Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Nat Commun ; 14(1): 8260, 2023 Dec 13.
Article En | MEDLINE | ID: mdl-38086839

Metabolic reprogramming in cancer and immune cells occurs to support their increasing energy needs in biological tissues. Here we propose Single Cell Spatially resolved Metabolic (scSpaMet) framework for joint protein-metabolite profiling of single immune and cancer cells in male human tissues by incorporating untargeted spatial metabolomics and targeted multiplexed protein imaging in a single pipeline. We utilized the scSpaMet to profile cell types and spatial metabolomic maps of 19507, 31156, and 8215 single cells in human lung cancer, tonsil, and endometrium tissues, respectively. The scSpaMet analysis revealed cell type-dependent metabolite profiles and local metabolite competition of neighboring single cells in human tissues. Deep learning-based joint embedding revealed unique metabolite states within cell types. Trajectory inference showed metabolic patterns along cell differentiation paths. Here we show scSpaMet's ability to quantify and visualize the cell-type specific and spatially resolved metabolic-protein mapping as an emerging tool for systems-level understanding of tissue biology.


Lung Neoplasms , Metabolomics , Female , Male , Humans , Metabolomics/methods , Systems Biology
2.
Am J Kidney Dis ; 81(6): 728-731, 2023 06.
Article En | MEDLINE | ID: mdl-36805076
3.
Cell Mol Life Sci ; 79(10): 516, 2022 Sep 14.
Article En | MEDLINE | ID: mdl-36102977

Diabetic nephropathy (DN) is a significant complication of diabetes and the leading cause of end-stage renal disease. Hyperglycemia-induced dysfunction of the glomerular podocytes is a major contributor to the deterioration of renal function in DN. Previously, we demonstrated that podocyte-specific disruption of the Src homology phosphatase 2 (Shp2) ameliorated lipopolysaccharide-induced renal injury. This study aims to evaluate the contribution of Shp2 to podocyte function under hyperglycemia and explore the molecular underpinnings. We report elevated Shp2 in the E11 podocyte cell line under high glucose and the kidney under streptozotocin- and high-fat diet-induced hyperglycemia. Consistently, Shp2 disruption in podocytes was associated with partial renoprotective effects under hyperglycemia, as evidenced by the preserved renal function. At the molecular level, Shp2 deficiency was associated with altered renal insulin signaling and diminished hyperglycemia-induced renal endoplasmic reticulum stress, inflammation, and fibrosis. Additionally, Shp2 knockdown in E11 podocytes mimicked the in vivo deficiency of this phosphatase and ameliorated the deleterious impact of high glucose, whereas Shp2 reconstitution reversed these effects. Moreover, Shp2 deficiency attenuated high glucose-induced E11 podocyte migration. Further, we identified the protein tyrosine kinase FYN as a putative mediator of Shp2 signaling in podocytes under high glucose. Collectively, these findings suggest that Shp2 inactivation may afford protection to podocytes under hyperglycemia and highlight this phosphatase as a potential target to ameliorate glomerular dysfunction in DN.


Diabetic Nephropathies , Hyperglycemia , Podocytes , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Glucose/metabolism , Hyperglycemia/complications , Hyperglycemia/genetics , Hyperglycemia/metabolism , Mice , Phosphoric Monoester Hydrolases/metabolism , Podocytes/metabolism
4.
Pediatr Nephrol ; 37(11): 2583-2597, 2022 11.
Article En | MEDLINE | ID: mdl-34913986

Diabetic kidney disease (DKD), previously encountered predominantly in adult patients, is rapidly gaining center stage as a childhood morbidity and one that pediatric nephrologists are likely to encounter with increasing frequency. This is in large part due to the obesity epidemic and the consequent rise in type 2 diabetes in children and adolescents, as well as the more aggressive diabetes phenotype in today's youth with more rapid ß-cell decline and faster development and progression of diabetes-related complications along with lower responsiveness to the treatments used in adults. DKD, an end-organ complication of diabetes, is at the very least a marker of, and more likely a predisposing factor for, the development of adverse cardiovascular outcomes and premature mortality in children with diabetes. On an optimistic note, several new therapeutic approaches are now available for the management of diabetes in adults, such as GLP1 receptor agonists, SGLT2 inhibitors, and DPP4 inhibitors, that have also been shown to have a favorable impact on cardiorenal outcomes. Also promising is the success of very low-energy diets in inducing remission of diabetes in adults. However, the addition of these pharmacological and dietary approaches to the management toolbox of diabetes and DKD in children and adolescents awaits thorough assessment of their safety and efficacy in this population. This review outlines the scope of diabetes and DKD, and new developments that may favorably impact the management of children and young adults with diabetes and DKD.


Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Dipeptidyl-Peptidase IV Inhibitors , Sodium-Glucose Transporter 2 Inhibitors , Biomarkers , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/therapy , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/etiology , Diabetic Nephropathies/therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Humans , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
5.
Metab Syndr Relat Disord ; 19(9): 491-497, 2021 11.
Article En | MEDLINE | ID: mdl-34448598

Background: Nonalcoholic steatohepatitis (NASH) is a severe form of fatty liver disease that has been shown to be associated with chronic kidney disease (CKD). Mechanism for the association of NASH with CKD remains unclear. In this study, we examined the association between NASH severity and kidney glucose uptake and the liver-secreted signaling molecule fibroblast growth factor 21 (FGF21). Methods: Kinetic parameters for kidney glucose transport rate (K1) and standardized uptake value (SUV) were determined using dynamic positron emission tomography after injection of 18F-fluorodeoxyglucose. Liver biopsies were scored for NASH activity (inflammation and ballooning), fibrosis, and steatosis FGF21 was measured from fasting serum samples. Patients were categorized by liver biopsy and multivariate analyses were performed to evaluate the associations. Results: Of 41 NASH patients 73% were females, 71% white, 51% with steatosis ≥2, 39% with NASH activity ≥4 and fibrosis ≥3. With severe NASH activity, kidney SUV significantly increased even when adjusted for underlying insulin-resistant (IR) state. Kidney K1 decreased significantly in higher liver activity in unadjusted models but not when adjusted for IR. FGF21 decreased with severe liver activity in adjusted models (P < 0.05) and associated with kidney K1 but not SUV. Conclusion: Our pilot data indicate that kidney glucose metabolism associates with NASH activity and FGF21 levels, suggesting a potential mechanism to NASH-induced CKD. Clinical Trials.gov ID: NCT02754037.


Fibroblast Growth Factors , Glucose , Kidney , Non-alcoholic Fatty Liver Disease , Female , Fibroblast Growth Factors/blood , Glucose/metabolism , Humans , Kidney/metabolism , Male , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/physiopathology , Renal Insufficiency, Chronic/epidemiology , Severity of Illness Index
6.
N Engl J Med ; 382(26): 2493-2503, 2020 06 25.
Article En | MEDLINE | ID: mdl-32579810

BACKGROUND: Higher serum urate levels are associated with an increased risk of diabetic kidney disease. Lowering of the serum urate level with allopurinol may slow the decrease in the glomerular filtration rate (GFR) in persons with type 1 diabetes and early-to-moderate diabetic kidney disease. METHODS: In a double-blind trial, we randomly assigned participants with type 1 diabetes, a serum urate level of at least 4.5 mg per deciliter, an estimated GFR of 40.0 to 99.9 ml per minute per 1.73 m2 of body-surface area, and evidence of diabetic kidney disease to receive allopurinol or placebo. The primary outcome was the baseline-adjusted GFR, as measured with iohexol, after 3 years plus a 2-month washout period. Secondary outcomes included the decrease in the iohexol-based GFR per year and the urinary albumin excretion rate after washout. Safety was also assessed. RESULTS: A total of 267 patients were assigned to receive allopurinol and 263 to receive placebo. The mean age was 51.1 years, the mean duration of diabetes 34.6 years, and the mean glycated hemoglobin level 8.2%. The mean baseline iohexol-based GFR was 68.7 ml per minute per 1.73 m2 in the allopurinol group and 67.3 ml per minute per 1.73 m2 in the placebo group. During the intervention period, the mean serum urate level decreased from 6.1 to 3.9 mg per deciliter with allopurinol and remained at 6.1 mg per deciliter with placebo. After washout, the between-group difference in the mean iohexol-based GFR was 0.001 ml per minute per 1.73 m2 (95% confidence interval [CI], -1.9 to 1.9; P = 0.99). The mean decrease in the iohexol-based GFR was -3.0 ml per minute per 1.73 m2 per year with allopurinol and -2.5 ml per minute per 1.73 m2 per year with placebo (between-group difference, -0.6 ml per minute per 1.73 m2 per year; 95% CI, -1.5 to 0.4). The mean urinary albumin excretion rate after washout was 40% (95% CI, 0 to 80) higher with allopurinol than with placebo. The frequency of serious adverse events was similar in the two groups. CONCLUSIONS: We found no evidence of clinically meaningful benefits of serum urate reduction with allopurinol on kidney outcomes among patients with type 1 diabetes and early-to-moderate diabetic kidney disease. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; PERL ClinicalTrials.gov number, NCT02017171.).


Allopurinol/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetic Nephropathies/prevention & control , Enzyme Inhibitors/therapeutic use , Glomerular Filtration Rate/drug effects , Uric Acid/blood , Xanthine Oxidase/antagonists & inhibitors , Adult , Aged , Allopurinol/adverse effects , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/physiopathology , Double-Blind Method , Enzyme Inhibitors/adverse effects , Female , Humans , Male , Middle Aged , Renin-Angiotensin System , Treatment Failure
7.
Diabetes Care ; 42(8): 1454-1463, 2019 08.
Article En | MEDLINE | ID: mdl-31186299

OBJECTIVE: Higher serum uric acid (SUA) is associated with diabetic kidney disease (DKD). Preventing Early Renal Loss in Diabetes (PERL) evaluates whether lowering SUA with allopurinol slows glomerular filtration rate (GFR) loss in people with type 1 diabetes (T1D) and mild to moderate DKD. We present the PERL rationale, design, and baseline characteristics. RESEARCH DESIGN AND METHODS: This double-blind, placebo-controlled, multicenter trial randomized 530 participants with T1D, estimated GFR (eGFR) of 40-99.9 mL/min/1.73 m2, SUA ≥4.5 m/dL, and micro- to macroalbuminuric DKD or normoalbuminuria with declining kidney function (NDKF) (defined as historical eGFR decline ≥3 mL/min/1.73 m2/year) to allopurinol or placebo. The primary outcome is baseline-adjusted iohexol GFR (iGFR) after 3 years of treatment plus a 2-month washout period. RESULTS: Participants are 66% male and 84% white. At baseline, median age was 52 years and diabetes duration was 35 years, 93% of participants had hypertension, and 90% were treated with renin-angiotensin system inhibitors (median blood pressure 127/71 mmHg). Median HbA1c was 8%, SUA 5.9 mg/dL, iGFR 68 mL/min/1.73 m2, and historical eGFR slope -3.5 mL/min/1.73 m2/year. Compared with participants with albuminuria (n = 419), those with NDKF (n = 94) were significantly older (56 vs. 52 years), had lower HbA1c (7.7 vs. 8.1%) and SUA (5.4 vs. 6.0 mg/dL), and had higher eGFR (82 vs. 74 mL/min/1.73 m2) and historical eGFR loss (-4.7 vs. -2.5 mL/min/1.73 m2/year). These differences persisted when comparing groups with similar rates of historical eGFR loss. CONCLUSIONS: PERL will determine the effect of allopurinol on mild to moderate DKD in T1D, with or without albuminuria. Participants with normoalbuminuria and rapid GFR loss manifested many DKD risk factors of those with albuminuria, but with less severity.


Allopurinol/therapeutic use , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/drug therapy , Glomerular Filtration Rate/drug effects , Uric Acid/blood , Aged , Albuminuria/drug therapy , Albuminuria/etiology , Albuminuria/physiopathology , Blood Pressure , Diabetes Mellitus, Type 1/blood , Diabetic Nephropathies/etiology , Diabetic Nephropathies/physiopathology , Disease Progression , Double-Blind Method , Female , Humans , Male , Middle Aged , Renin-Angiotensin System/drug effects , Risk Factors
8.
Hypertension ; 74(1): 83-94, 2019 07.
Article En | MEDLINE | ID: mdl-31079532

In patients with diabetic kidney disease (DKD), plasma renin activity is usually decreased, but there is limited information on urinary renin and its origin. Urinary renin was evaluated in samples from patients with longstanding type I diabetes mellitus and mice with streptozotocin-induced diabetes mellitus. Renin-reporter mouse model (Ren1d-Cre;mT/mG) was made diabetic with streptozotocin to examine whether the distribution of cells of the renin lineage was altered in a chronic diabetic environment. Active renin was increased in urine samples from patients with DKD (n=36), compared with those without DKD (n=38; 3.2 versus 1.3 pg/mg creatinine; P<0.001). In mice with streptozotocin-induced diabetes mellitus, urine renin was also increased compared with nondiabetic controls. By immunohistochemistry, in mice with streptozotocin-induced diabetes mellitus, juxtaglomerular apparatus and proximal tubular renin staining were reduced, whereas collecting tubule staining, by contrast, was increased. To examine the role of filtration and tubular reabsorption on urinary renin, mice were either infused with either mouse or human recombinant renin and lysine (a blocker of proximal tubular protein reabsorption). Infusion of either form of renin together with lysine markedly increased urinary renin such that it was no longer different between nondiabetic and diabetic mice. Megalin mRNA was reduced in the kidney cortex of streptozotocin-treated mice (0.70±0.09 versus 1.01±0.04 in controls, P=0.01) consistent with impaired tubular reabsorption. In Ren1d-Cre;mT/mG with streptozotocin-induced diabetes mellitus, the distribution of renin lineage cells within the kidney was similar to nondiabetic renin-reporter mice. No evidence for migration of cells of renin linage to the collecting duct in diabetic mice could be found. Renin mRNA in microdissected collecting ducts from streptozotocin-treated mice, moreover, was not significantly different than in controls, whereas in kidney cortex, largely reflecting juxtaglomerular apparatus renin, it was significantly reduced. In conclusion, in urine from patients with type 1 diabetes mellitus and DKD and from mice with streptozotocin-induced diabetes mellitus, renin is elevated. This cannot be attributed to production from cells of the renin lineage migrating to the collecting duct in a chronic hyperglycemic environment. Rather, the elevated levels of urinary renin found in DKD are best attributed to altered glomerular filteration and impaired proximal tubular reabsorption.


Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/urine , Kidney Tubules, Collecting/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Renin/urine , Animals , Biopsy, Needle , Cohort Studies , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Female , Glomerular Filtration Rate , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Random Allocation , Reference Values , Renin/blood , Sensitivity and Specificity , Urinalysis
10.
Science ; 364(6436)2019 04 12.
Article En | MEDLINE | ID: mdl-30975860

To understand the health impact of long-duration spaceflight, one identical twin astronaut was monitored before, during, and after a 1-year mission onboard the International Space Station; his twin served as a genetically matched ground control. Longitudinal assessments identified spaceflight-specific changes, including decreased body mass, telomere elongation, genome instability, carotid artery distension and increased intima-media thickness, altered ocular structure, transcriptional and metabolic changes, DNA methylation changes in immune and oxidative stress-related pathways, gastrointestinal microbiota alterations, and some cognitive decline postflight. Although average telomere length, global gene expression, and microbiome changes returned to near preflight levels within 6 months after return to Earth, increased numbers of short telomeres were observed and expression of some genes was still disrupted. These multiomic, molecular, physiological, and behavioral datasets provide a valuable roadmap of the putative health risks for future human spaceflight.


Adaptation, Physiological , Astronauts , Space Flight , Adaptive Immunity , Body Weight , Carotid Arteries/diagnostic imaging , Carotid Intima-Media Thickness , DNA Damage , DNA Methylation , Gastrointestinal Microbiome , Genomic Instability , Humans , Male , Telomere Homeostasis , Time Factors , United States , United States National Aeronautics and Space Administration
11.
J Diabetes Complications ; 32(12): 1160-1168, 2018 Dec.
Article En | MEDLINE | ID: mdl-30316542

AIMS: To determine among adolescents and young adults with youth-onset type 1 diabetes and type 2 diabetes the rates and risk factors for albuminuria regression and progression. METHODS: Data from SEARCH, a longitudinal observational study of youth-onset type 1 diabetes (N = 1316) and type 2 diabetes (N = 143) were analyzed. Urine albumin:creatinine ratio (UACR) was measured from random urine specimens at baseline and follow-up visits (mean 7 years later). Albuminuria regression was defined as halving of baseline UACR when baseline UACR was ≥30 µg/mg; progression was defined as doubling of baseline UACR when follow-up UACR was ≥30 µg/mg, respectively. Multivariable regression assessed risk factors associated with low-risk albuminuria category (combined persistently-low albuminuria and regression) versus moderate-risk albuminuria category (combined persistently-high albuminuria and progression). RESULTS: Albuminuria progression was more common in type 2 diabetes versus type 1 diabetes (15.4% versus 6.0%, p<0.001). Moderate-risk albuminuria was associated with increasing HbA1c (adjusted OR (aOR) = 1.3, 95% CI 1.1-1.6) and lack of private health insurance (aOR = 2.7, 95%CI 1.1-6.5) in type 1 diabetes; and African American race (OR = 4.6, 95% CI 1.2-14.2), lower estimated insulin sensitivity score (aOR = 2.1, 95% CI 1.4-3.3), baseline UACR (aOR = 3.2, 95% CI 1.7-5.8), and follow-up estimated glomerular filtration rate (eGFR) (10-unit increase aOR = 1.3, 95% CI 1.0, 1.5) in type 2 diabetes. CONCLUSIONS: In the first decade of diabetes duration, kidney complications in type 2 diabetes are significantly more aggressive than in type 1 diabetes and may be associated with less modifiable risk factors including race, insulin sensitivity, and eGFR. Early interventions may help reduce long-term kidney complications.


Albuminuria/epidemiology , Albuminuria/pathology , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Adolescent , Adult , Age of Onset , Albuminuria/complications , Albuminuria/diagnosis , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/pathology , Disease Progression , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Prevalence , Registries , Risk Factors , United States/epidemiology , Young Adult
12.
Diabetes Care ; 41(11): 2361-2369, 2018 11.
Article En | MEDLINE | ID: mdl-30150236

OBJECTIVE: We examined the association of urine complement proteins with progression to end-stage renal disease (ESRD) or death in people with type 2 diabetes and proteinuric diabetic kidney disease (DKD). RESEARCH DESIGN AND METHODS: Using targeted mass spectrometry, we quantified urinary abundance of 12 complement proteins in a predominantly Mexican American cohort with type 2 diabetes and proteinuric DKD (n = 141). The association of urine complement proteins with progression to ESRD or death was evaluated using time-to-event analyses. RESULTS: At baseline, median estimated glomerular filtration rate (eGFR) was 54 mL/min/1.73 m2 and urine protein-to-creatinine ratio 2.6 g/g. Sixty-seven participants developed ESRD or died, of whom 39 progressed to ESRD over a median of 3.1 years and 40 died over a median 3.6 years. Higher urine CD59, an inhibitor of terminal complement complex formation, was associated with a lower risk of ESRD (hazard ratio [HR] [95% CI per doubling] 0.50 [0.29-0.87]) and death (HR [95% CI] 0.56 [0.34-0.93]), after adjustment for demographic and clinical covariates, including baseline eGFR and proteinuria. Higher urine complement components 4 and 8 were associated with lower risk of death (HR [95% CI] 0.57 [0.41-0.79] and 0.66 [0.44-0.97], respectively); higher urine factor H-related protein 2, a positive regulator of the alternative complement pathway, was associated with greater risk of death (HR [95% CI] 1.61 [1.05-2.48]) in fully adjusted models. CONCLUSIONS: In a largely Mexican American cohort with type 2 diabetes and proteinuric DKD, urine abundance of several complement and complement regulatory proteins was strongly associated with progression to ESRD and death.


Complement System Proteins/urine , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/urine , Diabetic Nephropathies/diagnosis , Kidney Failure, Chronic/diagnosis , Aged , Cross-Sectional Studies , Diabetes Mellitus, Type 2/ethnology , Diabetic Nephropathies/ethnology , Diabetic Nephropathies/mortality , Diabetic Nephropathies/urine , Disease Progression , Female , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/ethnology , Kidney Failure, Chronic/mortality , Kidney Failure, Chronic/urine , Male , Mass Spectrometry , Mexican Americans , Middle Aged , Prognosis , Proteinuria/diagnosis , Proteinuria/ethnology , Proteinuria/mortality , Proteinuria/pathology , Risk Factors , Survival Analysis
13.
EBioMedicine ; 26: 68-77, 2017 Dec.
Article En | MEDLINE | ID: mdl-29128444

Chronic kidney disease (CKD) is a public health problem with very high prevalence and mortality. Yet, there is a paucity of effective treatment options, partly due to insufficient knowledge of underlying pathophysiology. We combined metabolomics (GCMS) with kidney gene expression studies to identify metabolic pathways that are altered in adults with non-diabetic stage 3-4 CKD versus healthy adults. Urinary excretion rate of 27 metabolites and plasma concentration of 33 metabolites differed significantly in CKD patients versus controls (estimate range-68% to +113%). Pathway analysis revealed that the citric acid cycle was the most significantly affected, with urinary excretion of citrate, cis-aconitate, isocitrate, 2-oxoglutarate and succinate reduced by 40-68%. Reduction of the citric acid cycle metabolites in urine was replicated in an independent cohort. Expression of genes regulating aconitate, isocitrate, 2-oxoglutarate and succinate were significantly reduced in kidney biopsies. We observed increased urine citrate excretion (+74%, p=0.00009) and plasma 2-oxoglutarate concentrations (+12%, p=0.002) in CKD patients during treatment with a vitamin-D receptor agonist in a randomized trial. In conclusion, urinary excretion of citric acid cycle metabolites and renal expression of genes regulating these metabolites were reduced in non-diabetic CKD. This supports the emerging view of CKD as a state of mitochondrial dysfunction.


Metabolomics , Mitochondria/metabolism , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/genetics , Aconitic Acid/metabolism , Aged , Biopsy , Citric Acid Cycle/genetics , Female , Gene Expression Regulation/genetics , Humans , Isocitrates/metabolism , Ketoglutaric Acids/metabolism , Kidney/metabolism , Kidney/pathology , Male , Metabolic Networks and Pathways/genetics , Middle Aged , Mitochondria/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/urine , Succinic Acid/metabolism
14.
Clin Chem ; 63(9): 1515-1526, 2017 Sep.
Article En | MEDLINE | ID: mdl-28667184

BACKGROUND: The prevalence of chronic kidney disease (CKD) is increasing, leading to significant morbidity and mortality. Kidney biopsy remains the gold standard for diagnosing the underlying etiology of CKD, but the procedure carries complication risks. The aim of this study was to identify novel noninvasive biomarkers correlating with kidney function and histopathology in biopsy-proven CKD patients. METHODS: We profiled 2402 urinary microRNAs (miRNAs) to identify and confirm differentially expressed miRNAs associated with kidney function and histopathology in patients with diabetic nephropathy (n = 58) or lupus nephritis (n = 89), important etiologies of CKD, compared with healthy controls (n = 93 and 119, respectively). Top performing miRNAs were then measured in 2 independent multi-institutional cohorts of patients with diabetes mellitus with (n = 74) or without nephropathy (n = 71) and systemic lupus erythematosus with (n = 86) or without (n = 37) nephritis. RESULTS: In patients with diabetic nephropathy, miR-2861, miR-1915-3p, and miR-4532 were down-regulated (>10-fold, P < 0.0001) and were associated with estimated glomerular filtration rate (P < 0.01) and interstitial fibrosis/tubular atrophy (P < 0.05). The c-statistics for miR-2861, miR-1915-3p, and miR-4532 were 0.91, 0.86, and 0.85, respectively. In lupus nephritis patients, miR-3201 and miR-1273e were down-regulated (>3-fold, P < 0.0001) and associated with endocapillary glomerular inflammation (P < 0.01), with c-statistics of 0.97 and 0.91, respectively. CONCLUSIONS: We have identified novel miRNAs that correlate with histopathological lesions and functional markers of kidney damage to facilitate sensitive, specific, and noninvasive detection of diabetic nephropathy and lupus nephritis.


Biomarkers/analysis , Diabetic Nephropathies/diagnosis , Lupus Nephritis/diagnosis , MicroRNAs/genetics , Adult , Biomarkers/urine , DNA Replication Timing , Diabetic Nephropathies/genetics , Diabetic Nephropathies/physiopathology , Female , Humans , Lupus Nephritis/genetics , Lupus Nephritis/physiopathology , Male , MicroRNAs/urine , Reproducibility of Results , Transcriptome
15.
Am J Physiol Renal Physiol ; 313(2): F487-F494, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28468961

The pathways implicated in diabetic kidney disease (DKD) are largely derived from animal models. To examine if alterations in renin-angiotensin system (RAS) in humans are concordant with those in rodent models, we measured concentration of angiotensinogen (AOG), cathepsin D (CTSD), angiotensin-converting enzyme (ACE), and ACE2 and enzymatic activities of ACE, ACE2, and aminopeptidase-A in FVB mice 13-20 wk after treatment with streptozotocin (n = 9) or vehicle (n = 15) and people with long-standing type 1 diabetes, with (n = 37) or without (n = 81) DKD. In streptozotocin-treated mice, urine AOG and CTSD were 10.4- and 3.0-fold higher than in controls, respectively (P < 0.001). Enzymatic activities of ACE, ACE2, and APA were 6.2-, 3.2-, and 18.8-fold higher, respectively, in diabetic animals (P < 0.001). Angiotensin II was 2.4-fold higher in diabetic animals (P = 0.017). Compared with people without DKD, those with DKD had higher urine AOG (170 vs. 15 µg/g) and CTSD (147 vs. 31 µg/g). In people with DKD, urine ACE concentration was 1.8-fold higher (1.4 vs. 0.8 µg/g in those without DKD), while its enzymatic activity was 0.6-fold lower (1.0 vs. 1.6 × 109 RFU/g in those without DKD). Lower ACE activity, but not ACE protein concentration, was associated with ACE inhibitor (ACEI) treatment. After adjustment for clinical covariates, AOG, CTSD, ACE concentration, and ACE activity remained associated with DKD. In conclusion, in mice with streptozotocin-induced diabetes and in humans with DKD, urine concentrations and enzymatic activities of several RAS components are concordantly increased, consistent with enhanced RAS activity and greater angiotensin II formation. ACEI use was associated with a specific reduction in urine ACE activity, not ACE protein concentration, suggesting that it may be a marker of exposure to this widely-used therapy.


Diabetes Mellitus, Experimental/urine , Diabetes Mellitus, Type 1/urine , Diabetic Nephropathies/urine , Enzymes/urine , Renal Insufficiency, Chronic/urine , Renin-Angiotensin System , Adult , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Biomarkers/urine , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/diagnosis , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Female , Humans , Male , Mice , Middle Aged , Predictive Value of Tests , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/etiology , Renin-Angiotensin System/drug effects , Up-Regulation , Urinalysis
16.
Horm Res Paediatr ; 87(6): 385-395, 2017.
Article En | MEDLINE | ID: mdl-28554178

BACKGROUND/AIMS: In adults, lower vitamin D has been associated with increased albuminuria. This association has not been extensively studied in youth with or without type 1 diabetes. METHODS: We examined the cross-sectional association between vitamin D and albuminuria (urine albumin to creatinine ratio ≥30 mg/g) in 8,789 participants of the National Health and Nutrition Survey 2001-2006 (NHANES), who were 6-19 years old. Further, we examined the association between vitamin D and albuminuria in 938 participants from the SEARCH Nutritional Ancillary Study (SNAS), a longitudinal cohort of youth with type 1 diabetes. RESULTS: Of the NHANES participants, 5.3, 19.5, and 53.7% had vitamin D levels <30, 50 and 80 nmol/L, respectively. Albuminuria was present in 12.8% and was more common in younger children, females, non-Hispanic whites, non-obese children, and children with hypertension. After adjustments, there was no association between vitamin D and albuminuria. Among the SNAS participants with type 1 diabetes, we also found no association between baseline vitamin D and subsequent albuminuria in unadjusted or adjusted analyses. CONCLUSION: We did not find an association between serum vitamin D and albuminuria in either non-diabetic youth or those with type 1 diabetes. Further research is needed to more fully understand this relationship.


Albuminuria , Diabetes Complications , Diabetes Mellitus, Type 1 , Vitamin D/blood , Adolescent , Adult , Age Factors , Albuminuria/blood , Albuminuria/epidemiology , Albuminuria/urine , Child , Diabetes Complications/blood , Diabetes Complications/epidemiology , Diabetes Complications/urine , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/urine , Female , Humans , Male , Sex Factors
17.
JAMA Cardiol ; 2(3): 314-318, 2017 03 01.
Article En | MEDLINE | ID: mdl-28002548

Importance: Cardiovascular disease is the leading cause of morbidity and mortality in patients with chronic kidney disease (CKD). Understanding the relative contributions of cardiovascular disease event types to the excess burden of cardiovascular disease is important for developing effective strategies to improve outcomes. Objective: To determine absolute rates and risk differences of incident heart failure (HF), coronary heart disease (CHD), and stroke in participants with vs without CKD. Design, Setting and Participants: We pooled participants without prevalent cardiovascular disease from 3 community-based cohort studies: the Jackson Heart Study, Cardiovascular Health Study, and Multi-Ethnic Study of Atherosclerosis. The Jackson Heart Study was conducted between 2000 and 2010, the Cardiovascular Health Study was conducted between 1989 and 2003, and the Multi-Ethnic Study of Atherosclerosis was conducted between 2000 and 2012. Exposures: Chronic kidney disease was defined as estimated glomerular filtration rate less than 60 mL/min/1.73 m2, calculated using the combined creatinine-cystatin C CKD-Epidemiology Collaboration Equation. Main Outcomes and Measures: Poisson regression was used to calculate incidence rates (IRs) and risk differences of adjudicated incident HF, CHD, and stroke, comparing participants with vs without CKD. Results: Among 14 462 participants, the mean (SD) age was 63 (12) years, 59% (n = 8533) were women, and 44% (n = 6363) were African American. Overall, 1461 (10%) had CKD (mean [SD] estimated glomerular filtration rate, 49 [10] mL/min/1.73 m2). Unadjusted IRs for participants with and without CKD, respectively, were 22.0 (95% CI, 19.3-24.8) and 6.2 (95% CI, 5.8-6.7) per 1000 person-years for HF; 24.5 (95% CI, 21.6-27.5) and 8.4 (95% CI, 7.9-9.0) per 1000 person-years for CHD; and 13.4 (95% CI, 11.3-15.5) and 4.8 (95% CI, 4.4-5.3) for stroke. Adjusting for demographics, cohort, hypertension, diabetes, hyperlipidemia, and tobacco use, risk differences comparing participants with vs without CKD (per 1000 person-years) were 2.3 (95% CI, 1.2-3.3) for HF, 2.3 (95% CI, 1.2-3.4) for CHD, and 0.8 (95% CI, 0.09-1.5) for stroke. Among African American and Hispanic participants, adjusted risk differences comparing participants with vs without CKD for HF were 3.5 (95% CI, 1.5-5.5) and 7.8 (95% CI, 2.2-13.3) per 1000 person-years, respectively. Conclusions and Relevance: Among 3 diverse community-based cohorts, CKD was associated with an increased risk of HF that was similar in magnitude to CHD and greater than stroke. The excess risk of HF associated with CKD was particularly large among African American and Hispanic individuals. Efforts to improve health outcomes for patients with CKD should prioritize HF in addition to CHD prevention.


Black or African American , Coronary Disease/ethnology , Heart Failure/ethnology , Renal Insufficiency, Chronic/complications , Risk Assessment , Stroke/ethnology , Coronary Disease/etiology , Female , Follow-Up Studies , Heart Failure/etiology , Humans , Male , Middle Aged , Morbidity/trends , Prognosis , Renal Insufficiency, Chronic/ethnology , Retrospective Studies , Risk Factors , Stroke/etiology , Survival Rate/trends , Time Factors , United States/epidemiology
18.
Am J Physiol Renal Physiol ; 312(4): F716-F731, 2017 04 01.
Article En | MEDLINE | ID: mdl-27558558

Increasing incidences of obesity and diabetes have made diabetic kidney disease (DKD) the leading cause of chronic kidney disease and end-stage renal disease worldwide. Despite current pharmacological treatments, including strategies for optimizing glycemic control and inhibitors of the renin-angiotensin system, DKD still makes up almost one-half of all cases of end-stage renal disease in the United States. Compelling and mounting evidence has clearly demonstrated that immunity and inflammation play a paramount role in the pathogenesis of DKD. This article reviews the involvement of the immune system in DKD and identifies important roles of key immune and inflammatory mediators. One of the most recently identified biomarkers is serum amyloid A, which appears to be relatively specific for DKD. Novel and evolving treatment approaches target protein kinases, transcription factors, chemokines, adhesion molecules, growth factors, advanced glycation end-products, and other inflammatory molecules. This is the beginning of a new era in the understanding and treatment of DKD, and we may have finally reached a tipping point in our fight against the growing burden of DKD.


Anti-Inflammatory Agents/therapeutic use , Diabetic Nephropathies/drug therapy , Drug Discovery/methods , Inflammation Mediators/antagonists & inhibitors , Kidney/drug effects , Nephritis/drug therapy , Receptors, Immunologic/drug effects , Animals , Biomarkers/metabolism , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Molecular Targeted Therapy , Nephritis/immunology , Nephritis/metabolism , Nephritis/pathology , Predictive Value of Tests , Receptors, Immunologic/metabolism , Signal Transduction/drug effects
19.
Drug Metab Dispos ; 44(10): 1692-6, 2016 10.
Article En | MEDLINE | ID: mdl-27481856

The existing biobanks of remnant tissue from clinically indicated kidney biopsies are attractive potential reservoirs for quantification of clinically relevant human tissue proteins by quantitative proteomics. However, a significant caveat of this strategy is that the tissues are often preserved in optimal cutting temperature (OCT) medium. Although OCT is an effective method of preserving the morphologic and immunohistological characteristics of tissues for later study, it significantly impacts efforts to quantify protein expression by liquid chromatography-tandem mass spectrometry methods. We report here a simple, reproducible, and cost-effective procedure to extract proteins from OCT-embedded tissue samples. Briefly, the excess frozen OCT medium was scraped before thawing from the tissue specimens stored at -80°C for ∼3 months. The tissue samples were homogenized and diethyl ether/methanol extraction was performed to remove the remaining OCT medium. The recovered protein was denatured, reduced, and alkylated. The second step of protein extraction and desalting was performed by chloroform/methanol/water extraction of denatured proteins. The resultant protein pellet was trypsin-digested and the marker proteins of various kidney cellular compartments were quantified by targeted selective reaction monitoring proteomics. Upon comparison of peptide signals from OCT-embedded tissue and flash-frozen tissue from the same donors, both individual protein quantities, and their interindividual variabilities, were similar. Therefore, the approach reported here can be applied to clinical reservoirs of OCT-preserved kidney tissue to be used for quantitative proteomics studies of clinically relevant proteins expressed in different parts of the kidney (including drug transporters and metabolizing enzymes).


Kidney/metabolism , Proteins/isolation & purification , Aged , Chromatography, Liquid , Female , Humans , Male , Middle Aged , Tandem Mass Spectrometry , Temperature
20.
J Diabetes Complications ; 30(8): 1467-1472, 2016.
Article En | MEDLINE | ID: mdl-27522272

AIMS: To determine if serum levels of serum amyloid A (SAA) predict death and end-stage renal disease in a cohort of people with diabetic kidney disease. METHODS: In a longitudinal cohort study of 135 participants with type 2 diabetes and diabetic kidney disease, serum samples were assayed for SAA. Censored time-to-event analyses in Cox-proportional hazard models were utilized to assess SAA as a predictor of the primary outcome of death and end-stage renal disease. RESULTS: Participants were 73% Mexican-American (99/135) and 55% men (75/135), with a mean±SD age of 57±7.5years. At baseline, participants had hemoglobin A1c of 8.6±2.3%, systolic blood pressure of 153±27mm Hg, body mass index of 31±9kg/m2, median urine-albumin-to-creatinine ratio of 1861mg/g (interquartile range 720-3912mg/g), and estimated glomerular filtration rate of 55.7±22.3ml/min/1.73m2. Over a median duration of follow-up of 3.5years, 44% (60/135) of participants experienced a primary outcome event. The hazards ratio for the primary outcome was 3.03 (95% CI 1.43-6.40, p=0.003) in the highest (>1.0 µg/ml) compared to the lowest (<0.55 µg/ml) SAA tertile in a model adjusted for urine-albumin-to-creatinine ratio, estimated glomerular filtration rate, age, sex, and race/ethnicity. Addition of SAA as a covariate improved the model C-statistic (Δ c=0.017). CONCLUSIONS: In a longitudinal cohort study of participants with type 2 diabetes and DKD, higher levels of serum SAA predicted higher risk of death and ESRD. SAA is a promising targetable biomarker for DKD.


Diabetes Mellitus, Type 2/mortality , Diabetic Nephropathies/mortality , Kidney Failure, Chronic/mortality , Serum Amyloid A Protein/analysis , Aged , California , Diabetes Mellitus, Type 2/blood , Diabetic Nephropathies/blood , Disease Progression , Female , Glomerular Filtration Rate , Humans , Kidney Failure, Chronic/blood , Longitudinal Studies , Male , Mexican Americans , Middle Aged , Risk Factors
...