Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Kidney Int ; 94(4): 741-755, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29935951

RESUMO

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.


Assuntos
Heme Oxigenase-1/metabolismo , Interleucina-10/metabolismo , Túbulos Renais/patologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor da Anafilatoxina C5a/genética , Traumatismo por Reperfusão/genética , Animais , Proliferação de Células/genética , Células Cultivadas , Células Epiteliais , Fibrose , Inflamação/etiologia , Rim/diagnóstico por imagem , Túbulos Renais/metabolismo , Túbulos Renais/fisiopatologia , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Imagem de Perfusão , Fosforilação , Fatores de Proteção , Receptor da Anafilatoxina C5a/metabolismo , Regeneração/genética , Traumatismo por Reperfusão/complicações , Regulação para Cima
2.
Am J Physiol Renal Physiol ; 314(5): F881-F892, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357437

RESUMO

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.


Assuntos
Injúria Renal Aguda/complicações , Hipertensão/etiologia , Rim/irrigação sanguínea , Circulação Renal , Insuficiência Renal Crônica/etiologia , Traumatismo por Reperfusão/complicações , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Velocidade do Fluxo Sanguíneo , Pressão Sanguínea , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Mesângio Glomerular/patologia , Hipertensão/metabolismo , Hipertensão/patologia , Hipertensão/fisiopatologia , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Lipocalina-2/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Fatores de Tempo
3.
J Pharm Pharmacol ; 69(9): 1125-1135, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28573734

RESUMO

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.


Assuntos
Injúria Renal Aguda/fisiopatologia , Interleucina-17/genética , Insuficiência Renal Crônica/prevenção & controle , Traumatismo por Reperfusão/fisiopatologia , Injúria Renal Aguda/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Citometria de Fluxo , Inflamação/imunologia , Inflamação/fisiopatologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Insuficiência Renal Crônica/imunologia , Traumatismo por Reperfusão/imunologia
4.
J Am Soc Nephrol ; 26(11): 2860-70, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25788529

RESUMO

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.


Assuntos
Proteínas ADAM/sangue , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/sangue , Interleucina-6/sangue , MicroRNAs/sangue , Mieloblastina/imunologia , Proteína ADAM17 , Adulto , Idoso , Doenças Cardiovasculares/sangue , Células Cultivadas , Citocinas/sangue , Endotélio Vascular/fisiologia , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imunoensaio , Inflamação , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Mieloblastina/sangue , Fenótipo
5.
Crit Care Med ; 42(10): e654-62, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25083983

RESUMO

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.


Assuntos
Angiopoietina-2/fisiologia , Pulmão/metabolismo , Insuficiência de Múltiplos Órgãos/etiologia , Interferência de RNA/fisiologia , Sepse/complicações , Angiopoietina-2/metabolismo , Animais , Modelos Animais de Doenças , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Insuficiência de Múltiplos Órgãos/metabolismo , Insuficiência de Múltiplos Órgãos/fisiopatologia , RNA Interferente Pequeno/metabolismo , Receptor TIE-2/metabolismo , Sepse/metabolismo , Sepse/mortalidade , Sepse/fisiopatologia
6.
Am J Physiol Renal Physiol ; 307(6): F718-26, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25056349

RESUMO

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.


Assuntos
Injúria Renal Aguda/enzimologia , Proteína Quinase C-épsilon/metabolismo , Traumatismo por Reperfusão/enzimologia , Aloenxertos/enzimologia , Animais , Apoptose , Sobrevivência de Enxerto , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Testes de Função Renal , Transplante de Rim , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Hypertension ; 62(4): 726-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23959558

RESUMO

Hypertension is one of the major risk factors for chronic kidney disease. Using quantitative trait loci analysis, we identified the gene of the F-BAR protein NOSTRIN in the center of an overlapping region in rat and human quantitative trait loci that are associated with hypertension. Immunohistochemical analysis revealed a predominantly podocytic expression pattern of NOSTRIN in human and mouse glomeruli. Further, NOSTRIN colocalizes with cell-cell contact-associated proteins ß-catenin and zonula occludens-1 and interacts with the slit-membrane-associated adaptor protein CD2AP. In zebrafish larvae, knockdown of nostrin alters the glomerular filtration barrier function, inducing proteinuria and leading to ultrastructural morphological changes on the endothelial and epithelial side and of the glomerular basement membrane of the glomerular capillary loop. We conclude that NOSTRIN expression is an important factor for the integrity of the glomerular filtration barrier. Disease-related alteration of NOSTRIN expression may not only affect the vascular endothelium and, therefore, contribute to endothelial cell dysfunction but might also contribute to the development of podocyte disease and proteinuria.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Membrana Basal Glomerular/fisiopatologia , Hipertensão/genética , Glomérulos Renais/fisiopatologia , Proteínas de Membrana/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/ultraestrutura , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Glomérulos Renais/metabolismo , Glomérulos Renais/ultraestrutura , Proteínas de Membrana/metabolismo , Podócitos/metabolismo , Proteinúria/genética , Proteinúria/metabolismo , Proteinúria/fisiopatologia , Peixe-Zebra
9.
Hypertension ; 61(2): 501-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23213194

RESUMO

Aldosterone (Aldo) is involved in vascular remodeling and inflammation; however, the mechanisms are imperfectly defined. We hypothesized that Aldo alters endothelial integrity and modifies paracellular permeability. Human umbilical vein endothelial cells were exposed to Aldo (10(-9) mol/L) and alterations in paracellular permeability, assembly of tight and adherens junctions and activation of intracellular signaling pathways were determined. Aldo increased endothelial permeability for molecules ≤ 70 kDa within 60 minutes. A transient loss of cortical actin with formation of actin stress fibers and disruption of continuous adherens and tight junction strands accompanied these changes. Mineralocorticoid receptor blockade, inhibition of RhoA, or disruption of extracellular-regulated protein kinase1/2 signaling pathways attenuated the Aldo-related effects. Moreover, Aldo-induced cytoskeletal rearrangement led to rapid dephosphorylation of protein kinase B and subsequent deactivation of endothelial nitric oxide synthase. Ex vivo tracer flux experiments with Evans blue-conjugated albumin demonstrated a concordant response to Aldo in freshly isolated umbilical arteries. Furthermore, low-dose cortisol (3 × 10(-10) to 3 × 10(-9) mol/L) mimics the effect of Aldo on endothelial integrity, and Aldo, by upregulating11ß-hydroxysteroid dehydrogenase type 2, might even aggravate this deleterious effect of low-dose cortisol. We suggest that these mechanisms may contribute to the vasculopathy induced by inappropriate mineralocorticoid receptor activation.


Assuntos
Actinas/metabolismo , Aldosterona/farmacologia , Citoesqueleto/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrocortisona/farmacologia , Permeabilidade/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
10.
Hypertension ; 58(3): 471-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21747041

RESUMO

In humans, dehydroepiandrosterone (DHEA), with its sulfate, is the most abundant adrenal steroid, whereas the rat adrenals are not capable of synthesizing this steroid. Circulating concentrations of DHEA sulfate lie in the millimolar range and those of DHEA in the subnanomolar range. DHEA exerts protective potential during vascular remodeling, although the underlying mechanisms of this protection are imperfectly defined. We hypothesized that physiological doses of DHEA alter signaling pathways that are of central importance for vascular integrity. We exposed human endothelial cells, vascular smooth muscle cells, and fibroblasts to DHEA (10(-6) to 10(-10) mol/L) and observed a dose- and time-dependent increase of extracellular signal-regulated kinases 1 and 2 activation. Similar results were observed in rat vascular smooth muscle cells. In addition, in rat vascular smooth muscle cells, we found altered phosphorylation and cellular translocation of the transcription factor FoxO1. Pharmacological blockade of the mineralocorticoid receptor (MR) with eplerenone or small interfering RNA-mediated MR-silencing prevented DHEA-induced extracellular signal-regulated kinase 1/2 phosphorylation and its effects on FoxO1. Of note, in a cell-based MR transactivation assay, we did not find any agonist effect of DHEA on MR activity. We conclude that DHEA induces early signaling events in vascular cells that might underlie the DHEA-mediated protection against vasculopathies. These effects are dependent on the MR, although the finding that DHEA fails to act as a direct MR agonist suggests that additional signaling proteins are involved. In this regard, DHEA may either interact with coeffectors to modify MR activity or serves as a ligand for a yet unknown receptor that might transactivate the MR.


Assuntos
Desidroepiandrosterona/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Mineralocorticoides/metabolismo , Animais , Western Blotting , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Eplerenona , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Proteína Forkhead Box O1 , Humanos , Microscopia Confocal , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Interferência de RNA , Ratos , Receptores de Mineralocorticoides/genética , Espironolactona/análogos & derivados , Espironolactona/farmacologia , Fatores de Tempo
11.
J Am Soc Nephrol ; 22(3): 484-95, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21355054

RESUMO

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.


Assuntos
Rim/irrigação sanguínea , Proteínas de Membrana/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Receptor Celular 1 do Vírus da Hepatite A , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Rim/patologia , Rim/cirurgia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Necrose , Nefrectomia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle
12.
Vasc Health Risk Manag ; 6: 1125-33, 2010 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-21191433

RESUMO

The clinical importance of microparticles resulting from vesiculation of platelets and other blood cells is increasingly recognized, although no standardized method exists for their measurement. Only a few studies have examined the analytical and preanalytical steps and variables affecting microparticle detection. We focused our analysis on microparticle detection by flow cytometry. The goal of our study was to analyze the effects of different centrifugation protocols looking at different durations of high and low centrifugation speeds. We also analyzed the effect of filtration of buffer and long-term freezing on microparticle quantification, as well as the role of Annexin V in the detection of microparticles. Absolute and platelet-derived microparticles were 10- to 15-fold higher using initial lower centrifugation speeds at 1500 × g compared with protocols using centrifugation speeds at 5000 × g (P < 0.01). A clear separation between true events and background noise was only achieved using higher centrifugation speeds. Filtration of buffer with a 0.2 µm filter reduced a significant amount of background noise. Storing samples for microparticle detection at -80°C decreased microparticle levels at days 28, 42, and 56 (P < 0.05 for all comparisons with fresh samples). We believe that staining with Annexin V is necessary to distinguish true events from cell debris or precipitates. Buffers should be filtered and fresh samples should be analyzed, or storage periods will have to be standardized. Higher centrifugation speeds should be used to minimize contamination by smaller size platelets.


Assuntos
Micropartículas Derivadas de Células , Citometria de Fluxo/métodos , Anexina A5 , Plaquetas/ultraestrutura , Centrifugação , Criopreservação , Feminino , Filtração , Congelamento , Humanos , Leucócitos/ultraestrutura , Masculino , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Complexo Glicoproteico GPIb-IX de Plaquetas/análise
13.
Kidney Int ; 78(12): 1252-62, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20827258

RESUMO

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.


Assuntos
Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/urina , Estado Terminal , Peptídeos/urina , Proteômica , Adulto , Idoso , Idoso de 80 Anos ou mais , Albuminas/metabolismo , Biomarcadores/urina , Estudos de Coortes , Colágeno Tipo I/urina , Feminino , Fibrinogênio/urina , Humanos , Unidades de Terapia Intensiva , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Prognóstico , Sensibilidade e Especificidade , alfa 1-Antitripsina/urina , Microglobulina beta-2/urina
14.
BMC Cell Biol ; 11: 68, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20822540

RESUMO

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.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , AMP Cíclico/farmacologia , Endotélio Vascular/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Junções Íntimas/efeitos dos fármacos , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Permeabilidade da Membrana Celular/efeitos dos fármacos , Células Cultivadas , Claudina-5 , AMP Cíclico/análogos & derivados , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Proteína 2 com Domínio MARVEL , Proteínas de Membrana/genética , Microscopia Confocal , Ocludina , Transporte Proteico/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Veias Umbilicais/patologia , Regulação para Cima
15.
J Cell Mol Med ; 14(7): 1922-34, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19508384

RESUMO

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.


Assuntos
Apoptose , Endotélio/fisiologia , Macrófagos/fisiologia , Trombospondina 1/fisiologia , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Humanos , Imuno-Histoquímica , Macrófagos/citologia , Camundongos , Reação em Cadeia da Polimerase
16.
Hypertension ; 54(2): 359-64, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19564549

RESUMO

Growth arrest-specific protein 6 (Gas 6) is involved in inflammatory kidney diseases, vascular remodeling, cell adhesion, and thrombus formation. We explored a role for Gas 6 in aldosterone-induced target organ damage. We observed that Gas 6 was upregulated in rats with high aldosterone levels. Mineralocorticoid receptor blockade prevented target organ damage and decreased the elevated Gas 6 expression. Vascular smooth muscle cells given aldosterone increased their Gas 6 expression in vitro. To test the pathophysiological relevance, we investigated the effects of deoxycorticosterone acetate (DOCA) on Gas 6 gene-deleted ((-/-)) mice. After 6 weeks DOCA, Gas 6(-/-) mice developed similar telemetric blood pressure elevations compared to wild-type mice but were protected from cardiac hypertrophy. Cardiac expression of interleukin 6 and collagen IV was blunted in Gas 6(-/-) mice, indicating reduced inflammation and fibrosis. Gas 6(-/-) mice also had an improved renal function with reduced albuminuria, compared to wild-type mice. Renal fibrosis and fibronectin deposition in the kidney were also reduced. Gas 6 deficiency reduces the detrimental effects of aldosterone on cardiac and renal remodeling independent of blood pressure reduction. Gas 6 appears to play a role in mineralocorticoid receptor-mediated target organ damage. Furthermore, because warfarin interferes with Gas 6 protein expression, the findings could be of clinical relevance for anticoagulant choices.


Assuntos
Injúria Renal Aguda/fisiopatologia , Cardiomegalia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Músculo Liso Vascular/citologia , Injúria Renal Aguda/patologia , Albuminúria , Aldosterona/farmacologia , Análise de Variância , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/patologia , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Desoxicorticosterona/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Knockout , Probabilidade , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
17.
Nephrol Dial Transplant ; 24(8): 2567-75, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19398767

RESUMO

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.


Assuntos
Atrofia/genética , Fibrose/genética , Perfilação da Expressão Gênica , Rejeição de Enxerto/genética , Transplante de Rim , Túbulos Renais/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Atrofia/metabolismo , Atrofia/patologia , Biomarcadores/metabolismo , Biópsia , Criança , Feminino , Fibrose/metabolismo , Fibrose/patologia , Genoma Humano , Rejeição de Enxerto/metabolismo , Humanos , Técnicas Imunoenzimáticas , Túbulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Transplante Homólogo , Adulto Jovem
18.
J Immunol ; 181(2): 1179-89, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606671

RESUMO

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.


Assuntos
Células Endoteliais/imunologia , Rejeição de Enxerto , Transplante de Rim/imunologia , Rim/imunologia , Receptores de Superfície Celular/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Sobrevivência de Enxerto , Molécula 1 de Adesão Intercelular/imunologia , Molécula 1 de Adesão Intercelular/metabolismo , Rim/irrigação sanguínea , Rim/citologia , Rim/metabolismo , Masculino , Camundongos , Infiltração de Neutrófilos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Superfície Celular/deficiência , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Traumatismo por Reperfusão/imunologia , Transplante Homólogo , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/deficiência , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
19.
Hypertension ; 51(5): 1372-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18347231

RESUMO

Mineralocorticoid receptor blockade protects from angiotensin II-induced target-organ damage. 11beta-Hydroxysteroid dehydrogenase type 2 protects the mineralocorticoid receptor from activation by glucocorticoids; however, high glucocorticoid concentrations and absent 11beta-hydroxysteroid dehydrogenase type 2 in some tissues make glucocorticoids highly relevant mineralocorticoid receptor ligands. We investigated the effects of corticosterone (10(-6) to 10(-12) mol/L) on early vascular mineralocorticoid receptor signaling by Western blotting, confocal microscopy, and myography. Corticosterone initiated extracellular signal-regulated kinase 1/2 phosphorylation in rat vascular smooth muscle cells at > or =10(-11) mol/L doses. Protein synthesis inhibitors had no effect, indicating a nongenomic action. Corticosterone also stimulated c-Jun N-terminal kinase, p38, Src, and Akt phosphorylation at 15 minutes and enhanced angiotensin II-induced signaling at 5 minutes. A specific epidermal growth factor receptor blocker, AG1478, as well as the Src inhibitor PP2, markedly reduced corticosterone-induced extracellular signal-regulated kinase 1/2 phosphorylation, as did preincubation of cells with the mineralocorticoid receptor antagonist spironolactone. Silencing mineralocorticoid receptor with small interfering RNA abolished corticosterone-induced effects. Corticosterone (10(-9) mol/L) enhanced phenylephrine-induced contraction of intact aortic rings. These effects were dependent on the intact endothelium, mineralocorticoid receptor, and mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase signaling. We conclude that corticosterone induces rapid mineralocorticoid receptor signaling in vascular smooth muscle cells that involves mitogen-activated protein kinase kinase/extracellular signal-regulated kinase-dependent pathways. These new mineralocorticoid receptor-dependent signaling pathways suggest that glucocorticoids may contribute to vascular disease via mineralocorticoid receptor signaling, independent of circulating aldosterone.


Assuntos
Aorta/efeitos dos fármacos , Corticosterona/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Receptores de Mineralocorticoides/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Proteína Tirosina Quinase CSK , Células Cultivadas , Relação Dose-Resposta a Droga , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais/fisiologia , Vasoconstrição/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Quinases da Família src
20.
Am J Obstet Gynecol ; 198(3): 317.e1-5, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18068139

RESUMO

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.


Assuntos
Células Endoteliais , Endotélio Vascular/patologia , Pré-Eclâmpsia/sangue , Adulto , Feminino , Humanos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA