosificación , Inyecciones Intraperitoneales , Masculino , Ratones , Pruebas de Mutagenicidad , Procarbazina/administración & dosificación
14.
Rinsho Byori ; 19: Suppl:284, 1971 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-5167975
16.
Gastroenterol Jpn ; 26 Suppl 3: 234-8, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1909272

RESUMEN

The efficacy of interferon therapy (IFN) was investigated in 46 patients with chronic non-A, non-B (NANB) hepatitis, of would 40 (87.0%) were positive for anti-HCV antibody (Ab) (C-100-3). Three kinds of IFN were used; human lymphoblastoid interferon (HLBI), interferon alpha-2b and interferon beta. Total doses of IFN ranged from 1 million units (MU) to 10 MU and treatment duration ranged from 2 weeks to 144 weeks. Of 46 patients 34 (73.9%) responded to IFN. Nine patients have maintained normal ALT levels and 5 patients have maintained near-normal ALT levels for more than 6 months after cessation of IFN treatment. In these cases the titers of anti-HCV Ab had decreased significantly at the end of IFN therapy and 6 months after IFN therapy respectively. The mean age was young and the mean disease duration was short in effective cases. As for doses and treatment duration of IFN, low doses of IFN requires long treatment duration to acquire continuous efficacy and high doses of IFN requires rather short treatment durations. Therefore, early IFN treatment, higher doses and longer periods of IFN treatment may improve the response rate of patients with chronic NANB hepatitis.


Asunto(s)
Alanina Transaminasa/sangre , Anticuerpos Antihepatitis/sangre , Hepatitis C/tratamiento farmacológico , Interferón Tipo I/uso terapéutico , Hígado/patología , Adulto , Femenino , Hepatitis C/enzimología , Hepatitis C/inmunología , Hepatitis C/patología , Humanos , Masculino , Persona de Mediana Edad
17.
Gastroenterol Jpn ; 24(2): 164-9, 1989 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2744332

RESUMEN

We evaluated the usefulness of serum 2'-5'oligoadenylate synthetase (2-5AS) activity assay in monitoring the anti-viral activity of chronic type B hepatitis patients during IFN therapy. The serum 2-5AS activity was rapidly increased during the above therapy and was maintained at a medium-to-high level throughout the therapy period, although the capacity for increase reflected differences among individuals. The kinetics of serum 2-5AS activity during the therapy was almost consistent with that of the PBMCs 2-5AS activity. 2-5AS activity had an inverse correlation with DNA-P; i.e. DNA-P often disappeared from serum after interferon treatment in patients with a marked response in serum 2-5AS activity. The enhancement of serum 2-5AS activity during IFN therapy seemed to correlate with an increase in anti-viral activity. The results suggest that the serum 2-5AS activity assay is a useful probe for monitoring the anti-viral activity of chronic type B hepatitis patients during interferon therapy.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/sangre , Hepatitis B/terapia , Hepatitis Crónica/terapia , Interferón Tipo I/uso terapéutico , Humanos , Proteínas Recombinantes
18.
Crit Rev Toxicol ; 30(6): 629-799, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11145306

RESUMEN

The comet assay is a microgel electrophoresis technique for detecting DNA damage at the level of the single cell. When this technique is applied to detect genotoxicity in experimental animals, the most important advantage is that DNA lesions can be measured in any organ, regardless of the extent of mitotic activity. The purpose of this article is to summarize the in vivo genotoxicity in eight organs of the mouse of 208 chemicals selected from International Agency for Research on Cancer (IARC) Groups 1, 2A, 2B, 3, and 4, and from the U.S. National Toxicology Program (NTP) Carcinogenicity Database, and to discuss the utility of the comet assay in genetic toxicology. Alkylating agents, amides, aromatic amines, azo compounds, cyclic nitro compounds, hydrazines, halides having reactive halogens, and polycyclic aromatic hydrocarbons were chemicals showing high positive effects in this assay. The responses detected reflected the ability of this assay to detect the fragmentation of DNA molecules produced by DNA single strand breaks induced chemically and those derived from alkali-labile sites developed from alkylated bases and bulky base adducts. The mouse or rat organs exhibiting increased levels of DNA damage were not necessarily the target organs for carcinogenicity. It was rare, in contrast, for the target organs not to show DNA damage. Therefore, organ-specific genotoxicity was necessary but not sufficient for the prediction of organ-specific carcinogenicity. It would be expected that DNA crosslinkers would be difficult to detect by this assay, because of the resulting inhibition of DNA unwinding. The proportion of 10 DNA crosslinkers that was positive, however, was high in the gastrointestinal mucosa, stomach, and colon, but less than 50% in the liver and lung. It was interesting that the genotoxicity of DNA crosslinkers could be detected in the gastrointestinal organs even though the agents were administered intraperitoneally. Chemical carcinogens can be classified as genotoxic (Ames test-positive) and putative nongenotoxic (Ames test-negative) carcinogens. The Ames test is generally used as a first screening method to assess chemical genotoxicity and has provided extensive information on DNA reactivity. Out of 208 chemicals studied, 117 are Ames test-positive rodent carcinogens, 43 are Ames test-negative rodent carcinogens, and 30 are rodent noncarcinogens (which include both Ames test-positive and negative noncarcinogens). High positive response ratio (110/117) for rodent genotoxic carcinogens and a high negative response ratio (6/30) for rodent noncarcinogens were shown in the comet assay. For Ames test-negative rodent carcinogens, less than 50% were positive in the comet assay, suggesting that the assay, which detects DNA lesions, is not suitable for identifying nongenotoxic carcinogens. In the safety evaluation of chemicals, it is important to demonstrate that Ames test-positive agents are not genotoxic in vivo. This assay had a high positive response ratio for rodent genotoxic carcinogens and a high negative response ratio for rodent genotoxic noncarcinogens, suggesting that the comet assay can be used to evaluate the in vivo genotoxicity of in vitro genotoxic chemicals. For chemicals whose in vivo genotoxicity has been tested in multiple organs by the comet assay, published data are summarized with unpublished data and compared with relevant genotoxicity and carcinogenicity data. Because it is clear that no single test is capable of detecting all relevant genotoxic agents, the usual approach should be to carry out a battery of in vitro and in vivo tests for genotoxicity. The conventional micronucleus test in the hematopoietic system is a simple method to assess in vivo clastogenicity of chemicals. Its performance is related to whether a chemical reaches the hematopoietic system. Among 208 chemicals studied (including 165 rodent carcinogens), 54 rodents carcinogens do not induce micronuclei in mouse hematopoietic system despite the positive finding with one or two in vitro tests. Forty-nine of 54 rodent carcinogens that do not induce micronuclei were positive in the comet assay, suggesting that the comet assay can be used as a further in vivo test apart from the cytogenetic assays in hematopoietic cells. In this review, we provide one recommendation for the in vivo comet assay protocol based on our own data.


Asunto(s)
Carcinógenos/efectos adversos , Ensayo Cometa , Daño del ADN , Animales , Ensayo Cometa/métodos , Ensayo Cometa/normas , Bases de Datos Factuales , Femenino , Masculino , Ratones , Proyectos de Investigación , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA