Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Exp Med ; 214(8): 2405-2420, 2017 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-28626071

RESUMEN

Mitochondrial dysfunction is the most prominent source of oxidative stress in acute and chronic kidney disease. NLRX1 is a receptor of the innate immune system that is ubiquitously expressed and localized in mitochondria. We investigated whether NLRX1 may act at the interface of metabolism and innate immunity in a model of oxidative stress. Using a chimeric mouse model for renal ischemia-reperfusion injury, we found that NLRX1 protects against mortality, mitochondrial damage, and epithelial cell apoptosis in an oxidative stress-dependent fashion. We found that NLRX1 regulates oxidative phosphorylation and cell integrity, whereas loss of NLRX1 results in increased oxygen consumption, oxidative stress, and subsequently apoptosis in epithelial cells during ischemia-reperfusion injury. In line, we found that NLRX1 expression in human kidneys decreased during acute renal ischemic injury and acute cellular rejection. Although first implicated in immune regulation, we propose that NLRX1 function extends to the control of mitochondrial activity and prevention of oxidative stress and apoptosis in tissue injury.


Asunto(s)
Apoptosis/fisiología , Mitocondrias/fisiología , Proteínas Mitocondriales/fisiología , Estrés Oxidativo/fisiología , Daño por Reperfusión/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Isquemia/fisiopatología , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL
3.
Kidney Int ; 91(2): 352-364, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27692564

RESUMEN

Acute kidney injury is often the result of ischemia reperfusion injury, which leads to activation of coagulation and inflammation, resulting in necrosis of renal tubular epithelial cells. Platelets play a central role in coagulation and inflammatory processes, and it has been shown that platelet activation exacerbates acute kidney injury. However, the mechanism of platelet activation during ischemia reperfusion injury and how platelet activation leads to tissue injury are largely unknown. Here we found that renal ischemia reperfusion injury in mice leads to increased platelet activation in immediate proximity of necrotic cell casts. Furthermore, platelet inhibition by clopidogrel decreased cell necrosis and inflammation, indicating a link between platelet activation and renal tissue damage. Necrotic tubular epithelial cells were found to release extracellular DNA, which, in turn, activated platelets, leading to platelet-granulocyte interaction and formation of neutrophil extracellular traps ex vivo. Renal ischemia reperfusion injury resulted in increased DNA-platelet and DNA-platelet-granulocyte colocalization in tissue and elevated levels of circulating extracellular DNA and platelet factor 4 in mice. After renal ischemia reperfusion injury, neutrophil extracellular traps were formed within renal tissue, which decreased when mice were treated with the platelet inhibitor clopidogrel. Thus, during renal ischemia reperfusion injury, necrotic cell-derived DNA leads to platelet activation, platelet-granulocyte interaction, and subsequent neutrophil extracellular trap formation, leading to renal inflammation and further increase in tissue injury.


Asunto(s)
Plaquetas/metabolismo , ADN/metabolismo , Células Epiteliales/metabolismo , Trampas Extracelulares/metabolismo , Necrosis Tubular Aguda/metabolismo , Túbulos Renales/metabolismo , Activación Plaquetaria , Daño por Reperfusión/metabolismo , Animales , Plaquetas/efectos de los fármacos , Línea Celular , Clopidogrel , ADN/genética , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Necrosis Tubular Aguda/genética , Necrosis Tubular Aguda/patología , Necrosis Tubular Aguda/prevención & control , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Nefritis/genética , Nefritis/metabolismo , Nefritis/patología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Factor Plaquetario 4/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Transducción de Señal , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Factores de Tiempo
4.
Sci Rep ; 6: 38275, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27928159

RESUMEN

Renal ischemia reperfusion (IR)-injury induces activation of innate immune response which sustains renal injury and contributes to the development of delayed graft function (DGF). Triggering receptor expressed on myeloid cells-1 (TREM-1) is a pro-inflammatory evolutionary conserved pattern recognition receptor expressed on a variety of innate immune cells. TREM-1 expression increases following acute and chronic renal injury. However, the function of TREM-1 in renal IR is still unclear. Here, we investigated expression and function of TREM-1 in a murine model of renal IR using different TREM-1 inhibitors: LP17, LR12 and TREM-1 fusion protein. In a human study, we analyzed the association of non-synonymous single nucleotide variants in the TREM1 gene in a cohort comprising 1263 matching donors and recipients with post-transplant outcomes, including DGF. Our findings demonstrated that, following murine IR, renal TREM-1 expression increased due to the influx of Trem1 mRNA expressing cells detected by in situ hybridization. However, TREM-1 interventions by means of LP17, LR12 and TREM-1 fusion protein did not ameliorate IR-induced injury. In the human renal transplant cohort, donor and recipient TREM1 gene variant p.Thr25Ser was not associated with DGF, nor with biopsy-proven rejection or death-censored graft failure. We conclude that TREM-1 does not play a major role during experimental renal IR and after kidney transplantation.


Asunto(s)
Funcionamiento Retardado del Injerto/genética , Inflamación/tratamiento farmacológico , Daño por Reperfusión/tratamiento farmacológico , Receptor Activador Expresado en Células Mieloides 1/genética , Animales , Funcionamiento Retardado del Injerto/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Riñón/efectos de los fármacos , Riñón/lesiones , Riñón/metabolismo , Riñón/patología , Trasplante de Riñón/efectos adversos , Ácidos Láuricos/administración & dosificación , Ratones , Oligopéptidos , Polimorfismo de Nucleótido Simple/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Rodaminas/administración & dosificación , Receptor Activador Expresado en Células Mieloides 1/antagonistas & inhibidores
5.
PLoS One ; 10(9): e0137511, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26361210

RESUMEN

Ischemia reperfusion injury is a common cause of acute kidney injury and is characterized by tubular damage. Mitochondrial DNA is released upon severe tissue injury and can act as a damage-associated molecular pattern via the innate immune receptor TLR9. Here, we investigated the role of TLR9 in the context of moderate or severe renal ischemia reperfusion injury using wild-type C57BL/6 mice or TLR9KO mice. Moderate renal ischemia induced renal dysfunction but did not decrease animal well-being and was not regulated by TLR9. In contrast, severe renal ischemia decreased animal well-being and survival in wild-type mice after respectively one or five days of reperfusion. TLR9 deficiency improved animal well-being and survival. TLR9 deficiency did not reduce renal inflammation or tubular necrosis. Rather, severe renal ischemia induced hepatic injury as seen by increased plasma ALAT and ASAT levels and focal hepatic necrosis which was prevented by TLR9 deficiency and correlated with reduced circulating mitochondrial DNA levels and plasma LDH. We conclude that TLR9 does not mediate renal dysfunction following either moderate or severe renal ischemia. In contrast, our data indicates that TLR9 is an important mediator of hepatic injury secondary to ischemic acute kidney injury.


Asunto(s)
Riñón/irrigación sanguínea , Hígado/patología , Daño por Reperfusión/metabolismo , Receptor Toll-Like 9/metabolismo , Alanina Transaminasa/sangre , Animales , Línea Celular , Riñón/metabolismo , Riñón/patología , L-Lactato Deshidrogenasa/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/patología , Receptor Toll-Like 9/genética
6.
PLoS One ; 10(4): e0123203, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875776

RESUMEN

Temporal expression of chemokines is a crucial factor in the regulation of renal ischemia/reperfusion (I/R) injury and repair. Beside their role in the migration and activation of inflammatory cells to sites of injury, chemokines are also involved in other processes such as angiogenesis, development and migration of stem cells. In the present study we investigated the role of the chemokine MCP-1 (monocyte chemoattractant protein-1 or CCL2), the main chemoattractant for monocytes, during renal I/R injury. MCP-1 expression peaks several days after inducing renal I/R injury coinciding with macrophage accumulation. However, MCP-1 deficient mice had a significant decreased survival and increased renal damage within the first two days, i.e. the acute inflammatory response, after renal I/R injury with no evidence of altered macrophage accumulation. Kidneys and primary tubular epithelial cells from MCP-1 deficient mice showed increased apoptosis after ischemia. Taken together, MCP-1 protects the kidney during the acute inflammatory response following renal I/R injury.


Asunto(s)
Quimiocina CCL2/genética , Túbulos Renales/metabolismo , Daño por Reperfusión/genética , Animales , Apoptosis/genética , Quimiocina CCL2/deficiencia , Quimiocina CCL2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Genes Letales , Túbulos Renales/patología , Leucocitos/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Peroxidasa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Factores de Tiempo , Regulación hacia Arriba
7.
Am J Pathol ; 184(7): 2013-22, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24823805

RESUMEN

Ischemia/reperfusion injury is a major cause of acute kidney injury. Improving renal repair would represent a therapeutic strategy to prevent renal dysfunction. The innate immune receptor Nlrp3 is involved in tissue injury, inflammation, and fibrosis; however, its role in repair after ischemia/reperfusion is unknown. We address the role of Nlrp3 in the repair phase of renal ischemia/reperfusion and investigate the relative contribution of leukocyte- versus renal-associated Nlrp3 by studying bone marrow chimeric mice. We found that Nlrp3 expression was most profound during the repair phase. Although Nlrp3 expression was primarily expressed by leukocytes, both leukocyte- and renal-associated Nlrp3 was detrimental to renal function after ischemia/reperfusion. The Nlrp3-dependent cytokine IL-1ß remained unchanged in kidneys of all mice. Leukocyte-associated Nlrp3 negatively affected tubular apoptosis in mice that lacked Nlrp3 expression on leukocytes, which correlated with reduced macrophage influx. Nlrp3-deficient (Nlrp3KO) mice with wild-type bone marrow showed an improved repair response, as seen by a profound increase in proliferating tubular epithelium, which coincided with increased hepatocyte growth factor expression. In addition, Nlrp3KO tubular epithelial cells had an increased repair response in vitro, as seen by an increased ability of an epithelial monolayer to restore its structural integrity. In conclusion, Nlrp3 shows a tissue-specific role in which leukocyte-associated Nlrp3 is associated with tubular apoptosis, whereas renal-associated Nlrp3 impaired wound healing.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Epiteliales/citología , Túbulos Renales/patología , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Apoptosis , Trasplante de Médula Ósea , Hipoxia de la Célula , Proliferación Celular , Interleucina-1beta/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Leucocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
8.
Kidney Int ; 86(3): 558-69, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24717295

RESUMEN

Chronic kidney diseases (CKDs) are characterized by tubular atrophy and interstitial fibrosis. We previously showed that in obstructive nephropathy de novo CD44 renal expression contributes to renal fibrosis but attenuates tubular damage/apoptosis. As CD44-standard (CD44s) has been linked to TGF-ß1-mediated actions and CD44-variant-3 (CD44v3) favors HGF-c-Met binding, we compared the functional properties of these CD44 isoforms in the progression of obstructive nephropathy, using specific CD44-variant knockout/knockin mice. The presence of CD44v3 diminished tubular damage during obstructive nephropathy, decreased apoptosis, and increased proliferation of tubular epithelial cells, and prevented renal fibrosis development. In contrast, expression of CD44s led to increased tubular damage and tubular epithelial cell apoptosis, and more renal fibrosis. A relative increase in renal ß-catenin expression, HGF production, and HGF/c-Met signaling, together with a relative inhibition of TGF-ß1 downstream signaling and TGF-ß type I receptor expression, was found in CD44v3 mice compared with CD44s littermates. In line with this, Wnt3a/HGF treatment of tubular cells resulted in higher ß-catenin/p-AKT levels in CD44v3(+) tubular epithelial cells, whereas TGF-ß1 induced a mild collagen I upregulation in CD44v3(+) mouse embryonic fibroblasts as compared with CD44s(+) cells. Thus, CD44s and CD44v3 exert opposite roles in the progression of obstructive nephropathy, with CD44v3-v10 being the protective isoform that delays evolution of the renal pathology.


Asunto(s)
Receptores de Hialuranos/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Animales , Apoptosis , Enfermedad Crónica , Colágeno Tipo I/metabolismo , Células Epiteliales , Fibroblastos/metabolismo , Fibrosis , Factor de Crecimiento de Hepatocito/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Receptores de Hialuranos/genética , Enfermedades Renales/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Isoformas de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Obstrucción Ureteral/complicaciones , Proteína Wnt3A/farmacología , beta Catenina/metabolismo
9.
J Am Soc Nephrol ; 25(7): 1474-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24511123

RESUMEN

Activation of Rap1 by exchange protein activated by cAMP (Epac) promotes cell adhesion and actin cytoskeletal polarization. Pharmacologic activation of Epac-Rap signaling by the Epac-selective cAMP analog 8-pCPT-2'-O-Me-cAMP during ischemia-reperfusion (IR) injury reduces renal failure and application of 8-pCPT-2'-O-Me-cAMP promotes renal cell survival during exposure to the nephrotoxicant cisplatin. Here, we found that activation of Epac by 8-pCPT-2'-O-Me-cAMP reduced production of reactive oxygen species during reoxygenation after hypoxia by decreasing mitochondrial superoxide production. Epac activation prevented disruption of tubular morphology during diethyl maleate-induced oxidative stress in an organotypic three-dimensional culture assay. In vivo renal targeting of 8-pCPT-2'-O-Me-cAMP to proximal tubules using a kidney-selective drug carrier approach resulted in prolonged activation of Rap1 compared with nonconjugated 8-pCPT-2'-O-Me-cAMP. Activation of Epac reduced antioxidant signaling during IR injury and prevented tubular epithelial injury, apoptosis, and renal failure. Our data suggest that Epac1 decreases reactive oxygen species production by preventing mitochondrial superoxide formation during IR injury, thus limiting the degree of oxidative stress. These findings indicate a new role for activation of Epac as a therapeutic application in renal injury associated with oxidative stress.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Túbulos Renales Proximales/metabolismo , Estrés Oxidativo , Urotelio/metabolismo , Animales , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Factores de Intercambio de Guanina Nucleótido/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Urotelio/efectos de los fármacos
10.
Kidney Int ; 85(5): 1112-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24352154

RESUMEN

Metabolic syndrome (MetSyn) is a major health concern and associates with the development of kidney disease. The mechanisms linking MetSyn to renal disease have not been fully elucidated but are known to involve hyperuricemia, inflammation, and fibrosis. Since the innate immune receptor Nlrp3 is an important mediator of obesity and inflammation, we sought to determine whether Nlrp3 is involved in the development of MetSyn-associated nephropathy by giving wild-type or Nlrp3-knockout mice a Western-style compared to a normal diet or water without or with fructose. A plausible driver of pathology, the Nlrp3-dependent cytokine IL-1ß was not increased in the kidney. Interestingly, Nlrp3-dependent renal cholesterol accumulation, another well-known driver of renal pathology, was enhanced during MetSyn. We also determined the role of Nlrp3 and fructose-fortified water on the development of MetSyn and kidney function since fructose is an important driver of obesity and kidney disease. Surprisingly, fructose did not induce MetSyn but, irrespective of this, did induce Nlrp3-dependent renal inflammation. The presence of Nlrp3 was crucial for the development of Western-style diet-induced renal pathology as reflected by the prevention of renal inflammation, fibrosis, steatosis, microalbuminuria, and hyperuricemia in the Nlrp3-knockout mice. Thus, Nlrp3 may mediate renal pathology in the context of diet-induced MetSyn.


Asunto(s)
Proteínas Portadoras/metabolismo , Colesterol en la Dieta/metabolismo , Dieta Alta en Grasa , Dieta Occidental , Enfermedades Renales/metabolismo , Riñón/metabolismo , Síndrome Metabólico/metabolismo , Transducción de Señal , Animales , Biomarcadores/sangre , Proteínas Portadoras/genética , Carbohidratos de la Dieta/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Fructosa/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Riñón/inmunología , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/genética , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/genética , Síndrome Metabólico/inmunología , Síndrome Metabólico/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de LDL/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
11.
PLoS One ; 8(12): e84479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376813

RESUMEN

Acute kidney injury (AKI) is a common complication during systemic inflammatory response syndrome (SIRS), a potentially deadly clinical condition characterized by whole-body inflammatory state and organ dysfunction. CD44 is a ubiquitously expressed cell-surface transmembrane receptor with multiple functions in inflammatory processes, including sterile renal inflammation. The present study aimed to assess the role of CD44 in endotoxic shock-induced kidney inflammation and dysfunction by using CD44 KO and WT mice exposed intraperitoneally to LPS for 2, 4, and 24 hours . Upon LPS administration, CD44 expression in WT kidneys was augmented at all time-points. At 2 and 4 hours, CD44 KO animals showed a preserved renal function in comparison to WT mice. In absence of CD44, the pro-inflammatory cytokine levels in plasma and kidneys were lower, while renal expression of the anti-inflammatory cytokine IL-10 was higher. The cytokine levels were associated with decreased leukocyte influx and endothelial activation in CD44 KO kidneys. Furthermore, in vitro assays demonstrated a role of CD44 in enhancing macrophage cytokine responses to LPS and leukocyte migration. In conclusion, our study demonstrates that lack of CD44 impairs the early pro-inflammatory cytokine response to LPS, diminishes leukocyte migration/chemotaxis and endothelial activation, hence, delays endotoxic shock-induced AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Receptores de Hialuranos/inmunología , Inflamación/prevención & control , Lipopolisacáridos/inmunología , Choque Séptico/inmunología , Lesión Renal Aguda/etiología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Receptores de Hialuranos/genética , Immunoblotting , Inmunohistoquímica , Inflamación/etiología , Interleucina-10/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Choque Séptico/complicaciones , Estadísticas no Paramétricas , Factores de Tiempo
12.
Am J Physiol Renal Physiol ; 305(10): F1445-54, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24026183

RESUMEN

CD44 family members are cell surface glycoproteins, which are expressed on tubular epithelial cells (TEC) solely upon kidney injury and are involved in renal fibrosis development. Renal interstitial fibrosis is the final manifestation of chronic kidney diseases and is regulated by a complex network of cytokines, including the profibrotic factor transforming growth factor-ß1 (TGF-ß1) and the two antifibrotic cytokines bone morphogenic protein-7 (BMP-7) and hepatocyte growth factor (HGF). The present study investigates the potential role of CD44 standard (CD44s) and CD44v3-v10 (CD44v3) isoforms as modulators of the balance between TGF-ß1 and HGF/BMP-7. CD44s is the shortest and most common isoform. CD44v3-v10 (CD44v3) has heparan sulfate moieties, which enable the binding to HGF/BMP-7, and hence, might exert renoprotective effects. Using transgenic mice overexpressing either CD44s or CD44v3 specifically on proximal TEC, we found that in vitro the overexpression of CD44v3 on primary TEC renders cells less susceptible to TGF-ß1 profibrotic actions and more sensitive to BMP-7 and HGF compared with TEC overexpressing CD44s. One day after unilateral ureteric obstruction, obstructed kidneys from CD44v3 transgenic mice showed less tubular damage and myofibroblasts accumulation, which was associated with decreased TGF-ß1 signaling and increased BMP-7 synthesis and signaling compared with kidneys from wild-type and CD44s transgenic mice. These data suggest that CD44v3 plays a renoprotective role in early stage of chronic obstructive nephropathy.


Asunto(s)
Receptores de Hialuranos/metabolismo , Túbulos Renales Proximales/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/prevención & control , Animales , Proteína Morfogenética Ósea 7/metabolismo , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Fibrosis , Factor de Crecimiento de Hepatocito/metabolismo , Receptores de Hialuranos/genética , Mediadores de Inflamación/metabolismo , Túbulos Renales Proximales/inmunología , Túbulos Renales Proximales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba , Obstrucción Ureteral/genética , Obstrucción Ureteral/inmunología , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
13.
Infect Immun ; 80(11): 3812-20, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22890991

RESUMEN

Our immune system has to constantly strike a balance between activation and inhibition of an inflammatory response to combat invading pathogens and avoid inflammation-induced collateral tissue damage. Toll interleukin-1 receptor 8 (IL-1R-8)/single Ig domain IL-1R-related molecule (TIR8/SIGIRR) is an inhibitor of Toll-like receptor (TLR)/IL-1R signaling, which is predominantly expressed in the kidney. The biological role of renal TIR8 during infection is, however, unknown. We therefore evaluated renal TIR8 expression during Escherichia coli pyelonephritis and explored its role in host defense using TIR8(-/-) versus TIR8(+/+) mice. We found that TIR8 protein is abundantly present in the majority of cortical tubular epithelial cells. Pyelonephritis resulted in a significant downregulation of TIR8 mRNA in kidneys of TIR8(+/+) mice. TIR8 inhibited an effective host response against E. coli, as indicated by diminished renal bacterial outgrowth and dysfunction in TIR8(-/-) mice. This correlated with increased amounts of circulating and intrarenal neutrophils at the early phase of infection. TIR8(-/-) tubular epithelial cells had increased cytokine/chemokine production when stimulated with lipopolysaccharide (LPS) or heat-killed E. coli, suggesting that TIR8 played an anti-inflammatory role during pathogen stimulation by inhibiting LPS signaling. These data suggest that TIR8 is an important negative regulator of an LPS-mediated inflammatory response in tubular epithelial cells and dampens an effective antibacterial host response during pyelonephritis caused by uropathogenic E. coli.


Asunto(s)
Inflamación/metabolismo , Túbulos Renales/inmunología , Pielonefritis/inmunología , Receptores de Interleucina-1/metabolismo , Animales , Células Cultivadas , Quimiocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Escherichia coli , Infecciones por Escherichia coli , Femenino , Inmunohistoquímica , Túbulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/metabolismo , Pielonefritis/metabolismo , Pielonefritis/microbiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
14.
J Clin Invest ; 122(1): 348-58, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22201679

RESUMEN

Podocytes of the kidney adhere tightly to the underlying glomerular basement membrane (GBM) in order to maintain a functional filtration barrier. The clinical importance of podocyte binding to the GBM via an integrin-laminin-actin axis has been illustrated in models with altered function of α3ß1 integrin, integrin-linked kinase, laminin-521, and α-actinin 4. Here we expanded on the podocyte-GBM binding model by showing that the main podocyte adhesion receptor, integrin α3ß1, interacts with the tetraspanin CD151 in situ in humans. Deletion of Cd151 in mouse glomerular epithelial cells led to reduced adhesive strength to laminin by redistributing α3ß1 at the cell-matrix interface. Moreover, in vivo podocyte-specific deletion of Cd151 led to glomerular nephropathy. Although global Cd151-null B6 mice were not susceptible to renal disease, as has been shown previously, increasing blood and transcapillary filtration pressure induced nephropathy in these mice. Importantly, blocking the angiotensin-converting enzyme in renal disease-susceptible global Cd151-null FVB mice prolonged their median life span. Together, these results establish CD151 as a crucial modifier of integrin-mediated adhesion of podocytes to the GBM and show that blood pressure is an important factor in the initiation and progression of Cd151 knockout-induced nephropathy.


Asunto(s)
Presión Sanguínea/fisiología , Fallo Renal Crónico/fisiopatología , Tetraspanina 24/deficiencia , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Células Epiteliales/patología , Células Epiteliales/fisiología , Membrana Basal Glomerular/patología , Membrana Basal Glomerular/fisiopatología , Humanos , Integrina alfa3beta1/fisiología , Fallo Renal Crónico/tratamiento farmacológico , Fallo Renal Crónico/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , Podocitos/fisiología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Tetraspanina 24/genética , Tetraspanina 24/metabolismo
15.
Nephrol Dial Transplant ; 25(12): 3852-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20519232

RESUMEN

BACKGROUND: The chemokine stromal cell-derived factor-1 (SDF-1) is thought to be involved in mediating tissue repair by promoting migration of bone marrow stem or progenitor cells to the site of injury. Increased levels of renal SDF-1 are found after kidney injury. However, recently, we showed that SDF-1 does not play an important role in the migration of haematopoietic stem cells to the post-ischaemic kidney. The function of increased post-ischaemic renal SDF-1 expression in modulating renal ischaemia/reperfusion injury remains, therefore, unknown. METHODS: We studied the role of SDF-1 in renal ischaemia/reperfusion injury by locally decreasing SDF-1 expression and subsequent SDF-1 signalling in the corticomedullary region of the kidney using antisense oligonucleotide treatment in mice. RESULTS: Renal SDF-1 protein increased significantly in the early phase of ischaemia/reperfusion injury. Antisense treatment resulted in a reduction of corticomedullary SDF-1 expression which was accompanied by severely increased tubular injury and decreased renal function. We did not observe any difference in mobilization or retention of CXCR4-positive haematopoietic stem or progenitor cells after induction of renal ischaemia. Rather, antisense-treated animals showed markedly increased apoptosis of the tubular epithelium accompanied by an increased renal inflammatory response. Conclusions. These data indicate a new role for SDF-1 in renal pathogenesis by mediating tubular epithelial protection against ischaemic injury and suggest that SDF-1 by itself is not crucial for the influx of haematopoietic stem or progenitor cells towards the ischaemic injured kidney.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/prevención & control , Quimiocina CXCL12/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/patología , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/fisiología , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Corteza Renal/patología , Médula Renal/efectos de los fármacos , Médula Renal/metabolismo , Médula Renal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Oligonucleótidos Antisentido/farmacología , Daño por Reperfusión/patología , Transducción de Señal/fisiología
16.
PLoS One ; 5(12): e14386, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21200435

RESUMEN

BACKGROUND: Renal ischemia leads to apoptosis of tubular epithelial cells and results in decreased renal function. Tissue repair involves re-epithelialization of the tubular basement membrane. Survival of the tubular epithelium following ischemia is therefore important in the successful regeneration of renal tissue. The cytokine stem cell factor (SCF) has been shown to protect the tubular epithelium against apoptosis. METHODOLOGY/PRINCIPAL FINDINGS: In a mouse model for renal ischemia/reperfusion injury, we studied how expression of c-KIT on tubular epithelium and its ligand SCF protect cells against apoptosis. Administration of SCF specific antisense oligonucleotides significantly decreased specific staining of SCF following ischemia. Reduced SCF expression resulted in impaired renal function, increased tubular damage and increased tubular epithelial apoptosis, independent of inflammation. In an in vitro hypoxia model, stimulation of tubular epithelial cells with SCF activated survival signaling and decreased apoptosis. CONCLUSIONS/SIGNIFICANCE: Our data indicate an important role for c-KIT and SCF in mediating tubular epithelial cell survival via an autocrine pathway.


Asunto(s)
Regulación de la Expresión Génica , Túbulos Renales/metabolismo , Riñón/patología , Factor de Células Madre/biosíntesis , Animales , Apoptosis , Membrana Basal/metabolismo , Supervivencia Celular , Células Epiteliales/citología , Isquemia/patología , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteínas Proto-Oncogénicas c-kit/metabolismo
17.
J Infect Dis ; 200(7): 1162-5, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19702507

RESUMEN

The role that the enterococcal surface protein Esp plays in the capacity of Enterococcus faecium to adhere to uroepithelial cells and the role that it plays in urinary tract infection and peritonitis was investigated in vitro and in vivo, respectively, using Esp-expressing E. faecium (E1162) and its isogenic Esp-deficient mutant (E1162 Delta esp). Esp expression enhanced in vitro binding to bladder and kidney epithelial cells. In mice, higher numbers of E1162 were cultured from kidneys and bladders after the induction of urinary tract infection, compared with E1162 Delta esp numbers. This was accompanied by a higher frequency of bacteremia, higher cytokine levels in kidney tissue, and renal insufficiency. Esp had no effect on the course of E. faecium peritonitis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Enterococcus faecium/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/toxicidad , Infecciones Urinarias/microbiología , Animales , Adhesión Bacteriana , Proteínas Bacterianas/genética , Enterococcus faecium/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas de la Membrana/genética , Ratones , Unión Proteica
18.
PLoS One ; 4(5): e5704, 2009 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-19479087

RESUMEN

Tissue fibrosis and chronic inflammation are common causes of progressive organ damage, including progressive renal disease, leading to loss of physiological functions. Recently, it was shown that Toll-like receptor 2 (TLR2) is expressed in the kidney and activated by endogenous danger signals. The expression and function of TLR2 during renal fibrosis and chronic inflammation has however not yet been elucidated. Therefore, we studied TLR2 expression in human and murine progressive renal diseases and explored its role by inducing obstructive nephropathy in TLR2(-/-) or TLR2(+/+) mice. We found that TLR2 is markedly upregulated on tubular and tubulointerstitial cells in patients with chronic renal injury. In mice with obstructive nephropathy, renal injury was associated with a marked upregulation and change in distribution of TLR2 and upregulation of murine TLR2 danger ligands Gp96, biglycan, and HMGB1. Notably, TLR2 enhanced inflammation as reflected by a significantly reduced influx of neutrophils and production of chemokines and TGF-beta in kidneys of TLR2(-/-) mice compared with TLR2(+/+) animals. Although, the obstructed kidneys of TLR2(-/-) mice had less interstitial myofibroblasts in the later phase of obstructive nephropathy, tubular injury and renal matrix accumulation was similar in both mouse strains. Together, these data demonstrate that TLR2 can initiate renal inflammation during progressive renal injury and that the absence of TLR2 does not affect the development of chronic renal injury and fibrosis.


Asunto(s)
Inflamación/inmunología , Inflamación/patología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Receptor Toll-Like 2/metabolismo , Animales , Apoptosis , Proliferación Celular , Quimiocinas/metabolismo , Progresión de la Enfermedad , Activación Enzimática , Matriz Extracelular/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Fibrosis , Humanos , Riñón/enzimología , Riñón/patología , Enfermedades Renales/enzimología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Ligandos , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor Toll-Like 2/deficiencia , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/patología
19.
Nephrol Dial Transplant ; 24(7): 2082-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19223274

RESUMEN

BACKGROUND: Haematopoietic stem cells (HSC) have been shown to migrate to the ischemic kidney. The factors that regulate the trafficking of HSC to the ischemic damaged kidney are not fully understood. The stromal cell-derived factor-1 (SDF-1)/CXCR4-axis has been identified as the central signalling axis regulating trafficking of HSC to the bone marrow. Therefore, we hypothesized that SDF-1/CXCR4 interactions are implicated in the migration of HSC to the injured kidney. METHODS: HSC were isolated from mouse bone marrow and labelled with a cell tracker. Acceptor mice were subjected to unilateral ischemia and received HSC intravenously directly after reperfusion. In addition, in separate groups of acceptor mice, endogenous SDF-1 or HSC-associated CXCR4 was blocked or kidneys were injected with SDF-1. RESULTS: Exogenous HSC could be detected in the tubules and interstitium of the kidney 24 h after ischemic injury. Importantly, the amount of HSC in the ischemic kidney was markedly higher compared to the contralateral kidney. Neutralizing endogenous SDF-1 or HSC-associated CXCR4 did not prevent the migration of HSC. No increase in the number of labelled HSC could be observed after local administration of SDF-1, as was also determined in bilateral kidney ischemia. CONCLUSION: In conclusion, systemically administered HSC preferentially migrate to the ischemic injured kidney. This migration could not be prevented by blocking the SDF-1/CXCR4-axis or increased after local administration of SDF-1.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/fisiología , Células Madre Hematopoyéticas/fisiología , Riñón/irrigación sanguínea , Riñón/citología , Receptores CXCR4/fisiología , Daño por Reperfusión , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/fisiopatología
20.
Nephrol Dial Transplant ; 24(3): 801-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18842674

RESUMEN

BACKGROUND: Acute pyelonephritis is one of the most common bacterial infections. Tissue-type plasminogen activator (tPA) is a potent fibrinolytic agent, but can play a role in inflammatory processes as well. METHODS: We induced pyelonephritis in tPA(-/-) and C57BL/6 wild-type (WT) mice by intravesical inoculation with 10(10) CFU uropathogenic Escherichia coli 1677. The mice were killed after 24 and 48 h, after which bacterial outgrowth and cytokine levels in kidney homogenates were determined. Influx of neutrophils was quantified by myeloperoxidase-ELISA. Neutrophil phagocytosis and oxidative burst were measured. RESULTS: The tPA(-/-) kidneys contained significantly higher numbers of E. coli CFU, accompanied by higher levels of interleukin-1beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha). The number of infiltrating neutrophils was similar in tPA(-/-) and WT mice at both time points, suggesting that tPA(-/-) neutrophils have a lower ability to eliminate E. coli. Phagocytosis of E. coli organisms was not diminished in tPA(-/-) neutrophils. Interestingly, tPA(-/-) neutrophils showed a significantly lower ability to generate an oxidative burst reaction upon stimulation with E. coli than WT neutrophils. Incubation with recombinant tPA reversed this effect completely. CONCLUSIONS: These results show that deletion of the tPA-gene in mice leads to lower bactericidal potential of tPA(-/-) neutrophils, which results in significantly more bacterial outgrowth during experimental pyelonephritis.


Asunto(s)
Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/metabolismo , Pielonefritis/etiología , Pielonefritis/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Fagocitosis , Pielonefritis/patología , ARN Mensajero/metabolismo , Estallido Respiratorio/fisiología , Activador de Tejido Plasminógeno/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...