Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Pathol ; 263(4-5): 496-507, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38934262

RESUMEN

Chronic kidney disease (CKD) has emerged as a significant global public health concern. Recent epidemiological studies have highlighted the link between exposure to fine particulate matter (PM2.5) and a decline in renal function. PM2.5 exerts harmful effects on various organs through oxidative stress and inflammation. Acute kidney injury (AKI) resulting from ischaemia-reperfusion injury (IRI) involves biological processes similar to those involved in PM2.5 toxicity and is a known risk factor for CKD. The objective of this study was to investigate the impact of PM2.5 exposure on IRI-induced AKI. Through a unique environmentally controlled setup, mice were exposed to urban PM2.5 or filtered air for 12 weeks before IRI followed by euthanasia 48 h after surgery. Animals exposed to PM2.5 and IRI exhibited reduced glomerular filtration, impaired urine concentration ability, and significant tubular damage. Further, PM2.5 aggravated local innate immune responses and mitochondrial dysfunction, as well as enhancing cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway activation. This increased renal senescence and suppressed the anti-ageing protein klotho, leading to early fibrotic changes. In vitro studies using proximal tubular epithelial cells exposed to PM2.5 and hypoxia/reoxygenation revealed heightened activation of the STING pathway triggered by cytoplasmic mitochondrial DNA, resulting in increased tubular damage and a pro-inflammatory phenotype. In summary, our findings imply a role for PM2.5 in sensitising proximal tubular epithelial cells to IRI-induced damage, suggesting a plausible association between PM2.5 exposure and heightened susceptibility to CKD in individuals experiencing AKI. Strategies aimed at reducing PM2.5 concentrations and implementing preventive measures may improve outcomes for AKI patients and mitigate the progression from AKI to CKD. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Lesión Renal Aguda , Ratones Endogámicos C57BL , Material Particulado , Daño por Reperfusión , Animales , Lesión Renal Aguda/patología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Daño por Reperfusión/patología , Material Particulado/efectos adversos , Material Particulado/toxicidad , Ratones , Masculino , Contaminación del Aire/efectos adversos , Modelos Animales de Enfermedad , Riñón/patología , Riñón/metabolismo , Transducción de Señal , Tasa de Filtración Glomerular
2.
Eur J Immunol ; 53(11): e2350562, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37597325

RESUMEN

Levamisole (LMS) is a small molecule used in the treatment of idiopathic nephrotic syndrome (INS). The pathogenesis of INS remains unknown, but evidence points toward an immunological basis of the disease. Recently, LMS has been shown to increase the relapse-free survival in INS patients. While LMS has been hypothesized to exert an immunomodulatory effect, its mechanism of action remains unknown. Here, we show that LMS decreased activation and proliferation of human T cells. T-cell activation-associated cytokines such as IL-2, TNF-α, and IFN-γ were reduced upon LMS treatment, whereas IL-4 and IL-13 were increased. Gene expression profiling confirmed that the suppressive effects of LMS as genes involved in cell cycle progression were downregulated. Furthermore, genes associated with p53 activation were upregulated by LMS. In agreement, LMS treatment resulted in p53 phosphorylation and increased expression of the p53 target gene FAS. Accordingly, LMS sensitized activated T cells for Fas-mediated apoptosis. LMS treatment resulted in a mid-S phase cell cycle arrest accompanied by γH2AX-foci formation and phosphorylation of CHK1. Our findings indicate that LMS acts as an immunosuppressive drug that directly affects the activation and proliferation of human T cells by induction of DNA damage and the activation of a p53-dependent DNA damage response.


Asunto(s)
Levamisol , Proteína p53 Supresora de Tumor , Humanos , Levamisol/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , División Celular , Apoptosis , Linfocitos T , Daño del ADN
3.
Cells ; 13(1)2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38201227

RESUMEN

BACKGROUND: Chronic kidney disease often leads to kidney dysfunction due to renal fibrosis, regardless of the initial cause of kidney damage. Macrophages are crucial players in the progression of renal fibrosis as they stimulate inflammation, activate fibroblasts, and contribute to extracellular matrix deposition, influenced by their metabolic state. Nucleotide-binding domain and LRR-containing protein X (NLRX1) is an innate immune receptor independent of inflammasomes and is found in mitochondria, and it plays a role in immune responses and cell metabolism. The specific impact of NLRX1 on macrophages and its involvement in renal fibrosis is not fully understood. METHODS: To explore the specific role of NLRX1 in macrophages, bone-marrow-derived macrophages (BMDMs) extracted from wild-type (WT) and NLRX1 knockout (KO) mice were stimulated with pro-inflammatory and pro-fibrotic factors to induce M1 and M2 polarization in vitro. The expression levels of macrophage polarization markers (Nos2, Mgl1, Arg1, and Mrc1), as well as the secretion of transforming growth factor ß (TGFß), were measured using RT-PCR and ELISA. Seahorse-based bioenergetics analysis was used to assess mitochondrial respiration in naïve and polarized BMDMs obtained from WT and NLRX1 KO mice. In vivo, WT and NLRX1 KO mice were subjected to unilateral ureter obstruction (UUO) surgery to induce renal fibrosis. Kidney injury, macrophage phenotypic profile, and fibrosis markers were assessed using RT-PCR. Histological staining (PASD and Sirius red) was used to quantify kidney injury and fibrosis. RESULTS: Compared to the WT group, an increased gene expression of M2 markers-including Mgl1 and Mrc1-and enhanced TGFß secretion were found in naïve BMDMs extracted from NLRX1 KO mice, indicating functional polarization towards the pro-fibrotic M2 subtype. NLRX1 KO naïve macrophages also showed a significantly enhanced oxygen consumption rate compared to WT cells and increased basal respiration and maximal respiration capacities that equal the level of M2-polarized macrophages. In vivo, we found that NLRX1 KO mice presented enhanced M2 polarization markers together with enhanced tubular injury and fibrosis demonstrated by augmented TGFß levels, fibronectin, and collagen accumulation. CONCLUSIONS: Our findings highlight the unique role of NLRX1 in regulating the metabolism and function of macrophages, ultimately protecting against excessive renal injury and fibrosis in UUO.


Asunto(s)
Insuficiencia Renal Crónica , Animales , Ratones , Macrófagos , Genes Reguladores , Fibrosis , Factor de Crecimiento Transformador beta , Proteínas Mitocondriales
4.
Int J Mol Sci ; 22(5)2021 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33802460

RESUMEN

The lysosomal storage disease Niemann-Pick type C (NPC) is caused by impaired cholesterol efflux from lysosomes, which is accompanied by secondary lysosomal accumulation of sphingomyelin and glucosylceramide (GlcCer). Similar to Gaucher disease (GD), patients deficient in glucocerebrosidase (GCase) degrading GlcCer, NPC patients show an elevated glucosylsphingosine and glucosylated cholesterol. In livers of mice lacking the lysosomal cholesterol efflux transporter NPC1, we investigated the expression of established biomarkers of lipid-laden macrophages of GD patients, their GCase status, and content on the cytosol facing glucosylceramidase GBA2 and lysosomal integral membrane protein type B (LIMP2), a transporter of newly formed GCase to lysosomes. Livers of 80-week-old Npc1-/- mice showed a partially reduced GCase protein and enzymatic activity. In contrast, GBA2 levels tended to be reciprocally increased with the GCase deficiency. In Npc1-/- liver, increased expression of lysosomal enzymes (cathepsin D, acid ceramidase) was observed as well as increased markers of lipid-stressed macrophages (GPNMB and galectin-3). Immunohistochemistry showed that the latter markers are expressed by lipid laden Kupffer cells. Earlier reported increase of LIMP2 in Npc1-/- liver was confirmed. Unexpectedly, immunohistochemistry showed that LIMP2 is particularly overexpressed in the hepatocytes of the Npc1-/- liver. LIMP2 in these hepatocytes seems not to only localize to (endo)lysosomes. The recent recognition that LIMP2 harbors a cholesterol channel prompts the speculation that LIMP2 in Npc1-/- hepatocytes might mediate export of cholesterol into the bile and thus protects the hepatocytes.


Asunto(s)
Glucosilceramidasa/metabolismo , Hígado/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Receptores Depuradores/metabolismo , Animales , Transporte Biológico/fisiología , Catepsina D/metabolismo , Línea Celular , Línea Celular Tumoral , Enfermedad de Gaucher/metabolismo , Glucosilceramidas/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Esfingomielinas/metabolismo
5.
FASEB J ; 34(10): 13750-13761, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32856376

RESUMEN

Diabetic nephropathy (DN) is a major complication of diabetes and is associated with high risk for cardiovascular mortality, which is partially related to elevated platelet activity. Platelets are also active players in inflammation and fibrosis. In this study, we examine the effect of ticagrelor-induced platelet inhibition on the development of DN. DN was induced by unilateral nephrectomy followed by streptozotocin injections for 5 days. Mice received ticagrelor (300 mg/kg) or vehicle every other day, for 16 weeks. Experimental groups: non-diabetic control, diabetic control, non-diabetic ticagrelor, and diabetic ticagrelor. Ticagrelor treatment in diabetic mice lowered urinary albumin excretion, it prevented diabetes-induced mesangial matrix expansion, podocyte effacement, and glomerular endothelial cell injury, which includes loss of endothelial fenestrations, ICAM-1 expression, and PECAM expression. In addition, ticagrelor treatment prevented collagen IV deposition and macrophage infiltration in the tubulointerstitium and these diabetic mice showed lower systemic and tubular inflammation and tubular apoptosis. This tubular protection is likely to be a result of protection to the glomerular endothelium by ticagrelor, which reduces albuminuria and albumin toxicity to the tubules and reduced tubular and interstitial inflammation and fibrosis. In conclusion, ticagrelor-induced platelet inhibition protects against renal injury in diabetic mice, likely by protecting the glomerular endothelial cells.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Ticagrelor/uso terapéutico , Animales , Apoptosis , Colágeno/metabolismo , Nefropatías Diabéticas/etiología , Células Endoteliales/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/metabolismo , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Podocitos/efectos de los fármacos , Ticagrelor/administración & dosificación , Ticagrelor/farmacología
6.
Transl Res ; 217: 23-32, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31883728

RESUMEN

Type 1 diabetes patients are more prone to have hypertension than healthy individuals, possibly mediated by increased blood pressure (BP) sensitivity to high salt intake. The classical concept proposes that the kidney is central in salt-mediated BP rises, by insufficient renal sodium excretion leading to extracellular fluid volume expansion. Recent animal-derived findings, however, propose a causal role for disturbance of macrophage-mediated lymphangiogenesis. Its relevance for humans, specifically type 1 diabetes patients, is unknown. The present study aimed to assess responses of type 1 diabetes patients to a dietary salt load with regard to BP, extracellular fluid volume (using precise iohexol measurements), and CD163+ macrophage and lymphatic capillary density in skin biopsies. Also, macrophage expression of HLA-DR (a proinflammatory marker) and CD206 (an anti-inflammatory marker) was assessed. Type 1 diabetes patients (n = 8) showed a salt-sensitive BP increase without extracellular fluid volume expansion. Whereas healthy controls (n = 12), who had no BP increase, showed increased skin CD163+ and HLA-DR+ macrophages and dilation of lymphatic skin vasculature after the dietary salt load, these changes were absent (and in case of HLA-DR more heterogenic) in type 1 diabetes patients. In conclusion, we show that salt sensitivity in type 1 diabetes patients cannot be explained by the classical concept of extracellular fluid volume expansion. Rather, we open up a potential role for macrophages and the lymphatic system. Future studies on hypertension and diabetes need to scrutinize these phenomena.


Asunto(s)
Presión Sanguínea/fisiología , Diabetes Mellitus Tipo 1/fisiopatología , Líquido Extracelular/fisiología , Vasos Linfáticos/fisiología , Macrófagos/fisiología , Piel/inmunología , Cloruro de Sodio Dietético/administración & dosificación , Adolescente , Adulto , Estudios Cruzados , Humanos , Masculino , Estudios Prospectivos , Piel/irrigación sanguínea , Adulto Joven
7.
Platelets ; 31(3): 383-391, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31364433

RESUMEN

The pathophysiology of renal ischemia/reperfusion (I/R) injury is characterized by excessive activation of inflammation and coagulation processes followed by abnormal renal tissue repair, resulting in renal injury and function loss. Platelets are important actors in these processes, however to what extent platelets contribute to the pathophysiology of renal I/R injury still needs to be elucidated. In the current study, we treated wild-type mice with a platelet depleting antibody, which caused thrombocytopenia. We then investigated the role of platelets during the pathophysiology of renal I/R by subjecting control wild-type mice with normal platelet counts and thrombocytopenic wild-type mice to renal I/R injury. Our results showed that in the early phase of renal I/R injury, thrombocytopenia 24 hours after ischemia reperfusion does not influence renal injury, neutrophil infiltration and accumulation of inflammatory chemokines (e.g. keratinocyte chemoattractant, monocyte chemoattractant protein 1, tumor necrosis factor alpha). In the recovery and regeneration phase of I/R injury, respectively 5 and 10 days post-ischemia, thrombocytopenia did also not affect the accumulation of intra-renal neutrophils and macrophages, renal injury, and renal fibrosis. Together, these results imply that lowering platelet counts do not impact the pathogenesis of I/R injury in mice.


Asunto(s)
Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Daño por Reperfusión/complicaciones , Trombocitopenia/complicaciones , Animales , Biomarcadores , Modelos Animales de Enfermedad , Inmunohistoquímica , Inmunofenotipificación , Masculino , Ratones , Daño por Reperfusión/etiología , Trombocitopenia/etiología
8.
Nephrol Dial Transplant ; 35(8): 1320-1327, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30590723

RESUMEN

BACKGROUND: Ischaemia-reperfusion (IR) injury is an important determinant of delayed graft function (DGF) affecting allograft function. Mitochondrial DNA (mtDNA) is released upon cell death and platelet activation into the extracellular environment and has been suggested to be a biomarker in several diseases. Whether extracellular mtDNA accumulates in plasma and/or urine upon renal IR and predisposes DGF is unknown. METHODS: C57BL/6J wild-type mice were subjected to renal IR. In addition, an observational case-control study was set up enrolling 43 patients who underwent kidney transplantation. One day post-IR in mice and a few days following renal transplantation in human, blood and urine were collected. Patients were stratified into DGF and non-DGF groups. RESULTS: mtDNA-encoded genes accumulate in urine and plasma in both mice subjected to renal IR injury and in humans following renal transplantation. In human renal transplant recipients, cold ischaemia time and renal function correlate with urinary mtDNA levels. Urinary mtDNA levels but not urinary nuclear DNA levels were significantly higher in the DGF group compared with the non-DGF group. Multiple receiver operating characteristic curves revealed significant diagnostic performance for mtDNA-encoded genes cytochrome c oxidase III (COXIII); nicotinamide adenine dinucleotide hydrogen subunit 1 (NADH-deh); mitochondrially encoded, mitochondrially encoded nicotinamide adenine dinucleotide dehydrogenase 2 (MT-ND2) with an area under the curve of, respectively, 0.71 [P = 0.03; 95% confidence interval (CI) 0.54-0.89], 0.75 (P = 0.01; 95% CI 0.58-0.91) and 0.74 (P = 0.02; 95% CI 0.58-0.89). CONCLUSIONS: These data suggest that renal ischaemia time determines the level of mtDNA accumulation in urine, which associates with renal allograft function and the diagnosis of DGF following renal transplantation.


Asunto(s)
Biomarcadores/orina , ADN Mitocondrial/orina , Funcionamiento Retardado del Injerto/diagnóstico , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/complicaciones , Animales , Estudios de Casos y Controles , Funcionamiento Retardado del Injerto/etiología , Funcionamiento Retardado del Injerto/orina , Femenino , Supervivencia de Injerto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Curva ROC , Receptores de Trasplantes , Trasplante Homólogo
9.
Sci Rep ; 9(1): 17633, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31776357

RESUMEN

Obesity has become a worldwide health crisis and is associated with a plethora of comorbidities. The multi-organ effects of obesity have been linked to ectopic lipid accumulation. Thus, there is an urgent need to tackle the obesity crisis by developing effective lipid-lowering therapies. 2-hydroxypropyl-ß-Cyclodextrin (2HP-ß-CD) has been previously shown to reduce lysosomal cholesterol accumulation in a murine model of Niemann Pick Type C (NPC) disease. Using a murine model of Western diet-induced obesity (DIO), we report the effects of 2HP-ß-CD in counteracting weight gain, expansion of adipose tissue mass and ectopic lipid accumulation. Interestingly, DIO caused intracellular storage of neutral lipids in hepatic tissues and of phospholipids in kidneys, both of which were prevented by 2HP-ß-CD. Importantly, this report brings attention to the nephrotoxic effects of 2HP-ß-CD: renal tubular damage, inflammation and fibrosis. These effects may be overlooked, as they are best appreciated upon assessment of renal histology.


Asunto(s)
Dieta Occidental/efectos adversos , Hipolipemiantes/uso terapéutico , Enfermedades Renales/inducido químicamente , Obesidad/etiología , beta-Ciclodextrinas/uso terapéutico , Animales , Colesterol/análisis , Modelos Animales de Enfermedad , Hipolipemiantes/efectos adversos , Riñón/química , Riñón/efectos de los fármacos , Hígado/química , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/prevención & control , Fosfolípidos/análisis , Triglicéridos/análisis , beta-Ciclodextrinas/efectos adversos
10.
JCI Insight ; 4(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31672939

RESUMEN

Inflammation may play a role in the link between high salt intake and its deleterious consequences. However, it is unknown whether salt can induce proinflammatory priming of monocytes and macrophages in humans. We investigated the effects of salt on monocytes and macrophages in vitro and in vivo by performing a randomized crossover trial in which 11 healthy human subjects adhered to a 2-week low-salt and high-salt diet. We demonstrate that salt increases monocyte expression of CCR2, a chemokine receptor that mediates monocyte infiltration in inflammatory diseases. In line with this, we show a salt-induced increase of plasma MCP-1, transendothelial migration of monocytes, and skin macrophage density after high-salt diet. Macrophages demonstrate signs of an increased proinflammatory phenotype after salt exposure, as represented by boosted LPS-induced cytokine secretion of IL-6, TNF, and IL-10 in vitro, and by increased HLA-DR expression and decreased CD206 expression on skin macrophages after high-salt diet. Taken together, our data open up the possibility for inflammatory monocyte and macrophage responses as potential contributors to the deleterious effects of high salt intake.


Asunto(s)
Inflamación/metabolismo , Monocitos/efectos de los fármacos , Receptores CCR2/metabolismo , Cloruro de Sodio Dietético/farmacología , Adulto , Estudios Cruzados , Citocinas/metabolismo , Femenino , Humanos , Masculino , Monocitos/metabolismo , Cloruro de Sodio Dietético/metabolismo , Adulto Joven
11.
Front Immunol ; 10: 1469, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31354698

RESUMEN

Long-term sequelae of acute kidney injury (AKI) are associated with incomplete recovery of renal function and the development of chronic kidney disease (CKD), which can be mediated by aberrant innate immune activation, mitochondrial pathology, and accumulation of senescent tubular epithelial cells (TECs). Herein, we show that the innate immune receptor Triggering receptor expressed on myeloid cells-1 (TREM-1) links mitochondrial metabolism to tubular epithelial senescence. TREM-1 is expressed by inflammatory and epithelial cells, both players in renal repair after ischemia/reperfusion (IR)-induced AKI. Hence, we subjected WT and TREM1/3 KO mice to different models of renal IR. TREM1/3 KO mice displayed no major differences during the acute phase of injury, but increased mortality was observed in the recovery phase. This detrimental effect was associated with maladaptive repair, characterized by persistent tubular damage, inflammation, fibrosis, and TEC senescence. In vitro, we observed an altered mitochondrial homeostasis and cellular metabolism in TREM1/3 KO primary TECs. This was associated with G2/M arrest and increased ROS accumulation. Further exposure of cells to ROS-generating triggers drove the cells into a stress-induced senescent state, resulting in decreased wound healing capacity. Treatment with a mitochondria anti-oxidant partly prevented the senescent phenotype, suggesting a role for mitochondria herein. In summary, we have unraveled a novel (metabolic) mechanism by which TREM1/3 deficiency drives senescence in TECs. This involves redox imbalance, mitochondrial dysfunction and a decline in cellular metabolic activities. These finding suggest a novel role for TREM-1 in maintaining tubular homeostasis through regulation of mitochondrial metabolic flexibility.


Asunto(s)
Lesión Renal Aguda/patología , Túbulos Renales/citología , Mitocondrias/metabolismo , Receptor Activador Expresado en Células Mieloides 1/genética , Animales , Apoptosis/inmunología , Hipoxia de la Célula/genética , Células Cultivadas , Senescencia Celular/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Fibrosis/patología , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Receptor Activador Expresado en Células Mieloides 1/deficiencia
12.
PLoS One ; 14(3): e0214437, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30908533

RESUMEN

Diabetic nephropathy (DN) is a microvascular complication of diabetes mellitus that results in both tubular and glomerular injury. Low-grade inflammation and oxidative stress are two mechanisms known to drive the progression of DN. Nucleotide-binding leucine-rich repeat containing family member X1 (NLRX1) is an innate immune receptor, uniquely located in mitochondria, that has been found to regulate inflammatory responses and to dampen renal oxidative stress by regulating oxidative phosphorylation. For this reason, we investigated the role of NLRX1 in the development of DN in a Type 1 Diabetes mouse model. We analyzed the effect of NLRX1 deficiency on diabetes development and the accompanied renal damage, inflammation, and fibrosis. We found that multiple low doses of streptozotocin induced body weight loss, polydipsia, hyperglycemia, glycosuria, and a mild DN phenotype in wildtype and NLRX1-deficient mice, without significant differences between these mouse strains. Despite increased NLRX1 expression in diabetic wildtype mice, NLRX1 deficiency did not affect the diabetic phenotype induced by streptozotocin treatment, as reflected by similar levels of polyuria, microalbuminuria, and increased renal markers of oxidative stress and inflammation in wildtype and NLRX1-deficient mice. The present findings show that NLRX1 does not mediate the development of streptozotocin-induced diabetes and diabetic-induced nephropathy in mice after multiple low doses of streptozotocin. This data implies that, while NLRX1 can be triggered by cellular stress, its regulatory and functional effects may be dependent on the specific physiological conditions. In the case of DN, NLRX1 may be neither helpful nor harmful, but rather a marker of metabolic stress.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Riñón/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Estreptozocina/farmacología , Animales , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Relación Dosis-Respuesta a Droga , Fibrosis , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/deficiencia , Estrés Oxidativo/efectos de los fármacos , Fenotipo
13.
Sci Rep ; 9(1): 106, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30643171

RESUMEN

Calcineurin inhibitor Tacrolimus, is a potent immunosuppressive drug widely used in order to prevent acute graft rejection. Urinary tract infection (UTI) is the most frequent infectious complication in renal transplant patients and long-term use of Tacrolimus might be involved in higher susceptibility to bacterial infections. It remains largely unknown how Tacrolimus affects the host innate immune response against lower and upper UTI. To address this issue, we used experimental UTI model by intravesical inoculation of uropathogenic E.coli in female wild-type mice pre-treated with Tacrolimus or solvent (CTR). We found that Tacrolimus pre-treated mice displayed higher bacterial loads (cystitis, pyelonephritis and bacteremia) than CTR mice. Granulocytes from Tacrolimus pre-treated mice phagocytized less E. coli, released less MPO and expressed decreased levels of CXCR2 receptor upon infection. Moreover, Tacrolimus reduced TLR5 expression in bladder macrophages during UTI. This immunosuppressive state can be explained by the upregulation of TLR-signaling negative regulators (A20, ATF3, IRAK-M and SOCS1) and parallel downregulation of TLR5 as observed in Tacrolimus treated granulocytes and macrophages. We conclude that Tacrolimus impairs host innate immune responses against UTI.


Asunto(s)
Inhibidores de la Calcineurina/efectos adversos , Infecciones por Escherichia coli/patología , Inmunosupresores/efectos adversos , Tacrolimus/efectos adversos , Infecciones Urinarias/patología , Escherichia coli Uropatógena/crecimiento & desarrollo , Animales , Carga Bacteriana , Inhibidores de la Calcineurina/administración & dosificación , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Femenino , Granulocitos/efectos de los fármacos , Granulocitos/inmunología , Factores Inmunológicos/metabolismo , Inmunosupresores/administración & dosificación , Ratones , Peroxidasa/metabolismo , Fagocitosis/efectos de los fármacos , Tacrolimus/administración & dosificación , Infecciones Urinarias/inmunología , Escherichia coli Uropatógena/inmunología
14.
J Pathol ; 246(4): 470-484, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30073645

RESUMEN

Obesity and dyslipidaemia are features of the metabolic syndrome and risk factors for chronic kidney disease. The cellular mechanisms connecting metabolic syndrome with chronic kidney disease onset and progression remain largely unclear. We show that proximal tubular epithelium is a target site for lipid deposition upon overnutrition with a cholesterol-rich Western-type diet. Affected proximal tubule epithelial cells displayed giant vacuoles of lysosomal or autophagosomal origin, harbouring oxidised lipoproteins and concentric membrane layer structures (multilamellar bodies), reminiscent of lysosomal storage diseases. Additionally, lipidomic analysis revealed renal deposition of cholesterol and phospholipids, including lysosomal phospholipids. Proteomic profiles of renal multilamellar bodies were distinct from those of epidermis or lung multilamellar bodies and of cytoplasmic lipid droplets. Tubular multilamellar bodies were observed in kidney biopsies of obese hypercholesterolaemic patients, and the concentration of the phospholipidosis marker di-docosahexaenoyl (22:6)-bis(monoacylglycerol) phosphate was doubled in urine from individuals with metabolic syndrome and chronic kidney disease. The enrichment of proximal tubule epithelial cells with phospholipids and multilamellar bodies was accompanied by enhanced inflammation, fibrosis, tubular damage markers, and higher urinary electrolyte content. Concomitantly to the intralysosomal lipid storage, a renal transcriptional response was initiated to enhance lysosomal degradation and lipid synthesis. In cultured proximal tubule epithelial cells, inhibition of cholesterol efflux transport or oxysterol treatment induced effects very similar to the in vivo situation, such as multilamellar body and phospholipid amassing, and induction of damage, inflammatory, fibrotic, and lipogenic molecules. The onset of phospholipidosis in proximal tubule epithelial cells is a novel pathological trait in metabolic syndrome-related chronic kidney disease, and emphasises the importance of healthy lysosomes and nutrition for kidney well-being. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Colesterol en la Dieta/efectos adversos , Dieta Alta en Grasa/efectos adversos , Hipercolesterolemia/complicaciones , Túbulos Renales Proximales/metabolismo , Lisosomas/metabolismo , Obesidad/complicaciones , Fosfolípidos/efectos adversos , Insuficiencia Renal Crónica/etiología , Animales , Estudios de Casos y Controles , Línea Celular , Colesterol en la Dieta/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Túbulos Renales Proximales/ultraestructura , Lisosomas/ultraestructura , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfolípidos/metabolismo , Proteómica/métodos , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
15.
BMC Nephrol ; 19(1): 78, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29609537

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) is characterized by sustained tissue damage and ongoing tubulo-interstitial inflammation and fibrosis. Pattern recognition receptors (PRRs) including Toll-like receptors (TLRs) and NOD-like receptors (NLRs) can sense endogenous ligands released upon tissue damage, leading to sterile inflammation and eventually irreversible kidney disease. It is known that NOD1 and NOD2 contribute to the pathogenesis of various inflammatory diseases, including acute kidney injury. However their role in chronic kidney disease is largely unknown. The aim of this study was therefore to investigate the contribution of NOD1 and NOD2 in renal interstitial fibrosis and obstructive nephropathy. METHODS: To do so, we performed unilateral ureteral obstruction (UUO) in wild type (WT) and NOD1/NOD2 double deficient (DKO) mice and analysed renal damage, fibrosis and inflammation. Data were analysed using the non-parametric Mann-Whitney U-test. RESULTS: Minor changes in inflammatory response were observed in NOD1/2 DKO mice, while no effects were observed on renal injury and the development of fibrosis. CONCLUSION: No difference in renal injury and fibrosis between WT and NOD1/NOD2 DKO mice following obstructive nephropathy induced by ureteral obstruction.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína Adaptadora de Señalización NOD1/deficiencia , Proteína Adaptadora de Señalización NOD2/deficiencia , Insuficiencia Renal Crónica/metabolismo , Obstrucción Ureteral/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/genética , Animales , Femenino , Fibrosis/etiología , Fibrosis/genética , Fibrosis/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/genética , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1883-1895, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29514047

RESUMEN

NOD-like receptor (NLR)X1 (NLRX1) is an ubiquitously expressed inflammasome-independent NLR that is uniquely localized in mitochondria with as yet unknown effects on metabolic diseases. Here, we report that NLRX1 is essential in regulating cellular metabolism in non-immune parenchymal hepatocytes by decreasing mitochondrial fatty acid-dependent oxidative phosphorylation (OXPHOS) and promoting glycolysis. NLRX1 loss in mice has a profound impact on the prevention of diet-induced metabolic syndrome parameters, non-alcoholic fatty liver disease (NAFLD) progression, and renal dysfunction. Despite enhanced caloric intake, NLRX1 deletion in mice fed a western diet (WD) results in protection from liver steatosis, hepatic fibrosis, obesity, insulin resistance, glycosuria and kidney dysfunction parameters independent from inflammation. While mitochondrial content was equal, NLRX1 loss in hepatocytes leads to increased fatty acid oxidation and decreased steatosis. In contrast, glycolysis was decreased in NLRX1-deficient cells versus controls. Thus, although first implicated in immune regulation, we show that NLRX1 function extends to the control of hepatocyte energy metabolism via the restriction of mitochondrial fatty acid-dependent OXPHOS and enhancement of glycolysis. As such NLRX1 may be an attractive novel therapeutic target for NAFLD and metabolic syndrome.


Asunto(s)
Grasas de la Dieta/efectos adversos , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Síndrome Metabólico/metabolismo , Proteínas Mitocondriales/deficiencia , Animales , Grasas de la Dieta/farmacología , Ácidos Grasos/genética , Hígado Graso/inducido químicamente , Hígado Graso/genética , Hígado Graso/patología , Eliminación de Gen , Hepatocitos/patología , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/genética , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología
17.
Transpl Immunol ; 47: 18-21, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29305091

RESUMEN

BACKGROUND: BK virus nephropathy (BKPyVN) is a major complication after renal transplantation. Little is known about the intra renal immune response during BKPyVN. The role of macrophages remains elusive. The activation of aryl hydrocarbon receptor (AHR) - a transcription factor involved in drug metabolism - plays a key role in inflammation and viral tolerance through modulation of macrophages polarization. Since AHR has not been studied in kidney transplantation, our aim was to compare the AHR expression within renal grafts in BKPyVN with T-cell mediated rejection (TCMR) as a control. METHODS: We evaluated AHR expression in kidney grafts from BKPyVN (n=8) with TCMR as control (n=6) among cases with available frozen material for AHR gene intragraft transcription measurement and stainings for AHR, CD68 and CD45. RESULTS: AHR transcription was higher in BKPyVN grafts versus TCMR (p=0.03). While CD68+ or CD45+ cell expression did not differ within infiltrates (median score=3 in both groups; p=1.0 and 0.69, respectively), a higher proportion of nuclear AHR expression was found in BKPyVN for CD68+ and CD45+ cells when compared with TCMR (score median 2 vs 0; p=0.007 and 1 vs 0; p=0.013, respectively). CONCLUSIONS: We describe for the first time a higher expression of AHR in inflammatory cell infiltrates from BKPyVN versus TCMR renal biopsies. Further studies are required to explore AHR as a potential target in the modulation of inflammatory response in BKPyVN with known modulating ligands.


Asunto(s)
Virus BK/fisiología , Rechazo de Injerto/inmunología , Enfermedades Renales/inmunología , Trasplante de Riñón , Riñón/inmunología , Macrófagos/inmunología , Infecciones por Polyomavirus/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T/inmunología , Infecciones Tumorales por Virus/inmunología , Adulto , Anciano , Diferenciación Celular , Citocinas/metabolismo , Femenino , Humanos , Inmunohistoquímica , Riñón/virología , Masculino , Persona de Mediana Edad , Trasplante Homólogo
18.
Histopathology ; 72(4): 601-608, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28881045

RESUMEN

AIMS: The technique used for classification of membranoproliferative glomerulonephritis (MPGN) has been changed from an electron microscopy-based to an immunofluorescence (IF)-based semiquantitative technique with immunoperoxidase (IP) staining as a backup option when IF is not possible. Since data on that matter is lacking, our aims were to study the interobserver variability, the correlation and the reclassification of MPGN based on these two techniques. METHODS AND RESULTS: We retrospectively analysed cases of type 1 MPGN. We repeated IF staining and performed IP staining for IgG, kappa, lambda, C3c and C4d in 35 renal biopsies, among which 19 biopsies had matched IP and IF samples. We observed substantial to near-perfect agreement among the seven observers for both IF and IP (W coefficients from 0.66 for IF lambda to 0.89 for IF C4d). Of the 19 cases with matched IP and IF samples, five (26%) turned out to have different diagnoses on IF and on IP. Also, the ability of C4d to discriminate immune complex-mediated glomerulonephritis (ICGN) from C3 glomerulopathy (C3G) was poor, with areas under the curve of 0.44 [95% confidence interval (CI) 0.24-0.63] and 0.66 (95% CI 0.50-0.81) for the receiver operating characteristic curves of IF and IP respectively. Limitations include the fact that no clinical data regarding complement activation were available. CONCLUSION: The diagnosis of ICGN versus C3GN depends on the immunochemical technique used. Also, the use of C4d failed to discriminate ICGN from C3G in our study. Further validation studies are required to avoid misdiagnosis based on kidney biopsy.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Glomerulonefritis Membranoproliferativa/diagnóstico , Técnicas para Inmunoenzimas/métodos , Adolescente , Adulto , Anciano , Complejo Antígeno-Anticuerpo/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad
19.
Am J Pathol ; 187(9): 1971-1983, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28668213

RESUMEN

TAM receptors (Tyro3, Axl, and Mer) have been implicated in innate immunity. Circulating TAM receptor soluble forms (sTyro3, sAxl, sMer) are related to autoimmune disorders. We investigated TAM and their ligand protein S in patients with diabetes. Urinary and plasma levels of protein S, sTyro3, sAxl, and sMer were determined in 126 patients with diabetes assigned to a normoalbuminuric or macroalbuminuric (urinary albumin excretion <30 mg/24 hours and >300 mg/24 hours, respectively) study group and 18 healthy volunteers. TAM and protein S immunostaining was performed on kidney biopsy specimens from patients with diabetic nephropathy (n = 9) and controls (n = 6). TAM expression and shedding by tubular epithelial cells were investigated by PCR and enzyme-linked immunosorbent assay in an in vitro diabetes model. Patients with macroalbuminuria diabetes had higher circulating levels of sMer and more urinary sTyro3 and sMer than normoalbuminuric diabetics. Increased clearance of sTyro3 and sMer was associated with loss of tubular Tyro3 and Mer expression in diabetic nephropathy tissue and glomerular depositions of protein S. During in vitro diabetes, human kidney cells had down-regulation of Tyro3 and Mer mRNA and increased shedding of sTyro3 and sMer. Renal injury in diabetes is associated with elevated systemic and urine levels of sMer and sTyro3. This is the first study reporting excretion of sTAM receptors in urine, identifying the kidney as a source of sTAM.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Proteína S/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/orina , Femenino , Humanos , Glomérulos Renales/metabolismo , Masculino , Persona de Mediana Edad , Proteína S/orina , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/orina , Proteínas Tirosina Quinasas Receptoras/sangre , Proteínas Tirosina Quinasas Receptoras/orina , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...