Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Biochem Biophys Res Commun ; 378(4): 826-31, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19070587

RESUMEN

We have recently shown that 3-deazaadenosine (c3Ado) inhibits atherogenesis in mice. We studied whether its anti-inflammatory capacity would also affect neointima-formation after balloon injury. Sprague Dawley rats underwent balloon angioplasty. C3Ado was administered orally, starting 5 days prior to the balloon injury and continued for 2 weeks. Fourteen days after balloon injury the intima/media ratio in the c3Ado-treated group was reduced by 67% (p<0.001) and luminal stenosis by 50% (p<0.001). Neointimal cellular density was decreased by 25% (p<0.001) and the induction of c-Jun and ki67 was markedly lower. The reduction of the intima/media ratio was still observed 3 months after balloon injury. Furthermore, a c3Ado-dependent inhibition of PDGF-mediated ERK-activation and proliferation could be demonstrated. Short-term administration of C3Ado inhibits neointima-formation in rats for at least 3 months after injury. The present findings implicate that c3Ado may be useful as an inhibitor of restenosis-formation after balloon angioplasty in humans.


Asunto(s)
Angioplastia de Balón/efectos adversos , Reestenosis Coronaria/prevención & control , Tubercidina/administración & dosificación , Túnica Íntima/efectos de los fármacos , Administración Oral , Animales , Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reestenosis Coronaria/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hígado/efectos de los fármacos , Masculino , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tubercidina/efectos adversos , Túnica Íntima/lesiones , Túnica Íntima/patología
2.
Circ Res ; 97(4): 305-13, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-16037570

RESUMEN

Atherosclerosis is considered to be an inflammatory disease. Tissue factor (TF), a prothrombotic molecule expressed by various cell types within atherosclerotic plaques, is thought to play an essential role in thrombus formation after atherosclerotic plaque rupture. Recent studies suggest that the antiinflammatory cytokine interleukin-10 (IL-10) has many antiatherosclerotic properties. Therefore, the effects of IL-10 on TF expression in response to inflammation were investigated. Mouse macrophages were stimulated with lipopolysaccharide (LPS) in the presence or absence of IL-10. Pretreatment with IL-10 resulted in a 50% decrease in TF mRNA expression and TF promoter activity. Binding of early growth response gene-1 (Egr-1) to the consensus DNA sequence, a key transcriptional activator of TF expression in response to inflammation, and the expression of Egr-1 mRNA were also inhibited by IL-10. This inhibition was independent of the induction of suppressor of cytokine signaling protein-3 by IL-10. Macrophages that had been transfected with luciferase reporter constructs containing the murine Egr-1 5'-flanking sequence exhibited reduced reporter gene activity in response to LPS stimulation with IL-10 pretreatment. Studies with deletion constructs of the Egr-1 promoter identified the proximal serum response element SRE3 as a potential regulatory site for the IL-10 mediated suppression of Egr-1 expression. Furthermore, activation of the upstream signal-transduction elements, such as mitogen-activated protein kinase kinase (MEK) 1/2, extracellular signal-regulated kinase 1/2, and Elk-1 were also inhibited by IL-10 pretreatment. Taken together, these results demonstrate a pathway for the IL-10 mediated inhibition of TF expression during inflammation and may explain the antiatherosclerotic effects of IL-10.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Interleucina-10/farmacología , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 1/fisiología , MAP Quinasa Quinasa 2/fisiología , Macrófagos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Elemento de Respuesta al Suero/fisiología , Tromboplastina/genética , Factores de Transcripción/antagonistas & inhibidores , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Endogámicos C57BL , Fosforilación , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Receptores Inmunológicos/fisiología , Proteínas Represoras/fisiología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Receptor Toll-Like 4 , Factores de Transcripción/genética , Factores de Transcripción/fisiología
3.
Circulation ; 109(22): 2792-800, 2004 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-15159288

RESUMEN

BACKGROUND: In a double-transgenic human renin and human angiotensinogen rat model, we found that mineralocorticoid receptor (MR) blockade ameliorated angiotensin II (Ang II)-induced renal and cardiac damage. How Ang II and aldosterone (Ald) might interact is ill defined. METHODS AND RESULTS: We investigated the effects of Ang II (10(-7) mol/L) and Ald (10(-7) mol/L) on extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling in vascular smooth muscle cells (VSMCs) with Western blotting and confocal microscopy. Ang II induced ERK 1/2 and JNK phosphorylation by 2 minutes. Ald achieved the same at 10 minutes. Ang II+Ald had a potentiating effect by 2 minutes. Two oxygen radical scavengers and the epidermal growth factor receptor (EGFR) antagonist AG1478 reduced Ang II-, Ald-, and combination-induced ERK1/2 phosphorylation. Preincubating the cells with the MR blocker spironolactone (10(-6) mol/L) abolished Ang II-induced ROS generation, EGFR transactivation, and ERK1/2 phosphorylation. CONCLUSIONS: Ald potentiates Ang II-induced ERK-1/2 and JNK phosphorylation. Oxygen radicals, the MR, and the EGFR play a role in early signaling induced by Ang II and Ald in VSMCs. These in vitro data may help explain the effects of MR blockade on Ang II-induced end-organ damage in vivo.


Asunto(s)
Aldosterona/fisiología , Angiotensina II/fisiología , Músculo Liso Vascular/fisiología , Angiotensina II/antagonistas & inhibidores , Animales , Animales Modificados Genéticamente , Células Cultivadas , Receptores ErbB/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Espironolactona/farmacología
4.
Circulation ; 107(12): 1632-9, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12668498

RESUMEN

BACKGROUND: We recently identified agonistic autoantibodies directed against the angiotensin AT1 receptor (AT1-AA) in the plasma of preeclamptic women. To elucidate their role further, we studied the effects of AT1-AA on reactive oxygen species (ROS), NADPH oxidase expression, and nuclear factor-kappaB (NF-kappaB) activation. METHODS AND RESULTS: We investigated human vascular smooth muscle cells (VSMC) and trophoblasts, as well as placentas. AT1-AA were isolated from sera of preeclamptic women. Angiotensin II (Ang II) and AT1-AA increased ROS production and the NADPH oxidase components, p22, p47, and p67 phox in Western blotting. We next tested if AT1-AA lead to NF-kappaB activation in VSMC and trophoblasts. AT1-AA activated NF-kappaB. Inhibitor-kappaBalpha (I-kappaBalpha) expression was reduced in response to AT1-AA. AT1 receptor blockade with losartan, diphenylene iodonium, tiron, and antisense against p22 phox all reduced ROS production and NF-kappaB activation. VSMC from p47phox-/- mice showed markedly reduced ROS generation and NF-kappaB activation in response to Ang II and AT1-AA. The p22, p47, and p67 phox expression in placentas from preeclamptic patients was increased, compared with normal placentas. Furthermore, NF-kappaB was activated and I-kappaBalpha reduced in placentas from preeclamptic women. CONCLUSIONS: NADPH oxidase is potentially an important source of ROS that may upregulate NF-kappaB in preeclampsia. We suggest that AT1-AA through activation of NADPH oxidase could contribute to ROS production and inflammatory responses in preeclampsia.


Asunto(s)
Autoanticuerpos/farmacología , NADPH Oxidasas/metabolismo , Preeclampsia/enzimología , Preeclampsia/inmunología , Receptores de Angiotensina/inmunología , Adulto , Angiotensina II/farmacología , Animales , Células Cultivadas , Activación Enzimática , Femenino , Humanos , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Placenta/efectos de los fármacos , Placenta/enzimología , Placenta/metabolismo , Preeclampsia/metabolismo , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1 , Receptores de Angiotensina/agonistas , Trofoblastos/efectos de los fármacos , Trofoblastos/enzimología , Trofoblastos/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 22(6): 914-20, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12067898

RESUMEN

Inflammatory response and chemotaxis of vascular wall cells play an important pathogenic role in the development of atherosclerosis. Monocyte chemoattractant protein-1 (MCP-1) is a potent chemoattractant for monocytes. Besides the induction of monocyte recruitment, it has been suggested that MCP-1 may directly activate smooth muscle cells. We investigated whether MCP-1 affects the proliferation and cytokine production of human vascular smooth muscle cells (VSMCs) and determined the underlying signal transduction pathways. Stimulation of VSMCs with MCP-1 induced proliferation and resulted in a concentration- and time-dependent release of the proinflammatory cytokine interleukin-6 (IL-6). Pretreatment with pertussis toxin, GF109203X, and pyrrolidine dithiocarbamate inhibited MCP-1-dependent IL-6 release, suggesting the involvement of G(i) proteins, protein kinase C, and nuclear factor-kappaB (NF-kappaB). MCP-1 also induced extracellular signal-regulated kinase, which, along with IL-6 release, was inhibited by pertussis toxin. PD98059 prevented MCP-1-induced extracellular signal-regulated kinase activation and cell proliferation. MCP-1 stimulated the binding activity of NF-kappaB and of activator protein-1 (AP-1). As demonstrated by cis element double-stranded (decoy) oligodeoxynucleotides, NF-kappaB was involved in IL-6 release by MCP-1, whereas proliferation was dependent on AP-1. The results clearly demonstrate that MCP-1 induces differential activation of NF-kappaB and AP-1 in VSMCs. Thus, our data propose a new mechanism for the proatherogenic effect of MCP-1.


Asunto(s)
Quimiocina CCL2/fisiología , Sustancias de Crecimiento/fisiología , Interleucina-6/biosíntesis , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , FN-kappa B/fisiología , Factor de Transcripción AP-1/fisiología , División Celular/fisiología , Puente de Arteria Coronaria , Activación Enzimática/fisiología , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN Mensajero/biosíntesis , Receptores CCR2 , Receptores de Quimiocina/biosíntesis , Proteínas Recombinantes/metabolismo , Vena Safena/citología
6.
J Mol Med (Berl) ; 82(1): 31-8, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14652666

RESUMEN

Reactive oxygen species (ROS) are involved in the transcriptional response to angiotensin (ANG) II. In this setting the role of NAD(P)H oxidase, an important source of ROS as second messengers, is not completely understood. In particular in human cells detailed insights into this mechanism are lacking. We investigated the role of ANG II and platelet-derived growth factor (PDGF) AA induced ROS generation derived from p22phox-containing NAD(P)H oxidase in the activation of activator protein (AP) 1 in human vascular smooth muscle cells (SMCs). Both ANG II and PDGF AA induced ROS generation in SMCs which was angiotensin type 1 receptor and PDGF alpha receptor dependent. Specific inhibition of the p22phox subunit of the NAD(P)H oxidase using either p22phox neutralizing antibody or p22phox antisense oligodeoxynucleotides (ODNs) attenuated both ANG II and PDGF AA induced ROS generation. Furthermore, PDGF AA but not ANG II induced p22phox mRNA expression. ANG II and PDGF AA both activated the redox-sensitive transcription factor AP-1, which was inhibited by p22phox antisense ODNs. These findings demonstrate that AP-1 activation in human SMCs in response to ANG II and PDGF AA is mediated via generation of p22phox-dependent ROS. This highlights the crucial role of the p22phox-containing NAD(P)H oxidase in the ANG II and PDGF AA induced signal transduction pathway.


Asunto(s)
Angiotensina II/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , NADPH Deshidrogenasa/metabolismo , Fosfoproteínas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Membrana Celular/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , NADPH Deshidrogenasa/genética , NADPH Oxidasas , Oxidación-Reducción , Fosfoproteínas/genética , Subunidades de Proteína/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología
7.
J Am Soc Nephrol ; 13(6): 1534-47, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12039983

RESUMEN

Monocyte chemoattractant protein-1 (MCP-1) is a potent chemokine synthesized by several cell types, e.g., inflammatory cells, such as monocytes, and resident renal cells, such as human tubular epithelial cells (TECs). Besides induction of monocyte recruitment, MCP-1 has been suggested to induce non-leukocytes to produce cytokines and adhesion molecules. Inflammation of the tubulointerstitium is a hallmark of many renal diseases and contributes to progression of renal failure; the purpose therefore of this study was to investigate the influence of MCP-1 on markers of inflammatory activation in human TECs. MCP-1 stimulated interleukin-6 (IL-6) secretion and intercellular adhesion molecule-1 (ICAM-1) synthesis in a time- and dose-dependent manner. In parallel, MCP-1 increased IL-6 and ICAM-1 mRNA expression in human TECs. Pretreatment with pertussis toxin, GF109203X, BAPTA-AM, and pyrrolidine dithiocarbamate inhibited MCP-1-dependent IL-6 and ICAM-1 synthesis, suggesting the involvement of Gi-proteins, protein kinase C, intracellular Ca(2+), and nuclear factor-kappaB (NF-kappaB) in MCP-1 signaling. Using electrophoretic gel mobility shift assay, we observed that MCP-1 stimulated binding activity of NF-kappaB. Binding activity of the activator protein-1 (AP-1), which has been implicated to regulate induction of the IL-6 gene together with NF-kappaB, was also stimulated by MCP-1. In the present experiments, NF-kappaB and AP-1 were involved in the MCP-1-mediated induction of IL-6, as demonstrated by cis element double-stranded (decoy) oligonucleotides (ODN). In contrast to IL-6 release, MCP-1-induced ICAM-1 expression was predominantly dependent on NF-kappaB activation. These results document for the first time that MCP-1 induces an inflammatory response in human TECs. This may be an important new mechanism in the pathogenesis of tubulointerstitial inflammation.


Asunto(s)
Quimiocina CCL2/farmacología , Túbulos Renales/efectos de los fármacos , FN-kappa B/fisiología , Nefritis/etiología , Factor de Transcripción AP-1/fisiología , Células Cultivadas , Quimiocina CCL2/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Interleucina-6/biosíntesis , Interleucina-6/genética , Túbulos Renales/inmunología , ARN Mensajero/análisis , Receptores CCR2 , Receptores de Quimiocina/genética
8.
Kidney Int ; 61(6): 2090-8, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12028449

RESUMEN

BACKGROUND: Cigarette smoking increases the risk of progression of diabetic and non-diabetic renal diseases. The mechanisms underlying the adverse effects of smoking are largely unknown. We examined the subtotally nephrectomized rat (i) to investigate whether components of cigarette smoke dissolved in acetone (cigarette smoke condensate) aggravate structural renal damage and (ii) to establish whether this provides an animal model that can be used to investigate potential pathomechanisms of cigarette smoke-induced renal damage. Since nicotine activates the sympathetic nerve system in humans, we investigated whether interference with this system modulates the effects of cigarette smoke condensate on the damaged kidney. METHODS: One group of Sprague-Dawley rats was subtotally nephrectomized (SNX). Acetone (SNX + solvent) or cigarette smoke condensate (SNX + cigarette) was applied daily to the oral mucosa. Another group of Sprague-Dawley rats was sham-operated and received the same treatments (sham + solvent, sham + cigarette). To investigate whether increased activity of the sympathetic nerve system is involved, the remnant kidney was denervated by microsurgical technique in one SNX + cigarette group. The control group for this intervention was a solvent-treated SNX group with denervated remnant kidney. Blood pressure (BP) was measured weekly by tail plethysmography. The experiment was terminated after 12 weeks. Structural renal damage was assessed by morphometric techniques (indices of glomerulosclerosis, tubulointerstitial and vascular damage) and urinary albumin and endothelin-1 excretion were measured. RESULTS: Indices of structural renal damage were increased in all SNX-groups. Treatment with cigarette smoke condensate further increased the indices of glomerulosclerosis and tubulointerstitial damage in SNX, but not sham-operated rats. This increase was completely prevented by renal denervation. No differences in systemic blood pressure were observed in the different SNX groups. Urinary albumin excretion went in parallel with the indices of glomerulosclerosis and tubulointerstitial damage and urinary endothelin-1 excretion was significantly increased in SNX + cigarette animals. CONCLUSION: These findings document that acetone soluble components in cigarette smoke aggravate glomerulosclerosis and tubulointerstitial damage in the renal ablation model. Renal injury induced by cigarette smoke condensate in this model is reversed by renal denervation. We conclude that cigarette smoke-induced renal damage is due, at least in part, to activation of the sympathetic nerve system.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/patología , Riñón/patología , Nefrectomía/métodos , Nicotiana , Humo/efectos adversos , Albuminuria/orina , Animales , Colesterol/sangre , Cotinina/orina , Creatinina/sangre , Endotelina-1/orina , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Riñón/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/fisiopatología , Triglicéridos/sangre , Urea/sangre
9.
Hypertension ; 44(6): 944-51, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15492129

RESUMEN

Activation of vascular smooth muscle cells (SMCs) by platelet-derived growth factor (PDGF) is a seminal event in the initiation and progression of the atherosclerotic lesion and may contribute to atherosclerotic plaque instability with plaque rupture and thrombus formation. Tissue factor (TF), a prothrombotic molecule expressed by various cell types within atherosclerotic plaques, is thought to play a major role in thrombus formation after plaque rupture. This study examined intracellular signaling pathways leading to TF expression and Egr-1 activation, a key element in tissue factor transcription, by PDGF-BB in rat SMCs. PDGF-BB induced TF mRNA and protein expression in a time-dependent manner. Early growth response factor-1 (Egr-1) binding activity was also induced by PDGF-BB, as well as phosphorylation of extracellular signal-regulated kinase. PDGF-BB-induced Egr-1 activation was suppressed by inhibitors of 2 upstream activators of Egr-1, extracellular signal-regulated kinase (ERK) and Src family kinases, whereas antioxidants, phosphatidylinositol 3-phosphate kinase, and p38 MAPK inhibitors had no effect. PDGF-BB-stimulated expression of the transcriptional co-repressor NAB2 was time-dependent. Furthermore, transient transfections of SMCs with wild-type and mutated TF promoter constructs showed that the Egr-1 binding region is an important transcriptional regulator of PDGF-BB-induced TF expression. Taken together, the results suggest that PDGF-BB induces TF expression and activity in SMC by a Src family kinases/ERK/Egr-1 signaling pathway and may therefore contribute to thrombus formation in advanced atherosclerosis and restenosis.


Asunto(s)
Arteriosclerosis/fisiopatología , Proteínas de Unión al ADN/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Tromboplastina/metabolismo , Trombosis/fisiopatología , Factores de Transcripción/metabolismo , Animales , Becaplermina , Células Cultivadas , Proteína 1 de la Respuesta de Crecimiento Precoz , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-sis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Familia-src Quinasas/fisiología
10.
Nephrol Dial Transplant ; 17(5): 780-7, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11981063

RESUMEN

BACKGROUND: Specific endothelin A (ET(A))-receptor blockade and ACE inhibition attenuate chronic transplant nephropathy (CTN) in the 'Fisher-to-Lewis' rat model. It is unknown (i) which of both pharmacological interventions attenuates CTN more effectively and (ii) whether combination therapy exerts additive nephroprotection. METHODS: We compared (i) the effects of specific ET(A)-receptor blockade with LU 302146 (30 mg/kg bw/day) and ACE inhibition with trandolapril (0.3 mg/kg bw/day) and (ii) the effect of a combination therapy of both drugs on the development of CTN. Kidneys of Fisher rats were orthotopically grafted to Lewis rats. Untreated 'Fisher-to-Lewis' allografts served as controls (TX). All animals received low-dose cyclosporin A (1.5 mg/kg body weight) for 10 days post-transplant to inhibit early acute rejection episodes. The duration of the experiment was 36 weeks. Blood pressure (BP) was measured every other week by tail plethysmography. Indices of glomerulosclerosis (GS), tubulointerstitial and vascular damage, number of glomeruli, total glomerular volume and mean glomerular volume were measured using morphometric and stereological techniques, respectively. Albuminuria, blood chemistry and haematology were measured at the end of the experiment. RESULTS: LU 302146 did not affect systolic BP. In contrast, trandolapril and combination treatment significantly reduced systolic BP. Histological signs of CTN were almost completely prevented by LU 302146 and trandolapril as compared to TX, e.g. GS=0.8+/-0.08 and 0.9+/-0.20 vs 1.8+/-0.21* (arbitrary unit; *P<0.001 vs treated groups). Allograft weight was significantly lower in treated vs TX animals. Trandolapril and combination therapy, but not LU 302146 alone, abrogated glomerular hypertrophy, i.e. mean glomerular volume: TX 2.22+/-0.43, trandolapril 1.61+/-0.38**, LU 302146 2.22+/-0.11, trandolapril+LU 302146 1.78+/-0.28* (microm(3); *P<0.05 vs control and LU 302146, **P<0.01 vs control and LU 302146). Albuminuria was lower in treated compared to TX animals. Combination therapy did not confer additional benefit compared to the respective monotherapies. CONCLUSIONS: We conclude that ET(A)-receptor blockade abrogates GS, tubulointerstitial and vascular damage in the 'Fisher-to-Lewis' model of CTN to a similar extent as ACE inhibition. However, only ACE inhibition inhibits glomerular hypertrophy. In contrast to ACE inhibition, the effect of ET(A)-receptor blockade is independent of BP. This finding is consistent with the notion that ET(A)-receptor mediated events play a partly BP-independent role in the genesis of CTN. Combination therapy exerts no additive nephroprotection.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Antagonistas de los Receptores de Endotelina , Indoles/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/etiología , Trasplante de Riñón/efectos adversos , Animales , Presión Sanguínea/efectos de los fármacos , Enfermedad Crónica , Quimioterapia Combinada , Riñón/patología , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Masculino , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Receptor de Endotelina A
11.
Nephrol Dial Transplant ; 18(1): 62-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12480961

RESUMEN

BACKGROUND: The effect of the specific endothelin (A) (ET(A))-receptor antagonist LU 302146 (LU) was assessed in a normotensive model of chronic transplant vasculopathy, i.e. orthotopic allotransplantation of the infrarenal abdominal aorta from spontaneously hypertensive-to-Wistar-Kyoto (SHR-to-WKY) rats. A second experimental setting was used to confirm the results in a different model, which to some extent may also address the issue of blood pressure (BP) in transplant vasculopathy, i.e. orthotopic allotransplantation of infrarenal abdominal aorta from WKY-to-SHR rats. Untreated sham-operated and isografted WKY and SHR served as controls. Allotransplanted animals treated with the angiotensin-converting enzyme (ACE) inhibitor trandolapril served as positive treatment controls. METHODS: Rats were randomized to receive standard diet or a diet designed to deliver either 30 mg LU/kg bw/day, 0.3 mg/kg bw/day trandolapril or a combination of both. The duration of either experiment was 8 weeks. BP was measured by tail plethysmography. RESULTS: Treatment with LU did not affect systolic BP in either experimental setting. In contrast, trandolapril and combination treatment significantly reduced systolic BP in SHRs. The increase in aortic wall thickness (given in mm) was abrogated to a similar extent in the three treatment groups as compared with untreated allotransplanted animals in either experimental setting (e.g. WKY sham-operated 0.084 +/- 0.013, P < 0.05 vs treatment groups; WKY isotransplanted 0.100 +/- 0.010, P < 0.05 vs treatment groups; WKY allotransplanted 0.289 +/- 0.077, P < 0.05 vs all groups; WKY allotransplanted + trandolapril 0.185 +/- 0.025; WKY allotransplanted + LU 301246 0.192 +/- 0.049; WKY allotransplanted + LU 301246 + trandolapril 0.190 +/- 0.041). This was due to an attenuation of the increase of intima and media thickness. Treatment with LU and trandolapril were similarly effective in attenuating the increase of the number of proliferating cell nuclear antigen (PCNA)-positive cells in the intima. Again, combination treatment did not confer additional benefit. In contrast, trandolapril was more effective than LU in attenuating the increase in the number of PCNA-positive cells in the media. Trandolapril or combination treatment, but not LU, attenuated transforming growth factor-beta expression in aortic allografts. CONCLUSIONS: The ET(A)-receptor blockade abrogates allograft vasculopathy in two different aorta allotransplantation models to a similar extent as ACE inhibition even in the absence of concomitant immunosuppression. At least in SHRs the effect of ET(A)-receptor blockade is independent of BP. This finding is consistent with the notion that ET(A)-receptor mediated events play a partly BP-independent role in the genesis of chronic transplant vasculopathy.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Aorta/trasplante , Compuestos de Bencidrilo/uso terapéutico , Antagonistas de los Receptores de Endotelina , Indoles/uso terapéutico , Pirimidinas/uso terapéutico , Trasplante Homólogo/efectos adversos , Enfermedades Vasculares/etiología , Enfermedades Vasculares/prevención & control , Animales , Presión Sanguínea , Quimioterapia Combinada , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptor de Endotelina A , Trasplante Homólogo/patología , Trasplante Homólogo/fisiología
12.
Basic Res Cardiol ; 98(6): 353-61, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14556080

RESUMEN

The terminal complement complex C5b-9 is known to participate in inflammatory processes including atherosclerosis. Inflammation appears to be a direct consequence of C5b-9-mediated cell stimulation. 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors may exert anti-inflammatory effects on vascular cells independent of lowering plasma cholesterol. Thus, we studied activation of vascular smooth muscle cells (VSMCs) by C5b-9 focusing on whether inhibition of the HMG-CoA reductase can reduce the proinflammatory effects of C5b-9.C5b-9 in sublytic concentrations increased the proliferation of human VSMCs and induced a time-dependent activation of the mitogen-activated protein (MAP) kinase extracellular signal-regulated kinase (ERK). Proliferation and ERK1/2 activation could be inhibited by the specific ERK inhibitor PD98059. HMG-CoA inhibition with cerivastatin-reduced VSMC proliferation and C5b-9-induced ERK1/2 activation. Cerivastatin also reduced the C5b-9-induced synthesis of the proinflammatory interleukin-6 (IL-6). Furthermore, C5b-9 induced activation of the transcription factors activator protein- 1 (AP-1) and nuclear factor-kappaB (NF-kappaB), which could be inhibited by pretreatment of VSMCs with cerivastatin. L-mevalonate and geranylgeranylpyrophosphate reversed the inhibitory effects of cerivastatin. The present study in VSMCs shows that cerivastatin inhibits IL-6 synthesis and cell proliferation induced by the terminal complement complex C5b-9. This may be an important mechanism contributing to the beneficial effects of HMG-CoA reductase inhibitors beyond lowering of plasma cholesterol.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/farmacología , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Activación Enzimática , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Interleucina-6/biosíntesis , Lovastatina/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Piridinas/farmacología , Factor de Transcripción AP-1/metabolismo
13.
Am J Pathol ; 161(5): 1679-93, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12414515

RESUMEN

Angiotensin (Ang) II promotes renal infiltration by immunocompetent cells in double-transgenic rats (dTGRs) harboring both human renin and angiotensinogen genes. To elucidate disease mechanisms, we investigated whether or not dexamethasone (DEXA) immunosuppression ameliorates renal damage. Untreated dTGRs developed hypertension, renal damage, and 50% mortality at 7 weeks. DEXA reduced albuminuria, renal fibrosis, vascular reactive oxygen stress, and prevented mortality, independent of blood pressure. In dTGR kidneys, p22phox immunostaining co-localized with macrophages and partially with T cells. dTGR dendritic cells expressed major histocompatibility complex II and CD86, indicating maturation. DEXA suppressed major histocompatibility complex II+, CD86+, dendritic, and T-cell infiltration. In additional experiments, we treated dTGRs with mycophenolate mofetil to inhibit T- and B-cell proliferation. Reno-protective actions of mycophenolate mofetil and its effect on dendritic and T cells were similar to those obtained with DEXA. We next investigated whether or not Ang II directly promotes dendritic cell maturation in vitro. Ang II did not alter CD80, CD83, and MHC II expression, but increased CCR7 expression and cell migration. To explore the role of tumor necrosis factor (TNF)-alpha on dendritic cell maturation in vivo, we treated dTGRs with the soluble TNF-alpha receptor etanercept. This treatment had no effect on blood pressure, but decreased albuminuria, nuclear factor-kappaB activation, and infiltration of all immunocompetent cells. These data suggest that immunosuppression prevents dendritic cell maturation and T-cell infiltration in a nonimmune model of Ang II-induced renal damage. Ang II induces dendritic migration directly, whereas in vivo TNF-alpha is involved in dendritic cell infiltration and maturation. Thus, Ang II may initiate events leading to innate and acquired immune response.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Dexametasona/farmacología , Inmunosupresores/farmacología , Enfermedades Renales/inmunología , Proteínas de Transporte de Membrana , Ácido Micofenólico/análogos & derivados , Angiotensinógeno/genética , Animales , Animales Modificados Genéticamente , Antígenos CD/metabolismo , Antígeno B7-2 , Presión Sanguínea , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Etanercept , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunoglobulina G/farmacología , Inflamación/prevención & control , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/patología , Enfermedades Renales/patología , Enfermedades Renales/prevención & control , Cinética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Ácido Micofenólico/farmacología , NADPH Deshidrogenasa/análisis , NADPH Oxidasas , FN-kappa B/metabolismo , Fosfoproteínas/análisis , Sustancias Protectoras/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores del Factor de Necrosis Tumoral , Renina/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA