Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Cell Mol Gastroenterol Hepatol ; 16(4): 557-572, 2023.
Article En | MEDLINE | ID: mdl-37369278

BACKGROUND & AIMS: Metabolic syndrome (MetS) is characterized by obesity, glucose intolerance, and hepatic steatosis. Alterations in the gut microbiome play important roles in the development of MetS. However, the mechanisms by which this occurs are poorly understood. Dual oxidase 2 (DUOX2) is an antimicrobial reduced nicotinamide adenine dinucleotide phosphate oxidase expressed in the gut epithelium. Here, we posit that epithelial DUOX2 activity provides a mechanistic link between the gut microbiome and the development of MetS. METHODS: Mice carrying an intestinal epithelial-specific deletion of dual oxidase maturation factor 1/2 (DA IEC-KO), and wild-type littermates were fed a standard diet and killed at 24 weeks. Metabolic alterations were determined by glucose tolerance, lipid tests, and body and organ weight measurements. DUOX2 activity was determined by Amplex Red. Intestinal permeability was determined by fluorescein isothiocyanate-dextran, microbial translocation assessments, and portal vein lipopolysaccharide measurements. Metagenomic analysis of the stool microbiome was performed. The role of the microbiome was assessed in antibiotic-treated mice. RESULTS: DA IEC-KO males showed increased body and organ weights accompanied by glucose intolerance and increased plasma lipid and liver enzyme levels, and increased adiposity in the liver and adipose tissue. Expression of F4/80, CD68, uncoupling protein 1, carbohydrate response element binding protein, leptin, and adiponectin was altered in the liver and adipose tissue of DA IEC-KO males. DA IEC-KO males produced less epithelial H2O2, had altered relative abundance of Akkermansiaceae and Lachnospiraceae in stool, and showed increased portal vein lipopolysaccharides and intestinal permeability. Females were protected from barrier defects and MetS, despite producing less H2O2. Antibiotic depletion abrogated all MetS phenotypes observed. CONCLUSIONS: Intestinal epithelial inactivity of DUOX2 promotes MetS in a microbiome-dependent manner.


Gastrointestinal Microbiome , Glucose Intolerance , Metabolic Syndrome , Animals , Female , Male , Mice , Anti-Bacterial Agents , Dual Oxidases , Hydrogen Peroxide , Lipopolysaccharides , Obesity/metabolism
2.
JCI Insight ; 7(13)2022 07 08.
Article En | MEDLINE | ID: mdl-35653195

Apolipoprotein C-III (apoC-III) is a critical regulator of triglyceride metabolism and correlates positively with hypertriglyceridemia and cardiovascular disease (CVD). It remains unclear if therapeutic apoC-III lowering reduces CVD risk and if the CVD correlation depends on the lipid-lowering or antiinflammatory properties. We determined the impact of interventional apoC-III lowering on atherogenesis using an apoC-III antisense oligonucleotide (ASO) in 2 hypertriglyceridemic mouse models where the intervention lowers plasma triglycerides and in a third lipid-refractory model. On a high-cholesterol Western diet apoC-III ASO treatment did not alter atherosclerotic lesion size but did attenuate advanced and unstable plaque development in the triglyceride-responsive mouse models. No lesion size or composition improvement was observed with apoC-III ASO in the lipid-refractory mice. To circumvent confounding effects of continuous high-cholesterol feeding, we tested the impact of interventional apoC-III lowering when switching to a cholesterol-poor diet after 12 weeks of Western diet. In this diet switch regimen, apoC-III ASO treatment significantly reduced plasma triglycerides, atherosclerotic lesion progression, and necrotic core area and increased fibrous cap thickness in lipid-responsive mice. Again, apoC-III ASO treatment did not alter triglyceride levels, lesion development, and lesion composition in lipid-refractory mice after the diet switch. Our findings suggest that interventional apoC-III lowering might be an effective strategy to reduce atherosclerosis lesion size and improve plaque stability when lipid lowering is achieved.


Atherosclerosis , Hyperlipidemias , Plaque, Atherosclerotic , Animals , Apolipoprotein C-III , Carrier Proteins , Cholesterol , Mice , Oligonucleotides , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Triglycerides/metabolism
3.
Nat Commun ; 12(1): 4662, 2021 08 02.
Article En | MEDLINE | ID: mdl-34341345

Impaired cellular cholesterol efflux is a key factor in the progression of renal, cardiovascular, and autoimmune diseases. Here we describe a class of 5-arylnicotinamide compounds, identified through phenotypic drug discovery, that upregulate ABCA1-dependent cholesterol efflux by targeting Oxysterol Binding Protein Like 7 (OSBPL7). OSBPL7 was identified as the molecular target of these compounds through a chemical biology approach, employing a photoactivatable 5-arylnicotinamide derivative in a cellular cross-linking/immunoprecipitation assay. Further evaluation of two compounds (Cpd A and Cpd G) showed that they induced ABCA1 and cholesterol efflux from podocytes in vitro and normalized proteinuria and prevented renal function decline in mouse models of proteinuric kidney disease: Adriamycin-induced nephropathy and Alport Syndrome. In conclusion, we show that small molecule drugs targeting OSBPL7 reveal an alternative mechanism to upregulate ABCA1, and may represent a promising new therapeutic strategy for the treatment of renal diseases and other disorders of cellular cholesterol homeostasis.


ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Diabetic Nephropathies/metabolism , Organic Chemicals/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Receptors, Steroid/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , Animals , Biological Transport/drug effects , Cells, Cultured , Disease Models, Animal , HEK293 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice, 129 Strain , Mice, Knockout , Molecular Structure , Niacinamide/chemistry , Niacinamide/pharmacology , Organic Chemicals/chemical synthesis , Organic Chemicals/chemistry , Podocytes/cytology , RNA Interference , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , THP-1 Cells
4.
Hum Mol Genet ; 30(3-4): 182-197, 2021 04 26.
Article En | MEDLINE | ID: mdl-33517446

Lipotoxicity was recently reported in several forms of kidney disease, including focal segmental glomerulosclerosis (FSGS). Susceptibility to FSGS in African Americans is associated with the presence of genetic variants of the Apolipoprotein L1 gene (APOL1) named G1 and G2. If and how endogenous APOL1 may alter mitochondrial function by the modifying cellular lipid metabolism is unknown. Using transgenic mice expressing the APOL1 variants (G0, G1 or G2) under endogenous promoter, we show that APOL1 risk variant expression in transgenic mice does not impair kidney function at baseline. However, APOL1 G1 expression worsens proteinuria and kidney function in mice characterized by the podocyte inducible expression of nuclear factor of activated T-cells (NFAT), which we have found to cause FSGS. APOL1 G1 expression in this FSGS-model also results in increased triglyceride and cholesterol ester contents in kidney cortices, where lipid accumulation correlated with loss of renal function. In vitro, we show that the expression of endogenous APOL1 G1/G2 in human urinary podocytes is associated with increased cellular triglyceride content and is accompanied by mitochondrial dysfunction in the presence of compensatory oxidative phosphorylation (OXPHOS) complexes elevation. Our findings indicate that APOL1 risk variant expression increases the susceptibility to lipid-dependent podocyte injury, ultimately leading to mitochondrial dysfunction.


Apolipoprotein L1/genetics , Genetic Variation , Glomerulosclerosis, Focal Segmental/metabolism , Lipid Metabolism , Mitochondria/metabolism , Podocytes/metabolism , Black or African American/genetics , Animals , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/physiopathology , Homeostasis , Humans , Mice , Mice, Transgenic , Mitochondria/physiology , Podocytes/physiology , Proteinuria , Triglycerides/metabolism
5.
Front Immunol ; 11: 769, 2020.
Article En | MEDLINE | ID: mdl-32508807

Proteoglycans are a specific subset of glycoproteins found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in metabolic homeostasis and meta-inflammation. Over the last decade, new insights have emerged on the mechanism and biological significance of these interactions in the context of diet-induced disorders such as obesity and type-2 diabetes. Complications of energy metabolism drive most diet-induced metabolic disorders, which results in low-grade chronic inflammation, thereby affecting proper function of many vital organs involved in energy homeostasis, such as the brain, liver, kidney, heart and adipose tissue. Here, we discuss how heparan, chondroitin and keratan sulfate proteoglycans modulate obesity-induced metabolic dysfunction and low-grade inflammation that impact the initiation and progression of obesity-associated morbidities.


Obesity/immunology , Obesity/metabolism , Proteoglycans/immunology , Proteoglycans/metabolism , Adipocytes , Adipose Tissue , Animals , Diet , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans , Humans , Inflammation/metabolism , Mice
6.
Curr Diab Rep ; 19(12): 144, 2019 11 21.
Article En | MEDLINE | ID: mdl-31754839

PURPOSE OF REVIEW: The goal of this review is to review the role that renal parenchymal lipid accumulation plays in contributing to diabetic kidney disease (DKD), specifically contributing to the mitochondrial dysfunction observed in glomerular renal cells in the context of DKD development and progression. RECENT FINDINGS: Mitochondrial dysfunction has been observed in experimental and clinical DKD. Recently, Ayanga et al. demonstrate that podocyte-specific deletion of a protein involved in mitochondrial dynamics protects from DKD progression. Furthermore, our group has recently shown that ATP-binding cassette A1 (a protein involved in cholesterol and phospholipid efflux) is significantly reduced in clinical and experimental DKD and that genetic or pharmacological induction of ABCA1 is sufficient to protect from DKD. ABCA1 deficiency in podocytes leads to mitochondrial dysfunction observed with alterations of mitochondrial lipids, in particular, cardiolipin (a mitochondrial-specific phospholipid). However, through pharmacological reduction of cardiolipin peroxidation DKD progression is reverted. Lipid metabolism is significantly altered in the diabetic kidney and renders cellular components, such as the podocyte, susceptible to injury leading to worsened DKD progression. Dysfunction of the lipid metabolism pathway can also lead to mitochondrial dysfunction and mitochondrial lipid alteration. Future research aimed at targeting mitochondrial lipids content and function could prove to be beneficial for the treatment of DKD.


Diabetic Nephropathies/physiopathology , Kidney Glomerulus/physiopathology , Lipid Metabolism Disorders/physiopathology , Lipid Metabolism/physiology , Mitochondria/physiology , Receptor Cross-Talk/physiology , Diabetic Nephropathies/etiology , Humans , Kidney Glomerulus/pathology , Lipid Metabolism Disorders/etiology , Lipids/physiology , Podocytes/physiology
7.
J Clin Invest ; 129(8): 3387-3400, 2019 07 22.
Article En | MEDLINE | ID: mdl-31329164

Fibroblasts from patients with Tangier disease carrying ATP-binding cassette A1 (ABCA1) loss-of-function mutations are characterized by cardiolipin accumulation, a mitochondrial-specific phospholipid. Suppression of ABCA1 expression occurs in glomeruli from patients with diabetic kidney disease (DKD) and in human podocytes exposed to DKD sera collected prior to the development of DKD. We demonstrated that siRNA ABCA1 knockdown in podocytes led to reduced oxygen consumption capabilities associated with alterations in the oxidative phosphorylation (OXPHOS) complexes and with cardiolipin accumulation. Podocyte-specific deletion of Abca1 (Abca1fl/fl) rendered mice susceptible to DKD, and pharmacological induction of ABCA1 improved established DKD. This was not mediated by free cholesterol, as genetic deletion of sterol-o-acyltransferase-1 (SOAT1) in Abca1fl/fl mice was sufficient to cause free cholesterol accumulation but did not cause glomerular injury. Instead, cardiolipin mediates ABCA1-dependent susceptibility to podocyte injury, as inhibition of cardiolipin peroxidation with elamipretide improved DKD in vivo and prevented ABCA1-dependent podocyte injury in vitro and in vivo. Collectively, we describe a pathway definitively linking ABCA1 deficiency to cardiolipin-driven mitochondrial dysfunction. We demonstrated that this pathway is relevant to DKD and that ABCA1 inducers or inhibitors of cardiolipin peroxidation may each represent therapeutic strategies for the treatment of established DKD.


ATP Binding Cassette Transporter 1/deficiency , Cardiolipins/metabolism , Diabetic Nephropathies/metabolism , Lipid Peroxidation , Mitochondria/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Cardiolipins/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Humans , Mice , Mitochondria/genetics , Mitochondria/pathology , Podocytes , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
8.
Kidney Int ; 94(6): 1151-1159, 2018 12.
Article En | MEDLINE | ID: mdl-30301568

Studies suggest that altered renal lipid metabolism plays a role in the pathogenesis of diabetic kidney disease and that genetic or pharmacological induction of cholesterol efflux protects from the development of diabetic kidney disease and focal segmental glomerulosclerosis (FSGS). Here we tested whether altered lipid metabolism contributes to renal failure in the Col4a3 knockout mouse model for Alport Syndrome. There was an eight-fold increase in the cholesterol content in renal cortexes of mice with Alport Syndrome. This was associated with increased glomerular lipid droplets and cholesterol crystals. Treatment of mice with Alport Syndrome with hydroxypropyl-ß-cyclodextrin (HPßCD) reduced cholesterol content in the kidneys of mice with Alport Syndrome and protected from the development of albuminuria, renal failure, inflammation and tubulointerstitial fibrosis. Cholesterol efflux and trafficking-related genes were primarily affected in mice with Alport Syndrome and were differentially regulated in the kidney cortex and isolated glomeruli. HPßCD also protected from proteinuria and mesangial expansion in a second model of non-metabolic kidney disease, adriamycin-induced nephropathy. Consistent with our experimental findings, microarray analysis confirmed dysregulation of several lipid-related genes in glomeruli isolated from kidney biopsies of patients with primary FSGS enrolled in the NEPTUNE study. Thus, lipid dysmetabolism occurs in non-metabolic glomerular disorders such as Alport Syndrome and FSGS, and HPßCD improves renal function in experimental Alport Syndrome and FSGS.


2-Hydroxypropyl-beta-cyclodextrin/therapeutic use , Glomerulosclerosis, Focal Segmental/drug therapy , Kidney Glomerulus/pathology , Nephritis, Hereditary/drug therapy , 2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Animals , Autoantigens/genetics , Biopsy , Cholesterol/metabolism , Collagen Type IV/genetics , Doxorubicin/toxicity , Female , Glomerulosclerosis, Focal Segmental/chemically induced , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Lipid Metabolism/drug effects , Mice , Mice, Knockout , Nephritis, Hereditary/genetics , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , Observational Studies as Topic
...