Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 308(2): F122-30, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25428126

RESUMEN

Tissue fibrosis is a major cause of death in developed countries. It commonly occurs after either acute or chronic injury and affects diverse organs, including the heart, liver, lung, and kidney. Using the renal ablation model of chronic kidney disease, we previously found that the development of progressive renal fibrosis was dependent on p21(WAF1/Cip1) expression; the genetic knockout of the p21 gene greatly alleviated this disease. In the present study, we expanded on this observation and report that fibrosis induced by two different acute injuries to the kidney is also dependent on p21. In addition, when p21 expression was restricted only to the proximal tubule, fibrosis after injury was induced in the whole organ. One molecular fibrogenic switch we describe is transforming growth factor-ß induction, which occurred in vivo and in cultured kidney cells exposed to adenovirus expressing p21. Our data suggests that fibrosis is p21 dependent and that preventing p21 induction after stress could be a novel therapeutic target.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Túbulos Renales Proximales/metabolismo , Nefroesclerosis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Ácidos Aristolóquicos , Células Cultivadas , Femenino , Humanos , Ratones Noqueados
2.
Oncotarget ; 4(4): 622-35, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23599172

RESUMEN

Constitutively activated signaling molecules are often the primary drivers of malignancy, and are favored targets for therapeutic intervention. However, the effectiveness of targeted inhibition of cell signaling can be blunted by compensatory signaling which generates adaptive resistance mechanisms and reduces therapeutic responses. Therefore, it is important to identify and target these compensatory pathways with combinations of targeted agents to achieve durable clinical benefit. In this report, we demonstrate the use of high-throughput combinatorial drug screening as a discovery tool to identify compensatory pathways that generate resistance to the cytotoxic effects of targeted therapy. We screened 420 drug combinations in 14 different cell lines representing three cancer lineages, and assessed the ability of each combination to cause synergistic cytotoxicity. Drug substitution studies were used to validate the functionally important drug targets. Of the 84 combinations that caused robust synergy in multiple cell lines, none were synergistic in more than half of the lines tested, and we observed no pattern of lineage specificity in the observed synergies. This reflects the plasticity of cell signaling networks, even among cell lines of the same tissue of origin. Mechanistic analysis of one novel synergistic combination identified in the screen, the multi-kinase inhibitor Ro31-8220 and lapatinib, demonstrated compensatory crosstalk between the p70S6 kinase and EGF receptor pathways. In addition, we identified BAD as a node of convergence between these two pathways that may be playing a role in the enhanced apoptosis observed upon combination treatment.


Asunto(s)
Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Sinergismo Farmacológico , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoprecipitación , Transducción de Señal/fisiología
3.
Am J Physiol Renal Physiol ; 304(7): F875-82, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364800

RESUMEN

Endoplasmic reticulum (ER) stress contributes to acute kidney injury induced by several causes. Kidney dysfunction was shown to be influenced by gender differences. In this study we observed differences in the severity of kidney injury between male and female mice in response to tunicamycin, an ER stress agent. Tunicamycin-treated male mice showed a severe decline in kidney function and extensive kidney damage of proximal tubules in the kidney outer cortex (S1 and S2 segments). Interestingly, female tunicamycin-treated mice did not show a decline in kidney function, and their kidneys showed damage localized primarily to proximal tubules in the inner cortex (S3 segment). Protein markers of ER stress, glucose-regulated protein, and X-box binding protein 1 were also more elevated in male mice. Similarly, the induction of apoptosis was higher in tunicamycin-treated male mice, as measured by the activation of Bax and caspase-3. Testosterone administered to female mice before tunicamycin resulted in a phenotype similar to male mice with a comparable decline in renal function, tissue morphology, and induction of ER stress markers. We conclude that kidneys of male mice are much more susceptible to ER stress-induced acute kidney injury than those of females. Moreover, this sexual dimorphism could provide an interesting model to study the relation between kidney function and injury to a specific nephron segment.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Estrés del Retículo Endoplásmico/fisiología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Apoptosis/fisiología , Caspasa 3/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Factores de Transcripción del Factor Regulador X , Caracteres Sexuales , Testosterona/farmacología , Factor de Transcripción CHOP/biosíntesis , Factores de Transcripción/metabolismo , Tunicamicina , Proteína X Asociada a bcl-2/metabolismo
4.
Am J Physiol Renal Physiol ; 300(5): F1171-9, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325496

RESUMEN

Cisplatin cytotoxicity is dependent on cyclin-dependent kinase 2 (Cdk2) activity in vivo and in vitro. We found that an 18-kDa protein identified by mass spectrometry as p21(WAF1/Cip1) was phosphorylated by Cdk2 starting 12 h after cisplatin exposure. The analysis showed it was phosphorylated at serine 78, a site not previously identified. The adenoviral transduction of p21 before cisplatin exposure protects from cytotoxicity by inhibiting Cdk2. Although cisplatin causes induction of endogenous p21, the protection is inefficient. We hypothesized that phosphorylation of p21 at serine 78 could affect its role as a Cdk inhibitor, and thereby lessen its ability to protect from cisplatin cytotoxicity. To investigate the effect of serine 78 phosphorylation on p21 activity, we replaced serine 78 with aspartic acid, creating the phosphomimic p21(S78D). Mutant p21(S78D) was an inefficient inhibitor of Cdk2 and was inefficient at protecting TKPTS cells from cisplatin-induced cell death. We conclude that phosphorylation of p21 by Cdk2 limits the effectiveness of p21 to inhibit Cdk2, which is the mechanism for continued cisplatin cytotoxicity even after the induction of a protective protein.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/patología , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Ciclina A/genética , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Células HEK293 , Humanos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Ratones , Ratones de la Cepa 129 , Datos de Secuencia Molecular , Mutación , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Serina , Espectrometría de Masas en Tándem , Factores de Tiempo , Transducción Genética , Transfección
5.
Am J Physiol Renal Physiol ; 299(1): F112-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20444741

RESUMEN

Cisplatin cytotoxicity is dependent on cyclin-dependent kinase 2 (Cdk2) activity in vivo and in vitro. A Cdk2 mutant (Cdk2-F80G) was designed in which the ATP-binding pocket was altered. When expressed in mouse kidney cells, this protein was kinase inactive, did not inhibit endogenous Cdk2, but protected from cisplatin. The mutant was localized in the cytoplasm, but when coexpressed with cyclin A, it was activated, localized to the nucleus, and no longer protected from cisplatin cytotoxicity. Cells exposed to cisplatin in the presence of the activated mutant had an apoptotic phenotype, and endonuclease G was released from mitochondria similar to that mediated by endogenous Cdk2. But unlike apoptosis mediated by wild-type Cdk2, cisplatin exposure of cells expressing the activated mutant did not cause cytochrome c release or significant caspase-3 activation. We conclude that cisplatin likely activates both caspase-dependent and -independent cell death, and Cdk2 is required for both pathways. The mutant-inactive Cdk2 protected from both death pathways, but after activation by excess cyclin A, caspase-independent cell death predominated.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Quinasa 2 Dependiente de la Ciclina/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transporte Activo de Núcleo Celular , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Caspasa 3/metabolismo , Núcleo Celular/enzimología , Células Cultivadas , Ciclina A/genética , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Citocromos c/metabolismo , Citoplasma/enzimología , Citoprotección , Endodesoxirribonucleasas/metabolismo , Humanos , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/patología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mutación , Proteínas Recombinantes de Fusión/metabolismo , Transfección
6.
J Cell Biochem ; 105(3): 875-84, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18726893

RESUMEN

Ovarian cancer metastasis involves the sloughing of epithelial cells from the ovary into the peritoneal cavity, where the cells can survive and proliferate in peritoneal ascites under anchorage-independent conditions. For normal epithelial cells and fibroblasts, cell adhesion to the extracellular matrix is required to prevent apoptosis and for proper activation and nuclear signaling of the ERK MAP kinase. The mechanisms of ERK regulation by adhesion have been determined by our lab and others. In this report, we elucidate a novel means of ERK regulation by cellular adhesion in ovarian cancer cells. We demonstrate that ERK and its activator MEK are robustly stimulated after cell detachment from a substratum in several ovarian cancer cell lines, but not a benign ovarian cell line, independent of serum and FAK or PAK activity. MEK and ERK activation was sustained for 48 h after detachment, while activation by serum or growth factors in adherent cells was transient. Re-attachment of suspended ovarian cells to fibronectin restored basal levels of MEK and ERK activity. ERK activity in suspended cells was dynamically controlled through an autocrine stimulatory pathway and prevalent phosphatase activity. Suspended cells demonstrated higher levels of ERK nuclear signaling to Elk1 compared to adherent cells. Inhibition of ERK activation with the MEK inhibitor U0126 had minor effects on adherent cell growth, but greatly decreased growth in soft agar. These data demonstrate a unique regulation of ERK by cellular adhesion and suggest a mechanism by which ERK may regulate anchorage-independent growth of metastatic ovarian cancer cells.


Asunto(s)
Núcleo Celular/enzimología , Uniones Célula-Matriz/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Neoplasias Ováricas/enzimología , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/patología , Regulación hacia Arriba
7.
Mol Cell Biol ; 27(23): 8296-305, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17908799

RESUMEN

The extracellular signal-regulated kinase (ERK) cascade is activated in response to a multitude of extracellular signals and converts these signals into a variety of specific biological responses, including cell differentiation, cell movement, cell division, and apoptosis. The specificity of the biological response is likely to be controlled in large measure by the localization of signaling, thus enabling ERK activity to be directed towards specific targets. Here we show that the RACK1 scaffold protein functions specifically in integrin-mediated activation of the mitogen-activated protein kinase/ERK cascade and targets active ERK to focal adhesions. We found that RACK1 associated with the core kinases of the ERK pathway, Raf, MEK, and ERK, and that attenuation of RACK1 expression resulted in a decrease in ERK activity in response to adhesion but not in response to growth factors. RACK1 silencing also caused a reduction of active ERK in focal adhesions, an increase in focal adhesion length, a decreased rate of focal adhesion disassembly, and decreased motility. Our data further suggest that focal adhesion kinase is an upstream activator of the RACK1/ERK pathway. We suggest that RACK1 tethers the ERK pathway core kinases and channels signals from upstream activation by integrins to downstream targets at focal adhesions.


Asunto(s)
Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Adhesiones Focales/enzimología , Integrinas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuropéptidos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Pollos , Activación Enzimática , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Células 3T3 NIH , Péptidos , Transporte de Proteínas , Ratas , Receptores de Cinasa C Activada
8.
J Cell Biochem ; 94(4): 708-19, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15547943

RESUMEN

Specificity in signal transduction can be achieved through scaffolds, anchors, and adapters that assemble generic signal transduction components in specific combinations and locations. MEK Partner-1 (MP1) was identified as a potential "scaffold" protein for the mammalian extracellular signal-regulated kinase (ERK) pathway. To gain insight into the interactions of MP1 with the ERK pathway, we analyzed the ability of MP1 to bind to MEK1, ERK1, and to itself, and the regulation of these interactions. Gel filtration of cell lysates revealed two major MP1 peaks: a broad high molecular weight peak and a 28 kDa complex. An MP1 mutant that lost MEK1 binding no longer enhanced RasV12-stimulated ERK1 activity, and functioned as a dominant negative, consistent with the concept that MP1 function depends on facilitating these oligomerizations. Activation of the ERK pathway by serum or by RasV12 did not detectably affect MP1-MP1 dimerization or MP1-MEK1 interactions, but caused the dissociation of the MP1-ERK1 complex. Surprisingly, pharmacological inhibition of ERK activation did not restore the complex, suggesting that regulation of complex formation occurs independently of ERK phosphorylation. These results support the concept that MP1 functions as a regulator of MAP kinase signaling by binding to MEK1 and regulating its association with a larger signaling complex that may sequentially service multiple molecules of ERK.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Cricetinae , Activación Enzimática , Eliminación de Gen , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación/genética , Fosforilación , Unión Proteica , Suero , Proteínas ras/genética , Proteínas ras/metabolismo
9.
Proc Natl Acad Sci U S A ; 101(18): 6981-6, 2004 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-15118098

RESUMEN

Signal transduction occurs by the reversible assembly of oligomeric protein complexes that include both enzymatic proteins and proteins without known enzymatic activity. These nonenzymatic components can serve as scaffolds or anchors and regulate the efficiency, specificity, and localization of the signaling pathway. Here we report the identification of MORG1 (mitogen-activated protein kinase organizer 1), a member of the WD-40 protein family that was isolated as a binding partner of the extracellular signal-regulated kinase (ERK) pathway scaffold protein MP1. MORG1 specifically associates with several components of the ERK pathway, including MP1, Raf-1, MEK, and ERK, and stabilizes their assembly into an oligomeric complex. MORG1 facilitates ERK activation when cells are stimulated with lysophosphatidic acid, phorbol 12-myristate 13-acetate, or serum, but not in response to epidermal growth factor. Suppression of MORG1 by short interfering RNA leads to a marked reduction in ERK activity when cells are stimulated with serum. We propose that MORG1 is a component of a modular scaffold system that participates in the regulation of agonist-specific ERK signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Animales , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH
10.
Mol Cell Biol ; 24(6): 2308-17, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14993270

RESUMEN

Cell adhesion and spreading depend on activation of mitogen-activated kinase, which in turn is regulated both by growth factor and integrin signaling. Growth factors, such as epidermal growth factor, are capable of activating Ras and Raf, but integrin signaling is required to couple Raf to MEK and MEK to extracellular signal-regulated protein kinase (ERK). It was previously shown that Rac-p21-activated kinase (PAK) signaling regulated the physical association of MEK1 with ERK2 through phosphorylation sites in the proline-rich sequence (PRS) of MEK1. It was also shown that activation of MEK1 and ERK by integrins depends on PAK phosphorylation of S298 in the PRS. Here we report a novel MEK1-specific regulatory feedback mechanism that provides a means by which activated ERK can terminate continued PAK phosphorylation of MEK1. Activated ERK can phosphorylate T292 in the PRS, and this blocks the ability of PAK to phosphorylate S298 and of Rac-PAK signaling to enhance MEK1-ERK complex formation. Preventing ERK feedback phosphorylation on T292 during cellular adhesion prolonged phosphorylation of S298 by PAK and phosphorylation of S218 and S222, the MEK1 activating sites. We propose that activation of ERK during adhesion creates a feedback system in which ERK phosphorylates MEK1 on T292, and this in turn blocks additional S298 phosphorylation in response to integrin signaling.


Asunto(s)
Adhesión Celular/fisiología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Sitios de Unión/genética , Células COS , Activación Enzimática , Retroalimentación , MAP Quinasa Quinasa 1 , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Quinasas p21 Activadas
11.
J Cell Biol ; 162(2): 281-91, 2003 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-12876277

RESUMEN

Activation of the Ras-MAPK signal transduction pathway is necessary for biological responses both to growth factors and ECM. Here, we provide evidence that phosphorylation of S298 of MAPK kinase 1 (MEK1) by p21-activated kinase (PAK) is a site of convergence for integrin and growth factor signaling. We find that adhesion to fibronectin induces PAK1-dependent phosphorylation of MEK1 on S298 and that this phosphorylation is necessary for efficient activation of MEK1 and subsequent MAPK activation. The rapid and efficient activation of MEK and phosphorylation on S298 induced by cell adhesion to fibronectin is influenced by FAK and Src signaling and is paralleled by localization of phospho-S298 MEK1 and phospho-MAPK staining in peripheral membrane-proximal adhesion structures. We propose that FAK/Src-dependent, PAK1-mediated phosphorylation of MEK1 on S298 is central to the organization and localization of active Raf-MEK1-MAPK signaling complexes, and that formation of such complexes contributes to the adhesion dependence of growth factor signaling to MAPK.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células COS , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Chlorocebus aethiops , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Fibroblastos/citología , Fibroblastos/enzimología , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Adhesiones Focales/metabolismo , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/farmacología , MAP Quinasa Quinasa 1 , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Pirimidinas/farmacología , Ratas , Proteínas Recombinantes/metabolismo , Quinasas p21 Activadas , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/efectos de los fármacos , Familia-src Quinasas/metabolismo
12.
J Immunol ; 170(3): 1383-91, 2003 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-12538698

RESUMEN

IL-10 has proved to be a key cytokine in regulating inflammatory responses by controlling the production and function of various other cytokines. The suppressor of cytokine signaling (SOCS) gene products are a family of cytoplasmic molecules that are essential mediators for negatively regulating cytokine signaling. It has been previously shown that IL-10 induced SOCS3 expression and that forced constitutive expression of SOCS3 inhibits IL-10/STAT3 activation and LPS-induced macrophage activation. In this report, we show that, in addition to SOCS3 expression, IL-10 induces SOCS1 up-regulation in all cell lines tested, including Ba/F3 pro-B cells, MC/9 mast cells, M1 leukemia cells, U3A human fibroblasts, and primary mouse CD4(+) T cells. Induction of SOCS molecules is dependent on STAT3 activation by IL-10R1. Cell lines constitutively overexpressing SOCS proteins demonstrated that SOCS1 and SOCS3, but not SOCS2, are able to partially inhibit IL-10-mediated STAT3 activation and proliferative responses. Pretreatment of M1 cells with IFN-gamma resulted in SOCS1 induction and a reduction of IL-10-mediated STAT3 activation and cell growth inhibition. IL-10-induced SOCS is associated with the inhibition of IFN-gamma signaling in various cell types, and this inhibition is independent of C-terminal serine residues of the IL-10R, previously shown to be required for other anti-inflammatory responses. Thus, the present results show that both SOCS1 and SOCS3 are induced by IL-10 and may be important inhibitors of both IL-10 and IFN-gamma signaling. IL-10-induced SOCS1 may directly inhibit IL-10 IFN-gamma signaling, while inhibition of other proinflammatory cytokine responses may use additional IL-10R1-mediated mechanisms.


Asunto(s)
Proteínas Portadoras/fisiología , Inmunosupresores/farmacología , Interleucina-10/antagonistas & inhibidores , Interleucina-10/fisiología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Represoras , Transducción de Señal/inmunología , Factores de Transcripción , Animales , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , División Celular/inmunología , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Inhibidores de Crecimiento/fisiología , Humanos , Interferón gamma/antagonistas & inhibidores , Interferón gamma/farmacología , Ratones , Ratones Endogámicos CBA , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/biosíntesis , Factor de Transcripción STAT3 , Transducción de Señal/genética , Proteína 1 Supresora de la Señalización de Citocinas , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Transactivadores/fisiología , Transfección , Células Tumorales Cultivadas
13.
Transplantation ; 73(12): 1943-8, 2002 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12131693

RESUMEN

BACKGROUND: Intestinal transplantation has been hampered by high rates of intestinal allograft rejection. One mechanism of altering rejection in other organ transplant models has been blockade of second set T-cell costimulatory signals. AH.F5, a novel hamster anti-rat monoclonal antibody to CD154, blocks CD40-dependent T-cell costimulation. We hypothesized that blockade of this pathway might abrogate rejection in a rodent orthotopic survival model of intestinal transplantation. METHODS: Eight groups were studied with different dosing schema, including syngeneic transplants (group 1), untreated allogeneic transplants (group 2), allogeneic transplants plus multiple doses of AH.F5 alone given IV or s.c. (groups 3 and 4), allogeneic transplants plus donor splenocyte preconditioning with and without single dose AH.F5 (groups 5 and 6), and donor splenocyte preconditioning followed by multiple doses of AH.F5 with and without thymectomy (groups 7 and 8). RESULTS: Control animals all died within 12 days of transplantation, whereas antibody-alone and splenocytes-alone resulted in modest prolongation of survival to 16 days. Only animals treated with splenocytes before transplantation and AH.F5 survived long-term (>60 days, group 8). These animals tolerated donor-specific skin grafts, rejected third-party grafts, and fed normally. However, their weight gain was subnormal and they demonstrated intestinal muscular thickening, which might represent chronic rejection. Thymectomy prevented the induction of tolerance. CONCLUSIONS: AH.F5 prevents acute intestinal allograft rejection in combination with donor-specific splenocyte preconditioning. We achieved long-term survival and the animals appeared tolerant. Central conditioning is essential for success with this antibody when used alone. Further studies with different dosing regimens or second agents seem warranted.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Ligando de CD40/fisiología , Tolerancia Inmunológica , Intestino Delgado/trasplante , Animales , Ligando de CD40/inmunología , Cricetinae , Rechazo de Injerto , Masculino , Ratas , Ratas Endogámicas BN , Ratas Endogámicas Lew , Acondicionamiento Pretrasplante , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA