1
|
Gregg EW, Holman N, Sophiea M, Misra S, Pearson-Stuttard J, Valabhji J, Khunti K. Multiple long-term conditions as the next transition in the global diabetes epidemic. COMMUNICATIONS MEDICINE 2025; 5:42. [PMID: 39953177 PMCID: PMC11828996 DOI: 10.1038/s43856-025-00742-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/20/2023] [Accepted: 01/15/2025] [Indexed: 02/17/2025] Open
Abstract
Several transitions, or new patterns and dynamics in the contributors and health outcomes, have altered the character and burden of the multi-decade, worldwide growth in prevalence of type 2 diabetes (T2DM). These changes have led to different needs for prevention and care. These dynamics have been driven by diverse demographic, socio-economic, behavioural, and health system response factors. In this Perspective, we describe these transitions and how their attributes have set the stage for multimorbidity, or multiple long-term conditions (MLTCs), to be the next major challenge in the diabetes epidemic. We also describe how the timing and character of these stages differ in high-, middle-, and low-income countries. These challenges call for innovation and a stronger focus on MLTCs across the spectrum of cause, effectiveness, and implementation studies to guide prevention and treatment priorities.
Collapse
Affiliation(s)
- Edward W Gregg
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
- School of Public Health, Imperial College London, London, UK.
| | - Naomi Holman
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Public Health, Imperial College London, London, UK
- NHS England, Wellington House, London, UK
| | - Marisa Sophiea
- School of Public Health, Imperial College London, London, UK
| | - Shivani Misra
- Department of Diabetes and Endocrinology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
- Division of Metabolism, Digestion & Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | | | - Jonathan Valabhji
- NHS England, Wellington House, London, UK
- Division of Metabolism, Digestion & Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Chelsea & Westminster Hospital NHS Foundation Trust, London, UK
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
2
|
Kobayashi H, Looker HC, Ihara K, Md Dom ZI, Satake E, Tye SC, Duffin KL, Doria A, Nelson RG, Krolewski AS. Classification of Predictors of Rapid Development of Kidney Failure and Short-Term Changes in Concentration of Circulating Proteins. Clin J Am Soc Nephrol 2024; 20:01277230-990000000-00493. [PMID: 39480511 PMCID: PMC11835163 DOI: 10.2215/cjn.0000000603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/22/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
OBJECTIVE Limited knowledge exists regarding short-term changes/increases in concentrations of circulating proteins (referred here as deltas) and rapid development of kidney failure (rapid KF) in diabetes mellitus. RESEARCH DESIGN AND METHODS Concentrations of 452 circulating proteins were measured by OLINK proteomics platform at baseline and after a median interval of 3-4 years in 106 individuals with type 1 and 77 with type 2 diabetes in two case-control studies. During 10-year follow-up, 31 and 26 individuals, respectively, developed rapid KF. RESULTS Deltas for 40 proteins predicted rapid KF in both studies. All were better predictors than delta urine albumin-creatinine ratio, and half were better than delta glomerular filtration rate. Comparing the delta proteins with 46 circulating proteins of which elevated baseline concentrations were predictors of rapid KF risk in our previous study, 61 unique proteins were identified. Among these proteins, 21 were good predictors of rapid KF only when measured at baseline (predictors of initiation), 15 were good predictors when measured as deltas (predictors of progression) and 25 were good predictors when both baseline and delta concentrations were used (predictors of initiation and progression). An index score, developed for the latter 25 proteins, provided superior prediction of rapid KF. A subset of these latter proteins was associated with apoptotic processes/tumor necrosis factor (TNF) receptor signaling pathways. CONCLUSION Development of rapid KF in diabetes was preceded by elevated concentrations of multiple circulating proteins both at baseline and during short follow-up. Comparing baseline and short-term changes in concentrations of circulating proteins classified predictors of rapid KF risk into those associated with initiation, progression, or both. Predictors of both initiation & progression flagged apoptosis processes and TNF receptor signaling pathways. Multi-protein prognostic algorithms using proteins associated with both initiation and progression improved prediction of rapid KF risk beyond clinical variables.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Division of Nephrology, Hypertension, and Endocrinology, Nihon University School of Medicine, Tokyo, Japan
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
| | - Katsuhito Ihara
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Zaipul I. Md Dom
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Eiichiro Satake
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sok Cin Tye
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kevin L. Duffin
- Diabetes and Complication Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Robert G. Nelson
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
| | - Andrzej S. Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
3
|
Arreola EV, Coonrod DV, Roy Choudhury S, Knowler WC, Hoskin M, Wasak D, Williams R, Hanson RL, Pack E, Caballero R, Gonzalez A, Sinha M. Study protocol for Early Tracking of Childhood Health determinants (ETCHED): A longitudinal observational life course study. BMC Public Health 2024; 24:2661. [PMID: 39343891 PMCID: PMC11439307 DOI: 10.1186/s12889-024-20176-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/18/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The prevalence of childhood obesity and diabetes continues to rise in the United States (US), especially among minority populations. The objective of the Early Tracking of Childhood Health Determinants (ETCHED) study is to investigate the role of adverse fetal and early-life risk exposures that contribute to the development of childhood obesity and metabolic risk. METHODS ETCHED is a longitudinal observational study of American Indian/Alaska Native (AI/AN) and Hispanic pregnant woman and their offspring. Pregnant mothers ≥ 18 years old are enrolled at a large public hospital system in the southwestern US. Enrolled mothers are followed through pregnancy, delivery, and the maternal/offspring dyad will be followed until the child's 18th birthday. At each maternal visit, questionnaires assessing medical history, diet, physical activity, sleep, perceived stress, and socioeconomic and sociocultural information are obtained. Standard laboratory tests during maternal visits include glycemic measures, lipids, and renal function. Additional bio samples obtained include venous blood samples and cord blood for obesity/metabolic biomarkers and genetic/epigenetic testing, urinalysis, placental tissue for examining functional pathways, breast milk for metabolomics, and stool for metabolites and microbiome analysis. The offspring will have 6 infant/toddler visits at 6-12 weeks, 4 months, 6 months, 18 months, 2 and 3 years respectively. Thereafter, they will undergo comprehensive research visits (major visits) at 4-5 years, 6-9 years, 10-13 years, and 14-17 years. The major visits in children include detailed medical history, anthropometry, developmental assessment, socioeconomic and environmental assessments (food insecurity, family structure, and childcare), feeding and activity, biochemical tests, genetics/epigenetic testing, and ultrasound elastography. Electronic health records will be reviewed for additional clinical information. The primary analysis will constitute estimation of correlation coefficients between continuous variables. The planned study duration in this ongoing study is 23-years. DISCUSSION This is a life course study that that will examine biological and environmental risk factors for obesity and cardiometabolic risk from the intrauterine period to early childhood and adolescence in a population with high-risk of obesity and type 2 diabetes in the United States. The ETCHED study would also provide a unique opportunity to combine multi-omics and clinical data to create novel integrative models to predict the cardiometabolic risk associated with childhood obesity and possibly identify etiopathogenetic mechanisms and future targets of intervention. TRIAL REGISTRATION NUMBER ClinicalTrials.gov identifier: NCT03481829. Updated July 19, 2024, https://clinicaltrials.gov/study/NCT03481829?cond=ETCHED&rank=1 .
Collapse
Affiliation(s)
- Elsa Vazquez Arreola
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Dean V Coonrod
- Department of Obstetrics and Gynecology, Valleywise Health Medical Center, Phoenix, AZ, USA
- Department of Obstetrics and Gynecology, Affiliate Faculty Creighton, University School of Medicine Phoenix Regional Campus, Phoenix, AZ, USA
| | - Sourav Roy Choudhury
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - William C Knowler
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Mary Hoskin
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Dorota Wasak
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Rachel Williams
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Robert L Hanson
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Elena Pack
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Rachel Caballero
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Amanda Gonzalez
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA
| | - Madhumita Sinha
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, AZ, USA.
- National Institutes of Health (NIH), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix Epidemiology and Clinical Research Branch (PECRB), 1550 E. Indian School Rd., Phoenix, AZ, 85014, USA.
| |
Collapse
|
4
|
Saulnier PJ, Looker HC, Layton A, Lemley KV, Nelson RG, Bjornstad P. Loss of Glomerular Permselectivity in Type 2 Diabetes Associates With Progression to Kidney Failure. Diabetes 2023; 72:1682-1691. [PMID: 37586079 PMCID: PMC10588283 DOI: 10.2337/db23-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/21/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
We examined whether defects in glomerular size selectivity in type 2 diabetes are associated with progressive kidney disease. Glomerular filtration rate (GFR) and fractional clearances of dextrans of graded sizes were measured in 185 American Indians. The permselectivity model that best fit the dextran sieving data represented the glomerular capillary as being perforated by small restrictive pores and a parallel population of larger nonrestrictive pores characterized by ω0, the fraction of total filtrate volume passing through this shunt. The hazard ratio (HR) for kidney failure was expressed per 1-SD increase of ω0 by Cox regression after adjusting for age, sex, mean arterial pressure, HbA1c, GFR, and the urine albumin-to-creatinine ratio (ACR). Baseline mean ± SD age was 43 ± 10 years, HbA1c 8.9 ± 2.5%, GFR 147 ± 46 mL/min, and median (interquartile range) ACR 41 (11-230) mg/g. During a median follow-up of 17.7 years, 67 participants developed kidney failure. After adjustment, each 1-SD increment in ω0 was associated with a higher risk of kidney failure (HR 1.55 [95% CI 1.17, 2.05]). Enhanced transglomerular passage of test macromolecules was associated with progression to kidney failure, independent of albuminuria and GFR, suggesting that mechanisms associated with impaired glomerular permselectivity are important determinants of progressive kidney disease. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Pierre J. Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
- University of Poitiers, INSERM CIC1402, Poitiers, France
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
| | - Anita Layton
- University of Waterloo, Waterloo, Ontario, Canada
| | - Kevin V. Lemley
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ
| | | |
Collapse
|
5
|
Reynolds EL, Votruba K, Jack CR, Beare R, Reid RI, Preboske GM, Waseta C, Pop‐Busui R, Nelson RG, Callaghan BC, Feldman EL. Association between brain health outcomes and metabolic risk factors in persons with diabetes. Ann Clin Transl Neurol 2023; 10:1891-1898. [PMID: 37518982 PMCID: PMC10578900 DOI: 10.1002/acn3.51859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
We performed a cross-sectional study to determine associations between cognition and MRI-derived brain outcomes, with obesity, diabetes duration, and metabolic risk factors in 51 Pima American Indians with longstanding type 2 diabetes (T2d) (mean [SD] age: 48.4 [11.3] years, T2d duration: 20.1 [9.1] years). Participants had similar cognition (NIH Toolbox Cognition Battery composite: 45.3 [9.8], p = 0.64, n = 51) compared to normative data. T2d duration, but not other metabolic risk factors, associated with decreased cortical thickness (Point Estimate (PE): -0.0061, 95%CI: -0.0113, -0.0009, n = 45), gray matter volume (PE: -830.39, 95%CI: -1503.14, -157.64, n = 45), and increased white matter hyperintensity volume (PE: 0.0389, 95%CI: 0.0049, 0.0729, n = 45).
Collapse
Affiliation(s)
- Evan L. Reynolds
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kristen Votruba
- Department of PsychiatryUniversity of MichiganAnn ArborMichiganUSA
| | | | - Richard Beare
- Peninsula Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | | | | | - Camille Waseta
- Chronic Kidney Disease SectionNational Institute of Diabetes and Digestive and Kidney DiseasesPhoenixArizonaUSA
| | - Rodica Pop‐Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and DiabetesUniversity of MichiganAnn ArborMichiganUSA
| | - Robert G. Nelson
- Chronic Kidney Disease SectionNational Institute of Diabetes and Digestive and Kidney DiseasesPhoenixArizonaUSA
| | | | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
6
|
Chai X, Wang Y, Wang J, Gong Q, Zhang J, Shao R. Innovation in diabetes prevention research: The 36-year legacy of China Da Qing diabetes prevention study. CHINESE SCIENCE BULLETIN 2023; 68:3834-3845. [DOI: 10.1360/tb-2023-0489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2025]
|
7
|
Looker HC, Chang DC, Baier LJ, Hanson RL, Nelson RG. Diagnostic criteria and etiopathogenesis of type 2 diabetes and its complications: Lessons from the Pima Indians. Presse Med 2023; 52:104176. [PMID: 37783422 PMCID: PMC10805453 DOI: 10.1016/j.lpm.2023.104176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/17/2023] [Revised: 03/28/2023] [Accepted: 07/19/2023] [Indexed: 10/04/2023] Open
Abstract
The Phoenix Epidemiology and Clinical Research Branch of the National Institute of Diabetes and Digestive and Kidney Diseases has conducted prospective studies of diabetes and its complications in the Pima Indians living in Arizona, USA for over 50 years. In this review we highlight areas in which these studies provided vital insights into the criteria used to diagnose type 2 diabetes, the pathophysiologic changes that accompany the development of type 2 diabetes, and the course and determinants of diabetes complications-focusing specifically on diabetic kidney disease. We include data from our longitudinal population-based study of diabetes and its complications, studies on the role of insulin resistance and insulin secretion in the pathophysiology of type 2 diabetes, and in-depth studies of diabetic kidney disease that include measures of glomerular function and research kidney biopsies. We also focus on the emerging health threat posed by youth-onset type 2 diabetes, which was first seen in the Pima Indians in the 1960s and is becoming an increasing issue worldwide.
Collapse
Affiliation(s)
- Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Douglas C Chang
- Obesity and Diabetes Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Leslie J Baier
- Diabetes Molecular Genetics Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Robert L Hanson
- Diabetes Genetic Epidemiology Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA.
| |
Collapse
|
8
|
Kobayashi H, Looker HC, Satake E, D’Addio F, Wilson JM, Saulnier PJ, Md Dom ZI, O’Neil K, Ihara K, Krolewski B, Badger HS, Petrazzuolo A, Corradi D, Galecki A, Wilson P, Najafian B, Mauer M, Niewczas MA, Doria A, Humphreys B, Duffin KL, Fiorina P, Nelson RG, Krolewski AS. Neuroblastoma suppressor of tumorigenicity 1 is a circulating protein associated with progression to end-stage kidney disease in diabetes. Sci Transl Med 2022; 14:eabj2109. [PMID: 35947673 PMCID: PMC9531292 DOI: 10.1126/scitranslmed.abj2109] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/03/2022]
Abstract
Circulating proteins associated with transforming growth factor-β (TGF-β) signaling are implicated in the development of diabetic kidney disease (DKD). It remains to be comprehensively examined which of these proteins are involved in the pathogenesis of DKD and its progression to end-stage kidney disease (ESKD) in humans. Using the SOMAscan proteomic platform, we measured concentrations of 25 TGF-β signaling family proteins in four different cohorts composed in total of 754 Caucasian or Pima Indian individuals with type 1 or type 2 diabetes. Of these 25 circulating proteins, we identified neuroblastoma suppressor of tumorigenicity 1 (NBL1, aliases DAN and DAND1), a small secreted protein known to inhibit members of the bone morphogenic protein family, to be most strongly and independently associated with progression to ESKD during 10-year follow-up in all cohorts. The extent of damage to podocytes and other glomerular structures measured morphometrically in 105 research kidney biopsies correlated strongly with circulating NBL1 concentrations. Also, in vitro exposure to NBL1 induced apoptosis in podocytes. In conclusion, circulating NBL1 may be involved in the disease process underlying progression to ESKD, and its concentration in circulation may identify subjects with diabetes at increased risk of progression to ESKD.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Hypertension, and Endocrinology, Nihon University School of Medicine, Tokyo, Japan
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Eiichiro Satake
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Francesca D’Addio
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
| | - Jonathan M. Wilson
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Pierre Jean. Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
- CHU Poitiers, University of Poitiers, Inserm, Clinical Investigation Center CIC1402, Poitiers, France
| | - Zaipul I. Md Dom
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kristina O’Neil
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Katsuhito Ihara
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Bozena Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hannah S. Badger
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Adriana Petrazzuolo
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
| | - Domenico Corradi
- Department of Medicine and Surgery, Unit of Pathology, University of Parma, Parma, Italy
| | - Andrzej Galecki
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Parker Wilson
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University in Saint Louis School of Medicine, St. Louis, USA
| | - Behzad Najafian
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Michael Mauer
- Department of Pediatrics and Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Monika A. Niewczas
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin Humphreys
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Kevin L. Duffin
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Paolo Fiorina
- Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università di Milano and Endocrinology Division ASST Sacco-FBF, Milan, Italy
- Nephrology Division, Boston Children’s Hospital, Boston, MA, USA
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Andrzej S. Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Kobayashi H, Looker HC, Satake E, Saulnier PJ, Md Dom ZI, O'Neil K, Ihara K, Krolewski B, Galecki AT, Niewczas MA, Wilson JM, Doria A, Duffin KL, Nelson RG, Krolewski AS. Results of untargeted analysis using the SOMAscan proteomics platform indicates novel associations of circulating proteins with risk of progression to kidney failure in diabetes. Kidney Int 2022; 102:370-381. [PMID: 35618095 PMCID: PMC9333266 DOI: 10.1016/j.kint.2022.04.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2021] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
This study applies a large proteomics panel to search for new circulating biomarkers associated with progression to kidney failure in individuals with diabetic kidney disease. Four independent cohorts encompassing 754 individuals with type 1 and type 2 diabetes and early and late diabetic kidney disease were followed to ascertain progression to kidney failure. During ten years of follow-up, 227 of 754 individuals progressed to kidney failure. Using the SOMAscan proteomics platform, we measured baseline concentration of 1129 circulating proteins. In our previous publications, we analyzed 334 of these proteins that were members of specific candidate pathways involved in diabetic kidney disease and found 35 proteins strongly associated with risk of progression to kidney failure. Here, we examined the remaining 795 proteins using an untargeted approach. Of these remaining proteins, 11 were significantly associated with progression to kidney failure. Biological processes previously reported for these proteins were related to neuron development (DLL1, MATN2, NRX1B, KLK8, RTN4R and ROR1) and were implicated in the development of kidney fibrosis (LAYN, DLL1, MAPK11, MATN2, endostatin, and ROR1) in cellular and animal studies. Specific mechanisms that underlie involvement of these proteins in progression of diabetic kidney disease must be further investigated to assess their value as targets for kidney-protective therapies. Using multivariable LASSO regression analysis, five proteins (LAYN, ESAM, DLL1, MAPK11 and endostatin) were found independently associated with risk of progression to kidney failure. Thus, our study identified proteins that may be considered as new candidate prognostic biomarkers to predict risk of progression to kidney failure in diabetic kidney disease. Furthermore, three of these proteins (DLL1, ESAM, and MAPK11) were selected as candidate biomarkers when all SOMAscan results were evaluated.
Collapse
Affiliation(s)
- Hiroki Kobayashi
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology, Hypertension, and Endocrinology, Nihon University School of Medicine, Tokyo, Japan
| | - Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Eiichiro Satake
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Pierre Jean Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA; Centre Hospitalier Universitaire, Centre d Investigation Clinique Poitiers, France
| | - Zaipul I Md Dom
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kristina O'Neil
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Katsuhito Ihara
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Bozena Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrzej T Galecki
- Cognitive Health Services Research Program, University of Michigan, Ann Arbor, Michigan, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Monika A Niewczas
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan M Wilson
- Diabetes and Complication Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Alessandro Doria
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Kevin L Duffin
- Diabetes and Complication Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA.
| | - Andrzej S Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
10
|
Abstract
Diabetes mellitus (DM) is gradually attacking the health and life of people all over the world. Diabetic kidney disease (DKD) is one of the most common chronic microvascular complications of DM, whose mechanism is complex and still lacks research. Sirtuin family is a class III histone deacetylase with highly conserved NAD+ binding domain and catalytic functional domain, while different N-terminal and C-terminal structures enable them to bind different deacetylated substrates to participate in the cellular NAD+ metabolism. The kidney is an organ rich in NAD+ and database exploration of literature shows that the Sirtuin family has different expression localization in renal, cellular, and subcellular structures. With the progress of modern technology, a variety of animal models and reagents for the Sirtuin family and DKD emerged. Machine learning in the literature shows that the Sirtuin family can regulate pathophysiological injury mainly in the glomerular filtration membrane, renal tubular absorption, and immune inflammation through various mechanisms such as epigenetics, multiple signaling pathways, and mitochondrial function. These mechanisms are the key nodes participating in DKD. Thus, it is of great significance for target therapy to study biological functions of the Sirtuin family and DKD regulation mechanism in-depth.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
- *Correspondence: Huiwen Ren,
| |
Collapse
|
11
|
Bian C, Gao J, Wang Y, Li J, Luan Z, Lu H, Ren H. Association of SIRT6 circulating levels with urinary and glycometabolic markers in pre-diabetes and diabetes. Acta Diabetol 2021; 58:1551-1562. [PMID: 34148121 DOI: 10.1007/s00592-021-01759-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/17/2021] [Accepted: 06/06/2021] [Indexed: 12/30/2022]
Abstract
AIM The study is aimed to detect the expression of serum Sirtuin 6 (SIRT6) with different severities and urinary albumin creatinine ratios (UACR) in type 2 diabetes mellitus (T2DM) patients, thus exploring the association of SIRT6 together with glycolipid metabolism and urinary protein in the cross-sectional study. METHODS T2DM patients (313 cases), pre-diabetic patients (102 cases), and healthy volunteers (100 cases) were selected. T2DM patients were divided into the normal albuminuria (103 cases, UACR < 30 mg/g), micro-albuminuria (106 cases, UACR 30-300 mg/g), and large amount of albuminuria group (104 cases, UACR > 300 mg/g) based on different UACR levels. The medical history was asked, biochemical indicators were detected, hematuria samples were taken, serum SIRT6 levels were detected, and detailed statistical analysis was conducted. RESULTS FPG, 2 h-PG, HOMA-IR, HbA1c, and LDL-C increased, while ISI and HDL-C decreased with the aggravation of diabetic status (P < 0.05). HbA1c, UACR, TNFα, HIF1α, and SIRT6 increased with UACR in T2DM patients (P < 0.05). Correlation analysis demonstrated that SIRT6 was significantly positively correlated with glycolipid metabolism in the whole samples, and correlated with UACR, TNFα, and HIF1α in T2DM patients (P < 0.05). Ridge regression analysis showed that SIRT6 was a risk factor for both glycolipid metabolism and urinary protein (P < 0.05). CONCLUSION SIRT6 increases with biomarkers in glycolipid metabolism and urinary protein in different severities of diabetes and UACR, which is expected to be a potential biomarker for early prediction and diagnosis related to glycolipid metabolism disorders and related nephropathy. Trial number: ChiCTR2000039808.
Collapse
Affiliation(s)
- Che Bian
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Gao
- Department of Gerontology, Xin Hua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuxia Wang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Li
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Lvshun South Road west 9, Dalian, 116044, Liaoning, China
| | - Heyuan Lu
- Advanced Institute for Medical Sciences, Dalian Medical University, Lvshun South Road west 9, Dalian, 116044, Liaoning, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Lvshun South Road west 9, Dalian, 116044, Liaoning, China.
| |
Collapse
|
12
|
Saulnier PJ, Looker HC, Mauer M, Najafian B, Gand E, Ragot S, Nelson RG, Bjornstad P. Intraglomerular Dysfunction Predicts Kidney Failure in Type 2 Diabetes. Diabetes 2021; 70:2344-2352. [PMID: 34257069 PMCID: PMC8576505 DOI: 10.2337/db21-0154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/22/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022]
Abstract
No longitudinal data link intraglomerular hemodynamic dysfunction with end-stage kidney disease (ESKD) in people with type 2 diabetes (T2D). Afferent (RA) and efferent (RE) arteriolar resistance and intraglomerular pressure (PGLO) are not directly measurable in humans but are estimable from glomerular filtration rate (GFR), renal plasma flow (RPF), blood pressure, hematocrit, and plasma oncotic pressure. We examined the association of the RA-to-RE ratio and PGLO with ESKD incidence in 237 Pima Indian individuals with T2D who underwent serial measures of GFR (iothalamate) and RPF (p-aminohippurate). Their association with kidney structural lesions was also examined in a subset of 111 participants. Of the 237 participants (mean age 42 years, diabetes duration 11 years, and GFR 153 mL/min and median urine albumin-to-creatinine ratio 36 mg/g), 69 progressed to ESKD during a median follow-up of 17.5 years. In latent class analysis, distinct trajectories characterized by increasing RA-to-RE ratio (HR 4.60, 95% CI 2.55-8.31) or elevated PGLO followed by a rapid decline (HR 2.96, 95% CI 1.45-6.02) strongly predicted incident ESKD. PGLO (R 2 = 21%, P < 0.0001) and RA-to-RE ratio (R 2 = 15%, P < 0.0001) also correlated with mesangial fractional volume, a structural predictor of DKD progression. In conclusion, intraglomerular hemodynamic parameters associated strongly with incident ESKD and correlated with structural lesions of DKD.
Collapse
Affiliation(s)
- Pierre J Saulnier
- NIDDK, Phoenix, AZ
- INSERM CIC1402, University of Poitiers, Poitiers, France
| | | | | | | | - Elise Gand
- INSERM CIC1402, University of Poitiers, Poitiers, France
| | | | | | | |
Collapse
|
13
|
Renal outcomes in Asian patients with type 2 diabetes mellitus treated with SGLT2 inhibitors: a systematic review and meta-analysis of randomized controlled trials. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-021-00999-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 10/20/2022] Open
Abstract
Abstract
Aim
This study investigated the effects of sodium-glucose cotransporter 2 (SGLT2) inhibitors on renal outcomes in Asian patients with type 2 diabetes mellitus (T2DM).
Materials and methods
We searched Medline, EMBASE, and the Cochrane Library to identify randomized controlled trials published up to April 2020 that compared SGLT2 inhibitors with placebo or active comparator and reported any renal outcomes in Asian patients with T2DM. Random effects models and inverse variance weighting were used to calculate relative risks with 95% confidence intervals (CIs).
Results
We included 14 studies, totaling 3792 patients, in the analysis. In the short term, SGLT2 inhibitors significantly slowed estimated glomerular filtration rate (eGFR) decline (MD: 0.80; 95% CI: 0.66 to 0.94; p < 0.00001) and reduced Scr levels (SMD: − 0.17; 95% CI: − 0.23 to − 0.10; p < 0.00001) as compared with the control groups. The SGLT2 inhibitor group also had an advantage over the control group in lowering uric acid (UA) (SMD: − 1.2; 95% CI: − 1.30 to − 1.11; p < 0.00001). There was no significant difference in urinary albumin creatinine ratio (UACR) reduction between the SGLT2 inhibitor and control groups (MD: − 8.87; 95% CI: − 19.80 to 2.06; p = 0.11). However, dapagliflozin does appear to reduce albuminuria (p = 0.005). Lastly, SGLT2 inhibitors increased the incidence of adverse events (AEs) related to renal function (OR: 1.90; 95% CI: 1.24 to 2.91; p = 0.003), but did not increase the incidence of renal impairment (OR: 0.85; 95% CI: 0.40 to 1.81; p = 0.68).
Conclusion
The use of SGLT2 inhibitors in Asian patients with T2DM can help delay the decline of eGFR and reduce Scr and UA. Although SGLT2 inhibitors have no overall advantage in reducing albuminuria, dapagliflozin does appear to reduce albuminuria, and while they may increase the occurrence of AEs related to renal function, they do not increase the incidence of renal impairment.
Collapse
|
14
|
Satake E, Saulnier PJ, Kobayashi H, Gupta MK, Looker HC, Wilson JM, Md Dom ZI, Ihara K, O’Neil K, Krolewski B, Pipino C, Pavkov ME, Nair V, Bitzer M, Niewczas MA, Kretzler M, Mauer M, Doria A, Najafian B, Kulkarni RN, Duffin KL, Pezzolesi MG, Kahn CR, Nelson RG, Krolewski AS. Comprehensive Search for Novel Circulating miRNAs and Axon Guidance Pathway Proteins Associated with Risk of ESKD in Diabetes. J Am Soc Nephrol 2021; 32:2331-2351. [PMID: 34140396 PMCID: PMC8729832 DOI: 10.1681/asn.2021010105] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/26/2021] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Mechanisms underlying the pro gression of diabetic kidney disease to ESKD are not fully understood. METHODS We performed global microRNA (miRNA) analysis on plasma from two cohorts consisting of 375 individuals with type 1 and type 2 diabetes with late diabetic kidney disease, and targeted proteomics analysis on plasma from four cohorts consisting of 746 individuals with late and early diabetic kidney disease. We examined structural lesions in kidney biopsy specimens from the 105 individuals with early diabetic kidney disease. Human umbilical vein endothelial cells were used to assess the effects of miRNA mimics or inhibitors on regulation of candidate proteins. RESULTS In the late diabetic kidney disease cohorts, we identified 17 circulating miRNAs, represented by four exemplars (miR-1287-5p, miR-197-5p, miR-339-5p, and miR-328-3p), that were strongly associated with 10-year risk of ESKD. These miRNAs targeted proteins in the axon guidance pathway. Circulating levels of six of these proteins-most notably, EFNA4 and EPHA2-were strongly associated with 10-year risk of ESKD in all cohorts. Furthermore, circulating levels of these proteins correlated with severity of structural lesions in kidney biopsy specimens. In contrast, expression levels of genes encoding these proteins had no apparent effects on the lesions. In in vitro experiments, mimics of miR-1287-5p and miR-197-5p and inhibitors of miR-339-5p and miR-328-3p upregulated concentrations of EPHA2 in either cell lysate, supernatant, or both. CONCLUSIONS This study reveals novel mechanisms involved in progression to ESKD and points to the importance of systemic factors in the development of diabetic kidney disease. Some circulating miRNAs and axon guidance pathway proteins represent potential targets for new therapies to prevent and treat this condition.
Collapse
Affiliation(s)
- Eiichiro Satake
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Pierre-Jean Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
- Poitiers University Hospital, University of Poitiers, Institut National de la Santé et de la Recherche Médicale (INSERM), Clinical Investigation Center CIC1402, Poitiers, France
| | - Hiroki Kobayashi
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Manoj K. Gupta
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
| | - Jonathan M. Wilson
- Diabetes and Complication Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Zaipul I. Md Dom
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Katsuhito Ihara
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kristina O’Neil
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Bozena Krolewski
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Caterina Pipino
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology (CAST), University G. d’Annunzio, Chieti, Italy
| | - Meda E. Pavkov
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Viji Nair
- Nephrology/Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Markus Bitzer
- Nephrology/Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Monika A. Niewczas
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Matthias Kretzler
- Nephrology/Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Michael Mauer
- Department of Pediatrics and Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alessandro Doria
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Behzad Najafian
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Rohit N. Kulkarni
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Kevin L. Duffin
- Diabetes and Complication Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Marcus G. Pezzolesi
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Division of Nephrology and Hypertension, University of Utah, Salt Lake City, Utah
| | - C. Ronald Kahn
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
| | - Andrzej S. Krolewski
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Nelson RG, Knowler WC, Kretzler M, Lemley KV, Looker HC, Mauer M, Mitch WE, Najafian B, Bennett PH. Pima Indian Contributions to Our Understanding of Diabetic Kidney Disease. Diabetes 2021; 70:1603-1616. [PMID: 34285119 PMCID: PMC8385607 DOI: 10.2337/dbi20-0043] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/16/2021] [Accepted: 05/23/2021] [Indexed: 11/13/2022]
Abstract
Prospective studies in informative populations are crucial to increasing our knowledge of disease. In this perspective, we describe a half century of studies in an American Indian population that transformed our understanding of kidney disease in type 2 diabetes, now recognized as the leading cause of kidney failure worldwide. Serial examinations conducted for many years that included the collection of data and samples across multiple domains captured an unprecedented volume of clinical, physiologic, morphometric, genomic, and transcriptomic data. This work permitted us to extensively characterize the course and determinants of diabetic kidney disease, its pathophysiologic underpinnings, and important secular trends of urgent concern to populations worldwide, including the emergence of youth-onset type 2 diabetes and its effect on development of diabetic kidney disease in midlife. By combining these data using the tools of integrative biology, we are developing new mechanistic insights into the development and progression of diabetic kidney disease in type 2 diabetes. These insights have already contributed to the identification and successful therapeutic targeting of a novel pathway in DKD. We anticipate that this work will continue to expand our understanding of this complex disease and influence its management in the coming years.
Collapse
Affiliation(s)
- Robert G Nelson
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - William C Knowler
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kevin V Lemley
- Department of Pediatrics, University of Southern California Keck School of Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Helen C Looker
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| | - Michael Mauer
- Department of Pediatrics and Medicine, University of Minnesota, Minneapolis, MN
| | - William E Mitch
- Division of Nephrology, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Behzad Najafian
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA
| | - Peter H Bennett
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ
| |
Collapse
|
16
|
Reynolds EL, Akinci G, Banerjee M, Looker HC, Patterson A, Nelson RG, Feldman EL, Callaghan BC. The determinants of complication trajectories in American Indians with type 2 diabetes. JCI Insight 2021; 6:146849. [PMID: 34027894 PMCID: PMC8262294 DOI: 10.1172/jci.insight.146849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/11/2020] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUNDWe aimed to determine whether metabolic syndrome (MetS) affects longitudinal trajectories of diabetic complications, including neuropathy, cardiovascular autonomic neuropathy (CAN), and kidney disease in American Indians with type 2 diabetes.METHODSWe performed a prospective study where participants underwent annual metabolic phenotyping and outcome measurements. The updated National Cholesterol Education Program criteria were used to define MetS and its individual components, using BMI instead of waist circumference. Neuropathy was defined using the Michigan Neuropathy Screening Instrument index, CAN with the expiration/inspiration ratio, and kidney disease with glomerular filtration rate. Mixed-effects models were used to evaluate associations between MetS and these outcomes.RESULTSWe enrolled 141 participants: 73.1% female, a mean (±SD) age of 49.8 (12.3), and a diabetes duration of 19.6 years (9.7 years) who were followed for a mean of 3.1 years (1.7 years). MetS components were stable during follow-up except for declining obesity and cholesterol. Neuropathy (point estimate [PE]: 0.30, 95% CI: 0.24, 0.35) and kidney disease (PE: -14.2, 95% CI: -16.8, -11.4) worsened over time, but CAN did not (PE: -0.002, 95% CI: -0.006, 0.002). We found a significant interaction between the number of MetS components and time for neuropathy (PE: 0.05, 95% CI: 0.01-0.10) but not CAN (PE: -0.003, 95% CI: -0.007, 0.001) or kidney disease (PE: -0.69, 95% CI: -3.16, 1.76). Systolic blood pressure (SBP, unit = 10 mmHg) was associated with each complication: neuropathy (PE: 0.23, 95% CI: 0.07, 0.39), CAN (PE: -0.02, 95% CI: -0.03, -0.02), and kidney disease (PE: -10.2, 95% CI: -15.4, -5.1).CONCLUSIONIn participants with longstanding diabetes, neuropathy and kidney disease worsened during follow-up, despite stable to improving MetS components, suggesting that early metabolic intervention is necessary to prevent complications in such patients. Additionally, the number of MetS components was associated with an increased rate of neuropathy progression, and SBP was associated with each complication.FUNDINGThe following are funding sources: NIH T32NS0007222, NIH R24DK082841, NIH R21NS102924, NIH R01DK115687, the Intramural Program of the NIDDK, the NeuroNetwork for Emerging Therapies, the Robert and Katherine Jacobs Environmental Health Initiative, the Robert E. Nederlander Sr. Program for Alzheimer's Research, and the Sinai Medical Staff Foundation.TRIAL REGISTRATIONClinicalTrials.gov, NCT00340678.
Collapse
Affiliation(s)
- Evan L. Reynolds
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gulcin Akinci
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pediatric Neurology, Dr. Behcet Uz Children’s Hospital, Izmir, Turkey
| | - Mousumi Banerjee
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Helen C. Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, Arizona, USA
| | - Adam Patterson
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert G. Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Phoenix, Arizona, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Brian C. Callaghan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Fine A, Cox D, Bouw M. Higher Incidence of Peritonitis in Native Canadians on Continuous Ambulatory Peritoneal Dialysis. Perit Dial Int 2020. [DOI: 10.1177/089686089401400305] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022] Open
Abstract
Objective To determine if the rate of peritoneal dialysis (PD)-related infections in our large Native population was higher than in non-Natives. Design Prospective study of PD-related infections, 1987 to 1993. Patients Forty-eight Natives and 136 non-Natives were studied. Comparisons of infection rates were made as well as determinations of the effect of diabetes and of dialysis techniques on infection rate. Results The chance of remaining free of peritonitis was far lower at 6 and 12 months in Natives versus nonNatives, 40% versus 76% at 6 months, and 24% versus 54% at 12 months (p < 0.01). Having diabetes or adding intraperitoneal insulin did not confer additional risk of peritonitis. The Y-Iine reduced the risk of peritonitis in non-Natives only. Exit-site infection (ESI) was significantly higher in Natives versus non-Natives, 0.42 versus 0.19 episodes per patient year (p < 0.01) mainly due to Staph. aureus. However, less than 30% of episodes of peritonitis were due to that organism. Staph. epidermidis peritonitis episodes were not more common in Native patients, whereas infections due to most other organisms were. Conclusion The susceptibility to both peritonitis and exit-site infection is increased in Native Canadians compared to non-Natives. The non protective effect of the Yline combined with increased peritonitis due to most organisms except Staph. epidermidis in Natives suggests that host factors could be important in these patients.
Collapse
Affiliation(s)
- Adrian Fine
- Section of Nephrology, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Darlyne Cox
- Section of Nephrology, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Maria Bouw
- Section of Nephrology, St. Boniface Hospital, Winnipeg, Manitoba, Canada
| |
Collapse
|
18
|
Calcium dobesilate reduces VEGF signaling by interfering with heparan sulfate binding site and protects from vascular complications in diabetic mice. PLoS One 2020; 15:e0218494. [PMID: 31935212 PMCID: PMC6959593 DOI: 10.1371/journal.pone.0218494] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2019] [Accepted: 12/21/2019] [Indexed: 01/09/2023] Open
Abstract
Inhibiting vascular endothelial growth factor (VEGF) is a therapeutic option in diabetic microangiopathy. However, VEGF is needed at physiological concentrations to maintain glomerular integrity; complete VEGF blockade has deleterious effects on glomerular structure and function. Anti-VEGF therapy in diabetes raises the challenge of reducing VEGF-induced pathology without accelerating endothelial cell injury. Heparan sulfate (HS) act as a co-receptor for VEGF. Calcium dobesilate (CaD) is a small molecule with vasoprotective properties that has been used for the treatment of diabetic microangiopathy. Preliminary evidence suggests that CaD interferes with HS binding sites of fibroblast growth factor. We therefore tested the hypotheses that (1) CaD inhibits VEGF signaling in endothelial cells, (2) that this effect is mediated via interference between CaD and HS, and (3) that CaD ameliorates diabetic nephropathy in a streptozotocin-induced diabetic mouse model by VEGF inhibition. We found that CaD significantly inhibited VEGF165-induced endothelial cell migration, proliferation, and permeability. CaD significantly inhibited VEGF165-induced phosphorylation of VEGFR-2 and suppressed the activity of VEGFR-2 mediated signaling cascades. The effects of CaD in vitro were abrogated by heparin, suggesting the involvement of heparin-like domain in the interaction with CaD. In addition, VEGF121, an isoform which does not bind to heparin, was not inhibited by CaD. Using the proximity ligation approach, we detected inhibition of interaction in situ between HS and VEGF and between VEGF and VEGFR-2. Moreover, CaD reduced VEGF signaling in mice diabetic kidneys and ameliorated diabetic nephropathy and neuropathy, suggesting CaD as a VEGF inhibitor without the negative effects of complete VEGF blockade and therefore could be useful as a strategy in treating diabetic nephropathy.
Collapse
|
19
|
Chemouny JM, Sannier A, Hanouna G, Raimbourg Q, Daugas É, Vigneau C, Vrtovnsik F. [Criteria to indicate kidney biopsy in type 2 diabetic patients with proteinuria: Survey among French nephrologists]. Nephrol Ther 2019; 15:524-531. [PMID: 31718993 DOI: 10.1016/j.nephro.2019.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 11/16/2022]
Abstract
Diabetic nephropathy is usually a presumptive diagnosis based on clinical and biological evidence. Renal biopsies are performed in diabetic patients with atypical findings evoking non-diabetic renal disease who could benefit from specific therapies. French speaking nephrologists were asked which criteria they retain to indicate renal biopsy in patients with type 2 diabetes and albuminuria>0.5g/day or equivalent through an online anonymous questionnaire. Among the suggested criteria were absence of diabetic retinopathy, hematuria, rapid decrease in GFR, short diabetes duration or rapid raise of proteinuria. 188 people answered the poll among whom interns (12%), fellows (13%), university hospital practitioners (26%), general hospital practitioners (24%), practitioners in a non-profit organization (13%), practitioners on private activity (10%), multi-modal practitioners (3%) and people without clinical activity (2%). Increasing proteinuria was retained as an indication criterion for renal biopsy by 51% of respondents, nephrotic syndrome by 56% of respondents, absence of diabetic retinopathy by 57% of respondents, short diabetes duration by 65% of respondents, rapid GFR decline by 75% of respondents and hematuria by 78% of respondents. These data highlight the high diversity of opinions on this topic and their discrepancies with guidelines and current literature regarding the association between non-diabetic renal disease and clinical and biological features. The lack of adhesion of nephrologists to guidelines was especially noteworthy regarding the absence of diabetic retinopathy. These results emphasize the need for studies focusing on biopsy indication criteria in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Jonathan Maurice Chemouny
- Service de néphrologie, hôpital Pontchaillou, CHU de Rennes, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France; Faculté de médecine, université Rennes 1, 2, avenue du professeur Léon-Bernard, CS 34317, 35043 Rennes cedex, France; Institut Micalis, Inra, AgroParisTech, university Paris-Saclay, domaine de Vilvert, 78352 Jouy-en-Josas cedex, France.
| | - Aurélie Sannier
- Laboratoire d'anatomopathologie et de cytologie, hôpital Bichat-Claude-Bernard, 46, rue Henri-Huchard, 75877 Paris cedex 18, France; Université Paris-Diderot, Sorbonne Paris Cité, 16, rue Henri-Huchard, 75018 Paris, France
| | - Guillaume Hanouna
- Université Paris-Diderot, Sorbonne Paris Cité, 16, rue Henri-Huchard, 75018 Paris, France; Service de néphrologie, hôpital Bichat-Claude-Bernard, DHU Fire, 46, rue Henri-Huchard, 75877 Paris cedex 18, France
| | - Quentin Raimbourg
- Service de néphrologie, hôpital Bichat-Claude-Bernard, DHU Fire, 46, rue Henri-Huchard, 75877 Paris cedex 18, France
| | - Éric Daugas
- Université Paris-Diderot, Sorbonne Paris Cité, 16, rue Henri-Huchard, 75018 Paris, France; Service de néphrologie, hôpital Bichat-Claude-Bernard, DHU Fire, 46, rue Henri-Huchard, 75877 Paris cedex 18, France; Inserm 1149, Center for research on inflammation (CRI), 16, rue Henri-Huchard, 75018 Paris, France
| | - Cécile Vigneau
- Service de néphrologie, hôpital Pontchaillou, CHU de Rennes, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France; Faculté de médecine, université Rennes 1, 2, avenue du professeur Léon-Bernard, CS 34317, 35043 Rennes cedex, France; IRSET-UMR 1085, 9, avenue du professeur Léon-Bernard, 35000 Rennes, France
| | - François Vrtovnsik
- Université Paris-Diderot, Sorbonne Paris Cité, 16, rue Henri-Huchard, 75018 Paris, France; Service de néphrologie, hôpital Bichat-Claude-Bernard, DHU Fire, 46, rue Henri-Huchard, 75877 Paris cedex 18, France; Inserm 1149, Center for research on inflammation (CRI), 16, rue Henri-Huchard, 75018 Paris, France
| |
Collapse
|
20
|
Reinhardt M, Cushman TR, Thearle MS, Krakoff J. Epicardial adipose tissue is a predictor of decreased kidney function and coronary artery calcification in youth- and early adult onset type 2 diabetes mellitus. J Endocrinol Invest 2019; 42:979-986. [PMID: 30674009 DOI: 10.1007/s40618-019-1011-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/20/2018] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE To examine the association of epicardial and pericardial fat volume (EFV, PFV) with cardiovascular risk factors and kidney function in Native Americans of southwestern heritage with youth and early adult onset type 2 diabetes mellitus (T2DM) versus healthy controls. METHODS Using computed tomography, we quantified EFV and PFV in 149 Native Americans (92 women, 57 men), 95 of which had T2DM (38 diagnosed prior to age 20 years). Duration of T2DM, mean carotid arterial mass (AM), coronary artery calcification (CAC), IL-6, and estimated glomerular filtration rate eGFRcr(CKD-EPI) were measured. RESULTS EFV and PFV were associated with BMI (r = 0.37, p < 0.0001; r = 0.26, p = 0.001) and did not differ between onset age-groups and controls (p > 0.05). EFV was associated with AM only in controls (r = 0.51, p < 0.0001). After adjustment for BMI, T2DM duration, HbA1C, age, and sex, EFV was a predictor of CAC and IL-6 concentrations in early adult onset T2DM (β = 0.05 ± 0.02 cm3, p = 0.03; β = 0.05 ± 0.01 pg/ml/cm3, p = 0.002). EFV and PFV were independent predictors of reduced eGFRcr(CKD-EPI) in the youth onset T2DM group (β = -0.3 ± 0.08 ml/min/cm3, p = 0.001; β = -0.25 ± 0.05 ml/min/cm3, p < 0.0001). CONCLUSIONS Epicardial fat volume may be a risk factor for heart disease in individuals with early adult onset T2DM and a predictor of decreased kidney function in individuals with youth onset T2DM.
Collapse
Affiliation(s)
- M Reinhardt
- Obesity and Diabetes Clinical Research Section, Department of Health and Human Services, Phoenix Epidemiology and Clinical Research Branch National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 4212 N. 16th Street, Phoenix, AZ, 85016, USA.
- Department of Diagnostic and Interventional Radiology, University of Leipzig Medical Center, Liebigstraße 20, 04103, Leipzig, Germany.
| | - T R Cushman
- Obesity and Diabetes Clinical Research Section, Department of Health and Human Services, Phoenix Epidemiology and Clinical Research Branch National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 4212 N. 16th Street, Phoenix, AZ, 85016, USA
| | - M S Thearle
- Obesity and Diabetes Clinical Research Section, Department of Health and Human Services, Phoenix Epidemiology and Clinical Research Branch National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 4212 N. 16th Street, Phoenix, AZ, 85016, USA
| | - J Krakoff
- Obesity and Diabetes Clinical Research Section, Department of Health and Human Services, Phoenix Epidemiology and Clinical Research Branch National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 4212 N. 16th Street, Phoenix, AZ, 85016, USA
| |
Collapse
|
21
|
Niewczas MA, Pavkov ME, Skupien J, Smiles A, Md Dom ZI, Wilson JM, Park J, Nair V, Schlafly A, Saulnier PJ, Satake E, Simeone CA, Shah H, Qiu C, Looker HC, Fiorina P, Ware CF, Sun JK, Doria A, Kretzler M, Susztak K, Duffin KL, Nelson RG, Krolewski AS. A signature of circulating inflammatory proteins and development of end-stage renal disease in diabetes. Nat Med 2019; 25:805-813. [PMID: 31011203 PMCID: PMC6508971 DOI: 10.1038/s41591-019-0415-5] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2018] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
Chronic inflammation is postulated to be involved in development of end stage renal disease (ESRD) in diabetes, but which specific circulating inflammatory proteins contribute to this risk remains unknown. To study this we examined 194 circulating inflammatory proteins in subjects from three independent cohorts with Type 1 and Type 2 diabetes. In each cohort we identified an extremely robust Kidney Risk Inflammatory Signature (KRIS) consisting of 17 novel proteins enriched for TNF Receptor Superfamily members that was associated with a 10-year risk of ESRD. All these proteins had a systemic, non-kidney source. Our prospective study findings provide strong evidence that KRIS proteins contribute to the inflammatory process underlying ESRD development in both types of diabetes. These proteins may be used as new therapeutic targets, new prognostic tests for high risk of ESRD and as surrogate outcome measures where changes in KRIS levels during intervention can reflect the tested therapy’s effectiveness. Proteomic profiling of circulating proteins in subjects from three independent cohorts with type 1 and type 2 diabetes, identified an extremely robust inflammatory signature, consisting of 17 proteins enriched for TNF Receptor Superfamily members that was associated with a 10-year risk of end-stage renal disease.
Collapse
Affiliation(s)
- Monika A Niewczas
- Research Division, Joslin Diabetes Center, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Meda E Pavkov
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jan Skupien
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Metabolic Diseases, Jagiellonian University Medical College, Krakow, Poland
| | - Adam Smiles
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Zaipul I Md Dom
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jonathan M Wilson
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Viji Nair
- Nephrology/Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | | | - Pierre-Jean Saulnier
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA.,CHU Poitiers, University of Poitiers, Inserm, Clinical Investigation Center CIC1402, Poitiers, France
| | - Eiichiro Satake
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Hetal Shah
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Helen C Looker
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Romeo ed Enrica Invernizzi Pediatric Center, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Carl F Ware
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jennifer K Sun
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alessandro Doria
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Matthias Kretzler
- Nephrology/Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin L Duffin
- Diabetes and Complications Department, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Robert G Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Andrzej S Krolewski
- Research Division, Joslin Diabetes Center, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Establishment of Risk Prediction Model for Retinopathy in Type 2 Diabetic Patients. Brain Inform 2019. [DOI: 10.1007/978-3-030-37078-7_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2022] Open
|
23
|
Arslanian S, Bacha F, Grey M, Marcus MD, White NH, Zeitler P. Evaluation and Management of Youth-Onset Type 2 Diabetes: A Position Statement by the American Diabetes Association. Diabetes Care 2018; 41:2648-2668. [PMID: 30425094 PMCID: PMC7732108 DOI: 10.2337/dci18-0052] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/03/2023]
Affiliation(s)
- Silva Arslanian
- Division of Pediatric Endocrinology, Metabolism, and Diabetes Mellitus, University of Pittsburgh, Pittsburgh, PA
- Center for Pediatric Research in Obesity and Metabolism, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Fida Bacha
- Children's Nutrition Research Center, Texas Children's Hospital and Baylor College of Medicine, Houston, TX
| | - Margaret Grey
- Yale School of Nursing, New Haven, CT
- Yale School of Medicine, New Haven, CT
| | | | - Neil H White
- Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Philip Zeitler
- Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
24
|
Vart P, Grams ME, Ballew SH, Woodward M, Coresh J, Matsushita K. Socioeconomic status and risk of kidney dysfunction: the Atherosclerosis Risk in Communities study. Nephrol Dial Transplant 2018; 34:1361-1368. [DOI: 10.1093/ndt/gfy142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/16/2017] [Accepted: 04/22/2018] [Indexed: 11/12/2022] Open
Abstract
Abstract
Background
There is strong evidence of an association between socioeconomic status (SES) and end-stage renal disease (ESRD). However, the association of SES with the risk of chronic kidney disease (CKD) and the rate of change in kidney function is unclear.
Methods
A cohort of 14 086 participants with an estimated glomerular filtration rate (eGFR) ≥60 mL/min/1.73 m2 at baseline in the Atherosclerosis Risk in Communities study (1987–89) were studied. The association of annual household income, educational attainment and neighborhood deprivation with incident ESRD, incident CKD and change in eGFR using four measurements over ∼23 years was assessed.
Results
A total of 432 participants developed ESRD and 3510 developed CKD over a median follow-up time of ∼23 years. After adjustment for demographics and baseline eGFR, the hazard ratio (HR) for incident ESRD compared with the high-income group was 1.56 [95% confidence interval (CI) 1.22–1.99 in the medium-income group and 2.30 (95% CI 1.75–3.02) in the low-income group (P-trend < 0.001), and for CKD was 1.10 (95% CI 1.01–1.20) in the medium-income group and 1.30 (95% CI 1.17–1.44) in the low-income group (P-trend < 0.001). After full adjustments, the HR for ESRD was 1.33 (95% CI 1.03–1.70) in the medium-income group and 1.50 (95% CI 1.14–1.98) in the low-income group (P-trend = 0.003) and for CKD was 1.01 (95% CI 0.92–1.10) in the medium-income group and 1.04 (95% CI 0.93–1.16) in the low-income group (P-trend = 0.50). The eGFR decline was 5% and 15% steeper in the medium- and low-income groups, respectively, after full adjustment (P-trend < 0.001). Results were similar, with lower educational attainment and higher neighborhood deprivation being associated with adverse outcomes.
Conclusions
SES (annual household income, educational attainment or neighborhood deprivation) was associated not only with ESRD risk but also with eGFR decline, although the association with CKD appeared weaker.
Collapse
Affiliation(s)
- Priya Vart
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Morgan E Grams
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Mark Woodward
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- The George Institute for Global Health, University of Oxford, Oxford, UK
- The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Josef Coresh
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | |
Collapse
|
25
|
Qiu C, Hanson RL, Fufaa G, Kobes S, Gluck C, Huang J, Chen Y, Raj D, Nelson RG, Knowler WC, Susztak K. Cytosine methylation predicts renal function decline in American Indians. Kidney Int 2018; 93:1417-1431. [PMID: 29709239 PMCID: PMC5973533 DOI: 10.1016/j.kint.2018.01.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/04/2017] [Revised: 01/07/2018] [Accepted: 01/25/2018] [Indexed: 12/18/2022]
Abstract
Diabetic nephropathy accounts for most of the excess mortality in individuals with diabetes, but the molecular mechanisms by which nephropathy develops are largely unknown. Here we tested cytosine methylation levels at 397,063 genomic CpG sites for association with decline in the estimated glomerular filtration rate (eGFR) over a six year period in 181 diabetic Pima Indians. Methylation levels at 77 sites showed significant association with eGFR decline after correction for multiple comparisons. A model including methylation level at two probes (cg25799291 and cg22253401) improved prediction of eGFR decline in addition to baseline eGFR and the albumin to creatinine ratio with the percent of variance explained significantly improving from 23.1% to 42.2%. Cg22253401 was also significantly associated with eGFR decline in a case-control study derived from the Chronic Renal Insufficiency Cohort. Probes at which methylation significantly associated with eGFR decline were localized to gene regulatory regions and enriched for genes with metabolic functions and apoptosis. Three of the 77 probes that were associated with eGFR decline in blood samples showed directionally consistent and significant association with fibrosis in microdissected human kidney tissue, after correction for multiple comparisons. Thus, cytosine methylation levels may provide biomarkers of disease progression in diabetic nephropathy and epigenetic variations contribute to the development of diabetic kidney disease.
Collapse
MESH Headings
- Adult
- Aged
- Albuminuria/ethnology
- Albuminuria/genetics
- Albuminuria/physiopathology
- Apoptosis/genetics
- Case-Control Studies
- Cell Cycle/genetics
- CpG Islands
- Cytosine
- DNA Methylation
- Diabetic Nephropathies/diagnosis
- Diabetic Nephropathies/ethnology
- Diabetic Nephropathies/genetics
- Diabetic Nephropathies/physiopathology
- Disease Progression
- Energy Metabolism/genetics
- Epigenesis, Genetic
- Female
- Fibrosis
- Genetic Predisposition to Disease
- Glomerular Filtration Rate/genetics
- Humans
- Indians, North American/genetics
- Kidney/pathology
- Kidney/physiopathology
- Kidney Failure, Chronic/diagnosis
- Kidney Failure, Chronic/ethnology
- Kidney Failure, Chronic/genetics
- Kidney Failure, Chronic/physiopathology
- Male
- Middle Aged
- Phenotype
- Prognosis
- Renal Insufficiency, Chronic/diagnosis
- Renal Insufficiency, Chronic/ethnology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/physiopathology
- Risk Factors
Collapse
Affiliation(s)
- Chengxiang Qiu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robert L Hanson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA.
| | - Gudeta Fufaa
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Sayuko Kobes
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Caroline Gluck
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jing Huang
- Department of Biostatistics, Epidemiology and Informatics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yong Chen
- Department of Biostatistics, Epidemiology and Informatics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dominic Raj
- Division of Renal Diseases and Hypertension, The George Washington School of Medicine, Washington, DC, USA
| | - Robert G Nelson
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - William C Knowler
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Affiliation(s)
- F. Dumler
- Division of Nephrology and Hypertension Henry Ford Hospital, Detroit - U.S.A
| |
Collapse
|
27
|
Bedigian MP. Improving the prognosis of diabetic patients: Evaluating the role of intensive versus moderate blood pressure control with selective angiotensin II receptor blocker (ARB) therapy. J Renin Angiotensin Aldosterone Syst 2017; 1:S25-8. [PMID: 17199216 DOI: 10.3317/jraas.2000.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/01/2022] Open
Abstract
The ABCD (Appropriate Blood Pressure Control in Diabetes) and ABCD-2V (Part 2 with Valsartan) are prospective, randomised clinical trials which will provide important data on the impact of intensive vs. moderate blood pressure (BP) control on microvascular and macrovascular complications in normotensive and hypertensive patients with type 2 diabetes mellitus (DM). The ABCD trial was a five-year study that compared the effects of intensive vs. moderate BP control on the endpoints of nephropathy, retinopathy, neuropathy, and cardiovascular disease events using a calcium channel blocker (CCB) and an angiotensin-converting enzyme (ACE) inhibitor as the primary antihypertensive agents. The recently published results of the hypertensive cohort of ABCD are reviewed herein. The follow-up study, ABCD-2V, is ongoing and was designed to compare intensive vs. moderate BP control on the same endpoints as the ABCD study, using the highly selective angiotensin II receptor blocker (ARB) valsartan as the primary antihypertensive agent. First results of ABCD-2V are expected in 2004. The baseline characteristics for the patients enrolled thus far in the hypertensive cohort of ABCD-2V are reviewed. These studies will provide insight into the role of intensive vs. moderate BP control in the management of normotensive and hypertensive patients with type 2 DM.
Collapse
Affiliation(s)
- M P Bedigian
- Novartis Pharmaceuticals, East Hanover, NJ, USA.
| |
Collapse
|
28
|
Molecular Mechanisms and Treatment Strategies in Diabetic Nephropathy: New Avenues for Calcium Dobesilate-Free Radical Scavenger and Growth Factor Inhibition. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1909258. [PMID: 29082239 PMCID: PMC5634607 DOI: 10.1155/2017/1909258] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/13/2017] [Accepted: 06/21/2017] [Indexed: 01/15/2023]
Abstract
Diabetic nephropathy is one of the most important microvascular complications of diabetes mellitus and is responsible for 40–50% of all cases of end stage renal disease. The therapeutic strategies in diabetic nephropathy need to be targeted towards the pathophysiology of the disease. The earlier these therapeutic strategies can bring about positive effects on vascular changes and prevent the vasculature in patients with diabetes from deteriorating, the better the renal function can be preserved. Studies evaluating anti-inflammatory and antioxidative strategies in diabetic nephropathy demonstrate the need and value of these novel treatment avenues. CaD is an established vasoactive and angioprotective drug that has shown a unique, multitarget mode of action in several experimental studies and in different animal models of diabetic microvascular complications. On the molecular level, CaD reduces oxidative stress and inhibits growth factors such as fibroblast growth factor and vascular endothelial growth factors. Recent findings have demonstrated a strong rationale for its use in reducing urine albumin excretion rate and markers of inflammation as well as improving endothelial function. Its beneficial effects make it an attractive therapeutic compound especially in the early stages of the disease. These findings, although promising, need further confirmation in prospective clinical trials with CaD.
Collapse
|
29
|
Fu S, Zhou S, Luo L, Ye P. Relationships of pancreatic beta-cell function with microalbuminuria and glomerular filtration rate in middle-aged and elderly population without type 2 diabetes mellitus: a Chinese community-based analysis. Clin Interv Aging 2017; 12:753-757. [PMID: 28496313 PMCID: PMC5422324 DOI: 10.2147/cia.s134496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Relationships of pancreatic beta-cell function abnormality with microalbuminuria (MA) and glomerular filtration rate (GFR) may differ by age, ethnicity and accompanied diseases. Previous studies were generally conducted in Western adult patients with type 2 diabetes mellitus (T2DM), and it is uncertain whether pancreatic beta-cell function is associated with MA and GFR in Chinese community-dwelling middle-aged and elderly population without T2DM. We therefore examined the relationships of pancreatic beta-cell function with two indices of renal damage, MA and GFR, in Chinese community-dwelling middle-aged and elderly population without T2DM. METHODS This analysis focused on 380 Beijing residents older than 45 years who were free of T2DM and completed the evaluation of pancreatic beta-cell function. RESULTS Median age was 67 (49-80) years. Levels of triglyceride, diastolic blood pressure and homeostasis model assessment-beta (HOMA-beta) index were positively related to urine microalbumin (P<0.05 for all). Age, low-density lipoprotein cholesterol levels and HOMA-beta index were inversely correlated with GFR, while high-density lipoprotein cholesterol levels were positively correlated with GFR (P<0.05 for all). In all three adjustment models, there was a significant positive association between HOMA-beta index and MA; subjects with higher beta-cell function had higher odds of MA (P<0.05 for all). There was no association between HOMA-beta index and GFR <60 mL/min/1.73 m2 in any model (P>0.05 for all). CONCLUSION Modeling the pancreatic beta-cell function with different adjusted variables provided the same conclusion of association with MA; beta-cell function was positively associated with MA. Additionally, there was a specific difference in the adjusted associations of pancreatic beta-cell function with MA and GFR <60 mL/min/1.73 m2; beta-cell function was not independently associated with GFR <60 mL/min/1.73 m2. This result indicated that abnormal pancreatic beta-cell function plays an important role in the development of MA.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology.,Department of Cardiology and Hainan Branch
| | - Shanjing Zhou
- Department of Traditional Chinese Medicine and Hainan Branch, Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | | | - Ping Ye
- Department of Geriatric Cardiology
| |
Collapse
|
30
|
Jiang Y, Ma L, Han C, Liu Q, Cong X, Xu Y, Zhao T, Li P, Cao Y. Effects of Apolipoprotein E Isoforms in Diabetic Nephropathy of Chinese Type 2 Diabetic Patients. J Diabetes Res 2017; 2017:3560920. [PMID: 28326331 PMCID: PMC5343254 DOI: 10.1155/2017/3560920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/10/2016] [Revised: 01/02/2017] [Accepted: 01/29/2017] [Indexed: 01/19/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the major chronic complications of diabetes. Genetic polymorphism of Apolipoprotein E (ApoE) has been proposed to participating in DN. The purpose of the study was to evaluate the relationship between ApoE genetic polymorphism and the presence of DN in Chinese type 2 diabetic patients. We studied 845 diabetic patients who were divided into DN group (n = 429) and control group (n = 416). ApoE genotype was determined by ApoE genotyping chip and the plasmatic biochemical characterization was performed on all subjects. There were differences (P < 0.001) in HbA1c, creatinine, and urinary albumin between the two groups. The ApoE ε2 allelic frequency was 7.69% in DN group versus 3.49% in control group (OR = 2.22, 95% CI = 1.41-3.47, and P < 0.05), as expected, ApoE E2/E2 and E2/E3 genotype frequency were higher in DN group (13.75% versus 6.49%, P < 0.05). The ApoE ε4 allelic frequency was 7.93% in DN group versus 11.54% in control group (OR = 0.70, 95% CI = 0.50-0.97, and P < 0.05), and DN group presented a lower frequency of ApoE E3/E4 and E4/E4 genotype frequency (14.91% versus 19.96%, P < 0.05). These results suggest ApoE ε2 allele may be a risk factor; however ApoE ε4 allele may play a protective role of DN in Chinese type 2 diabetic patients.
Collapse
Affiliation(s)
- YongWei Jiang
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Liang Ma
- Graduate School of Peking Union Medical College, Beijing, China
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - ChengWu Han
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Qian Liu
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Xiao Cong
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - YaPing Xu
- Department of Pathophysiology, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - TingTing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Ping Li
- Graduate School of Peking Union Medical College, Beijing, China
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - YongTong Cao
- Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
31
|
Van JAD, Scholey JW, Konvalinka A. Insights into Diabetic Kidney Disease Using Urinary Proteomics and Bioinformatics. J Am Soc Nephrol 2017; 28:1050-1061. [PMID: 28159781 DOI: 10.1681/asn.2016091018] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
A number of proteomic and peptidomic analyses of urine from diabetic subjects have been published in the quest for a biomarker that predicts progression of nephropathy. Less attention has been paid to the relationships between urinary proteins and the underlying biological processes revealed by the analyses. In this review, we focus on the biological processes identified by studying urinary proteins and protein-protein interactions at each stage of diabetic nephropathy to provide an overview of the events underlying progression of kidney disease reflected in the urine. In uncomplicated diabetes, proteomic/peptidomic analyses indicate that early activation of fibrotic pathways in the kidney occurs before the onset of microalbuminuria. In incipient nephropathy, when albumin excretion rates are abnormal, proteomic/peptidomic analyses suggest that changes in glomerular permselectivity and tubular reabsorption account, at least in part, for the proteins and peptides that appear in the urine. Finally, overt nephropathy is characterized by proteins involved in wound healing, ongoing fibrosis, and inflammation. These findings suggest that there is a spectrum of biological processes in the diabetic kidney and that assessing protein networks may be more informative than individual markers with respect to the stage of disease and the risk of progression.
Collapse
Affiliation(s)
- Julie A D Van
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; and
| | - James W Scholey
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; and.,Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| | - Ana Konvalinka
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; and.,Department of Medicine, Division of Nephrology, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Diabetes Mellitus Affected Patients Classification and Diagnosis through Machine Learning Techniques. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.procs.2017.08.193] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/30/2023]
|
33
|
Narres M, Claessen H, Droste S, Kvitkina T, Koch M, Kuss O, Icks A. The Incidence of End-Stage Renal Disease in the Diabetic (Compared to the Non-Diabetic) Population: A Systematic Review. PLoS One 2016; 11:e0147329. [PMID: 26812415 PMCID: PMC4727808 DOI: 10.1371/journal.pone.0147329] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/10/2015] [Accepted: 01/01/2016] [Indexed: 12/15/2022] Open
Abstract
End-stage renal disease (ESRD) in diabetes is a life threatening complication resulting in a poor prognosis for patients as well as high medical costs. The aims of this systematic review were (1) to evaluate the incidence of ESRD due to all causes and due to diabetic nephropathy in the diabetic population and differences between incidences of ESRD with respect to sex, ethnicity, age and regions, (2) to compare incidence rates in the diabetic and non-diabetic population, and (3) to investigate time trends. The systematic review was conducted according to the PRISMA group guidelines by performing systematic literature searches in the biomedical databases until January 3rd 2015; thirty-two studies were included. Among patients with incident type 1 diabetes the 30-year cumulative incidence ranged from 3.3% to 7.8%. Among patients with prevalent diabetes, incidence rates of ESRD due to all causes ranged from 132.0 to 167.0 per 100,000 person-years, whereas incidence rates of ESRD due to diabetic nephropathy varied from 38.4 to 804.0 per 100,000 person-years. The incidence of ESRD in the diabetic population was higher compared to the non-diabetic population, and relative risks varied from 6.2 in the white population to 62.0 among Native Americans. The results regarding time trends were inconsistent. The review conducted demonstrates the considerable variation of incidences of ESRD among the diabetic population. Consistent findings included an excess risk when comparing the diabetic to the non-diabetic population and ethnic differences. We recommend that newly designed studies should use standardized methods for the determination of ESRD and population at risk.
Collapse
MESH Headings
- Databases, Factual
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 1/ethnology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/ethnology
- Diabetic Nephropathies/complications
- Diabetic Nephropathies/ethnology
- Humans
- Incidence
- Kidney Failure, Chronic/epidemiology
- Kidney Failure, Chronic/ethnology
- Kidney Failure, Chronic/etiology
- Risk Factors
Collapse
Affiliation(s)
- Maria Narres
- Institute for Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
| | - Heiner Claessen
- Institute for Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
| | - Sigrid Droste
- Department of Public Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tatjana Kvitkina
- Institute for Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
- Department of Public Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Koch
- Center of Nephrology, Mettmann, Germany
- Clinic of Nephrology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Oliver Kuss
- Institute for Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Andrea Icks
- Institute for Biometrics and Epidemiology, German Diabetes Center, Düsseldorf, Germany
- Department of Public Health, Heinrich-Heine-University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
34
|
Davoudi S, Sobrin L. Novel Genetic Actors of Diabetes-Associated Microvascular Complications: Retinopathy, Kidney Disease and Neuropathy. Rev Diabet Stud 2016; 12:243-59. [PMID: 26859656 DOI: 10.1900/rds.2015.12.243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023] Open
Abstract
Both type 1 and type 2 diabetes mellitus can lead to the common microvascular complications of diabetic retinopathy, kidney disease, and neuropathy. Diabetic patients do not universally develop these complications. Long duration of diabetes and poor glycemic control explain a lot of the variability in the development of microvascular complications, but not all. Genetic factors account for some of the remaining variability because of the heritability and familial clustering of these complications. There have been a large number of investigations, including linkage studies, candidate gene studies, and genome-wide association studies, all of which have sought to identify the specific variants that increase susceptibility. For retinopathy, several genome-wide association studies have been performed in small or midsize samples, but no reproducible loci across the studies have been identified. For diabetic kidney disease, genome-wide association studies in larger samples have been performed, and loci for this complication are beginning to emerge. However, validation of the existing discoveries, and further novel discoveries in larger samples is ongoing. The amount of genetic research into diabetic neuropathy has been very limited, and much is dedicated to the understanding of genetic risk factors only. Collaborations that pool samples and aim to detect phenotype classifications more precisely are promising avenues for a better explanation of the genetics of diabetic microvascular complications.
Collapse
Affiliation(s)
- Samaneh Davoudi
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| | - Lucia Sobrin
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA
| |
Collapse
|
35
|
Fufaa GD, Weil EJ, Lemley KV, Knowler WC, Brosius FC, Yee B, Mauer M, Nelson RG. Structural Predictors of Loss of Renal Function in American Indians with Type 2 Diabetes. Clin J Am Soc Nephrol 2016; 11:254-61. [PMID: 26792530 DOI: 10.2215/cjn.05760515] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/27/2015] [Accepted: 10/14/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Diabetes is the leading cause of kidney failure in the United States, but early structural determinants of renal function loss in type 2 diabetes are poorly defined. We examined the association between morphometrically determined renal structural variables and loss of renal function in 111 American Indians with type 2 diabetes who volunteered for a research kidney biopsy at the end of a 6-year clinical trial designed to test the renoprotective efficacy of losartan versus placebo. Participants were subsequently followed in an observational study, in which annual measurements of GFR (iothalamate) initiated during the clinical trial were continued. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Renal function loss was defined as ≥40% loss of GFR from the research examination performed at the time of kidney biopsy. Associations with renal function loss were evaluated by Cox proportional hazards regression. Hazard ratios (HRs) were reported per 1-SD increment for each morphometric variable. RESULTS Of 111 participants (82% women; baseline mean [±SD] age, 46 years old [±10]; diabetes duration, 16 years [±6]; hemoglobin A1c =9.4% [±2.2]; GFR=147 ml/min [±56]; median albumin-to-creatinine ratio, 41 mg/g [interquartile range, 13-158]), 51 (46%) developed renal function loss during a median follow-up of 6.6 years (interquartile range, 3.1-9.0). Fourteen had baseline GFR <90 ml/min, and three had baseline GFR <60 ml/min. Higher mesangial fractional volume (HR, 2.27; 95% confidence interval [95% CI], 1.58 to 3.26), percentage of global glomerular sclerosis (HR, 1.63; 95% CI, 1.21 to 2.21), nonpodocyte cell number per glomerulus (HR, 1.50; 95% CI, 1.10 to 2.05), glomerular basement membrane width (HR, 1.48; 95% CI, 1.05 to 2.08), mean glomerular volume (HR, 1.42; 95% CI, 1.02 to 1.96), and podocyte foot process width (HR, 1.28; 95% CI, 1.03 to 1.60); lower glomerular filtration surface density (HR, 0.62; 95% CI, 0.41 to 0.94); and fewer endothelial fenestrations (HR, 0.68; 95% CI, 0.48 to 0.95) were each associated with GFR decline after adjustment for baseline age, sex, duration of diabetes, hemoglobin A1c, GFR, and treatment assignment during the clinical trial. CONCLUSIONS Quantitative measures of glomerular structure predict loss of renal function in type 2 diabetes.
Collapse
Affiliation(s)
- Gudeta D Fufaa
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - E Jennifer Weil
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Kevin V Lemley
- Department of Pediatrics, University of Southern California Keck School of Medicine, Children's Hospital Los Angeles, Los Angeles, California
| | - William C Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona
| | - Frank C Brosius
- Division of Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Berne Yee
- Southwest Kidney Institute, Phoenix, Arizona; and
| | - Michael Mauer
- Department of Pediatrics and Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Robert G Nelson
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona;
| |
Collapse
|
36
|
Pavkov ME, Weil EJ, Fufaa GD, Nelson RG, Lemley KV, Knowler WC, Niewczas MA, Krolewski AS. Tumor necrosis factor receptors 1 and 2 are associated with early glomerular lesions in type 2 diabetes. Kidney Int 2016; 89:226-34. [PMID: 26398493 PMCID: PMC4805514 DOI: 10.1038/ki.2015.278] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2015] [Revised: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 01/28/2023]
Abstract
Elevated serum tumor necrosis factor receptor 1 (TNFR1) and 2 (TNFR2) concentrations are strongly associated with increased risk of end-stage renal disease in type 2 diabetes. However, little is known about the early glomerular structural lesions that develop in patients when these markers are elevated. Here we examined the relationships between TNFRs and glomerular structure in 83 American Indians with type 2 diabetes. Serum TNFRs and glomerular filtration rates (GFR, iothalamate) were measured during a research exam performed within a median of 0.9 months from a percutaneous kidney biopsy. Associations of TNFRs with glomerular structural variables were quantified by Spearman's correlations and by multivariable linear regression after adjustment for age, gender, diabetes duration, hemoglobin A1c, body mass index, and mean arterial pressure. The baseline mean age was 46 years, median GFR 130 ml/min, median albumin/creatinine ratio 26 mg/g, median TNFR1 1500 pg/ml, and median TNFR2 3284 pg/ml. After multivariable adjustment, TNFR1 and TNFR2 significantly correlated inversely with the percentage of endothelial cell fenestration and the total filtration surface per glomerulus. There were significant positive correlations with mesangial fractional volume glomerular basement membrane width, podocyte foot process width, and percent of global glomerular sclerosis. Thus, TNFRs may be involved in the pathogenesis of early glomerular lesions in diabetic nephropathy.
Collapse
Affiliation(s)
- Meda E Pavkov
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - E Jennifer Weil
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Gudeta D Fufaa
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Robert G Nelson
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA.
| | - Kevin V Lemley
- Department of Pediatrics, University of Southern California Keck School of Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - William C Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | - Monika A Niewczas
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrzej S Krolewski
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Abstract
The rising global prevalence of diabetes mellitus is accompanied by an increasing burden of morbidity and mortality that is attributable to the complications of chronic hyperglycaemia. These complications include blindness, renal failure and cardiovascular disease. Current therapeutic options for chronic hyperglycaemia reduce, but do not eradicate, the risk of these complications. Success in defining new preventative and therapeutic strategies hinges on an improved understanding of the molecular processes involved in the development of these complications. This Review explores the role of human genetics in delivering such insights, and describes progress in characterizing the sequence variants that influence individual predisposition to diabetic kidney disease, retinopathy, neuropathy and accelerated cardiovascular disease. Numerous risk variants for microvascular complications of diabetes have been reported, but very few have shown robust replication. Furthermore, only limited evidence exists of a difference in the repertoire of risk variants influencing macrovascular disease between those with and those without diabetes. Here, we outline the challenges associated with the genetic analysis of diabetic complications and highlight ongoing efforts to deliver biological insights that can drive translational benefits.
Collapse
|
38
|
Estacio RO. Renin-Angiotensin-Aldosterone System Blockade in Diabetes: Role of Direct Renin Inhibitors. Postgrad Med 2015; 121:33-44. [DOI: 10.3810/pgm.2009.05.2000] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/22/2023]
|
39
|
Nicholas SB, Iyengar SK. Does losartan prevent progression of early diabetic nephropathy in American Indians with type 2 diabetes? Diabetes 2013; 62:3014-6. [PMID: 23970520 PMCID: PMC3749328 DOI: 10.2337/db13-0748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Affiliation(s)
- Susanne B Nicholas
- Divisions of Nephrology and Endocrinology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA.
| | | |
Collapse
|
40
|
Do OH, Nguyen KT. The role of glycemia and blood pressure control on the rate of decline in glomerular filtration rate in Vietnamese type 2 diabetes patients. Int J Diabetes Dev Ctries 2013. [DOI: 10.1007/s13410-013-0112-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/21/2023] Open
|
41
|
Klein R, Klein BE. The Epidemiology of Diabetic Retinopathy. Retina 2013. [DOI: 10.1016/b978-1-4557-0737-9.00045-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/31/2023]
|
42
|
Abstract
Diabetic nephropathy (DN) is a devastating complication of type 1 and type 2 diabetes and leads to increased morbidity and premature mortality. Susceptibility to DN has an inherent genetic basis as evidenced by familial aggregation and ethnic-specific prevalence rates. Progress in identifying the underlying genetic architecture has been arduous with the realization that a single locus of large effect does not exist, unlike in predisposition to non-diabetic nephropathy in individuals with African ancestry. Numerous risk variants have been identified, each with a nominal effect, and they collectively contribute to disease. These results have identified loci targeting novel pathways for disease susceptibility. With continued technological advances and development of new analytic methods, additional genetic variants and mechanisms (e.g., epigenetic variation) will be identified and help to elucidate the pathogenesis of DN. These advances will lead to early detection and development of novel therapeutic strategies to decrease the incidence of disease.
Collapse
Affiliation(s)
- Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston Salem, NC USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston Salem, NC USA
| | - Barry I. Freedman
- Department of Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston Salem, NC USA
| |
Collapse
|
43
|
Williams WW, Salem RM, McKnight AJ, Sandholm N, Forsblom C, Taylor A, Guiducci C, McAteer JB, McKay GJ, Isakova T, Brennan EP, Sadlier DM, Palmer C, Söderlund J, Fagerholm E, Harjutsalo V, Lithovius R, Gordin D, Hietala K, Kytö J, Parkkonen M, Rosengård-Bärlund M, Thorn L, Syreeni A, Tolonen N, Saraheimo M, Wadén J, Pitkäniemi J, Sarti C, Tuomilehto J, Tryggvason K, Österholm AM, He B, Bain S, Martin F, Godson C, Hirschhorn JN, Maxwell AP, Groop PH, Florez JC. Association testing of previously reported variants in a large case-control meta-analysis of diabetic nephropathy. Diabetes 2012; 61:2187-94. [PMID: 22721967 PMCID: PMC3402313 DOI: 10.2337/db11-0751] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 01/13/2023]
Abstract
We formed the GEnetics of Nephropathy-an International Effort (GENIE) consortium to examine previously reported genetic associations with diabetic nephropathy (DN) in type 1 diabetes. GENIE consists of 6,366 similarly ascertained participants of European ancestry with type 1 diabetes, with and without DN, from the All Ireland-Warren 3-Genetics of Kidneys in Diabetes U.K. and Republic of Ireland (U.K.-R.O.I.) collection and the Finnish Diabetic Nephropathy Study (FinnDiane), combined with reanalyzed data from the Genetics of Kidneys in Diabetes U.S. Study (U.S. GoKinD). We found little evidence for the association of the EPO promoter polymorphism, rs161740, with the combined phenotype of proliferative retinopathy and end-stage renal disease in U.K.-R.O.I. (odds ratio [OR] 1.14, P = 0.19) or FinnDiane (OR 1.06, P = 0.60). However, a fixed-effects meta-analysis that included the previously reported cohorts retained a genome-wide significant association with that phenotype (OR 1.31, P = 2 × 10(-9)). An expanded investigation of the ELMO1 locus and genetic regions reported to be associated with DN in the U.S. GoKinD yielded only nominal statistical significance for these loci. Finally, top candidates identified in a recent meta-analysis failed to reach genome-wide significance. In conclusion, we were unable to replicate most of the previously reported genetic associations for DN, and significance for the EPO promoter association was attenuated.
Collapse
Affiliation(s)
- Winfred W. Williams
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Rany M. Salem
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Endocrine Research Unit, Department of Endocrinology, Children’s Hospital, Boston, Massachusetts
| | - Amy Jayne McKnight
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast, U.K
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
- Department of Biomedical Engineering and Computational Science, Aalto University, Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Andrew Taylor
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Candace Guiducci
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Jarred B. McAteer
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Gareth J. McKay
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast, U.K
| | - Tamara Isakova
- Division of Nephrology, University of Miami, Miller School of Medicine, Miami, Florida
| | - Eoin P. Brennan
- UCD Diabetes Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Denise M. Sadlier
- UCD Diabetes Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
- Mater University Hospital, Dublin, Ireland
| | - Cameron Palmer
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Endocrine Research Unit, Department of Endocrinology, Children’s Hospital, Boston, Massachusetts
| | - Jenny Söderlund
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Emma Fagerholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
- Department of Chronic Disease Prevention, Welfare and Health Promotion Division, National Institute for Health and Welfare, Helsinki, Finland
| | - Raija Lithovius
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Daniel Gordin
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Kustaa Hietala
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Department of Ophthalmology, Helsinki University Central Hospital, Helsinki, Finland
| | - Janne Kytö
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Department of Ophthalmology, Helsinki University Central Hospital, Helsinki, Finland
| | - Maija Parkkonen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Milla Rosengård-Bärlund
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Lena Thorn
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Nina Tolonen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Markku Saraheimo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Johan Wadén
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Janne Pitkäniemi
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Cinzia Sarti
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Chronic Disease Prevention, Welfare and Health Promotion Division, National Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- South Ostrobothnia Central Hospital, Seinäjoki, Finland
| | - Karl Tryggvason
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anne-May Österholm
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Bing He
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Steve Bain
- Institute of Life Sciences, Swansea University, Swansea, U.K
| | - Finian Martin
- UCD Diabetes Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- School of Biomolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Research Centre, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Joel N. Hirschhorn
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Endocrine Research Unit, Department of Endocrinology, Children’s Hospital, Boston, Massachusetts
| | - Alexander P. Maxwell
- Nephrology Research, Centre for Public Health, Queen’s University of Belfast, Belfast, U.K
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Jose C. Florez
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Corresponding author: Jose C. Florez,
| | | |
Collapse
|
44
|
Weil EJ, Lemley KV, Mason CC, Yee B, Jones LI, Blouch K, Lovato T, Richardson M, Myers BD, Nelson RG. Podocyte detachment and reduced glomerular capillary endothelial fenestration promote kidney disease in type 2 diabetic nephropathy. Kidney Int 2012; 82:1010-7. [PMID: 22718189 PMCID: PMC3472108 DOI: 10.1038/ki.2012.234] [Citation(s) in RCA: 216] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/15/2023]
Abstract
Podocyte detachment and reduced endothelial cell fenestration and relationships between these features and the classic structural changes of diabetic nephropathy have not been described in patients with type 2 diabetes. Here we studied these relationships in 37 Pima Indians with type 2 diabetes of whom 11 had normal albuminuria, 16 had microalbuminuria, and 10 had macroalbuminuria. Biopsies from 10 kidney donors (not American Indians) showed almost undetectable (0.03%) podocyte detachment and 43.5% endothelial cell fenestration. In patients with type 2 diabetes, by comparison, the mean percentage of podocyte detachment was significantly higher in macroalbuminuria (1.48%) than in normal albuminuria (0.41%) or microalbuminuria (0.37%). Podocyte detachment correlated significantly with podocyte number per glomerulus and albuminuria. The mean percentage of endothelial cell fenestration was significantly lower in macroalbuminuria (19.3%) than in normal albuminuria (27.4%) or microalbuminuria (27.2%) and correlated significantly with glomerular basement membrane thickness, albuminuria, fractional mesangial area, and the glomerular filtration rate (iothalamate clearance). Podocyte detachment and diminished endothelial cell fenestration were not correlated, but were related to classic lesions of diabetic nephropathy. Thus, our findings confirm the important role these injuries play in the development and progression of kidney disease in type 2 diabetes, just as they do in type 1 diabetes. Whether podocyte detachment creates conduits for proteins to escape the glomerular circulation and reduced endothelial fenestration lowers glomerular hydraulic permeability requires further study.
Collapse
Affiliation(s)
- E Jennifer Weil
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona 85014-4972, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pyram R, Kansara A, Banerji MA, Loney-Hutchinson L. Chronic kidney disease and diabetes. Maturitas 2012; 71:94-103. [DOI: 10.1016/j.maturitas.2011.11.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/28/2011] [Revised: 11/09/2011] [Accepted: 11/09/2011] [Indexed: 12/15/2022]
|
46
|
Berhane AM, Weil EJ, Knowler WC, Nelson RG, Hanson RL. Albuminuria and estimated glomerular filtration rate as predictors of diabetic end-stage renal disease and death. Clin J Am Soc Nephrol 2011; 6:2444-51. [PMID: 21852671 DOI: 10.2215/cjn.00580111] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES We investigated predictive value of albuminuria and estimated GFR (eGFR) for ESRD in Pima Indians with type 2 diabetes. DESIGN, SETTING, PARTICIPANTS AND MEASUREMENTS Beginning in 1982, 2420 diabetic Pima Indians ≥18 years old were followed until they developed ESRD or died or until December 31, 2005. Individuals were classified at baseline by urinary albumin-to-creatinine ratio (ACR) and by eGFR, calculated by the Chronic Kidney Disease Epidemiology Collaboration equation. Predictors of ESRD and mortality were examined by proportional hazards regression. RESULTS During a mean follow-up of 10.2 years, 287 individuals developed ESRD. Incidence of ESRD among individuals with macroalbuminuria (ACR ≥ 300 mg/g) was 9.3 times that of those with normoalbuminuria (ACR < 30 mg/g), controlled for age, gender, and duration of diabetes. Incidence among individuals with eGFR 15 to 29 ml/min per 1.73 m(2) was 81.9 times that of those with eGFR 90 to 119 ml/min per 1.73 m(2). Models that combined albuminuria and eGFR added significant predictive information about risk of ESRD or death compared with models containing eGFR or albuminuria alone. The hazard ratio for ESRD associated with a 10-ml/min per 1.73 m(2) lower eGFR was 1.36, whereas that associated with an increase in albuminuria category was 2.69; corresponding hazard ratios for death were 1.15 and 1.37. CONCLUSIONS These results suggest that incorporation of quantitative information about albuminuria into staging systems based on eGFR adds significant prognostic information about risk for diabetic ESRD and death.
Collapse
Affiliation(s)
- Abeba M Berhane
- Diabetes Epidemiology and Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, Arizona, USA
| | | | | | | | | |
Collapse
|
47
|
Chakkera HA, Hanson RL, Kobes S, Millis MP, Nelson RG, Knowler WC, Distefano JK. Association of variants in the carnosine peptidase 1 gene (CNDP1) with diabetic nephropathy in American Indians. Mol Genet Metab 2011; 103:185-90. [PMID: 21393041 PMCID: PMC3101283 DOI: 10.1016/j.ymgme.2011.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 02/15/2011] [Accepted: 02/15/2011] [Indexed: 01/11/2023]
Abstract
CNDP1 is located on 18q22.3, where linkage with diabetic nephropathy has been observed in several populations, including Pima Indians. However, evidence for association between CNDP1 alleles and diabetic nephropathy is equivocal and population-dependent. This study investigated CNDP1 as a candidate for diabetic kidney disease in Pima Indians. Nineteen tag single nucleotide polymorphisms spanning the CNDP1 locus were selected using genotype data from Chinese individuals in the HapMap resource along with 2 variants previously associated with diabetic nephropathy. All variants were genotyped in 3 different samples including a diabetic end-stage renal disease (ESRD) case-control study, a family-based study of diabetic individuals who participated in the linkage study for nephropathy, and a cohort of diabetic individuals in whom longitudinal measures of glomerular filtration rates (GFR) were performed. There was no statistically significant evidence for association with diabetic ESRD. However, nominal evidence for association was found in the family study, where markers rs12957330 (Odds ratio [OR]=0.29 per copy of G allele; p=0.04) and rs17817077 (OR=0.46 per copy of G allele; p=0.05) were associated with diabetic nephropathy. In addition, markers rs12964454, rs7244647, and rs7229005 were associated with changes in GFR (-8.5ml/min per copy of the G allele; p=0.04; 18.8ml/min per copy of the C allele; p=0.03; and -13.4ml/min per copy of the C allele; p=0.001, respectively). These findings provide nominal evidence supporting a role between CNDP1 variants and diabetic kidney disease.
Collapse
Affiliation(s)
- Harini A Chakkera
- Divisions of Nephrology and Transplantation, Mayo Clinic, Phoenix, AZ 85054, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Mooyaart AL, Valk EJJ, van Es LA, Bruijn JA, de Heer E, Freedman BI, Dekkers OM, Baelde HJ. Genetic associations in diabetic nephropathy: a meta-analysis. Diabetologia 2011; 54:544-53. [PMID: 21127830 PMCID: PMC3034040 DOI: 10.1007/s00125-010-1996-1] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/20/2010] [Accepted: 10/26/2010] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS This meta-analysis assessed the pooled effect of each genetic variant reproducibly associated with diabetic nephropathy. METHODS PubMed, EMBASE and Web of Science were searched for articles assessing the association between genes and diabetic nephropathy. All genetic variants statistically associated with diabetic nephropathy in an initial study, then independently reproduced in at least one additional study, were selected. Subsequently, all studies assessing these variants were included. The association between these variants and diabetic nephropathy (defined as macroalbuminuria/proteinuria or end-stage renal disease [ESRD]) was calculated at the allele level and the main measure of effect was a pooled odds ratio. Pre-specified subgroup analyses were performed, stratifying for type 1/type 2 diabetes mellitus, proteinuria/ESRD and ethnic group. RESULTS The literature search yielded 3,455 citations, of which 671 were genetic association studies investigating diabetic nephropathy. We identified 34 replicated genetic variants. Of these, 21 remained significantly associated with diabetic nephropathy in a random-effects meta-analysis. These variants were in or near the following genes: ACE, AKR1B1 (two variants), APOC1, APOE, EPO, NOS3 (two variants), HSPG2, VEGFA, FRMD3 (two variants), CARS (two variants), UNC13B, CPVL and CHN2, and GREM1, plus four variants not near genes. The odds ratios of associated genetic variants ranged from 0.48 to 1.70. Additional variants were detected in subgroup analyses: ELMO1 (Asians), CCR5 (Asians) and CNDP1 (type 2 diabetes). CONCLUSIONS/INTERPRETATION This meta-analysis found 24 genetic variants associated with diabetic nephropathy. The relative contribution and relevance of the identified genes in the pathogenesis of diabetic nephropathy should be the focus of future studies.
Collapse
Affiliation(s)
- A L Mooyaart
- Department of Pathology, Bldg.1, L1-Q, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Gong Q, Gregg EW, Wang J, An Y, Zhang P, Yang W, Li H, Li H, Jiang Y, Shuai Y, Zhang B, Zhang J, Gerzoff RB, Roglic G, Hu Y, Li G, Bennett PH. Long-term effects of a randomised trial of a 6-year lifestyle intervention in impaired glucose tolerance on diabetes-related microvascular complications: the China Da Qing Diabetes Prevention Outcome Study. Diabetologia 2011; 54:300-7. [PMID: 21046360 DOI: 10.1007/s00125-010-1948-9] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/24/2010] [Accepted: 09/20/2010] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS We determined the effects of 6 years of lifestyle intervention in persons with impaired glucose tolerance (IGT) on the development of retinopathy, nephropathy and neuropathy over a 20 year period. METHODS In 1986, 577 adults with IGT from 33 clinics in Da Qing, China were randomly assigned by clinic to a control group or one of three lifestyle intervention groups (diet, exercise, and diet plus exercise). Active intervention was carried out from 1986 to 1992. In 2006 we conducted a 20 year follow-up study of the original participants to compare the incidence of microvascular complications in the combined intervention group vs the control group. RESULTS Follow-up information was obtained on 542 (94%) of the 577 original participants. The cumulative incidence of severe retinopathy was 9.2% in the combined intervention group and 16.2% in the control group (p = 0.03, log-rank test). After adjusting for clinic and age, the incidence of severe retinopathy was 47% lower in the intervention group than the control group (hazard rate ratio 0.53, 95% CI 0.29-0.99, p = 0.048). No significant differences were found in the incidence of severe nephropathy (hazard rate ratio 1.05, 95% CI 0.16-7.05, intervention vs control, p = 0.96) or in the prevalence of neuropathy (8.6% vs 9.1%, p = 0.89) among the 20 year survivors. CONCLUSIONS/INTERPRETATION Lifestyle intervention for 6 years in IGT was associated with a 47% reduction in the incidence of severe, vision-threatening retinopathy over a 20 year interval, primarily due to the reduced incidence of diabetes in the intervention group. However, similar benefits were not seen for nephropathy or neuropathy.
Collapse
Affiliation(s)
- Q Gong
- Department of Endocrinology, China-Japan Friendship Hospital, Yinghua Dong Lu, Chaoyang District, Beijing, 100029, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hanson RL, Millis MP, Young NJ, Kobes S, Nelson RG, Knowler WC, DiStefano JK. ELMO1 variants and susceptibility to diabetic nephropathy in American Indians. Mol Genet Metab 2010; 101:383-90. [PMID: 20826100 PMCID: PMC6542634 DOI: 10.1016/j.ymgme.2010.08.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 07/06/2010] [Revised: 08/12/2010] [Accepted: 08/12/2010] [Indexed: 11/27/2022]
Abstract
Variants in the engulfment and cell motility 1 gene, ELMO1, have previously been associated with kidney disease attributed to type 2 diabetes. The Pima Indians of Arizona have high rates of diabetic nephropathy, which is strongly dependent on genetic determinants; thus, we sought to investigate the role of ELMO1 polymorphisms in mediating susceptibility to this disease in this population. Genotype distributions were compared among 141 individuals with nephropathy and 416 individuals without heavy proteinuria in a family study of 257 sibships, and 107 cases with diabetic ESRD and 108 controls with long duration diabetes and no nephropathy. We sequenced 17.4 kb of ELMO1 and identified 19 variants. We genotyped 12 markers, excluding those in 100% genotypic concordance with other variants or with a minor allele frequency <0.05, plus 21 additional markers showing association with ESRD in earlier studies. In the family study, the strongest evidence for association was with rs1345365 (odds ratio [OR]=2.42 per copy of A allele [1.35-4.32]; P=0.001) and rs10951509 (OR=2.42 per copy of A allele [1.31-4.48]; P=0.002), both of which are located in intron 13 and are in strong pairwise linkage disequilibrium (r(2)=0.97). These associations were in the opposite direction from those observed in African Americans, which suggests that the relationship between diabetic kidney disease and ELMO1 variation may involve as yet undiscovered functional variants or complex interactions with other biological variables.
Collapse
Affiliation(s)
- Robert L. Hanson
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 1550 East Indian School Road, Phoenix, AZ 85014
| | - Meredith P. Millis
- Translational Genomics Research Institute, Diabetes, Cardiovascular and Metabolic Diseases Division, 445 North Fifth Street, Phoenix, AZ 85004
| | - Naomi J. Young
- Translational Genomics Research Institute, Diabetes, Cardiovascular and Metabolic Diseases Division, 445 North Fifth Street, Phoenix, AZ 85004
| | - Sayuko Kobes
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 1550 East Indian School Road, Phoenix, AZ 85014
| | - Robert G. Nelson
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 1550 East Indian School Road, Phoenix, AZ 85014
| | - William C. Knowler
- Diabetes Epidemiology and Clinical Research Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 1550 East Indian School Road, Phoenix, AZ 85014
| | - Johanna K. DiStefano
- Translational Genomics Research Institute, Diabetes, Cardiovascular and Metabolic Diseases Division, 445 North Fifth Street, Phoenix, AZ 85004
- Corresponding author: Johanna K. DiStefano, Ph.D., Translational Genomics Research Institute, 445 North Fifth Street, Phoenix, AZ 85004, Tel: 602.343.8812, FAX: 602.343.8844,
| |
Collapse
|