Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 314(5): F881-F892, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357437

RESUMEN

Renal ischemia-reperfusion injury (IRI) is a severe complication of major surgery and a risk factor for increased morbidity and mortality. Here, we investigated mechanisms that might contribute to IRI-induced progression to chronic kidney disease (CKD). Acute kidney injury (AKI) was induced by unilateral IRI for 35 min in CD1 and C57BL/6 (B6) mice. Unilateral IRI was used to overcome early mortality. Renal morphology, NGAL upregulation, and neutrophil infiltration as well as peritubular capillary density were studied by immunohistochemistry. The composition of leukocyte infiltrates in the kidney after IRI was investigated by flow cytometry. Systemic blood pressure was measured with a tail cuff, and renal perfusion was quantified by functional magnetic resonance imaging (fMRI). Mesangial matrix expansion was assessed by silver staining. Following IRI, CD1 and B6 mice developed similar morphological signs of AKI and increases in NGAL expression, but neutrophil infiltration was greater in CD1 than B6 mice. IRI induced an increase in systemic blood pressure of 20 mmHg in CD1, but not in B6 mice; and CD1 mice also had a greater loss of renal perfusion and kidney volume than B6 mice ( P < 0.05). CD1 mice developed substantial interstitial fibrosis and decreased peritubular capillary (PTC) density by day 14 while B6 mice showed only mild renal scarring and almost normal PTC. Our results show that after IRI, CD1 mice develop more inflammation, hypertension, and later mesangial matrix expansion than B6 mice do. Subsequently, CD1 animals suffer from CKD due to impaired renal perfusion and pronounced permanent loss of peritubular capillaries.


Asunto(s)
Lesión Renal Aguda/complicaciones , Hipertensión/etiología , Riñón/irrigación sanguínea , Circulación Renal , Insuficiencia Renal Crónica/etiología , Daño por Reperfusión/complicaciones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Presión Sanguínea , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Mesangio Glomerular/patología , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/fisiopatología , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Lipocalina 2/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Infiltración Neutrófila , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Factores de Tiempo
2.
Kidney Int ; 94(4): 741-755, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29935951

RESUMEN

Severe ischemia reperfusion injury (IRI) results in rapid complement activation, acute kidney injury and progressive renal fibrosis. Little is known about the roles of the C5aR1 and C5aR2 complement receptors in IRI. In this study C5aR1-/- and C5aR2-/- mice were compared to the wild type in a renal IRI model leading to renal fibrosis. C5a receptor expression, kidney morphology, inflammation, and fibrosis were measured in different mouse strains one, seven and 21 days after IRI. Renal perfusion was evaluated by functional magnetic resonance imaging. Protein abundance and phosphorylation were assessed with high content antibody microarrays and Western blotting. C5aR1 and C5aR2 were increased in damaged tubuli and even more in infiltrating leukocytes after IRI in kidneys of wild-type mice. C5aR1-/- and C5aR2-/- animals developed less IRI-induced inflammation and showed better renal perfusion than wild-type mice following IRI. C5aR2-/- mice, in particular, had enhanced tubular and capillary regeneration with less renal fibrosis. Anti-inflammatory IL-10 and the survival/growth kinase AKT levels were especially high in kidneys of C5aR2-/- mice following IRI. LPS caused bone marrow-derived macrophages from C5aR2-/- mice to release IL-10 and to express the stress response enzyme heme oxygenase-1. Thus, C5aR1 and C5aR2 have overlapping actions in which the kidneys of C5aR2-/- mice regenerate better than those in C5aR1-/- mice following IRI. This is mediated, at least in part, by differential production of IL-10, heme oxygenase-1 and AKT.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Interleucina-10/metabolismo , Túbulos Renales/patología , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Anafilatoxina C5a/genética , Daño por Reperfusión/genética , Animales , Proliferación Celular/genética , Células Cultivadas , Células Epiteliales , Fibrosis , Inflamación/etiología , Riñón/diagnóstico por imagen , Túbulos Renales/metabolismo , Túbulos Renales/fisiopatología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Imagen de Perfusión , Fosforilación , Factores Protectores , Receptor de Anafilatoxina C5a/metabolismo , Regeneración/genética , Daño por Reperfusión/complicaciones , Regulación hacia Arriba
3.
J Am Soc Nephrol ; 26(11): 2860-70, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25788529

RESUMEN

ANCA-associated vasculitides are characterized by inflammatory destruction of small vessels accompanied by enhanced cleavage of membrane-bound proteins. One of the main proteases responsible for ectodomain shedding is disintegrin and metalloproteinase domain-containing protein 17 (ADAM17). Given its potential role in aggravating vascular dysfunction, we examined the role of ADAM17 in active proteinase-3 (PR3)-positive ANCA-associated vasculitis (AAV). ADAM17 concentration was significantly increased in plasma samples from patients with active PR3-AAV compared with samples from patients in remission or from other controls with renal nonvascular diseases. Comparably, plasma levels of the ADAM17 substrate syndecan-1 were significantly enhanced in active AAV. We also observed that plasma-derived ADAM17 retained its specific proteolytic activity and was partly located on extracellular microparticles. Transcript levels of ADAM17 were increased in blood samples of patients with active AAV, but those of ADAM10 or tissue inhibitor of metalloproteinases 3, which inhibits ADAMs, were not. We also performed a microRNA (miR) screen and identified miR-634 as significantly upregulated in blood samples from patients with active AAV. In vitro, miR-634 mimics induced a proinflammatory phenotype in monocyte-derived macrophages, with enhanced expression and release of ADAM17 and IL-6. These data suggest that ADAM17 has a prominent role in AAV and might account for the vascular complications associated with this disease.


Asunto(s)
Proteínas ADAM/sangre , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/sangre , Interleucina-6/sangre , MicroARNs/sangre , Mieloblastina/inmunología , Proteína ADAM17 , Adulto , Anciano , Enfermedades Cardiovasculares/sangre , Células Cultivadas , Citocinas/sangre , Endotelio Vascular/fisiología , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Inmunoensayo , Inflamación , Macrófagos/citología , Macrófagos/metabolismo , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Mieloblastina/sangre , Fenotipo
4.
Am J Physiol Renal Physiol ; 307(6): F718-26, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25056349

RESUMEN

Acute kidney injury (AKI) increases the risk of morbidity and mortality after major surgery and transplantation. We investigated the effect of PKC-ε deficiency on AKI and ischemic allograft damage after kidney transplantation. PKC-ε-deficient and wild type (WT) control mice were subjected to 35 min of renal pedicle clamping to induce AKI. PKC-ε deficiency was associated with a marked improvement in survival and an attenuated loss of kidney function. Furthermore, functional MRI experiments revealed better renal perfusion in PKC-ε-deficient mice than in WT mice one day after IRI. Acute tubular necrosis and neutrophil infiltration were markedly reduced in PKC-ε-deficient mice. To determine whether this resistance to ischemia-reperfusion injury resulted from changes in local renal cells or infiltrating leukocytes, we studied a life-supporting renal transplant model of ischemic graft injury. We transplanted kidneys from H(2b) PKC-ε-deficient mice (129/SV) and their corresponding WT littermates into major histocompatibility complex-incompatible H(2d) recipients (BALB/c) and induced ischemic graft injury by prolonged cold ischemia time. Recipients of WT allografts developed severe renal failure and died within 10 days of transplantation. Recipients of PKC-ε-deficient allografts had better renal function and survival; they had less generation of ROS and upregulation of proinflammatory proteins (i.e., ICAM-1, inducible nitric oxide synthase, and TNF-α) and showed less tubular epithelial cell apoptosis and inflammation in their allografts. These data suggest that local renal PKC-ε expression mediates proapoptotic and proinflammatory signaling and that an inhibitor of PKC-ε signaling could be used to prevent hypoxia-induced AKI.


Asunto(s)
Lesión Renal Aguda/enzimología , Proteína Quinasa C-epsilon/metabolismo , Daño por Reperfusión/enzimología , Aloinjertos/enzimología , Animales , Apoptosis , Supervivencia de Injerto , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Pruebas de Función Renal , Trasplante de Riñón , Masculino , Ratones , Ratones Endogámicos BALB C , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Crit Care Med ; 42(10): e654-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25083983

RESUMEN

OBJECTIVE: Angiopoietin-2, a protein secreted by stimulated endothelium and an antagonist of the endothelium-stabilizing receptor Tie2, contributes to the pathophysiology of septic multiple organ dysfunction. We tested the therapeutic potential of a pulmonary-endothelium-specific RNA interference-based angiopoietin-2 targeting strategy in sepsis. DESIGN: Laboratory and animal research. SETTINGS: Research laboratories of the Medical School Hannover, Department of Nephrology and Hypertension, Hannover and Silence Therapeutics GmbH, Berlin. SUBJECTS: C57Bl/6 mice. INTERVENTIONS: Lung-endothelium-specific angiopoietin-2 small interfering RNA was administered both before and after sepsis induction (cecal ligation and puncture or lipopolysaccharides) intravenously. MEASUREMENTS AND MAIN RESULTS: Angiopoietin-2 small interfering RNA was highly specific and reduced angiopoietin-2 expression in the septic murine lungs up to 73.8% (p = 0.01) and enhanced the phosphorylation of Tie2 both in control and septic animals. Angiopoietin-2 small interfering RNA reduced pulmonary interleukin-6 transcription, intercellular adhesion molecule expression, neutrophil infiltration, and vascular leakage. Manifestations of sepsis were also attenuated in distant organs, including the kidney, where renal function was improved without affecting local angiopoietin-2 production. Finally, angiopoietin-2 small interfering RNA ameliorated the severity of illness and improved survival in cecal ligation and puncture, both as a pretreatment and as a rescue intervention. CONCLUSION: The Tie2 antagonist angiopoietin-2 represents a promising target against sepsis-associated multiple organ dysfunction. A novel RNA interference therapeutic approach targeting gene expression in the pulmonary endothelium could be a clinically relevant pharmacological strategy to reduce injurious angiopoietin-2 synthesis.


Asunto(s)
Angiopoyetina 2/fisiología , Pulmón/metabolismo , Insuficiencia Multiorgánica/etiología , Interferencia de ARN/fisiología , Sepsis/complicaciones , Angiopoyetina 2/metabolismo , Animales , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Insuficiencia Multiorgánica/metabolismo , Insuficiencia Multiorgánica/fisiopatología , ARN Interferente Pequeño/metabolismo , Receptor TIE-2/metabolismo , Sepsis/metabolismo , Sepsis/mortalidad , Sepsis/fisiopatología
6.
J Am Soc Nephrol ; 22(3): 484-95, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21355054

RESUMEN

CD4+ T cells contribute to the pathogenesis of ischemia-reperfusion injury, which is the primary cause of delayed graft failure after kidney transplantation. The TIM-1:TIM-4 pathway participates in the activation/differentiation of CD4+ T cells, suggesting that it may modulate ischemia-reperfusion injury. Here, we studied the role of TIM-1 in a murine uninephrectomized renal ischemia-reperfusion injury model. Blocking the TIM-1:TIM-4 pathway with an antagonistic monoclonal antibody protected renal function and diminished reperfusion injury resulting from 30 minutes of ischemia. Histologic examination showed significantly less evidence of renal damage as evidenced by diminished tubular necrosis, preservation of the brush border, fewer cast formations, and less tubular dilation. Blocking TIM-1 also reduced the number of apoptotic cells and diminished local inflammation within ischemic kidneys, the latter shown by decreased recruitment of macrophages, neutrophils, and CD4+ T cells and by reduced local production of proinflammatory cytokines. Furthermore, TIM-1 blockade significantly improved survival after ischemia-reperfusion injury. Taken together, these data suggest that the TIM-1:TIM-4 pathway enhances injury after renal ischemia-reperfusion injury and may be a therapeutic target.


Asunto(s)
Riñón/irrigación sanguínea , Proteínas de la Membrana/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Receptor Celular 1 del Virus de la Hepatitis A , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Riñón/patología , Riñón/cirugía , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Necrosis , Nefrectomía , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control
7.
J Cell Mol Med ; 14(7): 1922-34, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19508384

RESUMEN

Rapid apoptotic cell engulfment is crucial for prevention of inflammation and autoimmune diseases and is conducted by special immunocompetent cells like macrophages or immature dendritic cells. We recently demonstrated that endothelial cells (ECs) also participate in apoptotic cell clearance. However, in contrast to conventional phagocytes they respond with an inflammatory phenotype. To further confirm these pro-inflammatory responses human ECs were exposed to apoptotic murine ECs and changes in thrombospondin-1 (TSP-1) expression and in activation of intracellular signalling cascades were determined by real-time qPCR, immunoblotting and immunocytochemistry. Human primary macrophages or monocytic lymphoma cells (U937) were incubated with conditioned supernatant of human ECs exposed to apoptotic cells and changes in activation, migration and phagocytosis were monitored. Finally, plasma levels of TSP-1 in patients with anti-neutrophil cytoplasmic antibody(ANCA)-associated vasculitis (AAV) were determined by ELISA. We provided evidence that apoptotic cells induce enhanced expression of TSP-1 in human ECs and that this increase in TSP-1 is mediated by the mitogen-activated protein kinases (MAPK) ERK1 and 2 and their upstream regulators MEK and B-Raf. We also showed that plasma TSP-1 levels are increased in patients with AAV. Finally, we showed that conditioned supernatant of ECs exposed to apoptotic cells induces pro-inflammatory responses in monocytes or U937 cells and demonstrated that increased TSP-1 expression enhances migration and facilitates engulfment of apoptotic cells by monocyte-derived macrophages or U937 cells. These findings suggest that under pathological conditions with high numbers of uncleared dying cells in the circulation endothelial-derived elevated TSP-1 level may serve as an attraction signal for phagocytes promoting enhanced recognition and clearance of apoptotic cells.


Asunto(s)
Apoptosis , Endotelio/fisiología , Macrófagos/fisiología , Trombospondina 1/fisiología , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Humanos , Inmunohistoquímica , Macrófagos/citología , Ratones , Reacción en Cadena de la Polimerasa
8.
BMC Cell Biol ; 11: 68, 2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20822540

RESUMEN

BACKGROUND: Endothelial tight and adherens junctions control a variety of physiological processes like adhesion, paracellular transport of solutes or trafficking of activated leukocytes. Formation and maintenance of endothelial junctions largely depend on the microenvironment of the specific vascular bed and on interactions of the endothelium with adjacent cell types. Consequently, primary cultures of endothelial cells often lose their specific junctional pattern and fail to establish tight monolayer in vitro. This is also true for endothelial cells isolated from the vein of human umbilical cords (HUVEC) which are widely used as model for endothelial cell-related studies. RESULTS: We here compared the effect of cyclic 3'-5'-adenosine monophosphate (cAMP) and its derivates on formation and stabilization of tight junctions and on alterations in paracellular permeability in HUVEC. We demonstrated by light and confocal laser microscopy that for shorter time periods the sodium salt of 8-bromoadenosine-cAMP (8-Br-cAMP/Na) and for longer incubation periods 8-(4-chlorophenylthio)-cAMP (pCPT-cAMP) exerted the greatest effects of all compounds tested here on formation of continuous tight junction strands in HUVEC. We further demonstrated that although all compounds induced protein kinase A-dependent expression of the tight junction proteins claudin-5 and occludin only pCPT-cAMP slightly enhanced paracellular barrier functions. Moreover, we showed that pCPT-cAMP and 8-Br-cAMP/Na induced expression and membrane translocation of tricellulin. CONCLUSIONS: pCPT-cAMP and, to a lesser extend, 8-Br-cAMP/Na improved formation of continuous tight junction strands and decreased paracellular permeability in primary HUVEC. We concluded that under these conditions HUVEC represent a feasible in vitro model to study formation and disassembly of endothelial tight junctions and to characterize tight junction-associated proteins.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/farmacología , AMP Cíclico/farmacología , Endotelio Vascular/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Uniones Estrechas/efectos de los fármacos , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Claudina-5 , AMP Cíclico/análogos & derivados , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Proteína 2 con Dominio MARVEL , Proteínas de la Membrana/genética , Microscopía Confocal , Ocludina , Transporte de Proteínas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Venas Umbilicales/patología , Regulación hacia Arriba
9.
Kidney Int ; 78(12): 1252-62, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20827258

RESUMEN

Early and accurate detection of acute kidney injury (AKI) is needed to prevent the progression to chronic kidney disease and to improve outcome. Here we used capillary electrophoresis-mass spectrometry to identify urinary peptides predictive of AKI in a training set of 87 urine samples longitudinally collected from patients in an intensive care unit. Within this patient cohort, 16 developed AKI while 14 maintained normal renal function. The sequence of twenty peptides significantly associated with AKI was identified. They were found to be degradation products of six proteins. These formed a diagnostic pattern. Peptides of albumin, α-1-antitrypsin, and ß-2-microglobulin were upregulated but fragments of fibrinogen α and collagens 1 α(I) and 1 α(III) were downregulated in AKI. After cross-validation of the training set, a good diagnostic performance of the marker pattern was found with an area under the ROC curve of 0.91. This was confirmed in a blinded validation set of 20 patients in the intensive care unit and 31 allogeneic hematopoietic stem cell transplantation patients, of which 13 had and 18 had not experienced an episode of AKI. In comparison to more established markers of AKI such as serum cystatin C and urinary kidney injury molecule-1, interleukin-18, and neutrophil gelatinase associated-lipocalin, the proteomic marker pattern was found to be of superior prognostic value, detecting AKI up to 5 days in advance of the rise in serum creatinine.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/orina , Enfermedad Crítica , Péptidos/orina , Proteómica , Adulto , Anciano , Anciano de 80 o más Años , Albúminas/metabolismo , Biomarcadores/orina , Estudios de Cohortes , Colágeno Tipo I/orina , Femenino , Fibrinógeno/orina , Humanos , Unidades de Cuidados Intensivos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pronóstico , Sensibilidad y Especificidad , alfa 1-Antitripsina/orina , Microglobulina beta-2/orina
10.
J Immunol ; 181(2): 1179-89, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606671

RESUMEN

Central mechanisms leading to ischemia induced allograft rejection are apoptosis and inflammation, processes highly regulated by the urokinase-type plasminogen activator (uPA) and its specific receptor (uPAR). Recently, up-regulation of uPA and uPAR has been shown to correlate with allograft rejection in human biopsies. However, the causal connection of uPA/uPAR in mediating transplant rejection and underlying molecular mechanisms remain poorly understood. In this study, we evaluated the role of uPA/uPAR in a mice model for kidney ischemia reperfusion (IR) injury and for acute kidney allograft rejection. uPAR but not uPA deficiency protected from IR injury. In the allogenic kidney transplant model, uPAR but not uPA deficiency of the allograft caused superior recipient survival and strongly attenuated loss of renal function. uPAR-deficient allografts showed reduced generation of reactive oxygen species and apoptosis. Moreover, neutrophil and monocyte/macrophage infiltration was strongly attenuated and up-regulation of the adhesion molecule ICAM-1 was completely abrogated in uPAR-deficient allografts. Inadequate ICAM-1 up-regulation in uPAR(-/-) primary aortic endothelial cells after C5a and TNF-alpha stimulation was confirmed by in vitro experiments. Our results demonstrate that the local renal uPAR plays an important role in the apoptotic and inflammatory responses mediating IR-injury and transplant rejection.


Asunto(s)
Células Endoteliales/inmunología , Rechazo de Injerto , Trasplante de Riñón/inmunología , Riñón/inmunología , Receptores de Superficie Celular/metabolismo , Daño por Reperfusión/metabolismo , Animales , Apoptosis , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Supervivencia de Injerto , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Riñón/irrigación sanguínea , Riñón/citología , Riñón/metabolismo , Masculino , Ratones , Infiltración Neutrófila , Especies Reactivas de Oxígeno/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Daño por Reperfusión/inmunología , Trasplante Homólogo , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/deficiencia , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
11.
Nephrol Dial Transplant ; 24(8): 2567-75, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19398767

RESUMEN

BACKGROUND: Interstitial fibrosis and tubular atrophy (IF/TA) in renal transplants are the major morphological correlates of progressive graft deterioration. Early diagnosis of IF/TA is a pre-requisite for a timely therapeutic intervention in patients at risk. To evaluate events occurring before the overt onset of IF/TA, gene expression profiling of 3-month protocol biopsies from patients with IF/TA was performed in a patient group (n = 8) who developed mild IF/TA [chronic allograft nephropathy (CAN) grade I, by the Banff scoring system] in the subsequent 6-month protocol biopsy ('progressors'), and in 12 patients without IF/TA at 6 months ('non-progressors'). METHODS: RNA was extracted, labelled and hybridized to human specific genome wide DNA microarrays. Normalized data were subjected to gene-centric and pathway-centric statistical methods. RESULTS: Compared to the non-progressors, the 3-month biopsies of the progressor group showed overexpression of several genes that are important in the T- and B-cell activation and immune response. Genes involved in pro-fibrotic processes were identified in the biopsies of the progressors that preceded the observed IF/TA at 6 months. Furthermore, several genes with transporter and metabolic functions were underrepresented in the progressors in the 3-month biopsies. CONCLUSION: Gene expression profiling of early protocol biopsies identified changes in the transcriptome of grafts, which may be important for the development of IF/TA. Such early detection of transcriptome changes can facilitate the identification of patients at risk shifting the intervention time point well before the histological diagnosis of irreversible IF/TA.


Asunto(s)
Atrofia/genética , Fibrosis/genética , Perfilación de la Expresión Génica , Rechazo de Injerto/genética , Trasplante de Riñón , Túbulos Renales/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Atrofia/metabolismo , Atrofia/patología , Biomarcadores/metabolismo , Biopsia , Niño , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Genoma Humano , Rechazo de Injerto/metabolismo , Humanos , Técnicas para Inmunoenzimas , Túbulos Renales/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Trasplante Homólogo , Adulto Joven
12.
Diabetes ; 56(2): 346-54, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17259378

RESUMEN

The protein kinase C (PKC)-beta isoform has been implicated to play a pivotal role in the development of diabetic kidney disease. We tested this hypothesis by inducing diabetic nephropathy in PKC-beta-deficient (PKC-beta(-/-)) mice. We studied nondiabetic and streptozotocin-induced diabetic PKC-beta(-/-) mice compared with appropriate 129/SV wild-type mice. After 8 weeks of diabetes, the high-glucose-induced renal and glomerular hypertrophy, as well as the increased expression of extracellular matrix proteins such as collagen and fibronectin, was reduced in PKC-beta(-/-) mice. Furthermore, the high-glucose-induced expression of the profibrotic cytokine transforming growth factor (TGF)-beta1 and connective tissue growth factor were significantly diminished in the diabetic PKC-beta(-/-) mice compared with diabetic wild-type mice, suggesting a role of the PKC-beta isoform in the regulation of renal hypertrophy. Notably, increased urinary albumin-to-creatinine ratio persisted in the diabetic PKC-beta(-/-) mice. The loss of the basement membrane proteoglycan perlecan and the podocyte protein nephrin in the diabetic state was not prevented in the PKC-beta(-/-) mice as previously demonstrated in the nonalbuminuric diabetic PKC-alpha(-/-) mice. In summary, the differential effects of PKC-beta deficiency on diabetes-induced renal hypertrophy and albuminuria suggest that PKC-beta contributes to high-glucose-induced TGF-beta1 expression and renal fibrosis, whereas perlecan, as well as nephrin, expression and albuminuria is regulated by other signaling pathways.


Asunto(s)
Albuminuria/genética , Nefropatías Diabéticas/enzimología , Riñón/patología , Proteína Quinasa C/deficiencia , Albuminuria/prevención & control , Animales , Deleción Cromosómica , Colágeno Tipo IV/metabolismo , Creatinina/metabolismo , Diabetes Mellitus Experimental , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Fibronectinas/metabolismo , Fibrosis , Proteoglicanos de Heparán Sulfato/metabolismo , Hipertrofia , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Tamaño de los Órganos , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Proteína Quinasa C/genética , ARN/metabolismo , Estreptozocina , Factor de Crecimiento Transformador beta1/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Am J Obstet Gynecol ; 198(3): 317.e1-5, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18068139

RESUMEN

OBJECTIVE: Preeclampsia is a disorder of endothelial cells, and novel markers of the disease are eagerly awaited. We tested the hypothesis that circulating endothelial cells (CECs) are elevated in preeclampsia and that cell numbers correlate with disease activity. STUDY DESIGN: CECs were measured in 10 patients with preeclampsia as well as pregnant and nonpregnant controls. Cells were enumerated prior to delivery, 1 and 3-5 days thereafter. Enumeration of CECs was performed with anti-CD 146-driven immunomagnetic isolation and subsequent Ulex lectin staining. RESULTS: Markedly elevated CEC numbers were detected in women with preeclampsia (median 88 cells/mL; P < .001) when compared with normal pregnancies (median 16 cells/mL) and healthy nonpregnant women (12 cells/mL). There was a significant correlation of CEC numbers and systolic blood pressure (P < .02). A rapid decline of cell numbers after delivery paralleled the clinical recovery. CONCLUSION: Circulating endothelial cells are a novel marker of vascular damage in preeclampsia.


Asunto(s)
Células Endoteliales , Endotelio Vascular/patología , Preeclampsia/sangre , Adulto , Femenino , Humanos , Embarazo
14.
FASEB J ; 20(7): 994-6, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16597674

RESUMEN

Long-acting third-generation dihydropyridine calcium channel blockers (CCBs) improve endothelial dysfunction and prevent cardiovascular events in humans, but their cellular and molecular mechanisms of tissue protection are not elucidated in detail. We assessed organ (renal) protection by the highly lipophilic CCB lercanidipine in a double-transgenic rat (dTGR) model with overexpression of human renin and angiotensinogen genes. We randomly treated dTGR with lercanidipine (2.5 mg/kg/day; n=20) or vehicle (n=20) for 3 wk. Furthermore, we explored the influence of lercanidipine on protein kinase C (PKC) signaling in vivo and in vitro using endothelial and vascular smooth muscle cell cultures. Cumulative mortality was 60% in untreated dTGR, whereas none of the lercanidipine-treated animals died (P<0.001). We found significantly less albuminuria and improved renal function in lercanidipine-treated dTGR (both P<0.05). Lercanidipine treatment also significantly (P<0.05) reduced blood levels of the endogenous NOS inhibitor asymmetric dimethylarginine. On histological examination, we observed significantly less tissue inflammation and fibrosis in lercanidipine-treated animals (both P<0.05). Lercanidipine significantly inhibited angiotensin (ANG) I-mediated PKC-alpha and -delta activation in vivo and in vitro, partly due to reduced intracellular calcium flux. As a result, lercanidipine improved endothelial cell permeability in vitro. Lercanidipine prevents tissue injury and improves survival in a model of progressive organ damage. These effects may result, at least in part, from inhibition of tissue inflammation as well as improved NO bioavailability. Modulation of PKC activity may be an important underlying intracellular mechanism.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/farmacología , Hipertensión/metabolismo , Albúminas/metabolismo , Amidohidrolasas/metabolismo , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Animales Modificados Genéticamente , Arginina/análogos & derivados , Arginina/sangre , Arginina/metabolismo , Regulación Enzimológica de la Expresión Génica , Hipertensión/tratamiento farmacológico , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratas , Ratas Sprague-Dawley , Renina/genética , Renina/metabolismo
15.
J Pharm Pharmacol ; 69(9): 1125-1135, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28573734

RESUMEN

OBJECTIVES: IL-17A contributes to acute kidney injury and fibrosis. Therefore, we asked whether IL-17A deficiency or treatment with a IL-17A blocking antibody impacts severe renal ischaemia reperfusion injury (IRI) and the progression to chronic kidney disease (CKD). METHODS: IL-17A-deficient and wild-type (WT) mice underwent transient unilateral renal pedicle clamping for 45 min to induce IRI and subsequent renal fibrosis. Furthermore, a neutralizing anti-IL-17A antibody (mAb) was injected into WT mice before induction of renal IRI intravenously. On days 1, 7 and 21, inflammation, fibrosis, leukocyte infiltration and pro-inflammatory and pro-fibrotic cytokine expression were assessed in kidneys using histology, qPCR and flow cytometry. KEY FINDINGS: IL-17A was significantly increased after renal IRI in WT kidneys. Levels of pro-inflammatory (MCP-1) cytokine and pro-fibrotic (collagen 1α1, fibronectin) transcripts were similar in the experimental groups studied. IL-17A deficiency had no effect on renal T-cell influx or the number, inflammatory phenotype, or spatial distribution of macrophages. Similarly, administration of an IL-17A blocking antibody did not attenuate inflammation. CONCLUSIONS: Despite the effects of IL-17 in other inflammation models, neither genetic IL-17A deficiency nor treatment with an IL-17A blocking antibody attenuated IRI and progression to CKD. We conclude that in severe renal IRI IL-17A is not crucially involved in disease progression.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Interleucina-17/genética , Insuficiencia Renal Crónica/prevención & control , Daño por Reperfusión/fisiopatología , Lesión Renal Aguda/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Citometría de Flujo , Inflamación/inmunología , Inflamación/fisiopatología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Insuficiencia Renal Crónica/inmunología , Daño por Reperfusión/inmunología
16.
Am J Kidney Dis ; 48(4): 564-70, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16997052

RESUMEN

BACKGROUND: Circulating endothelial cells (CECs) are a reliable marker of disease activity in a variety of vascular disorders. Damage to microvascular endothelial cells is a hallmark of thrombotic microangiopathy (TMA). The aim of this study is to identify and count CECs during the course of TMA and evaluate whether cell numbers may serve as a prognostic marker in patients undergoing plasma exchange. METHODS: Fifteen patients (8 women, 7 men) aged 31 to 66 years with TMA of different causes were studied before and after 4 sessions of plasma exchange. CECs were isolated by using anti-CD146-driven immunomagnetic isolation and counted after staining with Ulex Europaeus lectin-1. RESULTS: Numbers of CECs were markedly elevated in all patients before treatment (64 to 672 CEC/mL; mean, 320 +/- 205 CEC/mL) compared with healthy controls (0 to 16 CEC/mL; mean, 6.4 +/- 4.2 CEC/mL; P < 0.001). Patients with a favorable outcome had significantly greater initial CEC levels (mean, 426 +/- 175 CEC/mL; P < 0.001), and cell numbers decreased significantly after 4 treatments of plasma exchange (mean, 101 +/- 53 CEC/mL; P = 0.001). Patients with disease unresponsive to plasma exchange presented with lower initial CEC levels (mean, 108 +/- 36 CEC/mL), and numbers failed to decrease after plasma exchange (mean, 114 +/- 57 CEC/mL; P = 0.827). CONCLUSION: Markedly elevated numbers of CECs reflect severe and widespread endothelial damage in patients with TMA. Cell numbers at presentation and their degree of decrease after 4 sessions of plasma exchange could provide important prognostic clues.


Asunto(s)
Anemia Hemolítica/sangre , Células Endoteliales/patología , Trombosis/sangre , Trombosis/patología , Adulto , Anciano , Anemia Hemolítica/diagnóstico , Anemia Hemolítica/patología , Anemia Hemolítica/fisiopatología , Biomarcadores/sangre , Femenino , Humanos , Masculino , Microcirculación/patología , Persona de Mediana Edad , Agregación Plaquetaria/fisiología , Pronóstico , Trombocitopenia/diagnóstico , Trombocitopenia/patología , Trombocitopenia/fisiopatología , Trombosis/diagnóstico , Trombosis/fisiopatología
17.
Physiol Genomics ; 20(3): 256-67, 2005 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-15623567

RESUMEN

About one-half of double transgenic rats (dTGR) overexpressing the human renin and angiotensinogen genes die by age 7 wk of terminal heart failure (THF); the other (preterminal) one-half develop cardiac damage but survive. Our study's aim was to elucidate cardiac gene expression differences in dTGR-THF compared with dTGR showing compensated cardiac hypertrophy but not yet THF. dTGR treated with losartan (LOS) and nontransgenic rats (SD) served as controls. THF-dTGR body weight was significantly lower than for all other groups. At death, THF-dTGR had blood pressures of 228 +/- 7 mmHg (cardiac hypertrophy index 6.2 +/- 0.1 mg/g). Tissue Doppler showed reduced peak early (Ea) to late (Aa) diastolic expansion in THF-dTGR, indicating diastolic function. Preterminal dTGR had blood pressures of 197 +/- 5 mmHg (cardiac hypertrophy index 5.1 +/- 0.1 mg/g); Ea < Aa compared with LOS-dTGR (141 +/- 6 mmHg; 3.7+/-0.1 mg/g; Ea > Aa) and SD (112 +/- 4 mmHg; 3.6 +/- 0.1 mg/g; Ea > Aa). Left ventricular RNA was isolated for the Affymetrix system and TaqMan RT-PCR. THF-dTGR and dTGR showed upregulation of hypertrophy markers and alpha/beta-myosin heavy chain switch to the fetal isoform. THF-dTGR (vs. dTGR) showed upregulation of 239 and downregulation of 150 genes. Various genes of mitochodrial respiratory chain and lipid catabolism were reduced. In addition, genes encoding transcription factors (CEBP-beta, c-fos, Fra-1), coagulation, remodeling/repair components (HSP70, HSP27, heme oxygenase), immune system (complement components, IL-6), and metabolic pathway were differentially expressed. In contrast, LOS-dTGR and SD had similar expression profiles. These data demonstrate that THF-dTGR show an altered expression profile compared with preterminal dTGR.


Asunto(s)
Caquexia/genética , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/genética , Corazón/fisiopatología , Animales , Caquexia/complicaciones , Bases de Datos de Ácidos Nucleicos , Modelos Animales de Enfermedad , Ecocardiografía , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/diagnóstico por imagen , Masculino , Ratas , Ratas Sprague-Dawley
18.
Circulation ; 110(8): 1006-12, 2004 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-15302785

RESUMEN

BACKGROUND: The hematopoietic cytokine erythropoietin has cytoprotective effects in endothelial cells in vitro that are mediated through direct activation of the pro-survival Akt tyrosine kinase signaling pathway. We tested the hypothesis that low-dose therapy with the long-acting recombinant human erythropoietin analogue darbepoetin alpha protects vascular endothelium in vivo in a classic remnant kidney rat model characterized by severe endothelial damage, progressive vascular sclerosis, and ischemia-induced tissue fibrosis. METHODS AND RESULTS: Using a parallel group study design, we randomly assigned animals after 5/6 nephrectomy to treatment with either saline (n=36) or 0.1 microg/kg body wt darbepoetin (n=24) subcutaneously once weekly. We monitored hematocrit, blood pressure, and serum creatinine regularly and obtained renal tissue 6 weeks after nephrectomy for morphological and immunohistochemical analysis. Darbepoetin-treated animals had significantly improved survival compared with saline-treated controls (63% versus 33%; P<0.05), although hematocrit levels were similar in both groups. Darbepoetin treatment ameliorated endothelial damage; attenuated the composite tissue injury score (saline 1.9+/-0.4; darbepoetin 0.4+/-0.2; P<0.001), which included vascular sclerosis, glomerulosclerosis, and tubulointerstitial damage; and preserved renal function. We found persistent activation of the pro-survival Akt signaling pathway in endothelial and epithelial glomerular cells in darbepoetin-treated animals, accompanied by a significant reduction of apoptotic cell death in renal tissue. CONCLUSIONS: Low-dose darbepoetin treatment confers vascular and tissue protection that is associated with persistent stimulation of the pro-survival Akt signaling pathway. The use of recombinant human erythropoietin or analogues may have utility in preventing ischemia-related progressive vascular injury and organ failure.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Insuficiencia Multiorgánica/prevención & control , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Darbepoetina alfa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/patología , Hematócrito , Movilización de Célula Madre Hematopoyética , Hipertensión Renal/etiología , Hipertensión Renal/fisiopatología , Isquemia/prevención & control , Riñón/irrigación sanguínea , Tablas de Vida , Masculino , Insuficiencia Multiorgánica/etiología , Nefrectomía , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Proteínas Proto-Oncogénicas c-akt , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Diabetes ; 53(8): 2101-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15277392

RESUMEN

Activation of protein kinase C (PKC) isoforms has been implicated in the pathogenesis of diabetic nephropathy. We showed earlier that PKC-alpha is activated in the kidneys of hyperglycemic animals. We now used PKC-alpha(-/-) mice to test the hypothesis that this PKC isoform mediates streptozotocin-induced diabetic nephropathy. We observed that renal and glomerular hypertrophy was similar in diabetic wild-type and PKC-alpha(-/-) mice. However, the development of albuminuria was almost absent in the diabetic PKC-alpha(-/-) mice. The hyperglycemia-induced downregulation of the negatively charged basement membrane heparan sulfate proteoglycan perlecan was completely prevented in the PKC-alpha(-/-) mice, compared with controls. We then asked whether transforming growth factor-beta1 (TGF-beta1) and/or vascular endothelial growth factor (VEGF) is implicated in the PKC-alpha-mediated changes in the basement membrane. The hyperglycemia-induced expression of VEGF165 and its receptor VEGF receptor II (flk-1) was ameliorated in PKC-alpha(-/-) mice, whereas expression of TGF-beta1 was not affected by the lack of PKC-alpha. Our findings indicate that two important features of diabetic nephropathy-glomerular hypertrophy and albuminuria-are differentially regulated. The glucose-induced albuminuria seems to be mediated by PKC-alpha via downregulation of proteoglycans in the basement membrane and regulation of VEGF expression. Therefore, PKC-alpha is a possible therapeutic target for the prevention of diabetic albuminuria.


Asunto(s)
Nefropatías Diabéticas/genética , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Proteoglicanos/metabolismo , Albuminuria/orina , Animales , Secuencia de Bases , Glucemia/metabolismo , Peso Corporal , Cartilla de ADN , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/orina , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/orina , Hiperglucemia/genética , Hiperglucemia/fisiopatología , Riñón/anatomía & histología , Riñón/patología , Riñón/ultraestructura , Glomérulos Renales/anatomía & histología , Ratones , Ratones Endogámicos , Ratones Noqueados , Tamaño de los Órganos , Proteína Quinasa C-alfa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
20.
J Hypertens ; 20(5): 885-93, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12011649

RESUMEN

Cilnidipine is a novel dihydropyridine (DHP) antagonist. However, its pharmacological effects on vascular DHP-sensitive L-type channels and protein kinase C (PKC)-mediated arterial contraction is incompletely understood. To address this issue, we studied the effects of cilnidipine on multi-subunit, C-class L-type Ca2+ channels in rat aortic A7r5 cells, as well as on Ca2+ channel (L-type) alpha1C-b and (T-type) alpha1G subunits in the Xenopus oocyte expression system. Cilnidipine dose- and time-dependently inhibited Ba2+ currents in A7r5 cells, with half-maximal inhibitions (IC50) at 10 nmol/l after 10 min. Unlike classical pharmacological Ca2+ channel blockers, cilnidipine's block of Ca2+ currents did not reach steady-state levels within 10 min, indicating steady-state half-maximal inhibition of native, multi-subunit L-type channels at < 10 nmol/l. In contrast, smooth muscle alpha1Cb currents were blocked by cilnidipine at much higher doses (steady-state IC50, 20 micromol/l) whereas alpha1G currents were not inhibited by cilnidipine (30 micromol/l). Cilnidipine dose-dependently inhibited depolarization- and Ca2+-induced contractions of rat aortic rings, with an IC50 of 10 nmol/l at 10 min. However, the onset of the effects was very slow, with approximately 71% inhibition by 3 nmol/l cilnidipine after 90 min exposure to cilnidipine. In contrast, cilnidipine did not inhibit phorbol 12-myristate-13-acetate (100 nmol/l)-mediated contractions. We conclude that cilnidipine represents an extremely slow-acting DHP that targets multi-subunit L-type channels, but not PKC in arterial smooth muscle. Because cilnidipine is less potent in cells expressing the pore-forming alpha1C-b subunit, the data further suggest that this unique slow-acting mechanism of cilnidipine is mediated by a complex interaction of cilnidipine with alpha1C-b and accessory channel subunits.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Dihidropiridinas/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/fisiología , Calcio/farmacología , Canales de Calcio Tipo L/fisiología , Línea Celular , Conductividad Eléctrica , Electrofisiología , Isoformas de Proteínas/fisiología , Proteína Quinasa C/metabolismo , Ratas , Acetato de Tetradecanoilforbol/farmacología , Vasoconstricción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA