Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Transplant ; 10(2): 220-30, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19788504

RESUMEN

Cardiac transplantation is an effective treatment for multiple types of heart failure refractive to therapy. Although immunosuppressive therapeutics have increased survival rates within the first year posttransplant, chronic rejection (CR) remains a significant barrier to long-term graft survival. Indicators of CR include patchy interstitial fibrosis, vascular occlusion and progressive loss of graft function. Multiple factors have been implicated in the onset and progression of CR, including TGFbeta, IL-6 and connective tissue growth factor (CTGF). While associated with CR, the role of CTGF in CR and the factors necessary for CTGF induction in vivo are not understood. To this end, we utilized forced expression and neutralizing antibody approaches. Transduction of allografts with CTGF significantly increased fibrotic tissue development, though not to levels observed with TGFbeta transduction. Further, intragraft CTGF expression was inhibited by IL-6 neutralization whereas TGFbeta expression remained unchanged, indicating that IL-6 effects may potentiate TGFbeta-mediated induction of CTGF. Finally, neutralizing CTGF significantly reduced graft fibrosis without reducing TGFbeta and IL-6 expression levels. These findings indicate that CTGF functions as a downstream mediator of fibrosis in CR, and that CTGF neutralization may ameliorate fibrosis and hypertrophy associated with CR.


Asunto(s)
Trasplante de Corazón/inmunología , Interleucina-6/metabolismo , Factor de Crecimiento Transformador beta/inmunología , Animales , Anticuerpos Neutralizantes , Factor de Crecimiento del Tejido Conjuntivo , Progresión de la Enfermedad , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Supervivencia de Injerto , Corazón , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/metabolismo
2.
Am J Transplant ; 9(8): 1773-83, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19538487

RESUMEN

Chronic cardiac allograft rejection is the major barrier to long term graft survival. There is currently no effective treatment for chronic rejection except re-transplantation. Though neointimal development, fibrosis, and progressive deterioration of graft function are hallmarks of chronic rejection, the immunologic mechanisms driving this process are poorly understood. These experiments tested a functional role for IL-6 in chronic rejection by utilizing serial echocardiography to assess the progression of chronic rejection in vascularized mouse cardiac allografts. Cardiac allografts in mice transiently depleted of CD4+ cells that develop chronic rejection were compared with those receiving anti-CD40L therapy that do not develop chronic rejection. Echocardiography revealed the development of hypertrophy in grafts undergoing chronic rejection. Histologic analysis confirmed hypertrophy that coincided with graft fibrosis and elevated intragraft expression of IL-6. To elucidate the role of IL-6 in chronic rejection, cardiac allograft recipients depleted of CD4+ cells were treated with neutralizing anti-IL-6 mAb. IL-6 neutralization ameliorated cardiomyocyte hypertrophy, graft fibrosis, and prevented deterioration of graft contractility associated with chronic rejection. These observations reveal a new paradigm in which IL-6 drives development of pathologic hypertrophy and fibrosis in chronic cardiac allograft rejection and suggest that IL-6 could be a therapeutic target to prevent this disease.


Asunto(s)
Cardiomegalia/metabolismo , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Trasplante de Corazón/patología , Interleucina-6/metabolismo , Miocardio/metabolismo , Miocardio/patología , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD4-Positivos/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía , Femenino , Fibrosis , Rechazo de Injerto/diagnóstico por imagen , Corazón/efectos de los fármacos , Interleucina-6/antagonistas & inhibidores , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Homólogo
3.
Am J Transplant ; 8(10): 2037-48, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18828767

RESUMEN

Inductive therapy with anti-CD4 or anti-CD40L monoclonal antibodies (mAb) leads to long-term allograft acceptance but the immune parameters responsible for graft maintenance are not well understood. This study employed an adoptive transfer system in which cells from mice bearing long-term cardiac allografts following inductive anti-CD4 or anti-CD40L therapy were transferred into severe combined immunodeficiency (SCID) allograft recipients. SCID recipients of cells from anti-CD4-treated mice (anti-CD4 cells) did not reject allografts while those receiving cells from anti-CD40L-treated mice (anti-CD40L cells) did reject allografts. Carboxyfluorescein succinimidyl ester (CFSE) labeling of transferred cells revealed that this difference was not associated with differential proliferative capacities of these cells in SCID recipients. Like cells from naïve mice, anti-CD40L cells mounted a Th1 response following transfer while anti-CD4 cells mounted a dominant Th2 response. Early (day 10) T-cell priming was detectable in both groups of primary allograft recipients but persisted to day 30 only in recipients treated with anti-CD4 mAb. Thus, anti-CD40L therapy appears to result in graft-reactive T cells with a naïve phenotype while anti-CD4 therapy allows progression to an altered state of differentiation. Additional data herein support the notion that anti-CD40L mAb targets activated, but not memory, cells for removal or functional silencing.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/metabolismo , Linfocitos T/inmunología , Trasplante/métodos , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Fluoresceínas/farmacología , Memoria Inmunológica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Succinimidas/farmacología , Células Th2/metabolismo
4.
Am J Transplant ; 8(8): 1622-30, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18557731

RESUMEN

Though complement (C) deposition within the transplant is associated with allograft rejection, the pathways employed have not been established. In addition, evidence suggests that C-mediated cytolysis may be necessary for the tolerance-inducing activities of mAb therapies. Hence, we assessed the role of the classical C pathway in acute allograft rejection and its requirement for experimental mAb therapies. C1q-deficient (C1q-/-) recipients rejected allografts at a faster rate than wild-type (WT) recipients. This rejection was associated with exacerbated graft pathology but not with enhanced T-cell responses in C1q-/- recipients. However, the humoral response to donor alloantigens was accelerated in C1q-/- mice, as an early IgG response and IgG deposition within the graft were observed. Furthermore, deposition of C3d, but not C4d was observed in grafts isolated from C1q-/- recipients. To assess the role of the classical C pathway in inductive mAb therapies, C1q-/- recipients were treated with anti-CD4 or anti-CD40L mAb. The protective effects of anti-CD4 mAb were reduced in C1q-/- recipients, however, this effect did not correlate with ineffective depletion of CD4+ cells. In contrast, the protective effects of anti-CD40L mAb were less compromised in C1q-/- recipients. Hence, this study reveals unanticipated roles for C1q in the rejection process.


Asunto(s)
Complemento C1q/metabolismo , Vía Clásica del Complemento , Rechazo de Injerto/prevención & control , Inmunoglobulina G/metabolismo , Tolerancia al Trasplante/fisiología , Animales , Anticuerpos Monoclonales , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad
5.
Am J Transplant ; 6(5 Pt 1): 959-66, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16611331

RESUMEN

Late loss of allograft function is primarily attributed to chronic rejection (CR). There are no effective treatments for CR and the underlying cause of the disease is unknown. This study compared events that occurred within cardiac allografts placed in mice that received either anti-CD4 therapy and develop CR or anti-CD40L therapy and do not develop CR. Both TGFbeta and connective tissue growth factor (CTGF), which is induced by TGFbeta, were expressed in grafts with CR but were not expressed in grafts without CR. TGFbeta transfection of allografts in anti-CD40L-treated recipients resulted in CTGF expression and CR. However, TGFbeta transfection of syngeneic grafts did not result in CTGF expression or CR. These data indicate that TGFbeta alone is insufficient to induce CR and that CTGF is required. Further, antigenic stimulation is required for TGFbeta induction of CTGF. Thus, CTGF may serve as a therapeutic target for CR.


Asunto(s)
Rechazo de Injerto/patología , Trasplante de Corazón/inmunología , Proteínas Inmediatas-Precoces/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Ligando de CD40/inmunología , Factor de Crecimiento del Tejido Conjuntivo , Femenino , Trasplante de Corazón/patología , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/inmunología , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transfección , Factor de Crecimiento Transformador beta/inmunología , Trasplante Homólogo , Trasplante Isogénico
6.
Breast Cancer Res ; 3(5): 294-8, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11597317

RESUMEN

Two recent papers provide new evidence relevant to the role of the breast cancer susceptibility gene BRCA2 in DNA repair. Moynahan et al provide genetic data indicating a requirement for BRCA2 in homology-dependent (recombinational) repair of DNA double-strand breaks. The second paper, by Davies et al, begins to address the mechanism through which BRCA2 makes its contribution to recombinational repair. BRCA2 appears to function in recombination via interactions with the major eukaryotic recombinase RAD51 [1,2,3]. We briefly review the context in which the two studies were carried out, we comment on the results presented, and we discuss models designed to account for the role of BRCA2 in RAD51-mediated repair.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Neoplasias/genética , Factores de Transcripción/genética , Proteína BRCA2 , Reparación del ADN , Proteínas de Unión al ADN/genética , Femenino , Humanos , Recombinasa Rad51 , Recombinación Genética
7.
J Biol Chem ; 276(45): 41906-12, 2001 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-11551925

RESUMEN

Dmc1 and Rad51 are eukaryotic RecA homologues that are involved in meiotic recombination. The expression of Dmc1 is limited to meiosis, whereas Rad51 is expressed in mitosis and meiosis. Dmc1 and Rad51 have unique and overlapping functions during meiotic recombination. Here we report the purification of the Dmc1 protein from the budding yeast Saccharomyces cerevisiae and present basic characterization of its biochemical activity. The protein has a weak DNA-dependent ATPase activity and binds both single-strand DNA (ssDNA) and double-strand DNA. Electrophoretic mobility shift assays suggest that DNA binding by Dmc1 is cooperative. Dmc1 renatures linearized plasmid DNA with first order reaction kinetics and without requiring added nucleotide cofactor. In addition, Dmc1 catalyzes strand assimilation of ssDNA oligonucleotides into homologous supercoiled duplex DNA in a reaction promoted by ATP or the non-hydrolyzable ATP analogue AMP-PNP.


Asunto(s)
Proteínas de Ciclo Celular , ADN de Cadena Simple/química , ADN Superhelicoidal/química , Proteínas de Unión al ADN/farmacología , Renaturación de Ácido Nucleico , Saccharomyces cerevisiae/química , Adenosina Trifosfatasas/metabolismo , Catálisis , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae
8.
Proc Natl Acad Sci U S A ; 98(15): 8411-8, 2001 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-11459983

RESUMEN

Members of the RecA family of recombinases from bacteriophage T4, Escherichia coli, yeast, and higher eukaryotes function in recombination as higher-order oligomers assembled on tracts of single-strand DNA (ssDNA). Biochemical studies have shown that assembly of recombinase involves accessory factors. These studies have identified a class of proteins, called recombination mediator proteins, that act by promoting assembly of recombinase on ssDNA tracts that are bound by ssDNA-binding protein (ssb). In the absence of mediators, ssb inhibits recombination reactions by competing with recombinase for DNA-binding sites. Here we briefly review mediated recombinase assembly and present results of new in vivo experiments. Immuno-double-staining experiments in Saccharomyces cerevisiae suggest that Rad51, the eukaryotic recombinase, can assemble at or near sites containing ssb (replication protein A, RPA) during the response to DNA damage, consistent with a need for mediator activity. Correspondingly, mediator gene mutants display defects in Rad51 assembly after DNA damage and during meiosis, although the requirements for assembly are distinct in the two cases. In meiosis, both Rad52 and Rad55/57 are required, whereas either Rad52 or Rad55/57 is sufficient to promote assembly of Rad51 in irradiated mitotic cells. Rad52 promotes normal amounts of Rad51 assembly in the absence of Rad55 at 30 degrees C but not 20 degrees C, accounting for the cold sensitivity of rad55 null mutants. Finally, we show that assembly of Rad51 is induced by radiation during S phase but not during G(1), consistent with the role of Rad51 in repairing the spontaneous damage that occurs during DNA replication.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Meiosis/fisiología , Mitosis/fisiología , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Adenosina Trifosfatasas , Animales , Daño del ADN , Enzimas Reparadoras del ADN , ADN de Cadena Simple/metabolismo , Células Eucariotas , Exodesoxirribonucleasa V , Exodesoxirribonucleasas/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Recombinasa Rad51 , Rec A Recombinasas/metabolismo , Proteína de Replicación A
9.
Oncogene ; 20(32): 4433-8, 2001 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-11466627

RESUMEN

The breast cancer susceptibility gene BRCA1 encodes a multifunctional protein that is mutated in many hereditary breast and ovarian cancers. We have cloned a homologue of the human BRCA1 gene from chicken; the gene encodes a 1749 amino acid protein that is 33% identical to human BRCA1. Phylogenetic analyses of the chicken and mammalian proteins indicate that the chicken gene is a bona fide BRCA1 orthologue, the first to be described from a non-mammal. Most of the chicken protein has diverged considerably from its mammalian orthologues, although the RING and BRCT repeat regions are highly conserved. This marked overall sequence divergence has allowed us to identify nine additional highly-conserved motifs (ranging from 8--56 amino acids in length) which are likely important for BRCA1 function.


Asunto(s)
Proteína BRCA1/química , Proteína BRCA1/genética , Pollos/genética , Genes BRCA1 , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas Aviares , Embrión de Pollo , Clonación Molecular , Secuencia Conservada , Humanos , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido
10.
J Immunol ; 166(5): 3248-55, 2001 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-11207279

RESUMEN

Both wild-type (WT) and IFN-gamma-deficient (IFN-gamma(-/-)) C57BL/6 mice can rapidly reject BALB/c cardiac allografts. When depleted of CD8(+) cells, both WT and IFN-gamma(-/-) mice rejected their allografts, indicating that these mice share a common CD4-mediated, CD8-independent mechanism of rejection. However, when depleted of CD4(+) cells, WT mice accepted their allografts, while IFN-gamma(-/-) recipients rapidly rejected them. Hence, IFN-gamma(-/-), but not WT mice developed an unusual CD8-mediated, CD4-independent, mechanism of allograft rejection. Allograft rejection in IFN-gamma(-/-) mice was associated with intragraft accumulation of IL-4-producing cells, polymorphonuclear leukocytes, and eosinophils. Furthermore, this form of rejection was resistant to treatment with anti-CD40 ligand (CD40L) mAb, which markedly prolonged graft survival in WT mice. T cell depletion studies verified that anti-CD40L treatment failed to prevent CD8-mediated allograft rejection in IFN-gamma(-/-) mice. However, anti-CD40L treatment did prevent CD4-mediated rejection in IFN-gamma(-/-) mice, although grafts were eventually rejected when CD8(+) T cells repopulated the periphery. The IL-4 production and eosinophil influx into the graft that occurred during CD8-mediated rejection were apparently epiphenomenal, since treatment with anti-IL-4 mAb blocked intragraft accumulation of eosinophils, but did not interfere with allograft rejection. These studies demonstrate that a novel, CD8-mediated mechanism of allograft rejection, which is resistant to experimental immunosuppression, can develop when IFN-gamma is limiting. An understanding of this mechanism is confounded by its association with Th2-like immune events, which contribute unique histopathologic features to the graft but are apparently unnecessary for the process of allograft rejection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/metabolismo , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Interferón gamma/deficiencia , Interferón gamma/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Femenino , Rechazo de Injerto/genética , Rechazo de Injerto/patología , Trasplante de Corazón/inmunología , Trasplante de Corazón/patología , Interleucina-4/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Inmunología del Trasplante/genética , Trasplante Homólogo
11.
Transplantation ; 70(9): 1292-301, 2000 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11087143

RESUMEN

We have developed a model of transforming growth factor (TGF)beta1 gene transfer into mouse vascularized cardiac allografts to study the use of gene transfer as an immunosuppressive therapy in transplantation. Donor hearts were perfused with either DNA-liposome complexes or adenoviral vectors that encode the active form of human TGFbeta1. DNA-liposome mediated transfection prolonged allograft survival in approximately two-thirds of transplant recipients, while adenoviral delivery of TGFbeta1 was not protective. Protective TGFbeta1 gene transfer was associated with reduced Th1 responses and an inhibition of the alloantibody isotype switch. The protective effects of TGFbeta1 gene transfer were overridden by exogenous interleukin-12 administration. Interestingly, alloreactive CD4+ and CD8+ cells exhibited distinct sensitivities to TGFbeta1 gene transfer: CD4+ Th1 function was abrogated by this modality, although CD8+ Th1 function was not. Transient depletion of recipient CD8+ cells markedly prolonged the survival of grafts transfected with either DNA-liposome complexes or adenoviral vectors. Transgene expression persisted for at least 60 days, and Th1 responses were not detectable until CD8+ T cells repopulated the periphery. However, long-term transfected allografts appeared to exhibit exacerbated fibrosis and neointimal development. These manifestations of chronic rejection were absent in long-term transfected isografts, suggesting that long-term expression of active TGFbeta1 alone is not sufficient to induce fibrosis of the grafts. Collectively, these data illustrate the utility of immunosuppressive gene therapy as a treatment for transplantation when combined with additional conditioning regimens. Further, they illustrate that alloreactive CD4+ and CD8+ cells may be differentially influenced by cytokine manipulation strategies.


Asunto(s)
ADN/fisiología , Factor de Crecimiento Transformador beta/genética , Adenoviridae/fisiología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Vasos Coronarios/metabolismo , Femenino , Expresión Génica , Transferencia de Gen Horizontal , Trasplante de Corazón/fisiología , Interleucina-12/farmacología , Liposomas , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Sensibilidad y Especificidad , Células TH1/inmunología , Factor de Crecimiento Transformador beta1 , Transgenes/genética
12.
Proc Natl Acad Sci U S A ; 97(20): 10814-9, 2000 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-11005857

RESUMEN

Two RecA homologs, Rad51 and Dmc1, assemble as cytologically visible complexes (foci) at the same sites on meiotic chromosomes. Time course analysis confirms that co-foci appear and disappear as the single predominant form. A large fraction of co-foci are eliminated in a red1 mutant, which is expected as a characteristic of the interhomolog-specific recombination pathway. Previous studies suggested that normal Dmc1 loading depends on Rad51. We show here that a mutation in TID1/RDH54, encoding a RAD54 homolog, reduces Rad51-Dmc1 colocalization relative to WT. A rad54 mutation, in contrast, has relatively little effect on RecA homolog foci except when strains also contain a tid1/rdh54 mutation. The role of Tid1/Rdh54 in coordinating RecA homolog assembly may be very direct, because Tid1/Rdh54 is known to physically bind both Dmc1 and Rad51. Also, Dmc1 foci appear early in a tid1/rdh54 mutant. Thus, Tid1 may normally act with Rad51 to promote ordered RecA homolog assembly by blocking Dmc1 until Rad51 is present. Finally, whereas double-staining foci predominate in WT nuclei, a subset of nuclei with expanded chromatin exhibit individual Rad51 and Dmc1 foci side-by-side, suggesting that a Rad51 homo-oligomer and a Dmc1 homo-oligomer assemble next to one another at the site of a single double-strand break (DSB) recombination intermediate.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN/genética , Proteínas Fúngicas/genética , Meiosis/genética , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Animales , ADN Helicasas , Enzimas Reparadoras del ADN , ADN-Topoisomerasas , Regulación Fúngica de la Expresión Génica , Cobayas , Recombinasa Rad51 , Saccharomyces cerevisiae
13.
Cell ; 102(3): 279-91, 2000 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-10975519

RESUMEN

Phosphorylation of histone H3 at serine 10 occurs during mitosis and meiosis in a wide range of eukaryotes and has been shown to be required for proper chromosome transmission in Tetrahymena. Here we report that Ipl1/aurora kinase and its genetically interacting phosphatase, Glc7/PP1, are responsible for the balance of H3 phosphorylation during mitosis in Saccharomyces cerevisiae and Caenorhabditis elegans. In these models, both enzymes are required for H3 phosphorylation and chromosome segregation, although a causal link between the two processes has not been demonstrated. Deregulation of human aurora kinases has been implicated in oncogenesis as a consequence of chromosome missegregation. Our findings reveal an enzyme system that regulates chromosome dynamics and controls histone phosphorylation that is conserved among diverse eukaryotes.


Asunto(s)
Caenorhabditis elegans/citología , Proteínas Fúngicas/metabolismo , Histonas/metabolismo , Mitosis , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Saccharomyces cerevisiae/citología , Animales , Aurora Quinasas , Caenorhabditis elegans/metabolismo , Genoma , Proteínas del Helminto/metabolismo , Fenotipo , Fosforilación , ARN sin Sentido , ARN Interferente Pequeño , Saccharomyces cerevisiae/metabolismo , Serina/metabolismo , Especificidad de la Especie
14.
J Biol Chem ; 275(31): 23899-903, 2000 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-10843985

RESUMEN

Mutations in breast cancer tumor susceptibility genes, BRCA1 and BRCA2, predispose women to early onset breast cancer and other malignancies. The Brca genes are involved in multiple cellular processes in response to DNA damage including checkpoint activation, gene transcription, and DNA repair. Biochemical interaction with the recombinational repair protein Rad51 (Scully, R., Chen, J., Ochs, R. L., Keegan, K., Hoekstra, M., Feunteun, J., and Livingston, D. M. (1997) Cell 90, 425-435), as well as genetic evidence (Moynahan, M. E., Chiu, J. W., Koller, B. H., and Jasin, M. (1999) Mol. Cell 4, 511-518 and Snouwaert, J. N., Gowen, L. C., Latour, A. M., Mohn, A. R., Xiao, A., DiBiase, L., and Koller, B. H. (1999) Oncogene 18, 7900-7907), demonstrates that Brca1 is involved in recombinational repair of DNA double strand breaks. Using isogenic Brca1(+/+) and brca1(-/-) mouse embryonic stem (ES) cell lines, we investigated the role of Brca1 in the cellular response to two different categories of DNA damage: x-ray induced damage and cross-linking damage caused by the chemotherapeutic agent, cisplatinum. Immunoflourescence studies with normal and brca1(-/-) mutant mouse ES cell lines indicate that Brca1 promotes assembly of subnuclear Rad51 foci following both types of DNA damage. These foci are likely to be oligomeric complexes of Rad51 engaged in repair of DNA lesions or in processes that allow cells to tolerate such lesions during DNA replication. Clonogenic assays show that brca1(-/-) mutants are 5-fold more sensitive to cisplatinum compared with wild-type cells. Our studies suggest that Brca1 contributes to damage repair and/or tolerance by promoting assembly of Rad51. This function appears to be shared with Brca2.


Asunto(s)
Núcleo Celular/metabolismo , Cisplatino/farmacología , Proteínas de Unión al ADN/metabolismo , Genes BRCA1 , Neoplasias Mamarias Animales/genética , Rayos X/efectos adversos , Animales , Proteína BRCA2 , Supervivencia Celular , Reactivos de Enlaces Cruzados , Daño del ADN , Reparación del ADN , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Predisposición Genética a la Enfermedad , Ratones , Proteínas de Neoplasias/genética , Recombinasa Rad51 , Factores de Transcripción/genética
15.
Gene Ther ; 7(7): 575-82, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10819572

RESUMEN

Introducing immunomodulatory molecules into allografts by gene transfer may avoid the side-effects of systemic immunosuppression. vMIP-II and MC148 are two recently identified chemokine homologues encoded by human herpes virus 8 and Molluscum contagiosum, respectively, that have antagonistic activities against multiple different CC and CXC chemokine receptors. We hypothesized that introduction of these molecules into cardiac allografts may block leukocyte infiltration into the grafts and prolong survival. Vascularized and nonvascularized cardiac allografts in mice were performed and plasmid DNA encoding vMIP-II, MC148 and/or vIL-10 was transferred into the allograft at the time of transplantation. Gene transfer of either vMIP-II or MC148 into cardiac allografts markedly prolonged graft survival. Combining gene transfer of either one of these chemokine antagonists with vIL-10 gene transfer, which has a mechanistically different immunosuppressive action, further enhanced graft survival. vMIP-II and MC148 gene transfer both resulted in a marked decrease of donor-specific cytotoxic T lymphocytes (CTL) infiltrating the grafts and inhibited alloantibody production. These results demonstrate that plasmid-mediated gene transfer of virally encoded chemokine antagonists vMIP-II and MC148 can block donor-specific lymphocyte immunity within cardiac allografts and prolong graft survival. This is a new mechanistic approach to analyze, treat, and prevent graft rejection. Delivery of these or related molecules by gene transfer or conventional pharmacologic means may represent a novel therapeutic modality for alloactivation.


Asunto(s)
Terapia Genética/métodos , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Receptores de Quimiocina/antagonistas & inhibidores , Transfección/métodos , Animales , Formación de Anticuerpos , Femenino , Vectores Genéticos/administración & dosificación , Herpesvirus Humano 8/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Virus del Molusco Contagioso/genética , Receptores de Quimiocina/genética , Linfocitos T Citotóxicos/inmunología
16.
Hum Gene Ther ; 11(6): 839-50, 2000 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-10779161

RESUMEN

We performed a clinical study of five patients with melanoma to evaluate the immunobiological effects of retrovirally transduced autologous tumor cells given as a vaccine to prime draining lymph nodes. Patients were inoculated with both wild-type (WT) and GM-CSF gene-transduced tumor cells in different extremities. Approximately 7 days later, vaccine-primed lymph nodes (VPLNs) were removed. There was an increased infiltration of dendritic cells (DCs) in the GM-CSF-secreting vaccine sites compared with the WT vaccine sites. This resulted in a greater number of cells harvested from the GM-CSF-VPLNs compared with the WT-VPLNs at a time when serum levels of GM-CSF were not detectable. Four of five patients proceeded to have the adoptive transfer of GM-CSF-VPLN cells secondarily activated and expanded ex vivo with anti-CD3 MAb and IL-2. One patient had a durable complete remission of metastatic tumor. Utilizing cytokine (IFN-gamma, GM-CSF, IL-10) release assays, GM-CSF-VPLN T cells manifested diverse responses when exposed to tumor antigen in vitro. In two of two patients, GM-CSF-VPLN T cell responses were different from those of matched WT-VPLN cells. This study documents measurable immunobiologic differences of GM-CSF-transduced tumor cells given as a vaccine compared with WT tumor cells. The complete tumor remission in one patient provides a rationale to pursue this approach further.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Inmunoterapia Adoptiva , Ganglios Linfáticos/citología , Melanoma/terapia , Células Tumorales Cultivadas/metabolismo , Adulto , Anciano , Trasplante de Células , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunohistoquímica , Interleucina-2/uso terapéutico , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Transducción Genética , Trasplante Autólogo , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/trasplante
17.
Genes Cells ; 4(8): 425-44, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10526232

RESUMEN

BACKGROUND: DMC1, the meiosis-specific eukaryotic homologue of bacterial recA, is required for completion of meiotic recombination and cell cycle progression past prophase. In a dmc1 mutant, double strand break recombination intermediates accumulate and cells arrest in prophase. We isolated genes which, when present at high copy numbers, suppress the meiotic arrest phenotype conferred by dmc1 mutations. RESULTS: Among the genes isolated were two which suppress arrest by altering the recombination process. REC114 suppresses formation of double strand break (DSB) recombination intermediates. The low viability of spores produced by dmc1 mutants carrying high copy numbers of REC114 is rescued when reductional segregation is bypassed by mutation of spo13. High copy numbers of RAD54 suppress dmc1 arrest, promote DSB repair, and allow formation of viable spores following reductional segregation. Analysis of the combined effects of a null mutation in RED1, a gene required for meiotic chromosome structure, with null mutations in RAD54 and DMC1 shows that RAD54, while not normally important for repair of DSBs during meiosis, is required for efficient repair of breaks by the intersister recombination pathway that operates in red1 dmc1 double mutants. CONCLUSIONS: Over-expression of REC114 suppresses meiotic arrest by preventing formation of DSBs. High copy numbers of RAD54 activate a DMC1-independent mechanism that promotes repair of DSBs by homology-mediated recombination. The ability of RAD54 to promote DMC1-independent recombination is proposed to involve suppression of a constraint that normally promotes recombination between homologous chromatids rather than sisters.


Asunto(s)
Proteínas de Ciclo Celular , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas Fúngicas/genética , Meiosis , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Supresión Genética , ADN Helicasas , Enzimas Reparadoras del ADN , Biblioteca de Genes , Modelos Genéticos , Mutación , Plásmidos/metabolismo , Proteínas Recombinantes/metabolismo , Recombinasas , Saccharomyces cerevisiae/genética , Factores de Tiempo
18.
Genetics ; 153(2): 607-20, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10511543

RESUMEN

Checkpoint gene function prevents meiotic progression when recombination is blocked by mutations in the recA homologue DMC1. Bypass of dmc1 arrest by mutation of the DNA damage checkpoint genes MEC1, RAD17, or RAD24 results in a dramatic loss of spore viability, suggesting that these genes play an important role in monitoring the progression of recombination. We show here that the role of mitotic checkpoint genes in meiosis is not limited to maintaining arrest in abnormal meioses; mec1-1, rad24, and rad17 single mutants have additional meiotic defects. All three mutants display Zip1 polycomplexes in two- to threefold more nuclei than observed in wild-type controls, suggesting that synapsis may be aberrant. Additionally, all three mutants exhibit elevated levels of ectopic recombination in a novel physical assay. rad17 mutants also alter the fraction of recombination events that are accompanied by an exchange of flanking markers. Crossovers are associated with up to 90% of recombination events for one pair of alleles in rad17, as compared with 65% in wild type. Meiotic progression is not required to allow ectopic recombination in rad17 mutants, as it still occurs at elevated levels in ndt80 mutants that arrest in prophase regardless of checkpoint signaling. These observations support the suggestion that MEC1, RAD17, and RAD24, in addition to their proposed monitoring function, act to promote normal meiotic recombination.


Asunto(s)
Proteínas de Ciclo Celular/genética , Reparación del ADN/genética , Proteínas Fúngicas/genética , Genes Fúngicos , Recombinación Genética , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Proteínas de Ciclo Celular/metabolismo , Cromosomas Fúngicos/genética , Intercambio Genético , Daño del ADN , Proteínas de Unión al ADN , Endodesoxirribonucleasas/metabolismo , Proteínas Fúngicas/metabolismo , Genotipo , Péptidos y Proteínas de Señalización Intracelular , Meiosis , Modelos Genéticos , Mutagénesis , Mutagénesis Insercional , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas , Saccharomyces cerevisiae/citología
19.
Nat Med ; 5(10): 1143-9, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10502817

RESUMEN

The immune response to adenoviral vectors can induce inflammation and loss of transgene expression in transfected tissues. This would limit the use of adenovirus-mediated gene transfer in disease states in which long-term gene expression is required. While studying the effect of the anti-adenoviral immune response in transplantation, we found that transgene expression persisted in cardiac isografts transfected with an adenovirus encoding beta-galactosidase. Transfected grafts remained free of inflammation, despite the presence of an immune response to the vector. Thus, adenovirus-mediated gene transfer may have therapeutic value in cardiac transplantation and heart diseases. Furthermore, immunological limitations of adenoviral vectors for gene therapy are not universal for all tissue types.


Asunto(s)
Adenoviridae/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos/inmunología , Trasplante de Corazón/inmunología , Animales , Femenino , Trasplante de Corazón/efectos adversos , Infusiones Intravenosas , Ratones , Ratones Endogámicos C57BL , Transgenes , Trasplante Isogénico , beta-Galactosidasa/genética
20.
Transplantation ; 67(12): 1548-55, 1999 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-10401761

RESUMEN

BACKGROUND: Transient depletion of CD4+ T cells in cardiac allograft recipients prolongs allograft survival; however, grafts exhibit signs of chronic rejection characterized by collagen deposition and neointima development. Although it is believed that Th1 cells promote acute graft rejection, the role of these cells in chronic rejection remains unclear. Hence, our study evaluated whether Th1 cells are associated with the development of chronic cardiac allograft rejection. METHODS: Splenocytes obtained from C57BL/6 recipients bearing BALB/c hearts with signs of chronic rejection were adoptively transferred into C57BL/6 SCID cardiac allograft recipients. As a measure of Th1 function, interferon-y production was determined after restimulation of recipient splenocytes with donor alloantigens. RESULTS: Transfer of splenocytes in SCID allograft recipients resulted in accelerated chronic rejection in the majority of mice. Characterization of these cells before transfer revealed hyporesponsive Th1 function. However, donor-specific proliferative responses and precursor interleukin-2 producing helper and cytotoxic T lymphocyte frequencies were comparable to that of naive splenocytes. Further, splenocytes obtained from SCID recipients with advanced signs of chronic rejection remained deficient in Th1 function, suggesting that Th1 are not involved in this disease process. This possibility was further supported by the development of chronic rejection in IL-12 knockout recipients. Finally, when splenocytes used for adoptive transfer retained Th1 function, transfer of these cells into SCID recipients resulted in acute allograft rejection. CONCLUSIONS: We have established a model in which the mediators of chronic rejection may be further explored. In this system, the absence rather than the presence of donor-reactive Th1 is associated with chronic rejection. These data indicate that Th1-independent effector mechanisms are responsible for chronic rejection in this model.


Asunto(s)
Citocinas/fisiología , Trasplante de Corazón/inmunología , Adyuvantes Inmunológicos/fisiología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD4/inmunología , División Celular/fisiología , Enfermedad Crónica , Rechazo de Injerto/inmunología , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Bazo/citología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...