Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 552
Filtrar
1.
Am J Transplant ; 17(9): 2312-2325, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28276660

RESUMEN

Despite the introduction of novel and more targeted immunosuppressive drugs, the long-term survival of kidney transplants has not improved satisfactorily. Early antigen-independent intragraft inflammation plays a critical role in the initiation of the alloimmune response and impacts long-term graft function. Complement activation is a key player both in ischemia/reperfusion injury (IRI) as well as in adaptive antigraft immune response after kidney transplantation. Since the alternative pathway (AP) amplifies complement activation regardless of the initiation pathways and renal IR injured cells undergo uncontrolled complement activation, we speculated whether selective blockade of AP could be a strategy for prolonging kidney graft survival. Here we showed that Balb/c kidneys transplanted in factor b deficient C57 mice underwent reduced IRI and diminished T cell-mediated rejection. In in vitro studies, we found that fb deficiency in T cells and dendritic cells conferred intrinsic impaired alloreactive/allostimulatory functions, respectively, both in direct and indirect pathways of alloantigen presentation. By administering anti-fB antibody to C57 wt recipients in the early post Balb/c kidney transplant phases, we documented that inhibition of AP during both ischemia/reperfusion and early adaptive immune response is necessary for prolonging graft survival. These findings may have implication for the use of AP inhibitors in clinical kidney transplantation.


Asunto(s)
Activación de Complemento/inmunología , Factor B del Complemento/deficiencia , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/prevención & control , Linfocitos T/inmunología , Aloinjertos , Animales , Factor B del Complemento/genética , Rechazo de Injerto/etiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/etiología
2.
Gene Ther ; 24(5): 308-313, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28346435

RESUMEN

Ex vivo gene transfer to the graft before transplantation is an attractive option for circumventing systemic side effects of chronic antirejection therapy. Gene delivery of the immunomodulatory protein cytotoxic T-lymphocyte-associated protein 4-immunoglobulin (CTLA4-Ig) prevented chronic kidney rejection in a rat model of allotransplantation without the need for systemic immunosuppression. Here we generated adeno-associated virus type 2 (AAV2) and AAV9 vectors encoding for LEA29Y, an optimized version of CTLA4-Ig. Both LEA29Y vectors were equally efficient for reducing T-cell proliferation in vitro. Serotype 9 was chosen for in vivo experiments owing to a lower frequency of preformed antibodies against the AAV9 capsid in 16 non-human primate tested sera. AAV9-LEA29Y was able to transduce the kidney of non-human primates in an autotransplantation model. Expression of LEA29Y mRNA by renal cells translated into the production of the corresponding protein, which was confined to the graft but not detected in serum. Results in non-human primates represent a step forward in maintaining the portability of this strategy into clinics.


Asunto(s)
Abatacept/genética , Dependovirus/genética , Terapia Genética/métodos , Rechazo de Injerto/terapia , Trasplante de Riñón/efectos adversos , Abatacept/metabolismo , Animales , Línea Celular Tumoral , Vectores Genéticos/genética , Rechazo de Injerto/etiología , Células HEK293 , Humanos , Macaca fascicularis , Masculino , Ratas , Ratas Endogámicas Lew , Linfocitos T/inmunología , Trasplante Autólogo/efectos adversos
3.
Diabetes Obes Metab ; 18(1): 64-71, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26434564

RESUMEN

AIMS: To develop and validate a model to simulate progression of diabetic kidney disease (DKD) from early onset until end-stage renal disease (ESRD), and to assess the effect of renin-angiotensin system (RAS) intervention in early, intermediate and advanced stages of DKD. METHODS: We used data from the BENEDICT, IRMA-2, RENAAL and IDNT trials that assessed effects of RAS intervention in patients with type 2 diabetes. We built a model with discrete disease stages based on albuminuria and estimated glomerular filtration rate (eGFR). Using survival analyses, we assessed the effect of RAS intervention on delaying ESRD in early [eGFR>60 ml/min/1.73 m(2) and albumin:creatinine ratio (ACR) <30 mg/g], intermediate (eGFR 30-60 ml/min/1.73 m(2) or ACR 30-300 mg/g) and advanced (eGFR <30 ml/min/1.73 m(2) or ACR >300 mg/g) stages of DKD for patients in different age groups. RESULTS: For patients at early, intermediate and advanced stage of disease, whose mean age was 60 years and who received placebo, the median time to ESRD was 21.4, 10.8 and 4.7 years, respectively. RAS intervention delayed the predicted time to ESRD by 4.2, 3.6 and 1.4 years, respectively. The benefit of early RAS intervention was more pronounced in younger patients; for example, for patients with a mean age of 45 years, RAS intervention at early, intermediate or advanced stage delayed ESRD by 5.9, 4.0 and 1.1 years versus placebo. CONCLUSIONS: RAS intervention early in the course of proteinuric DKD is more beneficial than late intervention in delaying ESRD.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/tratamiento farmacológico , Fallo Renal Crónico/prevención & control , Tiempo de Tratamiento , Factores de Edad , Anciano , Albúminas/análisis , Albuminuria/complicaciones , Creatinina/análisis , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/patología , Progresión de la Enfermedad , Femenino , Tasa de Filtración Glomerular , Humanos , Fallo Renal Crónico/etiología , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Sistema Renina-Angiotensina/efectos de los fármacos , Factores de Riesgo , Análisis de Supervivencia , Factores de Tiempo
4.
Am J Transplant ; 14(5): 1046-60, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24731002

RESUMEN

Ischemia/reperfusion (I/R) injury is a major determinant of graft survival in kidney transplantation. Survivin, an inhibitor of apoptosis that participates in the control of mitosis and cell cycle progression, has been implicated in renal protection and repair after I/R injury; however, no study has been performed in the transplant setting. We investigated the role of survivin in modulating posttransplant I/R injury in syngeneic and allogeneic kidney grafts, and studied whether protection from I/R injury impacted on the recipient immune system, on chronic allograft nephropathy and rejection. We used genetically engineered mice with survivin haploinsufficiency and WT mice in which survivin over-expression was induced by gene-delivery. Survivin haploinsufficiency in syngeneic grafts was associated with exuberant I/R tissue injury, which triggered inflammation eventually resulting in graft loss. Conversely, survivin over-expression in the grafts minimized I/R injury and dysfunction in syngeneic grafts and in a clinically relevant fully MHC-mismatched allogeneic combination. In the latter, survivin over-expression translated into limited anti-donor adaptive immune response and less long-term allograft injury with protection from renal parenchymal damage. Our data support survivin over-expression in the graft as a novel target for protocols aimed at limiting tissue damage at the time of transplant ultimately modulating the recipient immune system.


Asunto(s)
Rechazo de Injerto/prevención & control , Supervivencia de Injerto/fisiología , Inflamación/prevención & control , Proteínas Inhibidoras de la Apoptosis/fisiología , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/prevención & control , Proteínas Represoras/fisiología , Animales , Apoptosis , Proliferación Celular , Femenino , Técnicas de Transferencia de Gen , Rechazo de Injerto/etiología , Rechazo de Injerto/patología , Inflamación/etiología , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Survivin , Donantes de Tejidos , Trasplante Homólogo
6.
Am J Transplant ; 13(8): 2201-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23731345

RESUMEN

Hemolytic uremic syndrome (HUS) is a disease of microangiopathic hemolytic anemia, thrombocytopenia and acute renal failure. About 90% of cases are secondary to infections by Escherichia coli strains producing Shiga-like toxins (STEC-HUS), while 10% are associated with mutations in genes encoding proteins of complement system (aHUS). We describe two patients with a clinical history of STEC-HUS, who developed end-stage renal disease (ESRD) soon after disease onset. They received a kidney transplant but lost the graft for HUS recurrence, a complication more commonly observed in aHUS. Before planning a second renal transplantation, the two patients underwent genetic screening for aHUS-associated mutations that revealed the presence of a heterozygous CFI mutation in patient #1 and a heterozygous MCP mutation in patient #2, and also in her mother who donated the kidney. This finding argues that the two cases originally diagnosed as STEC-HUS had indeed aHUS triggered by STEC infection on a genetic background of impaired complement regulation. Complement gene sequencing should be performed before kidney transplantation in patients who developed ESRD following STEC-HUS since they may be undiagnosed cases of aHUS, at risk of posttransplant recurrence. Furthermore, genetic analysis of donors is mandatory before living-related transplantation to exclude carriers of HUS-predisposing mutations.


Asunto(s)
Factor I de Complemento/genética , Infecciones por Escherichia coli/complicaciones , Síndrome Hemolítico-Urémico/complicaciones , Fallo Renal Crónico/etiología , Proteína Cofactora de Membrana/genética , Mutación/genética , Adulto , Estudios de Casos y Controles , Cartilla de ADN/química , Cartilla de ADN/genética , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Pruebas Genéticas , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/etiología , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/microbiología , Heterocigoto , Humanos , Fallo Renal Crónico/patología , Fallo Renal Crónico/terapia , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Linaje , Pronóstico , Recurrencia , Factores de Riesgo , Escherichia coli Shiga-Toxigénica , Trombocitopenia/complicaciones , Trombocitopenia/genética , Trombocitopenia/microbiología , Adulto Joven
7.
N Engl J Med ; 368(23): 2169-81, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23738544

RESUMEN

BACKGROUND: Atypical hemolytic-uremic syndrome is a genetic, life-threatening, chronic disease of complement-mediated thrombotic microangiopathy. Plasma exchange or infusion may transiently maintain normal levels of hematologic measures but does not treat the underlying systemic disease. METHODS: We conducted two prospective phase 2 trials in which patients with atypical hemolytic-uremic syndrome who were 12 years of age or older received eculizumab for 26 weeks and during long-term extension phases. Patients with low platelet counts and renal damage (in trial 1) and those with renal damage but no decrease in the platelet count of more than 25% for at least 8 weeks during plasma exchange or infusion (in trial 2) were recruited. The primary end points included a change in the platelet count (in trial 1) and thrombotic microangiopathy event-free status (no decrease in the platelet count of >25%, no plasma exchange or infusion, and no initiation of dialysis) (in trial 2). RESULTS: A total of 37 patients (17 in trial 1 and 20 in trial 2) received eculizumab for a median of 64 and 62 weeks, respectively. Eculizumab resulted in increases in the platelet count; in trial 1, the mean increase in the count from baseline to week 26 was 73×10(9) per liter (P<0.001). In trial 2, 80% of the patients had thrombotic microangiopathy event-free status. Eculizumab was associated with significant improvement in all secondary end points, with continuous, time-dependent increases in the estimated glomerular filtration rate (GFR). In trial 1, dialysis was discontinued in 4 of 5 patients. Earlier intervention with eculizumab was associated with significantly greater improvement in the estimated GFR. Eculizumab was also associated with improvement in health-related quality of life. No cumulative toxicity of therapy or serious infection-related adverse events, including meningococcal infections, were observed through the extension period. CONCLUSIONS: Eculizumab inhibited complement-mediated thrombotic microangiopathy and was associated with significant time-dependent improvement in renal function in patients with atypical hemolytic-uremic syndrome. (Funded by Alexion Pharmaceuticals; C08-002 ClinicalTrials.gov numbers, NCT00844545 [adults] and NCT00844844 [adolescents]; C08-003 ClinicalTrials.gov numbers, NCT00838513 [adults] and NCT00844428 [adolescents]).


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Complemento C5/antagonistas & inhibidores , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Microangiopatías Trombóticas/prevención & control , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacocinética , Terapia Combinada , Femenino , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/terapia , Humanos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/etiología , Masculino , Persona de Mediana Edad , Mutación , Intercambio Plasmático , Recuento de Plaquetas , Calidad de Vida , Adulto Joven
8.
Am J Transplant ; 13(2): 266-74, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23312002

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is a glomerular disease characterized by proteinuria, frequent progression to end-stage renal disease, and recurrence after kidney transplantation in ∼25% of patients, which negatively impacts long-term allograft survival. Experimental studies suggest that abnormalities in T and, possibly, B cells may represent one initial pathogenic trigger, leading to podocyte injury and progressive loss. New data also support the existence of circulating permeability factors able to damage the podocytes, but no single molecule has been consistently identified as the causal pathogenic element in FSGS recurrence. Unfortunately, major progress from mechanistic studies has not translated into substantial advancements in patient treatment, with plasmapheresis (PP) and high doses of cyclosporine (CsA) remaining the mainstays of therapy. Despite consistent experimental and clinical evidence that treatment of proteinuria slows renal function decline in proteinuric nephropathies, maximal use of antiproteinuric agents such as renin angiotensin system antagonists is not routine in the management of FSGS recurrence. More recently, encouraging results have been reported with anti-CD20 depleting antibody rituximab, but further studies are needed to establish its safety/efficacy profile.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Glomeruloesclerosis Focal y Segmentaria/terapia , Trasplante de Riñón/métodos , Nefrología/tendencias , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Biopsia , Ciclosporina/farmacología , Supervivencia de Injerto , Humanos , Nefrología/métodos , Podocitos/citología , Proteinuria/diagnóstico , Ratas , Recurrencia , Sistema Renina-Angiotensina , Rituximab
9.
Cell Transplant ; 22(3): 423-36, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22889699

RESUMEN

Mesenchymal stem cells (MSCs) of bone marrow origin appear to be an attractive candidate for cell-based therapies. However, the major barrier to the effective implementation of MSC-based therapies is the lack of specific homing of exogenously infused cells and overall the inability to drive them to the diseased or damaged tissue. In order to circumvent these limitations, we developed a preconditioning strategy to optimize MSC migration efficiency and potentiate their beneficial effect at the site of injury. Initially, we screened different molecules by using an in vitro injury-migration setting, and subsequently, we evaluated the effectiveness of the different strategies in mice with acute kidney injury (AKI). Our results showed that preconditioning of MSCs with IGF-1 before infusion improved cell migration capacity and restored normal renal function after AKI. The present study demonstrates that promoting migration of MSCs could increase their therapeutic potential and indicates a new therapeutic paradigm for organ repair.


Asunto(s)
Lesión Renal Aguda/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Lesión Renal Aguda/patología , Animales , Células de la Médula Ósea/citología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
10.
Am J Transplant ; 12(9): 2373-83, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22642544

RESUMEN

Multipotent mesenchymal stromal cells (MSC) have recently emerged as promising candidates for cell-based immunotherapy in solid-organ transplantation. However, optimal conditions and settings for fully harnessing MSC tolerogenic properties need to be defined. We recently reported that autologous MSC given posttransplant in kidney transplant patients was associated with transient renal insufficiency associated with intragraft recruitment of neutrophils and complement C3 deposition. Here, we moved back to a murine kidney transplant model with the aim to define the best timing of MSC infusion capable of promoting immune tolerance without negative effects on early graft function. We also investigated the mechanisms of the immunomodulatory and/or proinflammatory activities of MSC according to whether cells were given before or after transplant. Posttransplant MSC infusion in mice caused premature graft dysfunction and failed to prolong graft survival. In this setting, infused MSC localized mainly into the graft and associated with neutrophils and complement C3 deposition. By contrast, pretransplant MSC infusion induced a significant prolongation of kidney graft survival by a Treg-dependent mechanism. MSC-infused pretransplant localized into lymphoid organs where they promoted early expansion of Tregs. Thus, pretransplant MSC infusion may be a useful approach to fully exploit their immunomodulatory properties in kidney transplantation.


Asunto(s)
Trasplante de Riñón/inmunología , Células Madre Mesenquimatosas/inmunología , Animales , Supervivencia de Injerto , Inmunohistoquímica , Inmunoterapia , Ratones , Ratones Endogámicos BALB C
11.
Clin Exp Immunol ; 165(2): 202-10, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21649646

RESUMEN

Dendritic cells (DCs) are the most potent antigen-presenting cells and play a crucial role by modulating the T cell immune response against infective agents, tumour antigens and alloantigens. The current study shows that differentiating bone marrow (BM)-derived DCs but not fully differentiated DCs are targets of erythropoietin (EPO). Indeed, DCs emerging from rat bone marrow, but not splenic DCs, express the EPO receptor (Epo-R) and respond to EPO stimulation displaying a more activated phenotype with increased CD86, CD40 and interleukin (IL)-12 expression levels and a higher allostimulatory capacity on T cells than untreated DCs. Moreover, results here presented show that EPO up-regulates Toll-like receptor (TLR)-4 in differentiating DCs rendering these cells more sensitive to stimulation by the TLR-4 ligand lipopolysaccharide (LPS). Indeed, DCs treated with EPO and then stimulated by LPS were strongly allostimulatory and expressed CCR7, CD86, CD40, IL-12 and IL-23 at higher levels than those observed in DCs stimulated with LPS alone. It is tempting to speculate that EPO could act as an additional danger signal in concert with TLR-4 engagement. Thus, EPO, beyond its erythropoietic and cytoprotective effects, turns out to be an immune modulator.


Asunto(s)
Células Dendríticas/inmunología , Eritropoyetina/análogos & derivados , Factores Inmunológicos , Animales , Antígeno B7-2/biosíntesis , Western Blotting , Células de la Médula Ósea/metabolismo , Antígenos CD40/biosíntesis , Diferenciación Celular , Darbepoetina alfa , Células Dendríticas/metabolismo , Eritropoyetina/inmunología , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Interleucina-12/biosíntesis , Interleucina-23/biosíntesis , Lipopolisacáridos/inmunología , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas BN , Receptores CCR7/biosíntesis , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética
13.
Am J Transplant ; 10(7): 1517-23, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20642678

RESUMEN

Thrombotic microangiopathy (TMA) is a severe complication of kidney transplantation that often causes graft failure. TMA may occur de novo, often triggered by immunosuppressive drugs and acute antibody-mediated rejection, or recur in patients with previous history of hemolytic uremic syndrome (HUS). Recurrent TMA is very rare in patients who had developed end-stage renal failure following HUS caused by Shiga-toxin producing E. scherichia coli, whereas disease recurrence is common in patients with atypical HUS (aHUS). The underlying genetic defect greatly impacts the risk of posttransplant recurrence in aHUS. Indeed recurrence is almost the rule in patients with mutations in genes encoding factor H or factor I, whereas patients with a mutation in membrane-cofactor-protein gene have a good transplant outcome. Prophylactic and therapeutic options for posttransplant TMA, including plasma therapy, combined kidney and liver transplantation and targeted complement inhibitors are discussed in this review.


Asunto(s)
Trasplante de Riñón/efectos adversos , Microangiopatías Trombóticas/epidemiología , Autoanticuerpos/genética , Transfusión de Componentes Sanguíneos , Niño , Factor H de Complemento/genética , Vía Alternativa del Complemento/genética , Proteínas del Sistema Complemento/genética , Infecciones por Escherichia coli/epidemiología , Fibrinógeno/genética , Síndrome Hemolítico-Urémico/epidemiología , Síndrome Hemolítico-Urémico/genética , Humanos , Trasplante de Hígado/efectos adversos , Mutación , Plasma , Complicaciones Posoperatorias/epidemiología , Recurrencia , Insuficiencia del Tratamiento , Resultado del Tratamiento
16.
Gene Ther ; 16(11): 1373-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19675594

RESUMEN

ADAMTS13 is a plasma metalloprotease that regulates the size of the von Willebrand factor (VWF) multimers. Genetic or acquired deficiency of ADAMTS13 causes thrombotic thrombocytopenic purpura (TTP) in humans. Plasma infusion is the treatment of choice for patients with congenital ADAMTS13 deficiency. However, this practice exposes patients to the risk of infections, allergies and fluid volume overload. The search for alternative treatments is required. Here, we tested the ability of systemically administered adenovirus encoding human ADAMTS13 to restore the deficient protein in the circulation of Adamts13(-/-) mice. Injection of the adenovirus efficiently transduced the liver, kidney, lung, heart and spleen, resulting in the secretion of ADAMTS13 into plasma. A reduced area of thrombi was observed when blood from Ad-ADAMTS13-treated mice was perfused over a collagen-coated surface in a parallel plate flow chamber compared with blood of Ad-betaGal-treated controls. The secreted ADAMTS13 protein was functionally active even after 2 months from injection. The data provide the proof of principle for developing a novel therapy for the correction of ADAMTS13 deficiency in patients with hereditary TTP.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos , Metaloendopeptidasas/genética , Púrpura Trombocitopénica Trombótica/terapia , Proteína ADAMTS13 , Animales , Metaloendopeptidasas/sangre , Metaloendopeptidasas/deficiencia , Ratones , Ratones Noqueados , Púrpura Trombocitopénica Trombótica/sangre , Transducción Genética
17.
Am J Nephrol ; 30(3): 280-6, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19521070

RESUMEN

BACKGROUND: Patients with diabetic nephropathy are at high risk for further progressive renal function loss. Treatments that decrease albuminuria have been linked with renal and cardiovascular protection. However, even when taking optimal treatment, residual renal and cardiovascular risk remains high which correlates with the magnitude of residual albuminuria. Use of vitamin D receptor activators, such as calcitriol and paricalcitol, is associated with improved sur- vival. A small study with paricalcitol showed reductions in albuminuria. The VITAL study tests the hypothesis whether paricalcitol persistently reduces albuminuria in diabetic subjects already receiving angiotensin-converting enzyme inhibitor (ACEI) and/or angiotensin receptor blocker (ARB) therapy. METHODS: Randomization in this double-blind trial is equal allocation to paricalcitol 1 micro/day, 2 microg/day, or placebo. Inclusion criteria include: a diagnosis of type 2 diabetes, urinary albumin/creatinine ratio (UACR) between 100-3,000 mg/g, estimated glomerular filtration rate (eGFR) between 15-90 ml/min/1.73 m(2), serum calcium <9.8 mg/dl, and parathyroid hormone (PTH) between 35-500 pg/ml. RESULTS: Baseline characteristics of the 281 subjects are: 69% men, mean age 64.9 +/- 10.4 years, eGFR 40.7 +/- 16.7 ml/min, median UACR (interquartile range) 612.3 mg/g (281-1,181 mg/g) and PTH 98.4 +/- 63.8 pg/ml. CONCLUSION: This trial will be the first clinical test of the hypothesis that paricalcitol possesses pleiotropic effects and can modulate albuminuria in the setting of ACEI and/or ARB therapy. Results will have important clinical implications and are expected in November 2009.


Asunto(s)
Albuminuria/tratamiento farmacológico , Ergocalciferoles/uso terapéutico , Receptores de Calcitriol/efectos de los fármacos , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad
18.
Transplant Proc ; 41(5): 1797-800, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19545731

RESUMEN

Type 1 diabetes is associated with a progressive loss of beta cells and pancreatic islet transplantation could represent a cure for this disease. Herein we explored whether transplantation of bone marrow-derived mesenchymal stem cells (MSCs) allowed a reduced number of pancreatic islets to improve glycemic control in diabetic rats, by promoting islet vascularization. We transplanted 2000 syngenic islets alone or in combination with MSCs (10(6) cells) under the kidney capsules of diabetic Lewis rats. Animals transplanted with 2000 islets never reached normoglycemia. In contrast, rats transplanted with 2000 islets plus MSCs, showed a gradual fall in glycemia after transplantation, with normoglycemia maintained until killing. Comparable glycemic control was obtained with transplantation of 3000 islets alone. The MSC preparation used for in vivo experiments expressed high levels of vascular endothelial growth factor (VEGF(165)) and, at less extent, VEGF(189), as evaluated by reverse transcriptase polymerase chain reaction (RT-PCR). In transplanted animals, vascularization was quantified by morphometric analysis of islet grafts with anti-RECA and anti-insulin antibodies. MSCs were stained with PKH-26. Mean capillary density was 1002 +/- 55 capillaries/mm(2) in islets transplanted alone. Co-infusion of MSCs with islets significantly increased the number of capillaries to 1459 +/- 66 capillaries/mm(2). In conclusion, our study indicated that co-transplantation of MSCs with pancreatic islets improved islet graft function by promoting graft vascularization.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos/métodos , Trasplante de Islotes Pancreáticos/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Cartilla de ADN , Masculino , Neovascularización Fisiológica , Ratas , Ratas Endogámicas Lew , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayo de Capsula Subrrenal , Transcripción Genética
19.
Int J Artif Organs ; 32(1): 12-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19241359

RESUMEN

OBJECTIVES: Daily nocturnal hemodialysis (NHD) has been proposed as a valuable strategy to improve outcomes for patients on conventional hemodialysis (CHD), but it is burdened by high costs and logistic issues. Thrice NHD might represent a more affordable approach to improve hemodialysis patient outcome. METHODS: Here we retrospectively analyzed the data on blood pressure, body weight, and hematochemical parameters in a cohort of 7 patients (mean age 50.4-/+11.0 years, duration of CHD 14.3-/+11.5 years) who registered in the NHD program at the dialysis unit of Ospedali Riuniti, Bergamo, Italy. Data for the 2 first years of NHD were compared with those of the last year on CHD. RESULTS: At 2 years after start of NHD, we found a significant decrease in systolic (149.4-/+16.6 vs. 128.4+/-26.0 mm Hg, p<0.001) and diastolic (87.7-/+11.1 vs. 79.6-/+16.7 mm Hg, p<0.05) blood pressure, along with a significant reduction in the use of per-patient antihypertensive agents (1.17-/+1.19 vs. 0.47-/+0.89, p<0.05 and an increase in dry body weight (61.4-/+21.8 vs. 67.1-/+16.4 kg, p<0.001). Moreover, patients had a significant reduction in phosphate levels (6.2-/+2.4 vs. 5.4-/+3.0 mg/dL, p<0.01). The procedure was safe and well tolerated and did not require extra cost for ad hoc facilities. CONCLUSION: NHD is an effective approach to optimize chronic dialysis therapy.


Asunto(s)
Fallo Renal Crónico/terapia , Diálisis Renal/métodos , Adulto , Antihipertensivos/uso terapéutico , Biomarcadores/sangre , Presión Sanguínea/efectos de los fármacos , Peso Corporal , Calcio/sangre , Femenino , Unidades de Hemodiálisis en Hospital , Humanos , Italia , Fallo Renal Crónico/sangre , Fallo Renal Crónico/fisiopatología , Masculino , Persona de Mediana Edad , Fosfatos/sangre , Evaluación de Programas y Proyectos de Salud , Diálisis Renal/efectos adversos , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento
20.
Clin Exp Immunol ; 151(2): 199-209, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18070148

RESUMEN

Genetic and acquired abnormalities in complement factor H (CFH) have been associated with two different human renal diseases: haemolytic uraemic syndrome and membrano proliferative glomerulonephritis. The new genetic and pathogenetic findings in these diseases and their clinical implications for the management and cure of patients are reviewed in this paper.


Asunto(s)
Glomerulonefritis Membranoproliferativa/genética , Síndrome Hemolítico-Urémico/genética , Factor H de Complemento/genética , Glomerulonefritis Membranoproliferativa/inmunología , Glomerulonefritis Membranoproliferativa/terapia , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/terapia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA