Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Transplantation ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38995954

RESUMEN

BACKGROUND: The clinical standard for pancreas preservation for transplantation is static cold storage (SCS). Oxygenation during preservation has been shown to be advantageous in clinical studies. This study evaluates the efficiency of different oxygenation modalities during hypothermic pancreas preservation. METHODS: Thirty-two porcine pancreases were procured in a controlled donation after circulatory death model and were divided to be preserved in 8 groups: (1) SCS, (2) hypothermic machine perfusion (HMP), (3) hypothermic oxygenated machine perfusion (HOPE) with 21% oxygen, (4) HOPE and 100%, (5) SCS and oxygen carrier, M101, (6) HMP and M101, (7) HOPE 21% and M101, and (8) HOPE 100% and M101. All the groups underwent 24 h of hypothermic preservation, followed by 2 h of normothermic reperfusion. Oxygen partial pressures were assessed using parenchymal probes. Perfusion parameters, perfusate samples, and tissue biopsies were analyzed. RESULTS: This study showed that HMP was linked to higher tissue oxygen partial pressures, lower succinate levels, and better reperfusion parameters. Furthermore, the addition of M101 to either SCS or HMP was associated with lower succinate and creatinine phosphokinase accumulation, suggesting a protective effect against ischemia. CONCLUSIONS: Our research has demonstrated the efficacy of machine perfusion in hypothermic conditions in providing oxygen to the pancreas during preservation and conditioning the pancreatic microvasculature for reperfusion during transplantation. Furthermore, the addition of M101 suggests a protective effect on the graft from ischemia.

2.
Cell Mol Gastroenterol Hepatol ; 18(1): 133-153, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38428588

RESUMEN

BACKGROUND & AIMS: The presence of myenteric plexitis in the proximal resection margins is a predictive factor of early postoperative recurrence in Crohn's disease. To decipher the mechanisms leading to their formation, T-cell interactions with enteric neural cells were studied in vitro and in vivo. METHODS: T cells close to myenteric neural cells were retrospectively quantified in ileocolonic resections from 9 control subjects with cancer and 20 patients with Crohn's disease. The mechanisms involved in T-cell adhesion were then investigated in co-cultures of T lymphocytes with enteric glial cells (glia). Finally, the implication of adhesion molecules in the development of plexitis and colitis was studied in vitro but also in vivo in Winnie mice. RESULTS: The mean number of T cells close to glia, but not neurons, was significantly higher in the myenteric ganglia of relapsing patients with Crohn's disease (2.42 ± 0.5) as compared with controls (0.36 ± 0.08, P = .0007). Co-culture experiments showed that exposure to proinflammatory cytokines enhanced T-cell adhesion to glia and increased intercellular adhesion molecule-1 (ICAM-1) expression in glia. We next demonstrated that T-cell adhesion to glia was inhibited by an anti-ICAM-1 antibody. Finally, using the Winnie mouse model of colitis, we showed that the blockage of ICAM-1/lymphocyte function-associated antigen-1 (LFA-1) with lifitegrast reduced colitis severity and decreased T-cell infiltration in the myenteric plexus. CONCLUSIONS: Our present work argues for a role of glia-T-cell interaction in the development of myenteric plexitis through the adhesion molecules ICAM-1/LFA-1 and suggests that deciphering the functional consequences of glia-T-cell interaction is important to understand the mechanisms implicated in the development and recurrence of Crohn's disease.


Asunto(s)
Adhesión Celular , Técnicas de Cocultivo , Enfermedad de Crohn , Molécula 1 de Adhesión Intercelular , Plexo Mientérico , Neuroglía , Linfocitos T , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Enfermedad de Crohn/patología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Plexo Mientérico/patología , Plexo Mientérico/metabolismo , Plexo Mientérico/inmunología , Neuroglía/metabolismo , Neuroglía/patología , Neuroglía/inmunología , Estudios Retrospectivos , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
J Am Soc Nephrol ; 33(7): 1402-1410, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35728884

RESUMEN

BACKGROUND: Chronic hypomagnesemia is commonly due to diarrhea, alcoholism, and drugs. More rarely, it is caused by genetic defects in the effectors of renal magnesium reabsorption. METHODS: In an adult patient with acquired severe hypomagnesemia, hypocalcemia, tubulointerstitial nephropathy, and rapidly progressing kidney injury, similarities between the patient's presentation and features of genetic disorders of renal magnesium transport prompted us to investigate whether the patient had an acquired autoimmune cause of renal magnesium wasting. To determine if the patient's condition might be explained by autoantibodies directed against claudin-16 or claudin-19, transmembrane paracellular proteins involved in renal magnesium absorption, we conducted experiments with claudin knockout mice and transfected mouse kidney cells expressing human claudin-16 or claudin-19. We also examined effects on renal magnesium handling in rats given intravenous injections of IgG purified from sera from the patient or controls. RESULTS: Experiments with the knockout mice and in vitro transfected cells demonstrated that hypomagnesemia in the patient was causally linked to autoantibodies directed against claudin-16, which controls paracellular magnesium reabsorption in the thick ascending limb of Henle's loop. Intravenous injection of IgG purified from the patient's serum induced a marked urinary waste of magnesium in rats. Immunosuppressive treatment combining plasma exchange and rituximab was associated with improvement in the patient's GFR, but hypomagnesemia persisted. The patient was subsequently diagnosed with a renal carcinoma that expressed a high level of claudin-16 mRNA. CONCLUSIONS: Pathogenic claudin-16 autoantibodies represent a novel autoimmune cause of specific renal tubular transport disturbances and tubulointerstitial nephropathy. Screening for autoantibodies targeting claudin-16, and potentially other magnesium transporters or channels in the kidney, may be warranted in patients with acquired unexplained hypomagnesemia.


Asunto(s)
Hipocalcemia , Nefritis Intersticial , Animales , Autoanticuerpos , Claudinas/genética , Inmunoglobulina G , Magnesio , Ratones , Ratones Noqueados , Ratas
4.
J Clin Invest ; 130(11): 6109-6123, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33074246

RESUMEN

T cell exclusion causes resistance to cancer immunotherapies via immune checkpoint blockade (ICB). Myeloid cells contribute to resistance by expressing signal regulatory protein-α (SIRPα), an inhibitory membrane receptor that interacts with ubiquitous receptor CD47 to control macrophage phagocytosis in the tumor microenvironment. Although CD47/SIRPα-targeting drugs have been assessed in preclinical models, the therapeutic benefit of selectively blocking SIRPα, and not SIRPγ/CD47, in humans remains unknown. We report a potent synergy between selective SIRPα blockade and ICB in increasing memory T cell responses and reverting exclusion in syngeneic and orthotopic tumor models. Selective SIRPα blockade stimulated tumor nest T cell recruitment by restoring murine and human macrophage chemokine secretion and increased anti-tumor T cell responses by promoting tumor-antigen crosspresentation by dendritic cells. However, nonselective SIRPα/SIRPγ blockade targeting CD47 impaired human T cell activation, proliferation, and endothelial transmigration. Selective SIRPα inhibition opens an attractive avenue to overcoming ICB resistance in patients with elevated myeloid cell infiltration in solid tumors.


Asunto(s)
Memoria Inmunológica , Inmunoterapia , Neoplasias Mamarias Experimentales/terapia , Proteínas de Neoplasias/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T/inmunología , Animales , Femenino , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Receptores Inmunológicos/genética , Linfocitos T/patología
5.
Biochem Biophys Res Commun ; 519(4): 667-673, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31542230

RESUMEN

Calcineurin Inhibitors (CNIs) are routinely used for immunosuppression following solid organ transplantation. However, the prolonged use of these agents lead to organ fibrosis which limits their efficacy. CNIs induce TGFß expression, which is reported to augment endothelial-to-mesenchymal transition (EndMT), but their role in this process is not known. In these studies, we find that the CNIs FK506 and cyclosporine (CsA) are potent to increase endothelial cell (EC) proliferation using established in vitro assays (P < 0.05). Furthermore, using phosphokinase arrays, we find that each CNI activates the MAPK and Akt/mTOR signaling pathways, and that pharmacological inhibition of each pathway targets CNI-induced proliferative responses (P < 0.001). EndMT was evaluated by FACS for N-cadherin and CD31 expression and by qPCR for the expression of α-smooth muscle actin, N-cadherin and Snail. We find that CNIs do not directly induce dedifferentiation, while TGFß and hypoxia induce EndMT in small numbers of EC. In contrast, the treatment of EC with the inflammatory cytokine TNFα was potent to elicit an EndMT response, and its effects were most notably in EC following proliferation/doubling. Taken together, these observations suggest that CNIs elicit proliferative responses, which enhance EndMT in association with local inflammation. The clinical implications of these findings are that anti-proliferative therapeutics have high potential to target the initiation of this EndMT response.


Asunto(s)
Inhibidores de la Calcineurina/farmacología , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Células CHO , Cadherinas/metabolismo , Células Cultivadas , Cricetinae , Cricetulus , Ciclosporina/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Tacrolimus/farmacología
6.
Arthritis Rheumatol ; 71(4): 641-650, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30375745

RESUMEN

OBJECTIVE: To compare the effects of rituximab (RTX) and conventional immunosuppressants (CIs) on CD4+ T cells, Treg cells, and CD8+ T cells in antineutrophil cytoplasmic antibody-associated vasculitis (AAV). METHODS: A thorough immunophenotype analysis of CD4+, Treg, and CD8+ cells from 51 patients with AAV was performed. The production of cytokines and chemokines by CD8+ T cells stimulated in vitro was assessed using a multiplex immunoassay. The impact of AAV B cells on CD8+ T cell response was assessed using autologous and heterologous cocultures. RESULTS: CD4+ and Treg cell subsets were comparable among RTX-treated and CI-treated patients. In contrast, within the CD8+ T cell compartment, RTX, but not CIS, reduced CD45RA+CCR7- (TEMRA) cell frequency (from a median of 39% before RTX treatment to 10% after RTX treatment [P < 0.01]) and efficiently dampened cytokine/chemokine production (e.g., the median macrophage inflammatory protein 1α level was 815 pg/ml in patients treated with RTX versus 985 pg/ml in patients treated with CIs versus 970 pg/ml in those with active untreated AAV [P < 0.01]). CD8+ T cell subsets cocultured with autologous B cells produced more proinflammatory cytokines in AAV patients than in controls (e.g., for tumor necrosis factor-producing effector memory CD8+ T cells: 14% in AAV patients versus 9.2% in controls [P < 0.05]). In vitro disruption of AAV B cell-CD8+ T cell cross-talk reduced CD8+ T cell cytokine production, mirroring the reduced CD8+ response observed ex vivo after RTX treatment. CONCLUSION: The disruption of a pathogenic B cell/CD8+ T cell axis may contribute to the efficacy of RTX in AAV. Further studies are needed to determine the value of CD8+ T cell immunomonitoring in B cell-targeted therapies.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Inmunosupresores/farmacología , Rituximab/farmacología , Anciano , Linfocitos B/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunofenotipificación , Inmunosupresores/inmunología , Masculino , Persona de Mediana Edad , Rituximab/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento
7.
Pediatr Nephrol ; 31(1): 41-51, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25903640

RESUMEN

Advances in therapeutics have dramatically improved short-term graft survival, but the incidence of chronic rejection has not changed in the past 20 years. New insights into mechanism are sorely needed at this time and it is hoped that the development of predictive biomarkers will pave the way for the emergence of preventative therapeutics. In this review, we discuss a paradigm suggesting that sequential changes within graft endothelial cells (EC) lead to an intragraft microenvironment that favors the development of chronic rejection. Key initial events include EC injury, activation and uncontrolled leukocyte-induced angiogenesis. We propose that all of these early changes in the microvasculature lead to abnormal blood flow patterns, local tissue hypoxia, and an associated overexpression of HIF-1α-inducible genes, including vascular endothelial growth factor. We also discuss how cell intrinsic regulators of mTOR-mediated signaling within EC are of critical importance in microvascular stability and may thus have a role in the inhibition of chronic rejection. Finally, we discuss recent findings indicating that miRNAs may regulate EC stability, and we review their potential as novel non-invasive biomarkers of allograft rejection. Overall, this review provides insights into molecular events, genes, and signals that promote chronic rejection and their potential as biomarkers that serve to support the future development of interruption therapeutics.


Asunto(s)
Células Endoteliales/metabolismo , Rechazo de Injerto/etiología , Trasplante de Riñón/efectos adversos , Riñón/irrigación sanguínea , Microvasos/metabolismo , Investigación Biomédica Traslacional , Aloinjertos , Animales , Biomarcadores/metabolismo , Microambiente Celular , Enfermedad Crónica , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Microvasos/inmunología , Microvasos/fisiopatología , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento
8.
Sci Rep ; 5: 11789, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26156437

RESUMEN

Semaphorin 3F (SEMA3F) provides neuronal guidance cues via its ability to bind neuropilin 2 (NRP2) and Plexin A family molecules. Recent studies indicate that SEMA3F has biological effects in other cell types, however its mechanism(s) of function is poorly understood. Here, we analyze SEMA3F-NRP2 signaling responses in human endothelial, T cell and tumor cells using phosphokinase arrays, immunoprecipitation and Western blot analyses. Consistently, SEMA3F inhibits PI-3K and Akt activity, and responses are associated with the disruption of mTOR/rictor assembly and mTOR-dependent activation of the RhoA GTPase. We also find that the expression of vascular endothelial growth factor, as well as mTOR-inducible cellular activation responses and cytoskeleton stability are inhibited by SEMA3F-NRP2 interactions in vitro. In vivo, local and systemic overproduction of SEMA3F reduces tumor growth in NRP2-expressing xenografts. Taken together, SEMA3F regulates mTOR signaling in diverse human cell types, suggesting that it has broad therapeutic implications.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropilina-2/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Actinas/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Proteínas de la Membrana/genética , Ratones , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/genética , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Blood ; 122(10): 1833-42, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23881914

RESUMEN

The maintenance of normal tissue homeostasis and the prevention of chronic inflammatory disease are dependent on the active process of inflammation resolution. In endothelial cells (ECs), proinflammation results from the activation of intracellular signaling responses and/or the inhibition of endogenous regulatory/pro-resolution signaling networks that, to date, are poorly defined. In this study, we find that DEP domain containing mTOR interacting protein (DEPTOR) is expressed in different microvascular ECs in vitro and in vivo, and using a small interfering RNA (siRNA) knockdown approach, we find that it regulates mammalian target of rapamycin complex 1 (mTORC1), extracellular signal-regulated kinase 1/2, and signal transducer and activator of transcription 1 activation in part through independent mechanisms. Moreover, using limited gene arrays, we observed that DEPTOR regulates EC activation including mRNA expression of the T-cell chemoattractant chemokines CXCL9, CXCL10, CXCL11, CX3CL1, CCL5, and CCL20 and the adhesion molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 (P < .05). DEPTOR siRNA-transfected ECs also bound increased numbers of peripheral blood mononuclear cells (P < .005) and CD3+ T cells (P < .005) in adhesion assays in vitro and had increased migration and angiogenic responses in spheroid sprouting (P < .01) and wound healing (P < .01) assays. Collectively, these findings define DEPTOR as a critical upstream regulator of EC activation responses and suggest that it plays an important role in endogenous mechanisms of anti-inflammation and pro-resolution.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/patología , Inflamación/patología , Neovascularización Patológica/patología , Serina-Treonina Quinasas TOR/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular/genética , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Inflamación/genética , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/metabolismo , Neovascularización Patológica/genética , Unión Proteica , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/genética
10.
Hum Immunol ; 73(12): 1261-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22863981

RESUMEN

In this review, we discuss a paradigm whereby changes in the intragraft microenvironment promote or sustain the development of chronic allograft rejection. A key feature of this model involves the microvasculature including (a) endothelial cell (EC) destruction, and (b) EC proliferation, both of which result from alloimmune leukocyte- and/or alloantibody-induced responses. These changes in the microvasculature likely create abnormal blood flow patterns and thus promote local tissue hypoxia. Another feature of the chronic rejection microenvironment involves the overexpression of vascular endothelial growth factor (VEGF). VEGF stimulates EC activation and proliferation and it has potential to sustain inflammation via direct interactions with leukocytes. In this manner, VEGF may promote ongoing tissue injury. Finally, we review how these events can be targeted therapeutically using mTOR inhibitors. EC activation and proliferation as well as VEGF-VEGFR interactions require PI-3K/Akt/mTOR intracellular signaling. Thus, agents that inhibit this signaling pathway within the graft may also target the progression of chronic rejection and thus promote long-term graft survival.


Asunto(s)
Microambiente Celular/inmunología , Rechazo de Injerto/inmunología , Animales , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Trasplante Homólogo , Trasplantes , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Biochem Biophys Res Commun ; 425(2): 244-9, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22828505

RESUMEN

Agents that target the activity of the mammalian target of rapamycin (mTOR) kinase in humans are associated with proteinuria. However, the mechanisms underlying mTOR activity and signaling within the kidney are poorly understood. In this study, we developed a sensitive immunofluorescence technique for the evaluation of activated pmTOR and its associated signals in situ. While we find that pmTOR is rarely expressed in normal non-renal tissues, we consistently find intense expression in glomeruli within normal mouse and human kidneys. Using double staining, we find that the expression of pmTOR co-localizes with nephrin in podocytes and expression appears minimal within other cell types in the glomerulus. In addition, we found that pmTOR was expressed on occasional renal tubular cells within mouse and human kidney specimens. We also evaluated mTOR signaling in magnetic bead-isolated glomeruli from normal mice and, by Western blot analysis, we confirmed function of the pathway in glomerular cells vs. interstitial cells. Furthermore, we found that the activity of the pathway as well as the expression of VEGF, a target of mTOR-induced signaling, were reduced within glomeruli of mice following treatment with rapamycin. Collectively, these findings demonstrate that the mTOR signaling pathway is constitutively hyperactive within podocytes. We suggest that pmTOR signaling functions to regulate glomerular homeostasis in part via the inducible expression of VEGF.


Asunto(s)
Glomérulos Renales/enzimología , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Homeostasis , Humanos , Glomérulos Renales/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Podocitos/enzimología , Transducción de Señal , Sirolimus/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis
12.
Biochem Biophys Res Commun ; 419(1): 66-71, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22326918

RESUMEN

TNF-family molecules induce the expression Vascular Endothelial Growth Factor (VEGF) in endothelial cells (EC) and elicit signaling responses that result in angiogenesis. However, the role of TNF-receptor associated factors (TRAFs) as upstream regulators of VEGF expression or as mediators of angiogenesis is not known. In this study, HUVEC were cotransfected with a full-length VEGF promoter-luciferase construct and siRNAs to TRAF 1, -2, -3, -5, -6, and promoter activity was measured. Paradoxically, rather than inhibiting VEGF expression, we found that knockdown of TRAF6 resulted in a 4-6-fold increase in basal VEGF promoter activity compared to control siRNA-transfected EC (P<0.0001). In addition, knockdown of TRAF 1, -2, -3 or -5 resulted in a slight increase or no change in VEGF promoter activation. Using [(3)H]thymidine incorporation assays as well as the in vitro wound healing assay, we also found that basal rates of EC proliferation and migration were increased following TRAF6 knockdown; and this response was inhibited by the addition of a blocking anti-VEGF antibody into cell cultures. Using a limited protein array to gain insight into TRAF6-dependent intermediary signaling responses, we observed that TRAF6 knockdown resulted in an increase in the activity of Src family kinases. In addition, we found that treatment with AZD-0530, a pharmacological Src inhibitor, reduced the regulatory effect of TRAF6 knockdown on VEGF promoter activity. Collectively, these findings define a novel pro-angiogenic signaling response in EC that is regulated by TRAF6.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/fisiología , Neovascularización Fisiológica/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Activación Transcripcional , Benzodioxoles/farmacología , Movimiento Celular/genética , Proliferación Celular , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Luciferasas/genética , Regiones Promotoras Genéticas , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Factor 6 Asociado a Receptor de TNF/genética , Factor A de Crecimiento Endotelial Vascular/genética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
13.
Nephrol Dial Transplant ; 26(5): 1523-32, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20935016

RESUMEN

BACKGROUND: Buffalo/Mna rats spontaneously develop a nephrotic syndrome (NS). We have demonstrated that this rat nephropathy recurs after renal transplantation. We studied this recurrence by kinetic analysis of graft lesions, infiltrating cells and cytokines. METHODS: Kidneys from LEW.1 W rats were grafted into proteinuric Buff/Mna or healthy Wistar Furth recipients. Kidney samples were harvested before, during and after the occurrence of proteinuria and analysed for renal histology, cell populations and cytokine transcripts. Results were compared with the evolution of the initial disease studied previously. RESULTS: Both groups showed normal graft histology at Day 7 and an increasing podocyte swelling at Day 45 was seen only in the Buff/Mna recipients. At Day 80, glomerular atrophy with podocytosis and focal segmental glomerular sclerosis lesions, accompanied by tubular dilatation, appeared in the Buff/Mna group. At Day 122, the intensity of the tubular and glomerular lesions increased in Buff/Mna recipients but not in the control group. An analysis of desmin and Kim-1 (early markers of glomerular and tubular damage, respectively) transcripts expression showed that glomerular lesions precede tubular injury in this model. A monocyte infiltration associated with an increase in TNFα, IL1 and IL12 transcripts appeared before the recurrence. An early increase in Cbeta TCR transcripts with a predominant Th2 profile was observed, highlighting a Th2 polarization in the Buff/Mna recurrence. CONCLUSIONS: The comparison of profiles of recurrence and initial disease highlighted the same mediators for both events. We propose that initial Buff/Mna idiopathic nephrotic syndrome (INS) and post-transplantation recurrence represent the same entity and a valuable tool for the study of recurring INS.


Asunto(s)
Trasplante de Riñón/efectos adversos , Síndrome Nefrótico/patología , Complicaciones Posoperatorias , Animales , Síndrome Nefrótico/cirugía , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Recurrencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
Am J Kidney Dis ; 54(3): 522-32, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19520469

RESUMEN

BACKGROUND: Corticosteroid-resistant idiopathic nephrotic syndrome (INS) recurs rapidly after transplantation in 30% to 50% of transplant recipients, suggesting the presence of 1 or more circulating factors that alter the glomerular filtration barrier. We investigated the possible role in INS recurrence of soluble ST2 (sST2) protein, a marker of T helper type 2 (T(H)2) cells and a factor predicted to be regulated by the transcription factor c-Maf; involvement of sST2 protein would be consistent with the observation that both T(H)2 cells and c-Maf appear to be activated during INS relapse. STUDY DESIGN: Retrospective observational study. SETTING & PARTICIPANTS: Patients with biopsy-proven corticosteroid-resistant INS who had undergone kidney transplantation between September 1983 and April 2007 (n = 71). A control group consisting of proteinuric transplant recipients with kidney failure unrelated to INS (n = 34). PREDICTOR: Patients who developed INS recurrence after transplantation (n = 31) were compared with those in whom INS did not recur (n = 40) and the control group. Recurrence of INS was defined as urine protein excretion greater than 2 g/d immediately after transplantation that persisted at greater than 1 g/d despite treatment or a kidney graft biopsy showing minimal change glomerulonephritis or focal segmental glomerulosclerosis. OUTCOMES & MEASUREMENTS: Urine protein excretion in the 3 groups was 5.0 g/d (range, 1.3 to 10.5), 0.14 g/d (range, 0 to 0.46), and 4.3 g/d (range, 3 to 6.2). The sST2 protein was analyzed both quantitatively and qualitatively in patient sera, and its activity was tested in vitro on a mouse podocyte cell line and in vivo in rats. RESULTS: sST2 protein levels were significantly increased after transplantation in patients with INS recurrence compared with the 2 other groups (617.5 versus 23 pg/mL; P < 0.001 and 158.5 pg/mL; P < 0.01 respectively). However, patients with recurrence expressed a normal sST2 isoform, and the sST2 protein was unable to induce podocyte injury in vitro or trigger proteinuria in rats. LIMITATIONS: Pretransplantation and posttransplantation sera do not always represent paired samples. CONCLUSIONS: These data suggest that sST2 protein is a marker of INS recurrence that does not seem to be involved in the development of INS.


Asunto(s)
Trasplante de Riñón , Síndrome Nefrótico/sangre , Receptores de Superficie Celular/fisiología , Adolescente , Adulto , Animales , Biomarcadores/sangre , Células COS , Línea Celular Transformada , Niño , Chlorocebus aethiops , Femenino , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Trasplante de Riñón/efectos adversos , Masculino , Ratones , Persona de Mediana Edad , Síndrome Nefrótico/diagnóstico , Síndrome Nefrótico/prevención & control , Ratas , Ratas Endogámicas BUF , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Receptores de Superficie Celular/sangre , Estudios Retrospectivos , Prevención Secundaria , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA