Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cells ; 13(1)2023 12 21.
Article in English | MEDLINE | ID: mdl-38201227

ABSTRACT

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.


Subject(s)
Renal Insufficiency, Chronic , Animals , Mice , Macrophages , Genes, Regulator , Fibrosis , Transforming Growth Factor beta , Mitochondrial Proteins
2.
J Pathol ; 253(4): 404-414, 2021 04.
Article in English | MEDLINE | ID: mdl-33338266

ABSTRACT

Lipid accumulation is associated with various forms of acute renal injury; however, the causative factors and pathways underpinning this lipid accumulation have not been thoroughly investigated. In this study, we performed lipidomic profiling of renal tissue following ischaemia-reperfusion injury (IRI). We identified a significant accumulation of cholesterol and specific phospholipids and sphingolipids in kidneys 24 h after IRI. In light of these findings, we hypothesised that pathways involved in lipid metabolism may also be altered. Through the analysis of published microarray data, generated from sham and ischaemic kidneys, we identified nephron-specific metabolic pathways affected by IRI and validated these findings in ischaemic renal tissue. In silico analysis revealed the downregulation of several energy and lipid metabolism pathways, including mitochondrial fatty acid beta-oxidation (FAO), peroxisomal lipid metabolism, fatty acid (FA) metabolism, and glycolysis. The pentose phosphate pathway (PPP), which is fuelled by glycolysis, was the only metabolic pathway that was upregulated 24 h following IRI. In this study, we describe the effect of renal IRI on metabolic pathways and how this contributes to lipid accumulation. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Acute Kidney Injury/metabolism , Pentose Phosphate Pathway/physiology , Reperfusion Injury/metabolism , Animals , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL
3.
Sci Rep ; 9(1): 17633, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31776357

ABSTRACT

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.


Subject(s)
Diet, Western/adverse effects , Hypolipidemic Agents/therapeutic use , Kidney Diseases/chemically induced , Obesity/etiology , beta-Cyclodextrins/therapeutic use , Animals , Cholesterol/analysis , Disease Models, Animal , Hypolipidemic Agents/adverse effects , Kidney/chemistry , Kidney/drug effects , Liver/chemistry , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/prevention & control , Phospholipids/analysis , Triglycerides/analysis , beta-Cyclodextrins/adverse effects
4.
J Pathol ; 246(4): 470-484, 2018 12.
Article in English | MEDLINE | ID: mdl-30073645

ABSTRACT

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.


Subject(s)
Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Hypercholesterolemia/complications , Kidney Tubules, Proximal/metabolism , Lysosomes/metabolism , Obesity/complications , Phospholipids/adverse effects , Renal Insufficiency, Chronic/etiology , Animals , Case-Control Studies , Cell Line , Cholesterol, Dietary/metabolism , Disease Models, Animal , Fibrosis , Kidney Tubules, Proximal/ultrastructure , Lysosomes/ultrastructure , Male , Mice, Inbred C57BL , Mice, Transgenic , Phospholipids/metabolism , Proteomics/methods , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology
5.
BMC Public Health ; 18(1): 1033, 2018 Aug 20.
Article in English | MEDLINE | ID: mdl-30126399

ABSTRACT

BACKGROUND: Physical activity (PA) is important in combating childhood obesity. Parents, and thus parental PA, could influence PA in young children. We examined whether the time spent at different intensities of PA and the type of parental PA are associated with the PA of children aged 4-7 years, and whether the associations between child-parent pairs were sex-specific. METHODS: All the participants were recruited from the Groningen Expert Center for Kids with Obesity (GECKO) birth cohort (babies born between 1 April 2006 and 1 April 2007 in Drenthe province, the Netherlands) and were aged 4-7 years during measurement. PA in children was measured using the ActiGraph GT3X (worn at least 3 days, ≥10 h per day). PA in parents was assessed using the validated SQUASH questionnaire. RESULTS: Of the N = 1146 children with valid ActiGraph data and 838 mothers and 814 fathers with valid questionnaire data, 623 child-parent pairs with complete data were analysed. More leisure time PA in mothers was associated with more time spent in moderate-to-vigorous PA (MVPA) in children (Spearman r = 0.079, P < .05). Maternal PA was significantly related to PA in girls, but not boys. More time spent in maternal vigorous PA, in sports activity, and leisure time PA, were all related to higher MVPA in girls (Spearman r = 0.159, r = 0.133 and r = 0.127 respectively, Pall < .05). In fathers, PA levels were predominantly related to PA in sons. High MVPA in fathers was also related to high MVPA in sons (r = 0.132, P < 0.5). Spending more time in light PA was related to more sedentary time and less time in MVPA in sons. CONCLUSIONS: Higher PA in mothers, for instance in leisure activities, is related to higher PA in daughters, and more active fathers are related to more active sons. To support PA in young children, interventions could focus on the PA of the parent of the same sex as the child. Special attention may be needed for families where the parents have sedentary jobs, as children from these families seem to adopt more sedentary behaviour.


Subject(s)
Exercise/physiology , Exercise/psychology , Parent-Child Relations , Parents/psychology , Child , Child, Preschool , Cohort Studies , Female , Humans , Leisure Activities/psychology , Male , Netherlands , Pediatric Obesity/prevention & control , Sex Factors , Sports/psychology , Sports/statistics & numerical data , Surveys and Questionnaires , Time Factors
6.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1883-1895, 2018 May.
Article in English | MEDLINE | ID: mdl-29514047

ABSTRACT

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.


Subject(s)
Dietary Fats/adverse effects , Fatty Acids/metabolism , Fatty Liver/metabolism , Hepatocytes/metabolism , Metabolic Syndrome/metabolism , Mitochondrial Proteins/deficiency , Animals , Dietary Fats/pharmacology , Fatty Acids/genetics , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Gene Deletion , Hepatocytes/pathology , Metabolic Syndrome/chemically induced , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology
7.
J Exp Med ; 214(8): 2405-2420, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28626071

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Mitochondria/physiology , Mitochondrial Proteins/physiology , Oxidative Stress/physiology , Reperfusion Injury/physiopathology , Animals , Disease Models, Animal , Humans , Ischemia/physiopathology , Kidney/blood supply , Kidney/metabolism , Kidney/physiopathology , Male , Mice, Inbred C57BL
8.
Elife ; 52016 Sep 26.
Article in English | MEDLINE | ID: mdl-27668431

ABSTRACT

TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT ß-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.

9.
Nat Rev Nephrol ; 10(7): 398-414, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24890433

ABSTRACT

Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain receptors (NLRs) are families of pattern recognition receptors that, together with inflammasomes, sense and respond to highly conserved pathogen motifs and endogenous molecules released upon cell damage or stress. Evidence suggests that TLRs, NLRs and the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome have important roles in kidney diseases through regulation of inflammatory and tissue-repair responses to infection and injury. In this Review, we discuss the pathological mechanisms that are related to TLRs, NLRs and NLRP3 in various kidney diseases. In general, these receptors are protective in the host defence against urinary tract infection, but can sustain and self-perpetuate tissue damage in sterile inflammatory and immune-mediated kidney diseases. TLRs, NLRs and NLRP3, therefore, have become promising drug targets to enable specific modulation of kidney inflammation and suppression of immunopathology in kidney disease.


Subject(s)
Inflammasomes/immunology , Kidney Diseases/immunology , Receptors, Pattern Recognition/immunology , Toll-Like Receptors/immunology , Acute Kidney Injury/microbiology , Acute Kidney Injury/physiopathology , Adaptor Proteins, Signal Transducing/physiology , Animals , Carrier Proteins/immunology , Humans , Kidney/immunology , Kidney Transplantation , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Sepsis/complications , Toll-Like Receptors/genetics , Urinary Tract Infections/immunology
10.
Kidney Int ; 85(5): 1112-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24352154

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Cholesterol, Dietary/metabolism , Diet, High-Fat , Diet, Western , Kidney Diseases/metabolism , Kidney/metabolism , Metabolic Syndrome/metabolism , Signal Transduction , Animals , Biomarkers/blood , Carrier Proteins/genetics , Dietary Carbohydrates/metabolism , Disease Models, Animal , Fibrosis , Fructose/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Kidney/immunology , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/genetics , Kidney Diseases/immunology , Kidney Diseases/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/genetics , Metabolic Syndrome/immunology , Metabolic Syndrome/pathology , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...