Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(11): 1440-1451, 2021 11.
Article in English | MEDLINE | ID: mdl-34686860

ABSTRACT

Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases.


Subject(s)
Colitis/enzymology , Colon/enzymology , Cytotoxicity, Immunologic , Electron Transport Complex II/metabolism , Epithelial Cells/enzymology , Graft vs Host Disease/enzymology , Intestinal Mucosa/enzymology , Mitochondria/enzymology , T-Lymphocytes/immunology , Animals , Case-Control Studies , Cell Communication , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/pathology , Colon/immunology , Colon/ultrastructure , Disease Models, Animal , Electron Transport Complex II/genetics , Epithelial Cells/immunology , Epithelial Cells/ultrastructure , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/ultrastructure , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/immunology , Mitochondria/ultrastructure , Oxidative Phosphorylation , Succinic Acid/metabolism , T-Lymphocytes/metabolism
2.
Nat Immunol ; 17(5): 505-513, 2016 05.
Article in English | MEDLINE | ID: mdl-26998764

ABSTRACT

The effect of alterations in intestinal microbiota on microbial metabolites and on disease processes such as graft-versus-host disease (GVHD) is not known. Here we carried out an unbiased analysis to identify previously unidentified alterations in gastrointestinal microbiota-derived short-chain fatty acids (SCFAs) after allogeneic bone marrow transplant (allo-BMT). Alterations in the amount of only one SCFA, butyrate, were observed only in the intestinal tissue. The reduced butyrate in CD326(+) intestinal epithelial cells (IECs) after allo-BMT resulted in decreased histone acetylation, which was restored after local administration of exogenous butyrate. Butyrate restoration improved IEC junctional integrity, decreased apoptosis and mitigated GVHD. Furthermore, alteration of the indigenous microbiota with 17 rationally selected strains of high butyrate-producing Clostridia also decreased GVHD. These data demonstrate a heretofore unrecognized role of microbial metabolites and suggest that local and specific alteration of microbial metabolites has direct salutary effects on GVHD target tissues and can mitigate disease severity.


Subject(s)
Epithelial Cells/immunology , Gastrointestinal Microbiome/immunology , Graft vs Host Disease/immunology , Intestines/immunology , Metabolome/immunology , Acetylation/drug effects , Animals , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Butyrates/immunology , Butyrates/metabolism , Butyrates/pharmacology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Female , Gas Chromatography-Mass Spectrometry , Gastrointestinal Microbiome/physiology , Gene Expression/immunology , Graft vs Host Disease/etiology , Graft vs Host Disease/microbiology , Histone Acetyltransferases/genetics , Histone Acetyltransferases/immunology , Histone Acetyltransferases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Histone Deacetylases/metabolism , Histones/immunology , Histones/metabolism , Immunoblotting , Intestines/cytology , Intestines/microbiology , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
3.
Am J Physiol Renal Physiol ; 326(1): F30-F38, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37916286

ABSTRACT

Plasma nucleosides-pseudouridine (PU) and N2N2-dimethyl guanosine (DMG) predict the progression of type 2 diabetic kidney disease (DKD) to end-stage renal disease, but the mechanisms underlying this relationship are not well understood. We used a well-characterized model of type 2 diabetes (db/db mice) and control nondiabetic mice (db/m mice) to characterize the production and excretion of PU and DMG levels using liquid chromatography-mass spectrometry. The fractional excretion of PU and DMG was decreased in db/db mice compared with control mice at 24 wk before any changes to renal function. We then examined the dynamic changes in nucleoside metabolism using in vivo metabolic flux analysis with the injection of labeled nucleoside precursors. Metabolic flux analysis revealed significant decreases in the ratio of urine-to-plasma labeling of PU and DMG in db/db mice compared with db/m mice, indicating significant tubular dysfunction in diabetic kidney disease. We observed that the gene and protein expression of the renal tubular transporters involved with nucleoside transport in diabetic kidneys in mice and humans was reduced. In conclusion, this study strongly suggests that tubular handling of nucleosides is altered in early DKD, in part explaining the association of PU and DMG with human DKD progression observed in previous studies.NEW & NOTEWORTHY Tubular dysfunction explains the association between the nucleosides pseudouridine and N2N2-dimethyl guanosine and diabetic kidney disease.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Humans , Mice , Animals , Diabetic Nephropathies/metabolism , Pseudouridine/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Nucleosides/metabolism , Renal Elimination , Kidney/metabolism , Guanosine/metabolism
4.
J Lipid Res ; 64(6): 100381, 2023 06.
Article in English | MEDLINE | ID: mdl-37100172

ABSTRACT

Patients with chronic kidney disease (CKD) are at high risk for CVD. However, traditional CVD risk factors cannot completely explain the increased risk. Altered HDL proteome is linked with incident CVD in CKD patients, but it is unclear whether other HDL metrics are associated with incident CVD in this population. In the current study, we analyzed samples from two independent prospective case-control cohorts of CKD patients, the Clinical Phenotyping and Resource Biobank Core (CPROBE) and the Chronic Renal Insufficiency Cohort (CRIC). We measured HDL particle sizes and concentrations (HDL-P) by calibrated ion mobility analysis and HDL cholesterol efflux capacity (CEC) by cAMP-stimulated J774 macrophages in 92 subjects from the CPROBE cohort (46 CVD and 46 controls) and in 91 subjects from the CRIC cohort (34 CVD and 57 controls). We tested associations of HDL metrics with incident CVD using logistic regression analysis. No significant associations were found for HDL-C or HDL-CEC in either cohort. Total HDL-P was only negatively associated with incident CVD in the CRIC cohort in unadjusted analysis. Among the six sized HDL subspecies, only medium-sized HDL-P was significantly and negatively associated with incident CVD in both cohorts after adjusting for clinical confounders and lipid risk factors with odds ratios (per 1-SD) of 0.45 (0.22-0.93, P = 0.032) and 0.42 (0.20-0.87, P = 0.019) for CPROBE and CRIC cohorts, respectively. Our observations indicate that medium-sized HDL-P-but not other-sized HDL-P or total HDL-P, HDL-C, or HDL-CEC-may be a prognostic cardiovascular risk marker in CKD.


Subject(s)
Cardiovascular Diseases , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/complications , Cholesterol, HDL , Risk Factors , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology
6.
J Biol Chem ; 296: 100120, 2021.
Article in English | MEDLINE | ID: mdl-33234591

ABSTRACT

Increased myeloperoxidase (MPO) expression and activity are associated with atherosclerotic disease in patients with chronic kidney disease (CKD). However, the causal relationship between MPO and the development and progression of atherosclerosis in patients with CKD is unknown. Eight-week-old male low-density-lipoprotein-receptor-deficient mice were subjected to 5/6 nephrectomy, irradiated, and transplanted with bone marrow from MPO-deficient mice to induce bone marrow MPO deletion (CKD-bMPOKO) or bone marrow from WT mice as a control to maintain preserved bone marrow MPO(CKD-bMPOWT). The mice were maintained on a high-fat/high-cholesterol diet for 16 weeks. As anticipated, both groups of mice exhibited all features of moderate CKD, including elevated plasma creatinine, lower hematocrit, and increased intact parathyroid hormone but did not demonstrate any differences between the groups. Irradiation and bone marrow transplantation did not further affect body weight, blood pressure, creatinine, or hematocrit in either group. The absence of MPO expression in the bone marrow and atherosclerotic lesions of the aorta in the CKD-bMPOKO mice was confirmed by immunoblot and immunohistochemistry, respectively. Decreased MPO activity was substantiated by the absence of 3-chlorotyrosine, a specific by-product of MPO, in aortic atherosclerotic lesions as determined by both immunohistochemistry and highly sensitive LC-MS. Quantification of the aortic lesional area stained with oil red O revealed that CKD-bMPOKO mice had significantly decreased aortic plaque area as compared with CKD-bMPOWT mice. This study demonstrates the reduction of atherosclerosis in CKD mice with the deletion of MPO in bone marrow cells, strongly implicating bone-marrow-derived MPO in the pathogenesis of CKD atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Bone Marrow/metabolism , Peroxidase/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Atherosclerosis/pathology , Macrophages/metabolism , Male , Mice , Nephrectomy , Renal Insufficiency, Chronic/pathology , Serum Amyloid A Protein/metabolism
7.
Kidney Int ; 101(4): 814-823, 2022 04.
Article in English | MEDLINE | ID: mdl-35120996

ABSTRACT

Metabolomics genome wide association study (GWAS) help outline the genetic contribution to human metabolism. However, studies to date have focused on relatively healthy, population-based samples of White individuals. Here, we conducted a GWAS of 537 blood metabolites measured in the Chronic Renal Insufficiency Cohort (CRIC) Study, with separate analyses in 822 White and 687 Black study participants. Trans-ethnic meta-analysis was then applied to improve fine-mapping of potential causal variants. Mean estimated glomerular filtration rate was 44.4 and 41.5 mL/min/1.73m2 in the White and Black participants, respectively. There were 45 significant metabolite associations at 19 loci, including novel associations at PYROXD2, PHYHD1, FADS1-3, ACOT2, MYRF, FAAH, and LIPC. The strength of associations was unchanged in models additionally adjusted for estimated glomerular filtration rate and proteinuria, consistent with a direct biochemical effect of gene products on associated metabolites. At several loci, trans-ethnic meta-analysis, which leverages differences in linkage disequilibrium across populations, reduced the number and/or genomic interval spanned by potentially causal single nucleotide polymorphisms compared to fine-mapping in the White participant cohort alone. Across all validated associations, we found strong concordance in effect sizes of the potentially causal single nucleotide polymorphisms between White and Black study participants. Thus, our study identifies novel genetic determinants of blood metabolites in chronic kidney disease, demonstrates the value of diverse cohorts to improve causal inference in metabolomics GWAS, and underscores the shared genetic basis of metabolism across race.


Subject(s)
Genome-Wide Association Study , Renal Insufficiency, Chronic , Cohort Studies , Ethnicity , Female , Humans , Linkage Disequilibrium , Male , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/genetics
8.
J Lipid Res ; 62: 100135, 2021.
Article in English | MEDLINE | ID: mdl-34634315

ABSTRACT

Patients with chronic kidney disease (CKD) are at high risk for CVD. However, traditional lipid risk factors, including low HDL levels, cannot completely explain the increased risk. Altered HDL proteome is linked with both CVD and CKD, but the role of HDL proteins in incident CVD events in patients with CKD is unknown. In this prospective case-control study, we used targeted proteomics to quantify 31 HDL proteins in 92 subjects (46 incident new CVD and 46 one-to-one matched controls) at various stages of CKD. We tested associations of HDL proteins with incident CVD using matched logistic regression analysis. In the model fully adjusted for clinical confounders, lipid levels, C-reactive protein, and proteinuria, no significant associations were found for HDL-C, but we observed inverse associations between levels of HDL proteins paraoxonase/arylesterase 1 (PON1), paraoxonase/arylesterase 3 (PON3), and LCAT and incident CVD. Odds ratios (per 1 SD) were 0.38 (0.18-0.97, P = 0.042), 0.42 (0.20-0.92, P = 0.031), and 0.30 (0.11-0.83, P = 0.020) for PON1, PON3, and LCAT, respectively. Apolipoprotein A-IV remained associated with incident CVD in CKD patients in models adjusted for clinical confounders and lipid levels but lost significance with the addition of C-reactive protein and proteinuria to the model. In conclusion, levels of four HDL proteins, PON1, PON3, LCAT, and apolipoprotein A-IV, were found to be inversely associated with incident CVD events in CKD patients. Our observations indicate that HDLs' protein cargo, but not HDL-C levels, can serve as a marker-and perhaps mediator-for elevated CVD risk in CKD patients.


Subject(s)
Cardiovascular Diseases/metabolism , Lipoproteins, HDL/metabolism , Renal Insufficiency, Chronic/metabolism , Adult , Aged , Aged, 80 and over , Apolipoproteins A/metabolism , Aryldialkylphosphatase/metabolism , Female , Humans , Male , Middle Aged , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Regression Analysis
9.
Int J Mol Sci ; 21(21)2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33121167

ABSTRACT

Chronic kidney disease (CKD) is prevalent in 9.1% of the global population and is a significant public health problem associated with increased morbidity and mortality. CKD is associated with highly prevalent physiological and metabolic disturbances such as hypertension, obesity, insulin resistance, cardiovascular disease, and aging, which are also risk factors for CKD pathogenesis and progression. Podocytes and proximal tubular cells of the kidney strongly express AMP-activated protein kinase (AMPK). AMPK plays essential roles in glucose and lipid metabolism, cell survival, growth, and inflammation. Thus, metabolic disease-induced renal diseases like obesity-related and diabetic chronic kidney disease demonstrate dysregulated AMPK in the kidney. Activating AMPK ameliorates the pathological and phenotypical features of both diseases. As a metabolic sensor, AMPK regulates active tubular transport and helps renal cells to survive low energy states. AMPK also exerts a key role in mitochondrial homeostasis and is known to regulate autophagy in mammalian cells. While the nutrient-sensing role of AMPK is critical in determining the fate of renal cells, the role of AMPK in kidney autophagy and mitochondrial quality control leading to pathology in metabolic disease-related CKD is not very clear and needs further investigation. This review highlights the crucial role of AMPK in renal cell dysfunction associated with metabolic diseases and aims to expand therapeutic strategies by understanding the molecular and cellular processes underlying CKD.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Metabolic Diseases/complications , Renal Insufficiency, Chronic/metabolism , Disease Progression , Gene Expression Regulation , Humans , Lipid Metabolism , Metabolic Diseases/metabolism , Mitochondria/metabolism , Renal Insufficiency, Chronic/etiology
10.
J Lipid Res ; 60(5): 937-952, 2019 05.
Article in English | MEDLINE | ID: mdl-30862696

ABSTRACT

High-fat diet (HFD) causes renal lipotoxicity that is ameliorated with AMP-activated protein kinase (AMPK) activation. Although bioactive eicosanoids increase with HFD and are essential in regulation of renal disease, their role in the inflammatory response to HFD-induced kidney disease and their modulation by AMPK activation remain unexplored. In a mouse model, we explored the effects of HFD on eicosanoid synthesis and the role of AMPK activation in ameliorating these changes. We used targeted lipidomic profiling with quantitative MS to determine PUFA and eicosanoid content in kidneys, urine, and renal arterial and venous circulation. HFD increased phospholipase expression as well as the total and free pro-inflammatory arachidonic acid (AA) and anti-inflammatory DHA in kidneys. Consistent with the parent PUFA levels, the AA- and DHA-derived lipoxygenase (LOX), cytochrome P450, and nonenzymatic degradation (NE) metabolites increased in kidneys with HFD, while EPA-derived LOX and NE metabolites decreased. Conversely, treatment with 5-aminoimidazole-4-carboxamide-1-ß-D-furanosyl 5'-monophosphate (AICAR), an AMPK activator, reduced the free AA and DHA content and the DHA-derived metabolites in kidney. Interestingly, kidney and circulating AA, AA metabolites, EPA-derived LOX, and NE metabolites are increased with HFD; whereas, DHA metabolites are increased in kidney in contrast to their decreased circulating levels with HFD. Together, these changes showcase HFD-induced pro- and anti-inflammatory eicosanoid dysregulation and highlight the role of AMPK in correcting HFD-induced dysregulated eicosanoid pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diet, High-Fat/adverse effects , Eicosanoids/metabolism , Kidney Diseases/metabolism , Animals , Kidney Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL
11.
J Biol Chem ; 293(19): 7238-7249, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29581235

ABSTRACT

Increased myeloperoxidase (MPO) levels and activity are associated with increased cardiovascular risk among individuals with chronic kidney disease (CKD). However, a lack of good animal models for examining the presence and catalytic activity of MPO in vascular lesions has impeded mechanistic studies into CKD-associated cardiovascular diseases. Here, we show for the first time that exaggerated atherosclerosis in a pathophysiologically relevant CKD mouse model is associated with increased macrophage-derived MPO activity. Male 7-week-old LDL receptor-deficient mice underwent sham (control mice) or 5/6 nephrectomy and were fed either a low-fat or high-fat, high-cholesterol diet for 24 weeks, and the extents of atherosclerosis and vascular reactivity were assessed. MPO expression and oxidation products-protein-bound oxidized tyrosine moieties 3-chlorotyrosine, 3-nitrotyrosine, and o,o'-dityrosine-were examined with immunoassays and confirmed with mass spectrometry (MS). As anticipated, the CKD mice had significantly higher plasma creatinine, urea nitrogen, and intact parathyroid hormone along with lower hematocrit and body weight. On both the diet regimens, CKD mice did not have hypertension but had lower cholesterol and triglyceride levels than the control mice. Despite the lower cholesterol levels, CKD mice had increased aortic plaque areas, fibrosis, and luminal narrowing. They also exhibited increased MPO expression and activity (i.e. increased oxidized tyrosines) that co-localized with infiltrating lesional macrophages and diminished vascular reactivity. In summary, unlike non-CKD mouse models of atherosclerosis, CKD mice exhibit increased MPO expression and catalytic activity in atherosclerotic lesions, which co-localize with lesional macrophages. These results implicate macrophage-derived MPO in CKD-accelerated atherosclerosis.


Subject(s)
Aorta/metabolism , Atherosclerosis/metabolism , Kidney Failure, Chronic/complications , Muscle Proteins/metabolism , Oxidants/metabolism , Peroxidase/metabolism , Animals , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Blood Urea Nitrogen , Creatinine/blood , Diet, Fat-Restricted , Diet, High-Fat , Dietary Fats/administration & dosage , Disease Models, Animal , Disease Progression , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/physiopathology , Kidney Function Tests , Lipids/blood , Lipoproteins/blood , Macrophages/enzymology , Male , Mice , Mice, Knockout , Nephrectomy , Oxidative Stress , Parathyroid Hormone/blood , Receptors, LDL/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasodilation
12.
Am J Nephrol ; 48(4): 269-277, 2018.
Article in English | MEDLINE | ID: mdl-30326477

ABSTRACT

BACKGROUND: The gut microbiota is altered in patients with chronic kidney disease (CKD), and cardiovascular risk increases with progressive CKD. This study examined the potential link between short chain fatty acids (SCFAs), which are produced by the gut microbiota, and cardiovascular outcomes in patients with CKD. METHODS: SCFAs were measured using a targeted liquid chromatography-mass spectrometry platform in baseline plasma samples from 214 patients with CKD enrolled in the Clinical Phenotyping Resource and Biobank Core; 81 patients with coronary artery disease (CAD) and 133 without CAD were randomly assigned to training and validation subsets. The primary outcome was a history of CAD and the secondary outcome was a composite history of cardiovascular disease (CVD) at enrollment. RESULTS: We found significantly higher levels of the SCFA valerate among patients with CAD as compared with patients without CAD in the training set (p < 0.001). The valerate concentrations were also significantly higher among subjects with composite outcomes of CVD compared to those without CVD (p = 0.006). These results were subsequently replicated in the validation set. Logistic regression analysis revealed a strong independent association between plasma valerate levels and CVD in both training and validation sets. When valerate was added to the base clinical model comprising of diabetes, hypertension, urinary protein-creatinine ratio, and estimated glomerular filtration rate, it increased the c-statistics for predicting CVD from 0.68 to 0.79 (p = 0.02) in the training set, an observation which was confirmed in the validation set. -Conclusion: This study provides evidence for alterations in gut-microbiota-derived SCFAs with advancing CKD, demonstrates the association of higher plasma valerate levels with pre-existing CVD, and reveals areas for future exploration of cardiovascular risk in patients with CKD.


Subject(s)
Coronary Artery Disease/diagnosis , Gastrointestinal Microbiome/physiology , Pentanoic Acids/blood , Renal Insufficiency, Chronic/complications , Adult , Aged , Coronary Artery Disease/blood , Coronary Artery Disease/etiology , Disease Progression , Female , Glomerular Filtration Rate , Humans , Kidney/physiopathology , Male , Middle Aged , Pentanoic Acids/metabolism , Predictive Value of Tests , Prognosis , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , Risk Assessment/methods , Risk Factors
13.
Arterioscler Thromb Vasc Biol ; 37(6): 1068-1075, 2017 06.
Article in English | MEDLINE | ID: mdl-28428219

ABSTRACT

OBJECTIVE: CETP (cholesteryl ester transfer protein) plays an important role in lipoprotein metabolism; however, whether inhibition of CETP activity can prevent cardiovascular disease remains controversial. APPROACH AND RESULTS: We generated CETP knockout (KO) rabbits by zinc finger nuclease gene editing and compared their susceptibility to cholesterol diet-induced atherosclerosis to that of wild-type (WT) rabbits. On a chow diet, KO rabbits showed higher plasma levels of high-density lipoprotein (HDL) cholesterol than WT controls, and HDL particles of KO rabbits were essentially rich in apolipoprotein AI and apolipoprotein E contents. When challenged with a cholesterol-rich diet for 18 weeks, KO rabbits not only had higher HDL cholesterol levels but also lower total cholesterol levels than WT rabbits. Analysis of plasma lipoproteins revealed that reduced plasma total cholesterol in KO rabbits was attributable to decreased apolipoprotein B-containing particles, while HDLs remained higher than that in WT rabbits. Both aortic and coronary atherosclerosis was significantly reduced in KO rabbits compared with WT rabbits. Apolipoprotein B-depleted plasma isolated from CETP KO rabbits showed significantly higher capacity for cholesterol efflux from macrophages than that from WT rabbits. Furthermore, HDLs isolated from CETP KO rabbits suppressed tumor necrosis factor-α-induced vascular cell adhesion molecule 1 and E-selectin expression in cultured endothelial cells. CONCLUSIONS: These results provide evidence that genetic ablation of CETP activity protects against cholesterol diet-induced atherosclerosis in rabbits.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Cholesterol Ester Transfer Proteins/deficiency , Cholesterol, Dietary , Coronary Artery Disease/prevention & control , Lipid Metabolism, Inborn Errors/metabolism , Macrophages/metabolism , Animals , Animals, Genetically Modified , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoprotein A-I/blood , Apolipoproteins B/blood , Apolipoproteins E/blood , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Line , Cholesterol Ester Transfer Proteins/blood , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol, HDL/blood , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Models, Animal , E-Selectin/metabolism , Female , Gene Editing , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Lipid Metabolism, Inborn Errors/blood , Lipid Metabolism, Inborn Errors/genetics , Male , Mice , Rabbits , Time Factors , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/metabolism
14.
Physiol Genomics ; 47(6): 187-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25829393

ABSTRACT

The gut microbiota plays a critical role in maintaining physiological homeostasis. This study was designed to evaluate whether gut microbial composition affects hypertension. 16S rRNA genes obtained from cecal samples of Dahl salt-sensitive (S) and Dahl salt-resistant (R) rats were sequenced. Bacteria of the phylum Bacteroidetes were higher in the S rats compared with the R rats. Furthermore, the family S24-7 of the phylum Bacteroidetes and the family Veillonellaceae of the phylum Firmicutes were higher in the S rats compared with the R rats. Analyses of the various phylogenetic groups of cecal microbiota revealed significant differences between S and R rats. Both strains were maintained on a high-salt diet, administered antibiotics for ablation of microbiota, transplanted with S or R rat cecal contents, and monitored for blood pressure (BP). Systolic BP of the R rats remained unaltered irrespective of S or R rat cecal transplantation. Surprisingly, compared with the S rats given S rat cecal content, systolic BP of the S rats given a single bolus of cecal content from R rats was consistently and significantly elevated during the rest of their life, and they had a shorter lifespan. A lower level of fecal bacteria of the family Veillonellaceae and increased plasma acetate and heptanoate were features associated with the increased BP observed in the S rats given R rat microbiota compared with the S rats given S rat microbiota. These data demonstrate a link between microbial content and BP regulation and, because the S and R rats differ in their genomic composition, provide the necessary basis to further examine the relationship between the host genome and microbiome in the context of BP regulation in the Dahl rats.


Subject(s)
Gastrointestinal Microbiome , Hypertension/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Blood Pressure/drug effects , Cecum/drug effects , Cecum/microbiology , Cecum/transplantation , Fatty Acids/blood , Gastrointestinal Microbiome/drug effects , Genetic Variation , Genome , Hypertension/blood , Hypertension/physiopathology , Kaplan-Meier Estimate , Longevity , Metabolomics , Rats, Inbred Dahl , Sodium/blood , Sodium/urine , Systole/drug effects
15.
J Card Fail ; 21(12): 963-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26497755

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is increasingly recognized as a distinct entity with unique pathophysiology. In the Dietary Approaches to Stop Hypertension in Diastolic Heart Failure (DASH-DHF) study, the sodium-restricted Dietary Approaches to Stop Hypertension diet (DASH/SRD) was associated with improved blood pressure and cardiovascular function in 13 hypertensive patients with HFpEF. With the use of targeted metabolomics, we explored metabolite changes and their relationship with energy-dependent measures of cardiac function in DASH-DHF. METHODS AND RESULTS: With the use of chromatography and mass spectrometry, 152 metabolites including amino acids, free fatty acids, phospholipids, diglycerides, triglycerides, cholesterol esters, and acyl carnitines were measured. Comparison of baseline and post-DASH/SRD samples revealed increases in short-chain acetyl, butryl, and propionyl carnitines (P values .02, .03, .03, respectively). Increases in propionyl carnitine correlated with ventricular-arterial coupling ratio (Ees:Ea; r = 0.78; P = .005) and ventricular contractility (maximum rate of change of pressure-normalized stress [dσ*/dtmax]; r = 0.66; P = .03). Changes in L-carnitine also correlated with Ees:Ea (r = 0.62; P = .04) and dσ*/dtmax (r = 0.60; P = .05) and inversely with ventricular stiffness (r = -0.63; P = .03). CONCLUSIONS: Metabolite profile changes of patients with HFpEF during dietary modification with the use of DASH/SRD suggest improved energy substrate utilization. Additional studies are needed to clarify connections between diet, metabolic changes, and myocardial function in HFpEF.


Subject(s)
Diet, Sodium-Restricted , Heart Failure, Diastolic/blood , Heart Failure, Diastolic/diet therapy , Hypertension/diet therapy , Metabolome/physiology , Stroke Volume/physiology , Adult , Age Distribution , Aged , Chromatography/methods , Echocardiography, Doppler , Female , Heart Failure, Diastolic/diagnostic imaging , Heart Failure, Diastolic/epidemiology , Heart Failure, Diastolic/physiopathology , Humans , Hypertension/epidemiology , Hypertension/physiopathology , Incidence , Male , Mass Spectrometry/methods , Middle Aged , Pilot Projects , Prognosis , Risk Assessment , Sampling Studies , Severity of Illness Index , Sex Distribution
16.
Kidney Int ; 85(5): 1214-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24429397

ABSTRACT

Here we studied plasma metabolomic profiles as determinants of progression to end-stage renal disease (ESRD) in patients with type 2 diabetes (T2D). This nested case-control study evaluated 40 cases who progressed to ESRD during 8-12 years of follow-up and 40 controls who remained alive without ESRD from the Joslin Kidney Study cohort. Controls were matched with cases for baseline clinical characteristics, although controls had slightly higher eGFR and lower levels of urinary albumin excretion than cases. Plasma metabolites at baseline were measured by mass spectrometry-based global metabolomic profiling. Of the named metabolites in the library, 262 were detected in at least 80% of the study patients. The metabolomic platform recognized 78 metabolites previously reported to be elevated in ESRD (uremic solutes). Sixteen were already elevated in the baseline plasma of our cases years before ESRD developed. Other uremic solutes were either not different or not commonly detectable. Essential amino acids and their derivatives were significantly depleted in the cases, whereas certain amino acid-derived acylcarnitines were increased. All findings remained statistically significant after adjustment for differences between study groups in albumin excretion rate, eGFR, or HbA1c. Uremic solute differences were confirmed by quantitative measurements. Thus, abnormal plasma concentrations of putative uremic solutes and essential amino acids either contribute to progression to ESRD or are a manifestation of an early stage(s) of the disease process that leads to ESRD in T2D.


Subject(s)
Amino Acids, Essential/blood , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/etiology , Kidney Failure, Chronic/etiology , Metabolomics , Uremia/etiology , Aged , Biomarkers/blood , Boston , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetic Nephropathies/blood , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/physiopathology , Disease Progression , Female , Glomerular Filtration Rate , Glycated Hemoglobin/analysis , Humans , Kidney/physiopathology , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/physiopathology , Male , Mass Spectrometry , Metabolomics/methods , Middle Aged , Time Factors , Uremia/blood , Uremia/diagnosis , Uremia/physiopathology
17.
Cardiovasc Diabetol ; 13: 82, 2014 Apr 21.
Article in English | MEDLINE | ID: mdl-24751283

ABSTRACT

BACKGROUND: Glycation of high-density lipoprotein (HDL) decreases its ability to induce cyclooxygenase-2 (COX-2) expression and prostacyclin I-2 (PGI-2) release in endothelial cells. Whether lipid content of HDL, especially sphingosine-1-phosphate (S1P), plays any specific role in restoring the protective function of HDL in type 2 diabetes mellitus (T2DM) is still unknown. METHODS AND RESULTS: Immunochemical techniques demonstrated that glycated HDL loses its protective function of regulating COX-2 expression compared with diabetic HDL. We proved that the lipid content, especially phospholipid content differed between diabetic HDL and glycated HDL. Levels of HDL-c-bound S1P were increased in T2DM compared with control subjects as detected by UPLC-MS/MS (HDL-c-bound S1P in control subjects vs. T2DM: 309.1 ± 13.71 pmol/mg vs. 382.1 ± 24.45 pmol/mg, P < 0.05). Additionally, mRNA levels of S1P lyase enzymes and S1P phosphatase 1/2 were decreased in peripheral blood by real-time PCR. Antagonist of S1P receptor 1 and 3 (S1PR1/3) diminished the functional difference between apoHDL&PL (HDL containing the protein components and phospholipids) and diabetic apoHDL&PL (diabetic HDL containing the protein components and phospholipids). With different doses of S1P reconstituted on glycated HDL, its function in inducing the COX-2 expression was restored to the same level as diabetic HDL. The mechanism of S1P reconstituted HDL (rHDL) in the process of regulating COX-2 expression involved the phosphorylation of ERK/MAPK-CREB signal pathway. CONCLUSION/SIGNIFICANCE: S1P harbored on HDL is the main factor which restores its protective function in endothelial cells in T2DM. S1P and its receptors are potential therapeutic targets in ameliorating the vascular dysfunction in T2DM.


Subject(s)
Cytoprotection/physiology , Diabetes Mellitus, Type 2/blood , Glycosylation , Lipoproteins, HDL/blood , Lysophospholipids/blood , Sphingosine/analogs & derivatives , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Sphingosine/blood
18.
J Am Soc Nephrol ; 24(11): 1901-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23949796

ABSTRACT

Diabetic kidney disease is the leading cause of ESRD, but few biomarkers of diabetic kidney disease are available. This study used gas chromatography-mass spectrometry to quantify 94 urine metabolites in screening and validation cohorts of patients with diabetes mellitus (DM) and CKD(DM+CKD), in patients with DM without CKD (DM-CKD), and in healthy controls. Compared with levels in healthy controls, 13 metabolites were significantly reduced in the DM+CKD cohorts (P≤0.001), and 12 of the 13 remained significant when compared with the DM-CKD cohort. Many of the differentially expressed metabolites were water-soluble organic anions. Notably, organic anion transporter-1 (OAT1) knockout mice expressed a similar pattern of reduced levels of urinary organic acids, and human kidney tissue from patients with diabetic nephropathy demonstrated lower gene expression of OAT1 and OAT3. Analysis of bioinformatics data indicated that 12 of the 13 differentially expressed metabolites are linked to mitochondrial metabolism and suggested global suppression of mitochondrial activity in diabetic kidney disease. Supporting this analysis, human diabetic kidney sections expressed less mitochondrial protein, urine exosomes from patients with diabetes and CKD had less mitochondrial DNA, and kidney tissues from patients with diabetic kidney disease had lower gene expression of PGC1α (a master regulator of mitochondrial biogenesis). We conclude that urine metabolomics is a reliable source for biomarkers of diabetic complications, and our data suggest that renal organic ion transport and mitochondrial function are dysregulated in diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/metabolism , Metabolomics/methods , Mitochondrial Diseases/etiology , Adult , Aged , Female , Glomerular Filtration Rate , Humans , Ion Transport , Male , Middle Aged , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Renal Insufficiency, Chronic/metabolism , Transcription Factors/genetics
19.
J Am Soc Nephrol ; 22(10): 1846-55, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921143

ABSTRACT

The mechanisms underlying the association between obesity and progressive renal disease are not well understood. Exposure to a high-fat diet decreases levels of the cellular energy sensor AMPK in many organs, including the kidney, but whether AMPK contributes to the pathophysiology of kidney disease induced by a high-fat diet is unknown. In this study, we randomly assigned C57BL/6J mice to a standard or high-fat diet. After 1 week, mice fed a high-fat diet exhibited an increase in body weight, renal hypertrophy, an increase in urine H(2)O(2) and urine MCP-1, and a decrease in circulating adiponectin levels and renal AMPK activity. Urine ACR progressively increased after 4 weeks of a high-fat diet. After 12 weeks, kidneys of mice fed a high-fat diet demonstrated a marked increase in markers of fibrosis and inflammation, and AMPK activity remained significantly suppressed. To determine whether inhibition of AMPK activity explained these renal effects, we administered an AMPK activator along with a high-fat diet for 1 week. Although AMPK activation did not abrogate the weight gain, it reduced the renal hypertrophy, urine H(2)O(2), and urine and renal MCP-1. In vitro, AMPK activation completely inhibited the induction of MCP-1 by palmitic acid in mesangial cells. In conclusion, these data suggest that the energy sensor AMPK mediates the early renal effects of a high-fat diet.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Dietary Fats/adverse effects , Kidney Diseases/metabolism , Obesity/enzymology , Albuminuria/etiology , Animals , Biomarkers/urine , Chemokine CCL2/metabolism , Diet, High-Fat/adverse effects , Enzyme Activation , Hyperglycemia/etiology , Hypertrophy/etiology , Insulin Resistance , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Mice , Mice, Inbred C57BL , Obesity/complications
20.
J Am Soc Nephrol ; 22(6): 1144-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21511828

ABSTRACT

Pirfenidone is an oral antifibrotic agent that benefits diabetic nephropathy in animal models, but whether it is effective for human diabetic nephropathy is unknown. We conducted a randomized, double-blind, placebo-controlled study in 77 subjects with diabetic nephropathy who had elevated albuminuria and reduced estimated GFR (eGFR) (20 to 75 ml/min per 1.73 m²). The prespecified primary outcome was a change in eGFR after 1 year of therapy. We randomly assigned 26 subjects to placebo, 26 to pirfenidone at 1200 mg/d, and 25 to pirfenidone at 2400 mg/d. Among the 52 subjects who completed the study, the mean eGFR increased in the pirfenidone 1200-mg/d group (+3.3 ± 8.5 ml/min per 1.73 m²) whereas the mean eGFR decreased in the placebo group (-2.2 ± 4.8 ml/min per 1.73 m²; P = 0.026 versus pirfenidone at 1200 mg/d). The dropout rate was high (11 of 25) in the pirfenidone 2400-mg/d group, and the change in eGFR was not significantly different from placebo (-1.9 ± 6.7 ml/min per 1.73 m²). Of the 77 subjects, 4 initiated hemodialysis in the placebo group, 1 in the pirfenidone 2400-mg/d group, and none in the pirfenidone 1200-mg/d group during the study (P = 0.25). Baseline levels of plasma biomarkers of inflammation and fibrosis significantly correlated with baseline eGFR but did not predict response to therapy. In conclusion, these results suggest that pirfenidone is a promising agent for individuals with overt diabetic nephropathy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/physiopathology , Disease Progression , Pyridones/therapeutic use , Adult , Aged , Albuminuria/urine , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Biomarkers/urine , Creatinine/urine , Diabetic Nephropathies/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibrosis , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/physiology , Humans , Kidney/pathology , Male , Middle Aged , Pyridones/pharmacology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL