Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Transplantation ; 86(3): 436-44, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18698248

RESUMEN

BACKGROUND: Hypomagnesemia is a common finding in patients receiving cyclosporine A (CsA) therapy. The relationship between CsA-induced hypomagnesemia and nephrotoxicity and the effects of oral magnesium (Mg) supplementation remain unclear. After a retrospective analysis of the time-course of plasma Mg and creatinine levels in lung allograft recipients treated with both CsA and oral Mg supplementation, we investigated the effects of CsA treatment on Mg homeostasis in mice with normal or Mg-deficient diet and the effects of oral Mg supplementation on plasma Mg levels. METHODS: Thirty lung-allograft recipients entered the retrospective study. One thousand two hundred twenty-eight blood samples were analyzed for blood and creatinine levels. Cyclosporine A (50 mg/kg/day by intraperitoneal injection) was administered to mice maintained on normal diet (1400 ppm) or Mg-deficient (50 ppm) diet. Magnesium levels were determined in plasma, urine, feces and femur, and creatinine levels were determined in plasma and urine. RESULTS: Plasma Mg concentration declines from the day of transplantation in 36.7% of the patients despite Mg supplementation, without correlation with creatinine changes. In mice, CsA induced an early moderate hypomagnesemia, which could not be ameliorated by oral Mg supplementation and was aggravated by low-Mg dietary, late increase in plasma creatinine and decrease in urine creatinine without histological signs of renal injury, decrease in intestinal Mg absorption and Mg mobilization from bone. CONCLUSION: Cyclosporine A treatment may induce moderate hypomagnesemia that is aggravated by inadequate Mg intake and is not ameliorated by Mg supplementation. Because of the clinical complications of hypomagnesemia, Mg should be monitored regularly in allograft recipients receiving CsA.


Asunto(s)
Ciclosporina/efectos adversos , Suplementos Dietéticos , Inmunosupresores/efectos adversos , Enfermedades Renales/inducido químicamente , Trasplante de Pulmón , Deficiencia de Magnesio/inducido químicamente , Magnesio/sangre , Ácido Pirrolidona Carboxílico/uso terapéutico , Administración Oral , Adolescente , Adulto , Animales , Creatinina/sangre , Modelos Animales de Enfermedad , Femenino , Homeostasis , Humanos , Enfermedades Renales/sangre , Deficiencia de Magnesio/sangre , Deficiencia de Magnesio/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ácido Pirrolidona Carboxílico/administración & dosificación , Estudios Retrospectivos , Factores de Tiempo , Insuficiencia del Tratamiento
2.
Transpl Immunol ; 20(1-2): 83-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18707001

RESUMEN

INTRODUCTION: Magnesium (Mg) plays an essential role in a wide range of fundamental cellular reactions. It has been reported that in rodents Mg-deficient diet-induced hypomagnesemia results in an early inflammation. We have previously shown that chronic severe hypomagnesemia was associated neither with endothelial cell activation nor with an inflammatory process which are crucial in the allograft rejection process. T cell allogeneic stimulation activates the phosphatase calcineurin which triggers the signaling pathways leading to IL-2 synthesis and lymphocyte proliferation. Full activation of calcineurin requires Mg. Surveys suggest that a significant number of people consume less Mg than the international dietary reference intakes leading to hypomagnesemia in 2.5% to 15% of the general population. OBJECTIVE: The aim of the study was to investigate the effects of hypomagnesemia on lymphocyte allogeneic activation and proliferation in a murine model of dietary-induced hypomagnesemia. METHODS: C57BL/6J (H-2(b), Mls(b)) mice were given normal Mg-containing diet (1400 ppm Mg, control mice), or synthetic Mg-deficient diets containing either 50 ppm Mg or 150 ppm Mg for 28 days. Serum Mg levels were determined at days 5, 14 and 28. In parallel, complete urine and faeces were collected by using metabolic cages during a 24 h period for Mg determinations. Splenocytes from C57BL/6 mice fed either normal diet or 50 ppm Mg-diet were used as responder cells in mixed lymphocyte reaction (MLR) performed with splenocytes from C3H/He mice (H-2(k), Mls(IIa)) and C57BL/6 mice fed normal diet as stimulators for allogeneic and isogeneic conditions, respectively. TGF-beta and IL-2 productions were quantified in the supernates of mixed splenocytes cultures. 3x10(6) splenocytes from mice fed 50 ppm Mg-diet were used for calcineurin activity determination at day 28. RESULTS: In mice fed 150 ppm Mg-diet, moderate hypomagnesemia was observed from day 5 to day 28. Oral supplementation with Mg pidolate (5 or 20 mg Mg/kg/day) could not restore normal serum Mg levels. Serum Mg concentration early decreased in mice fed 50 ppm Mg-diet to achieve stabilized severe hypomagnesemia at days 14 and 28. Urine Mg concentration early dramatically fell down then stabilized in mice fed Mg-deficient diets. In MLR performed at day 28 with splenocytes from mice fed 50 ppm Mg-diet, proliferation and IL-2 production in allogeneic conditions were similar to control mice. No TGF-beta production was detected in any group. Lastly, calcineurin activity measured at day 28 was significantly lower in splenocyes from mice fed 50 ppm Mg-diet than in mice fed control diet. CONCLUSION: Mg-deficiency does not alter splenocyte allogeneic activation and proliferation and IL-2 production in vitro, although it partially inhibits calcineurin activity. We hypothesize that the remaining activity is sufficient for IL-2 gene normal activation. Alternatively, Mg-deficiency may trigger other signaling pathways leading to IL-2 production.


Asunto(s)
Activación de Linfocitos/inmunología , Deficiencia de Magnesio/inmunología , Magnesio/metabolismo , Bazo/inmunología , Animales , Calcineurina/inmunología , Dieta , Femenino , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Prueba de Cultivo Mixto de Linfocitos , Magnesio/sangre , Deficiencia de Magnesio/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Bazo/metabolismo , Factor de Crecimiento Transformador beta/inmunología
3.
Biomed Pharmacother ; 62(4): 264-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18400454

RESUMEN

BACKGROUND: Magnesium (Mg) deficiency may lead to serious metabolic, biological and organic dysfunctions, and cause various clinical disorders. In the current study, we explore endothelial cell activation, inflammation and cell death induced in the brain of adult mice by Mg-deficient diet. METHODS AND RESULTS: Neither TNFalpha, substance P, sTNFRI, sTNFRII proteins (ELISA), nor TNFalpha, adherence molecules and prolactin mRNAs, nor NK1R (immunohistochemistry on brain sections) were up-regulated. No inflammatory infiltrates and no apoptotic cells were observed. Using cDNA assay, we showed a neuroprotective, anti-apoptotic and neurotrophic gene expression profile in the brain at early stage of hypomagnesemia. As a model for neuronal injury, mild sound stimulation of Mg-deficient mice without convulsive seizures triggers neither the release of substance P, nor the development of an inflammatory process or cell death in the brain. CONCLUSION: Our results suggest that Mg-deficiency in mice favours the development of a neuroprotective environment in the brain.


Asunto(s)
Encéfalo/metabolismo , Perfilación de la Expresión Génica , Deficiencia de Magnesio/metabolismo , Animales , Apoptosis , Femenino , Inmunohistoquímica , Inflamación/etiología , Molécula 1 de Adhesión Intercelular/análisis , Molécula 1 de Adhesión Intercelular/genética , Interleucina-2/análisis , Magnesio/metabolismo , Ratones , Prolactina/genética , Receptores de Cannabinoides/genética , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores del Factor de Necrosis Tumoral/análisis
4.
Transpl Immunol ; 16(3-4): 200-7, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17138054

RESUMEN

Hypomagnesemia, which is frequently observed in patients treated with calcineurin inhibitors to prevent rejection after allogeneic transplantation, has been associated with a faster rate of decline in allograft function. The effect of hypomagnesemia on lung allograft has not been reported yet. In our model of isolated mouse lung, we have evaluated the early effects of allogeneic lung perfusion with blood from magnesium (Mg)-deficient mice for 3 h on lung activation and remodelling, compared to isogeneic perfusion. Hypomagnesemia (0.21+/-0.07 mmol Mg(2+)/l) was observed in blood from Mg-deficient mice, but no inflammatory pattern. The mRNA level of the intercellular adhesion molecule (ICAM)-1, but neither of the vascular cell adhesion molecule (VCAM)-1, nor of the cytokines tumor necrosis factor (TNF)alpha and interleukin (IL)-2, was enhanced (p<0.05). Although caspase-3 mRNA was transiently enhanced, no apoptotic cells were evidenced in lung tissues even after 3 h. Using cDNA array, we found that the genes encoding RANKL, RANK, TNFR2, NFATX, IL-1R2, IL-6R gp130, SOCS3, PDGFRB, P63, CSF3R, CXCL1, CXCL5, CX3CL1, CSF1, which are involved in inflammation and apoptosis regulation, were markedly up-regulated in allogeneic conditions. Our results support a limited allogeneic activation and an early stage of the inflammatory process in lung, at the time of inflammatory cell recruitment without lung tissue remodelling, as a result of hypomagnesemia. These findings suggest that cyclosporine-related hypomagnesemia, observed in most of the transplanted patients, does not constitute an additional risk for lung allograft outcome.


Asunto(s)
Rechazo de Injerto/etiología , Inflamación/etiología , Trasplante de Pulmón , Pulmón/patología , Deficiencia de Magnesio/complicaciones , Animales , Apoptosis/fisiología , Moléculas de Adhesión Celular/metabolismo , Citocinas/metabolismo , Femenino , Rechazo de Injerto/prevención & control , Inmunohistoquímica , Inmunosupresores/efectos adversos , Trasplante de Pulmón/patología , Deficiencia de Magnesio/inducido químicamente , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Técnicas de Cultivo de Órganos , Perfusión , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Homólogo
5.
Transpl Immunol ; 26(1): 55-61, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21964379

RESUMEN

INTRODUCTION: Tacrolimus (TAC) and cyclosporin (CsA) are commonly responsible for hypomagnesemia that predisposes in turn for hypertension, renal impairment and encephalopathy. OBJECTIVE: The effects of TAC on Mg(2+)-homeostasis and of pre-existing Mg(2+)-deficiency on TAC immunosuppressive activity were compared to CsA in mice. METHODS: Mg(2+) was quantified in plasma, erythrocytes, urine, feces, and femurs from mice treated with TAC 5mg/kg/day. Immunosuppression was assessed in splenocytes by mixed lymphocyte reaction, IL-2 quantification and CN activity determination. RESULTS: Plasma and urine Mg(2+) levels in TAC-treated mice were significantly lower from day 7 until day 21 (p<0.05 versus control) and returned to control value at day 28. Mg(2+) levels were unchanged in erythrocyte, feces and femur. Inhibition of allogeneic proliferation, IL-2 production and CN activity were 68, 56 and 30% lower (p<0.01) after 7 days of TAC-treatment, and 72, 68 and 51% lower (p<0.01) after 7 days of CsA-treatment with a dose of 50mg/kg/day. Dietary-induced hypomagnesemia resulted in significant inhibition of CN activity (p<0.01) without alteration of IL-2 production or allogeneic proliferation. However, it did not alter the effects observed with CsA- or TAC-treatment on allogeneic proliferation, IL-2 production and CN activity. CONCLUSION: By contrast with CsA, long-course TAC-treatment induced an early, but transient, and moderate hypomagnesemia without alteration of bone or erythrocyte stocks, intestinal absorption or renal function. Therefore, in clinical use, TAC should be preferred to CsA in patients with pathological or pharmacological conditions which favor Mg(2+)-deficiency. However, dietary-induced hypomagnesemia did not alter the immunosuppressive effects of TAC and CsA.


Asunto(s)
Inhibidores de la Calcineurina , Ciclosporina/efectos adversos , Inmunosupresores/efectos adversos , Deficiencia de Magnesio/inducido químicamente , Tacrolimus/efectos adversos , Animales , Ciclosporina/administración & dosificación , Suplementos Dietéticos , Eritrocitos/química , Heces/química , Femenino , Fémur/química , Humanos , Terapia de Inmunosupresión/métodos , Inmunosupresores/administración & dosificación , Interleucina-2/biosíntesis , Prueba de Cultivo Mixto de Linfocitos , Magnesio/sangre , Magnesio/orina , Ratones , Ratones Endogámicos C57BL , Bazo/citología , Bazo/efectos de los fármacos , Tacrolimus/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA