1
|
Wang W, Li Y, Hu Y, Wang J, Zhang Y, Fan L, Dai H, Guo H, Ding X, Chen F. Population pharmacokinetics of valproic acid in children with epilepsy: Implications for dose tailoring when switching from oral syrup to sustained-release tablets. CPT Pharmacometrics Syst Pharmacol 2024; 13:1554-1569. [PMID: 38923247 PMCID: PMC11533106 DOI: 10.1002/psp4.13191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Significant pharmacokinetic (PK) differences exist between different forms of valproic acid (VPA), such as syrup and sustained-release (SR) tablets. This study aimed to develop a population pharmacokinetic (PopPK) model for VPA in children with epilepsy and offer dose adjustment recommendation for switching dosage forms as needed. The study collected 1411 VPA steady-state trough concentrations (Ctrough) from 617 children with epilepsy. Using NONMEM software, a PopPK model was developed, employing a stepwise approach to identify possible variables such as demographic information and concomitant medications. The final model underwent internal and external evaluation via graphical and statistical methods. Moreover, Monte Carlo simulations were used to generate a dose tailoring strategy for typical patients weighting 20-50 kg. As a result, the PK characteristics of VPA were described using a one-compartment model with first-order absorption. The absorption rate constant (ka) was set at 2.64 and 0.46 h-1 for syrup and SR tablets. Body weight and sex were identified as significant factors affecting VPA's pharmacokinetics. The final PopPK model demonstrated acceptable prediction performance and stability during internal and external evaluation. For children taking syrup, a daily dose of 25 mg/kg resulted in the highest probability of achieving the desired target Ctrough, while a dose of 20 mg/kg/day was appropriate for those taking SR tablets. In conclusion, we established a PopPK model for VPA in children with epilepsy to tailor VPA dosage when switching between syrup and SR tablets, aiming to improve plasma VPA concentrations fluctuations.
Collapse
Affiliation(s)
- Wei‐Jun Wang
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Yue Li
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Ya‐Hui Hu
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Jie Wang
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuan‐Yuan Zhang
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Lin Fan
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Hao‐Ran Dai
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Hong‐Li Guo
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Xuan‐Sheng Ding
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingChina
| | - Feng Chen
- Department of Pharmacy, Pharmaceutical Sciences Research CenterChildren's Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
2
|
Escamilla-Cabrera B, Luis-Lima S, Gallego-Valcarce E, Sánchez-Dorta NV, Negrín-Mena N, Díaz-Martín L, Cruz-Perera C, Hernández-Valles AM, González-Rinne F, Rodríguez-Gamboa MJ, Estupiñán-Torres S, Miquel-Rodríguez R, Cobo-Caso MÁ, Delgado-Mallén P, Fernández-Suárez G, González-Rinne A, Hernández-Barroso G, González-Delgado A, Torres-Ramírez A, Jiménez-Sosa A, Ortiz A, Gaspari F, Hernández-Marrero D, Porrini EL. The error of estimated GFR in predialysis care. Sci Rep 2024; 14:5219. [PMID: 38433228 PMCID: PMC10909958 DOI: 10.1038/s41598-024-55022-8] [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] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024] Open
Abstract
The error of estimated glomerular filtration rate (eGFR) and its consequences in predialysis are unknown. In this prospective multicentre study, 315 predialysis patients underwent measured GFR (mGFR) by the clearance of iohexol and eGFR by 52 formulas. Agreement between eGFR and mGFR was evaluated by concordance correlation coefficient (CCC), total deviation index (TDI) and coverage probability (CP). In a sub-analysis we assessed the impact of eGFR error on decision-making as (i) initiating dialysis, (ii) preparation for renal replacement therapy (RRT) and (iii) continuing clinical follow-up. For this sub-analysis, patients who started RRT due to clinical indications (uremia, fluid overload, etc.) were excluded. eGFR had scarce precision and accuracy in reflecting mGFR (average CCC 0.6, TDI 70% and cp 22%) both in creatinine- and cystatin-based formulas. Variations -larger than 10 ml/min- between mGFR and eGFR were frequent. The error of formulas would have suggested (a) premature preparation for RTT in 14% of stable patients evaluated by mGFR; (b) to continue clinical follow-up in 59% of subjects with indication for RTT preparation due to low GFRm and (c) to delay dialysis in all asymptomatic patients (n = 6) in whom RRT was indicated based on very low mGFR. The error of formulas in predialysis was frequent and large and may have consequences in clinical care.
Collapse
Affiliation(s)
- Beatriz Escamilla-Cabrera
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
- Facultad de Medicina, Universidad de La Laguna, La Laguna, Spain
| | - Sergio Luis-Lima
- Department of Laboratory Medicine, Complejo Hospitalario Universitario de Canarias, Tenerife, Spain
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | | | | | - Natalia Negrín-Mena
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | - Laura Díaz-Martín
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | - Coriolano Cruz-Perera
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | | | - Federico González-Rinne
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | | | - Sara Estupiñán-Torres
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
| | - Rosa Miquel-Rodríguez
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
| | | | | | | | - Ana González-Rinne
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
| | | | | | - Armando Torres-Ramírez
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
- Facultad de Medicina, Universidad de La Laguna, La Laguna, Spain
| | | | - Alberto Ortiz
- Faculty of Medicine, Universidad Autónoma de Madrid. IIS-Fundación Jiménez Díaz. RICORS, Madrid, Spain
| | - Flavio Gaspari
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain
| | - Domingo Hernández-Marrero
- Nephrology Department, Complejo Hospitalario Universitario de Canarias, La Laguna, Spain
- Instituto de Tecnologías Biomédicas (ITB), Faculty of Medicine, University of La Laguna, La Laguna, Spain
- Facultad de Medicina, Universidad de La Laguna, La Laguna, Spain
| | - Esteban Luis Porrini
- Laboratory of Renal Function (LFR), Faculty of Medicine, Complejo Hospitalario Universitario de Canarias, University of La Laguna, La Laguna, Spain.
- Instituto de Tecnologías Biomédicas (ITB), Faculty of Medicine, University of La Laguna, La Laguna, Spain.
| |
Collapse
|
3
|
Barker AJ, Berthusen A, Vigers T, Schafer M, Browne LP, Bjornstad P. Estimation of glomerular filtration rate in a pediatric population using non-contrast kidney phase contrast magnetic resonance imaging. Pediatr Nephrol 2023; 38:2877-2881. [PMID: 36459246 PMCID: PMC10235207 DOI: 10.1007/s00467-022-05832-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Glomerular filtration rate (GFR) is a key measure of kidney function but often inaccurately ascertained by serum creatinine and cystatin C in pediatrics. In this pilot trial, we evaluated the relationship between GFR calculated by using phase-contrast MRI (PC-MRI) biomarkers and GFR by 125I-iothalamate clearance in youth undergoing bone marrow transplantation (BMT). METHODS A total of twenty-one pediatric BMT candidates (8-21 years of age) were recruited for a research kidney PC-MRI. After completion of 125I-iothalamate clearance, same-day PC-MRI measurements were completed of the kidney circulation without a gadolinium-based contrast agent. MRI included a non-contrast balanced-SSFP-triggered angiography to position ECG-gated breath-held 2D PC-MRI flow measurements (1.2 × 1.2 × 6 mm3). A multivariate model of MRI biomarkers estimating GFR (GFR-MRI) was selected using the elastic net approach. RESULTS The GFR-MRI variables selected by elastic net included average heart rate during imaging (bpm), peak aorta flow below the kidney artery take-offs (ml/s), average kidney artery blood flow, average peak kidney vein blood flow, and average kidney vein blood flow (ml/s). The GFR-MRI model demonstrated strong agreement with GFR by 125I-iothalamate (R2 = 0.65), which was stronger than what was observed with eGFR by the full age spectrum and Chronic Kidney Disease in Children under 25 (CKiD U25) approaches. CONCLUSION In this pilot study, noninvasive GFR-MRI showed strong agreement with gold standard GFR in youth scheduled for BMT. Further work is needed to evaluate whether non-contrast GFR-MRI holds promise to become a superior alternative to eGFR and GFR by clearance techniques. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Alex J Barker
- Department of Radiology: Pediatric Radiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| | - Alexander Berthusen
- Department of Radiology: Pediatric Radiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Tim Vigers
- Department of Pediatrics: Endocrinology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics: Section of Biostatistics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michal Schafer
- Department of Pediatrics: Cardiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Lorna P Browne
- Department of Radiology: Pediatric Radiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Petter Bjornstad
- Department of Pediatrics: Endocrinology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Medicine: Renal Diseases and Hypertension, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
4
|
Shafiee G, Gharibzadeh S, Panahi N, Razi F, Arzaghi SM, Haghpanah V, Ostovar A, Raeisi A, Mahdavi-Hezareh A, Larijani B, Esfahani EN, Heshmat R. Management goal achievements of diabetes care in Iran: study profile and main findings of DiaCare survey. J Diabetes Metab Disord 2023; 22:355-366. [PMID: 37255823 PMCID: PMC10225398 DOI: 10.1007/s40200-022-01149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 10/12/2022] [Indexed: 06/01/2023]
Abstract
Aim This paper presented the methodology and main findings of a population-based survey to determine diabetes care status among type 2 diabetic subjects in Iran. The current study assessed treatment goal achievements in type 2 diabetics, diabetes care service utilization, prevalence of diabetes complications, and psychological effects of diabetes in a representative sample of Iranian population in urban and rural areas. Materials and Methods This nationwide study was conducted between 2018 and 2020 as the observational survey entitled "Diabetes Care (DiaCare)". We studied a representative sample of participants with type 2 diabetes, aged 35-75 years, living in urban and rural areas in all thirty- one provinces of Iran. Data were collected by an interviewer in a form of a questionnaire that includes demographic and socioeconomic status, family and drug history, lifestyle, and self-reported psychological status according to a Patient's Health Questionnaire (PHQ). Management goal achievements, diabetes care service utilization, diabetes complications and psychological effects of diabetes were also assessed. Physical measurements were measured based on standard protocol. Fasting blood glucose (FBG), HbA1c, lipid profile, and also urine albumin to creatinine ratio were obtained from all participants of the study. Results Overall, 13,334 people with type 2 diabetes in 31 provinces of Iran completed the survey (response rate: 99.6%). In total 13,321 participants, 6683(50.17%) women and 6638(49.83%) men were included in our analysis. Thirteen recruited patients refused after the consenting process and did not respond. The mean age (SD) of total participants was 54.86 ± 9.44 years and 71.50% were from the urban areas. 13.66% of diabetic patients had achieved the triple target of management [controlled HbA1c, blood pressure, and Low-Density Lipoprotein-Cholesterol (LDL-C)] in the whole country. While 28.74% of people had controlled HbA1c and 33.40% of them had controlled FBG. Diabetic subjects living in rural areas had less controlled HbA1c (23.93 vs. 29.48), controlled FBG (29.50 vs. 34.20) and controlled triple targets (10.45 vs. 14.32) than those living in urban areas. Diabetic neuropathy and diabetic foot were more common in women than men, while end-stage of renal disease (ESRD) was more common in men than women. Conclusions This population-based study provided representative information about diabetes care in Iran. The high prevalence of diabetes and low proportion of diabetes control in Iran implies that it is necessary to identify factors associated with poor treatment goal achievements. Besides, general improvements in management and care of diabetes are mandatory.
Collapse
Affiliation(s)
- Gita Shafiee
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, NO 10, Jalale-Al-Ahmad Ave, Chamran Highway, Tehran, Iran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Pasteur Institute of Iran, Tehran, Iran
| | - Nekoo Panahi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Shahrivar St., North Kargar Ave, Tehran, Iran
| | - Seyed Masoud Arzaghi
- Elderly Health Research Center, Endocrinology and Metabolism Population Science Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Biobank Research Infrastructure, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Raeisi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Mahdavi-Hezareh
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, NO 10, Jalale-Al-Ahmad Ave, Chamran Highway, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Shahrivar St., North Kargar Ave, Tehran, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, NO 10, Jalale-Al-Ahmad Ave, Chamran Highway, Tehran, Iran
| |
Collapse
|
5
|
Sauder KA, Glueck DH, Harrall KK, D'Agostino R, Dolan LM, Lane AD, Liese AD, Lustigova E, Malik FS, Marcovina S, Mayer‐Davis E, Mottl A, Pihoker C, Reynolds K, Shah AS, Urbina EM, Wagenknecht LE, Daniels SR, Dabelea D. Exploring Racial and Ethnic Differences in Arterial Stiffness Among Youth and Young Adults With Type 1 Diabetes. J Am Heart Assoc 2023; 12:e028529. [PMID: 36994741 PMCID: PMC10122883 DOI: 10.1161/jaha.122.028529] [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] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/06/2023] [Indexed: 03/31/2023]
Abstract
Background We examined arterial stiffness in individuals with type 1 diabetes, and explored whether differences between Hispanic, non-Hispanic Black (NHB), and non-Hispanic White (NHW) individuals were attributable to modifiable clinical and social factors. Methods and Results Participants (n=1162; 22% Hispanic, 18% NHB, and 60% NHW) completed 2 to 3 research visits from ≈10 months to ≈11 years post type 1 diabetes diagnosis (mean ages of ≈9 to ≈20 years, respectively) providing data on socioeconomic factors, type 1 diabetes characteristics, cardiovascular risk factors, health behaviors, quality of clinical care, and perception of clinical care. Arterial stiffness (carotid-femoral pulse wave velocity [PWV], m/s) was measured at ≈20 years of age. We analyzed differences in PWV by race and ethnicity, then explored the individual and combined impact of the clinical and social factors on these differences. PWV did not differ between Hispanic (adjusted mean 6.18 [SE 0.12]) and NHW (6.04 [0.11]) participants after adjustment for cardiovascular risks (P=0.06) and socioeconomic factors (P=0.12), or between Hispanic and NHB participants (6.36 [0.12]) after adjustment for all factors (P=0.08). PWV was higher in NHB versus NHW participants in all models (all P<0.001). Adjustment for modifiable factors reduced the difference in PWV by 15% for Hispanic versus NHW participants; by 25% for Hispanic versus NHB; and by 21% for NHB versus NHW. Conclusions Cardiovascular and socioeconomic factors explain one-quarter of the racial and ethnic differences in PWV of young people with type 1 diabetes, but NHB individuals still experienced greater PWV. Exploration of pervasive inequities potentially driving these persistent differences is needed.
Collapse
Affiliation(s)
- Katherine A. Sauder
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) CenterUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Deborah H. Glueck
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) CenterUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Kylie K. Harrall
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) CenterUniversity of Colorado Anschutz Medical CampusAuroraCO
| | - Ralph D'Agostino
- Biostatistics and Data SciencesWake Forest University School of MedicineWinston‐SalemNC
| | - Lawrence M. Dolan
- Pediatrics, Cincinnati Children’s Hospital Medical Center Department of Pediatrics & The University of CincinnatiCincinnatiOH
| | - Abbi D. Lane
- Exercise ScienceUniversity of South Carolina Arnold School of Public HealthColumbiaSC
| | - Angela D. Liese
- Epidemiology and BiostatisticsUniversity of South Carolina Arnold School of Public HealthColumbiaSC
| | - Eva Lustigova
- Research & EvaluationKaiser Permanente Southern CaliforniaPasadenaCA
| | | | | | | | - Amy Mottl
- MedicineUniversity of North Carolina at Chapel HillChapel HillNC
| | | | - Kristi Reynolds
- Research & EvaluationKaiser Permanente Southern CaliforniaPasadenaCA
| | - Amy S. Shah
- Pediatrics, Cincinnati Children’s Hospital Medical Center Department of Pediatrics & The University of CincinnatiCincinnatiOH
| | - Elaine M. Urbina
- Pediatrics, Cincinnati Children’s Hospital Medical Center Department of Pediatrics & The University of CincinnatiCincinnatiOH
| | | | - Stephen R. Daniels
- PediatricsPediatrics, University of Colorado Anschutz Medical CampusAuroraCO
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) CenterUniversity of Colorado Anschutz Medical CampusAuroraCO
| |
Collapse
|
6
|
Heerspink HJ, Apperloo E, Davies M, Dicker D, Kandler K, Rosenstock J, Sørrig R, Lawson J, Zeuthen N, Cherney D. Effects of Semaglutide on Albuminuria and Kidney Function in People With Overweight or Obesity With or Without Type 2 Diabetes: Exploratory Analysis From the STEP 1, 2, and 3 Trials. Diabetes Care 2023; 46:801-810. [PMID: 36801984 PMCID: PMC10090901 DOI: 10.2337/dc22-1889] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/11/2023] [Indexed: 02/19/2023]
Abstract
OBJECTIVE These post hoc analyses of the Semaglutide Treatment Effect in People with obesity (STEP) 1-3 trials (NCT03548935, NCT03552757, and NCT03611582) explored the effects of semaglutide (up to 2.4 mg) on kidney function. RESEARCH DESIGN AND METHODS STEP 1-3 included adults with overweight/obesity; STEP 2 patients also had type 2 diabetes. Participants received once-weekly subcutaneous semaglutide 1.0 mg (STEP 2 only), 2.4 mg, or placebo for 68 weeks, plus lifestyle intervention (STEP 1 and 2) or intensive behavioral therapy (STEP 3). Changes in urine albumin-to-creatinine ratio (UACR) and UACR status from baseline to week 68 were assessed for STEP 2. Changes in estimated glomerular filtration rate (eGFR) were assessed from pooled STEP 1-3 data. RESULTS In STEP 2, 1,205 (99.6% total cohort) patients had UACR data; geometric mean baseline UACR was 13.7, 12.5, and 13.2 mg/g with semaglutide 1.0 mg, 2.4 mg, and placebo, respectively. At week 68, UACR changes were -14.8% and -20.6% with semaglutide 1.0 mg and 2.4 mg, respectively, and +18.3% with placebo (between-group differences [95% CI] vs. placebo: -28.0% [-37.3, -17.3], P < 0.0001 for semaglutide 1.0 mg; -32.9% [-41.6, -23.0], P = 0.003 for semaglutide 2.4 mg). UACR status improved in greater proportions of patients with semaglutide 1.0 mg and 2.4 mg versus placebo (P = 0.0004 and P = 0.0014, respectively). In the pooled STEP 1-3 analyses, 3,379 participants had eGFR data; there was no difference between semaglutide 2.4 mg and placebo in eGFR trajectories at week 68. CONCLUSIONS Semaglutide improved UACR in adults with overweight/obesity and type 2 diabetes. In participants with normal kidney function, semaglutide did not have an effect on eGFR decline.
Collapse
Affiliation(s)
- Hiddo J.L. Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Ellen Apperloo
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Melanie Davies
- Diabetes Research Centre, University of Leicester, Leicester, U.K
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester, U.K
- NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Dror Dicker
- Internal Medicine D, Hasharon Hospital-Rabin Medical Center, Petach-Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | | | | | - Rasmus Sørrig
- Global Medical Affairs, Novo Nordisk A/S, Søborg, Denmark
| | - Jack Lawson
- Global Medical Affairs, Novo Nordisk A/S, Søborg, Denmark
| | - Niels Zeuthen
- Biostatistics Obesity & Metabolism, Novo Nordisk A/S, Søborg, Denmark
| | - David Cherney
- Division of Nephrology, Department of Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Li B, Zhao X, Xie W, Hong Z, Zhang Y. Integrative analyses of biomarkers and pathways for diabetic nephropathy. Front Genet 2023; 14:1128136. [PMID: 37113991 PMCID: PMC10127684 DOI: 10.3389/fgene.2023.1128136] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Background: Diabetic nephropathy (DN) is a widespread diabetic complication and a major cause of terminal kidney disease. There is no doubt that DN is a chronic disease that imposes substantial health and economic burdens on the world's populations. By now, several important and exciting advances have been made in research on etiopathogenesis. Therefore, the genetic mechanisms underlying these effects remain unknown. Methods: The GSE30122, GSE30528, and GSE30529 microarray datasets were downloaded from the Gene Expression Omnibus database (GEO). Analyses of differentially expressed genes (DEGs), enrichment of gene ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were performed. Protein-protein interaction (PPI) network construction was completed by the STRING database. Hub genes were identified by Cytoscape software, and common hub genes were identified by taking intersection sets. The diagnostic value of common hub genes was then predicted in the GSE30529 and GSE30528 datasets. Further analysis was carried out on the modules to identify transcription factors and miRNA networks. As well, a comparative toxicogenomics database was used to assess interactions between potential key genes and diseases associated upstream of DN. Results: Samples from 19 DNs and 50 normal controls were identified in the GSE30122 dataset. 86 upregulated genes and 34 downregulated genes (a total of 120 DEGs). GO analysis showed significant enrichment in humoral immune response, protein activation cascade, complement activation, extracellular matrix, glycosaminoglycan binding, and antigen binding. KEGG analysis showed significant enrichment in complement and coagulation cascades, phagosomes, the Rap1 signaling pathway, the PI3K-Akt signaling pathway, and infection. GSEA was mainly enriched in the TYROBP causal network, the inflammatory response pathway, chemokine receptor binding, the interferon signaling pathway, ECM receptor interaction, and the integrin 1 pathway. Meanwhile, mRNA-miRNA and mRNA-TF networks were constructed for common hub genes. Nine pivotal genes were identified by taking the intersection. After validating the expression differences and diagnostic values of the GSE30528 and GSE30529 datasets, eight pivotal genes (TYROBP, ITGB2, CD53, IL10RA, LAPTM5, CD48, C1QA, and IRF8) were finally identified as having diagnostic values. Conclusion: Pathway enrichment analysis scores provide insight into the genetic phenotype and may propose molecular mechanisms of DN. The target genes TYROBP, ITGB2, CD53, IL10RA, LAPTM5, CD48, C1QA, and IRF8 are promising new targets for DN. SPI1, HIF1A, STAT1, KLF5, RUNX1, MBD1, SP1, and WT1 may be involved in the regulatory mechanisms of DN development. Our study may provide a potential biomarker or therapeutic locus for the study of DN.
Collapse
Affiliation(s)
- Bo Li
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Xu Zhao
- Emergency and Critical Care Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Wanrun Xie
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Zhenzhen Hong
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yi Zhang
- Department of Endocrinology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
8
|
Li B, Ye S, Fan Y, Lin Y, Li S, Peng H, Diao H, Chen W. Identification of novel key genes and potential candidate small molecule drugs in diabetic kidney disease using comprehensive bioinformatics analysis. Front Genet 2022; 13:934555. [PMID: 36035169 PMCID: PMC9411649 DOI: 10.3389/fgene.2022.934555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: The currently established diagnostic and prognostic tools for diabetic kidney disease (DKD) have limitations, which demands the necessity to find new genes and pathways associated with diagnosis and treatment. Our study aims to reveal the gene expression alteration and discover critical genes involved in the development of DKD, thus providing novel diagnostic molecular markers and therapeutic targets. Materials and methods: The differences of infiltrating immune cells within kidney were compared between healthy living donors and DKD patients. Besides, differentially expressed genes (DEGs) within kidney from healthy living donor, early stage DKD and advanced stage DKD samples were detected. Furthermore, the weighted co-expressed network (WGCNA) and protein-protein interaction (PPI) network were constructed, followed by recognition of core hub genes and module analysis. Receiver operating characteristic (ROC) curve analysis was implemented to determine the diagnostic value of hub genes, correlation analysis was employed to explore the association between hub genes and infiltrating immune cells, and certain hub genes was validated by quantitative real-time PCR and immunohistochemistry staining in cultured tubule cells and diabetic mice kidney. Finally, the candidate small molecules as potential drugs to treat DKD were anticipated through utilizing virtual screening and molecular docking investigation. Results: Our study revealed significantly higher proportion of infiltrating immune cells within kidney from DKD patients via probing the immune landscape by single-cell transcriptomics. Besides, 126 commonly shared DEGs identified among three group samples were enriched in immune biological process. In addition, the ROC curve analysis demonstrated the strong diagnostic accuracy of recognized hub genes (NFKB1, DYRK2, ATAD2, YAP1, and CHD3) from PPI network. Correlation analysis further confirmed the positive association between these hub genes with infiltrating natural killer cells. More importantly, the mRNA transcripts and protein abundance of YAP1 were significantly higher in high glucose-treated renal tubule cells and diabetic mice kidney, and the small molecules exhibiting the best binding affinities with YAP1 were predicted and acquired. Conclusion: Our findings for the first time indicate that NFKB1, DYRK2, ATAD2, YAP1, and CHD3 might be potential novel biomarkers and therapeutic targets for DKD, providing insights into the molecular mechanisms underlying the pathogenesis of DKD.
Collapse
Affiliation(s)
- Bin Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Siyang Ye
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuting Fan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yi Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Suchun Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Huajing Peng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Hui Diao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
- *Correspondence: Wei Chen,
| |
Collapse
|
9
|
Sauder KA, Stafford JM, Ehrlich S, Lawrence JM, Liese AD, Marcovina S, Mottl AK, Pihoker C, Saydah S, Shah AS, D'Agostino RB, Dabelea D. Disparities in Hemoglobin A 1c Testing During the Transition to Adulthood and Association With Diabetes Outcomes in Youth-Onset Type 1 and Type 2 Diabetes: The SEARCH for Diabetes in Youth Study. Diabetes Care 2021; 44:dc202983. [PMID: 34376501 PMCID: PMC8929181 DOI: 10.2337/dc20-2983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/12/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To identify correlates of hemoglobin A1c (HbA1c) testing frequency and associations with HbA1c levels and microvascular complications in youth-onset diabetes. RESEARCH DESIGN AND METHODS The SEARCH for Diabetes in Youth study collected data from individuals diagnosed with diabetes before age 20 at 8 years (n=1,885 type 1, n=230 type 2) and 13 years (n=649 type 1, n = 84 type 2) diabetes duration. We identified correlates of reporting ≥3 HbA1c tests/year using logistic regression. We examined associations of HbA1c testing with HbA1c levels and microvascular complications (retinopathy, neuropathy, or nephropathy) using sequentially adjusted linear and logistic regression. RESULTS For type 1 diabetes, odds of reporting ≥3 HbA1c tests/year at 8 and 13 years diabetes duration decreased with older age at diagnosis (odds ratio [OR] 0.91 [95% CI 0.88-0.95]), longer duration of diabetes (OR 0.90 [0.82-0.99]), not having a personal doctor (OR 0.44 [0.30-0.65]), and lapses in health insurance (OR 0.51 [0.27-0.96]). HbA1c testing ≥3 times/year over time was associated with lower HbA1c levels (OR -0.36% [-0.65 to -0.06]) and lower odds of microvascular complications (OR 0.64 [0.43-0.97]) at 13 years duration, but associations were attenuated after adjustment for HbA1c testing correlates (OR -0.17 [-0.46 to 0.13] and 0.70 [0.46-1.07], respectively). For type 2 diabetes, not seeing an endocrinologist decreased the odds of reporting ≥3 HbA1c tests/year over time (OR 0.19 [0.06-0.63]), but HbA1c testing frequency was not associated with HbA1c levels or microvascular complications. CONCLUSIONS We observed disparities in HbA1c testing frequency predominately by health care-related factors, which were associated with diabetes outcomes in type 1 diabetes.
Collapse
Affiliation(s)
- Katherine A Sauder
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado, Aurora, CO
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO
| | - Jeanette M Stafford
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | - Shelley Ehrlich
- Cincinnati Children's Hospital Medical Center and The University of Cincinnati, Cincinnati, OH
| | - Jean M Lawrence
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Angela D Liese
- Department of Epidemiology and Biostatistics, University of South Carolina Arnold School of Public Health, Columbia, SC
| | - Santica Marcovina
- Northwest Lipid Research Laboratory, University of Washington, Seattle, WA
| | - Amy K Mottl
- Division of Nephrology and Hypertension, University of North Carolina School of Medicine, Chapel Hill, NC
| | | | - Sharon Saydah
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, GA
| | - Amy S Shah
- Cincinnati Children's Hospital Medical Center and The University of Cincinnati, Cincinnati, OH
| | - Ralph B D'Agostino
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC
| | | |
Collapse
|
10
|
Inusa BPD, Liguoro I, Tayo B, Booth C, Turner C, Dalton NR. Reliability of different estimated glomerular filtration rate as measures of renal function in children with sickle cell disease. Blood Cells Mol Dis 2021; 91:102590. [PMID: 34256313 DOI: 10.1016/j.bcmd.2021.102590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/12/2021] [Accepted: 06/24/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND There is no reliable marker for detecting early renal disease in early children with sickle cell disease (SCD). Estimation of glomerular filtration rate (eGFR) as derived from the height/plasma creatinine formula is dependent on the accuracy of the creatinine analytical method used. The aim of this study was to evaluate different equations for eGFR. METHODS Children aged 5-16 years recruited. mGFR was obtained using plasma disappearance of Inutest/Iohexol, serum creatinine (SCr) was measured either by standard laboratory method or by tandem mass spectrometry (MSMS). Estimated GFR was then calculated either by "Bedside Schwartz method" or by the full-age spectrum (FAS) equation. FINDINGS A total of 79 patients (mean age 9.8 ± 4.0 years). A revised eGFR constant was calculated for Schwartz equation from the slope of the plot of height/plasma creatinine versus mGFR. Mean values for mGFR (132.7 ± 32.1 ml/min/1.73m2) and eGFR methods compared: eGFR from standard SCr was significantly higher (144.2 ± 37.3 ml/min/1.73m2, p = 0.008). The MSMS eGFR showed the lowest SD (SD = 27.5), while both FAS eGFR and FAS-height eGFR showed the highest correlation coefficient (r = 0.67). INTERPRETATION eGFR calculation based on height and SCr determined with MSMS traceable creatinine is more reliable than Schwartz formula using jaffe/enzymatic methods in SCD children.
Collapse
Affiliation(s)
- Baba Psalm Duniya Inusa
- Paediatric Haematology, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK.
| | - Ilaria Liguoro
- Department of Medicine, Division of Pediatrics, University of Udine, Italy
| | - Bamidele Tayo
- Department of Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, USA
| | - Caroline Booth
- Department of Nephrology, Evelina London Children's Hospital, Guy's and St Thomas NHS Foundation Trust, United Kingdom
| | - Charles Turner
- WellChild Laboratory, Evelina London Children's Hospital, Guy's and St Thomas NHS Foundation Trust, United Kingdom
| | - Neil R Dalton
- WellChild Laboratory, Evelina London Children's Hospital, Guy's and St Thomas NHS Foundation Trust, United Kingdom
| |
Collapse
|
11
|
Mahwish UN, Pasha M, Heera B, Raju SB, Jahan P. Implication of podocin promoter variant haplotype in south Indian diabetic kidney patients. Meta Gene 2021. [DOI: 10.1016/j.mgene.2020.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
12
|
|
13
|
Zafari N, Lotfaliany M, O'Keefe GJ, Kishore K, Torkamani N, MacIsaac RJ, Churilov L, Ekinci EI. Performance of 4 Creatinine-based Equations in Assessing Glomerular Filtration Rate in Adults with Diabetes. J Clin Endocrinol Metab 2021; 106:e61-e73. [PMID: 33090207 DOI: 10.1210/clinem/dgaa722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/16/2020] [Indexed: 01/30/2023]
Abstract
AIMS To evaluate diagnostic performance of glomerular filtration rate (GFR) estimated by modification of diet in renal disease (MDRD), chronic kidney disease epidemiology collaboration (CKD-EPI), full age spectrum (FAS), and revised Lund-Malmö (r-LM) equations in adults with diabetes. METHODS Individuals were included in this cross-sectional study if they had at least 1 measurement of technetium-99m diethylenetriamine-pentaacetic acid (99mTc-DTPA) GFR (mGFR) and serum creatinine (1487 patients with 2703 measures). GFR calculated by estimation equations was compared with mGFR. Diagnostic performance was assessed using concordance correlation coefficient (CCC), bias, precision, accuracy, reduced major axis regression (RMAR), and Bland-Altman plot. Analysis was repeated in subgroups based on sex, diabetes type, Hemoglobin A1C, and GFR level. RESULTS Of all patients, 1189 (86%) had type 2 diabetes. Mean mGFR, MDRD, CKD-EPI, FAS, and revised Lund-Malmö eGFR were 66, 72, 74, 71, and 67 mL/min/1.73m2, respectively. Overall, the r-LM had the highest CCC (0.83), lowest bias (-1.4 mL/min/1.73 m2), highest precision (16.2 mL/min/1.73 m2), and highest accuracy (P10 = 39%). The RMAR (slope, intercept) in r-LM, FAS, MDRD, and CKD-EPI was 1.18, -13.35; 0.97, -2.9; 1, -6.4, and 1.04, -11.3, respectively. The Bland-Altman plot showed that r-LM had the lowest mean difference and the narrowest 95% limit of agreement (-1.0, 54.1 mL/min/1.73 m2), while mean difference was more than 5-fold higher in FAS, MDRD, and CKD-EPI (-5.2, -6.3, and -8.2, respectively). CONCLUSIONS In adults with diabetes the revised Lund-Malmö performs better than MDRD, CKD-EPI, and FAS in calculating point estimates of GFR.
Collapse
Affiliation(s)
- Neda Zafari
- Melbourne Medical School, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Mojtaba Lotfaliany
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Geelong, Victoria, Australia
| | - Graeme J O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, and University of Melbourne, Melbourne, Victoria, Australia
| | - Kartik Kishore
- Data Analytics Research and Evaluation (DARE) Centre, Austin Health and The University of Melbourne, Heidelberg, Victoria, Australia
| | - Niloufar Torkamani
- Melbourne Medical School, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
| | - Richard J MacIsaac
- Department of Endocrinology & Diabetes, St Vincent's Hospital Melbourne and University of Melbourne, Victoria, Australia
| | - Leonid Churilov
- Melbourne Medical School, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Elif I Ekinci
- Melbourne Medical School, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Sauriasari R, Safitri DD, Azmi NU. Current updates on protein as biomarkers for diabetic kidney disease: a systematic review. Ther Adv Endocrinol Metab 2021; 12:20420188211049612. [PMID: 34721837 PMCID: PMC8554552 DOI: 10.1177/20420188211049612] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 09/12/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In the past decade, researchers have been focused on discovering protein biomarkers for diabetic kidney disease. This paper aims to search for, analyze, and synthesize current updates regarding the development of these efforts. METHODS We systematically searched the ScienceDirect, SpringerLink, and PubMed databases for observational studies of protein biomarkers in patients with diabetes mellitus. We included studies published between January 2018 and April 2020, that were based on a population of patients with type-1 or type-2 diabetes mellitus aged ⩾18 years, with an observational design such as cross-sectional, case-control, or cohort studies. The dependent variable of the research results was in the form of protein biomarkers from urine, plasma, or serum. RESULTS Following the screening process, 20 research articles with available full text met the inclusion criteria. These could be categorized as glomerular biomarkers (ANGPTL4, beta-2 microglobulin, Smad1, and glypican-5); inflammatory biomarkers (MCP-1 and adiponectin); and tubular biomarkers (NGAL, VDBP, megalin, sKlotho, and KIM-1). The development of a panel of biomarkers showed more promising results than those for a single biomarker in diagnosing diabetic kidney disease. CONCLUSION All the biomarkers discussed in this review showed promising results for predicting diabetic kidney disease because they correlate with albuminuria, eGFR, or both. However, of the 11 protein biomarkers, none have prognostic value beyond albuminuria and eGFR.
Collapse
Affiliation(s)
| | | | - Nuriza Ulul Azmi
- Faculty of Pharmacy, Universitas Indonesia, Depok, 16424, Indonesia
| |
Collapse
|
15
|
Zafari N, Churilov L, Wong LYL, Lotfaliany M, Hachem M, Kiburg KV, Kong L, Torkamani N, Baxter H, MacIsaac RJ, Ekinci EI. Evaluation of the diagnostic performance of the creatinine-based Chronic Kidney Disease Epidemiology Collaboration equation in people with diabetes: A systematic review. Diabet Med 2021; 38:e14391. [PMID: 32810875 DOI: 10.1111/dme.14391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/27/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022]
Abstract
AIMS GFR estimated with the creatinine-based Chronic Kidney Disease Epidemiology Collaboration (CKD-EPICr ) equation is used to screen for diabetic kidney disease and assess its severity. We systematically reviewed the process and outcome of evaluating CKD-EPICr in estimating point GFR or GFR decline over time in adults with type 1 or type 2 diabetes. METHODS In this systematic review, MEDLINE, Embase and Cochrane Central Register of Controlled Trials were searched up to August 2019. Observational studies comparing CKD-EPICr with measured GFR (mGFR) in adults with diabetes were included. Studies on people with kidney transplant, non-diabetes related kidney disease, pregnancy, potential kidney donors, and those with critical or other systematic illnesses were excluded. Two independent reviewers extracted data from published papers and disagreements were resolved by consensus. Risk-of-bias was assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 tool. (PROSPERO registration number: CRD42018108776). RESULTS From the 2820 records identified, 29 studies (14 704 participants) were included. All studies were at risk of bias. Bias (eight different forms) ranged from -26 to 35 ml min-1 1.73 m-2 ; precision (five different forms) ranged between 9 and 63 ml min-1 1.73 m-2 ; accuracy (five different forms) ranged between 16% and 96%; the correlation coefficient between CKD-EPICr and mGFR (four different forms) ranged between 0.38 and 0.86; and the reduced major axis regression slope ranged between 0.8 and 1.8. CONCLUSIONS Qualitative synthesis of data suggested CKD-EPICr was inaccurate in estimating point GFR or GFR decline over time. Furthermore, a lack of consistency in the methods and processes of evaluating the diagnostic performance of CKD-EPICr limits reliable quantitative assessment. The equation needs to be improved in adults with diabetes.
Collapse
Affiliation(s)
- N Zafari
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - L Churilov
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - L Y-L Wong
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - M Lotfaliany
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - M Hachem
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - K V Kiburg
- Department of Endocrinology & Diabetes, St Vincent's Hospital Melbourne and The University of Melbourne, Melbourne, Victoria, Australia
| | - L Kong
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - N Torkamani
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Department of Endocrinology Austin Health, Heidelberg, Victoria, Australia
| | - H Baxter
- Austin Health Sciences Library, Austin Health, Heidelberg, Victoria, Australia
| | - R J MacIsaac
- Department of Endocrinology & Diabetes, St Vincent's Hospital Melbourne and The University of Melbourne, Melbourne, Victoria, Australia
| | - E I Ekinci
- Department of Medicine, Austin health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Department of Endocrinology Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
16
|
Alzahrani S, Zaitone SA, Said E, El-Sherbiny M, Ajwah S, Alsharif SY, Elsherbiny NM. Protective effect of isoliquiritigenin on experimental diabetic nephropathy in rats: Impact on Sirt-1/NFκB balance and NLRP3 expression. Int Immunopharmacol 2020; 87:106813. [PMID: 32707499 DOI: 10.1016/j.intimp.2020.106813] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/29/2022]
Abstract
The prevalence of diabetes mellitus (DM) drastically increases worldwide. Persistent hyperglycemia affects body microvasculature causing injuries to kidney producing diabetic nephropathy (DNE). Manifestation of these microvascular complications is associated with disturbed redox homeostasis. The current study evaluated the effect of isoliquiritigenin (ISLQ), a bioactive chalcone found in licorice which is known for its antioxidant effect, on diabetes-induced renal injury. DM was prompted in male rats by streptozotocin (STZ, 50 mg/kg, intraperitoneally). ISLQ was administrated by oral gavage for 8 weeks at a dose (20 mg/kg/day). Features of renal injury were observed in kidneys of diabetic rats including, albuminuria and deteriorated renal function. Renal dysfunction was associated with reduced sirtuin-1 (Sirt-1) expression, increased renal oxidative stress, nucleotide-binding domain and leucine-rich repeat containing protein-3 (NLRP3), nuclear factor-κB (NFκB) and inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Moreover, there was significant downregulation of anti-inflammatory cytokine interleukin-10 (IL-10), glomerular and tubular injury and collagen accumulation. ISLQ administration preserved renal function and architecture, restored Sirt1 and renal oxidant-antioxidant balance, dampened inflammation and attenuated collagen accumulation. It can be inferred that ISLQ possess a protective effect and could have a potential as a food supplement to halt development and progression of DNE.
Collapse
Affiliation(s)
- Sharifa Alzahrani
- Pharmacology Department, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Sherbiny
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt; Department of Anatomy, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Sadeem Ajwah
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | | | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
17
|
Sekercioglu N, Lovblom LE, Bjornstad P, Lovshin JA, Lytvyn Y, Boulet G, Farooqi MA, Orszag A, Lai V, Tse J, Cham L, Keenan HA, Brent MH, Paul N, Bril V, Perkins BA, Cherney DZI. Risk factors for diabetic kidney disease in adults with longstanding type 1 diabetes: results from the Canadian Study of Longevity in Diabetes. Ren Fail 2020; 41:427-433. [PMID: 31162987 PMCID: PMC6566893 DOI: 10.1080/0886022x.2019.1614057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objectives: Diabetic kidney disease (DKD) is an independent predictor of cardiovascular morbidity and mortality in type 1 diabetes (T1D). We aimed to explore clinical and biochemical factors, including the achievement of American Diabetes Association (ADA) recommended targets associated with DKD in people living with T1D for ≥50 years. Methods: This was a post hoc analysis of a cross-sectional study of 75 participants enrolled in the Canadian Study of Longevity in T1D. We explored diabetes-related complications, including neuropathy, retinopathy, cardiovascular disease, and DKD. Study participants were dichotomized based on the achievement of ADA recommended targets as the low-target group (achieving ≤4 targets, n = 31) and high-target group (achieving >4 targets, n = 44). The outcome of interest was DKD defined by estimated glomerular filtration rate (eGFR) values <60/mL/min/1.73 m2 and/or 24-h albumin excretion >30 mg. Multivariable logistic regression models were employed to estimate odds ratios (ORs) for DKD with 95% confidence intervals (CIs). Results: Of the 75 participants with prolonged T1D duration (45% male, mean age 66 years), 25 participants had DKD and 50 did not. There was no statistical difference between the high- and low-target groups in terms of age and body mass index. eGFR was significantly higher and the prevalence of diabetic retinopathy was significantly lower in the high-target group. Older age at diagnosis of T1D and lower frequency component to high-frequency component ratio increased the odds of having DKD. Conclusions: In adults with prolonged T1D duration, older age at diagnosis and lower heart rate variability may be associated with DKD.
Collapse
Affiliation(s)
- Nigar Sekercioglu
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada.,b Department of Health Research Methods, Evidence, and Impact , McMaster University , Hamilton , Canada
| | - Leif Erik Lovblom
- c Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto , Canada
| | - Petter Bjornstad
- d Department of Pediatrics, Division of Endocrinology and Department of Medicine, Division of Nephrology , University of Colorado School of Medicine , Aurora , CO , USA
| | - Julie A Lovshin
- e Department of Medicine, Division of Endocrinology and Metabolism , Sunnybrook Health Sciences Centre, University of Toronto , Toronto , Canada
| | - Yuliya Lytvyn
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada
| | - Geneviève Boulet
- c Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto , Canada
| | - Mohammed A Farooqi
- c Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto , Canada
| | - Andrej Orszag
- c Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto , Canada
| | - Vesta Lai
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada
| | - Josephine Tse
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada
| | - Leslie Cham
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada
| | - Hillary A Keenan
- f Research Division , Joslin Diabetes Center , Boston , MA , USA
| | - Michael H Brent
- g Department of Ophthalmology and Vision Sciences Faculty of Medicine , University of Toronto , Toronto , Canada
| | - Narinder Paul
- h Joint Department of Medical Imaging, University of Toronto, Toronto, Canada and Department of Medical Imaging , Western University , London , Canada
| | - Vera Bril
- i Department of Medicine, Division of Neurology , University Health Network, University of Toronto , Toronto , Canada
| | - Bruce A Perkins
- c Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital , Toronto , Canada.,j Department of Medicine, Division of Endocrinology and Metabolism , Mount Sinai Hospital, University of Toronto , Toronto , Canada
| | - David Z I Cherney
- a Department of Medicine, Division of Nephrology , University of Toronto , Toronto , Canada.,k Department of Physiology , University of Toronto , Toronto , Canada.,l Department of Physiology and Banting and Best Diabetes Centre , University of Toronto , Toronto , Canada
| |
Collapse
|
18
|
Abstract
Since 1957, over 70 equations based on creatinine and/or cystatin C levels have been developed to estimate glomerular filtration rate (GFR). However, whether these equations accurately reflect renal function is debated. In this Perspectives article, we discuss >70 studies that compared estimated GFR (eGFR) with measured GFR (mGFR), involving ~40,000 renal transplant recipients and patients with chronic kidney disease (CKD), type 2 diabetes mellitus or polycystic kidney disease. Their results show that eGFR often differed from mGFR by ±30% or more, that eGFR values incorrectly staged CKD in 30-60% of patients, and that eGFR and mGFR gave different rates of GFR decline. Errors were unpredictable, and comparable for equations based on creatinine and/or cystatin C. We argue, therefore, that the persistence of these errors (despite intensive research) suggests that the problem lies with using creatinine and/or cystatin C as markers of renal function, rather than with the mathematical methods used for GFR estimation.
Collapse
|
19
|
Zafari N, Churilov L, MacIsaac RJ, Torkamani N, Baxter H, Kiburg KV, Ekinci E. Diagnostic performance of the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation at estimating glomerular filtration rate in adults with diabetes mellitus: a systematic review and meta-analysis protocol. BMJ Open 2019; 9:e031558. [PMID: 31473625 PMCID: PMC6720477 DOI: 10.1136/bmjopen-2019-031558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Timely detection leading to the implementation of reno-protective measures reduces the progression of diabetic kidney disease. Estimated glomerular filtration rate (eGFR) is a major surrogate of kidney function. The Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) Equation is a tool to estimate GFR. This protocol outlines a systematic-review, assessing the diagnostic accuracy of the CKD-EPI equation in adults with diabetes. METHODS AND ANALYSIS MEDLINE, Embase, Cochrane Central Register of Controlled Trials and grey literature will be searched for publications in English, Farsi, Dutch and Chinese from 2009 (when CKD-EPI was first introduced) to January 2019. Bridging searches will be conducted to capture literature published from January 2019 until final review publication. The inclusion criteria will be (1) study participants with diabetes; (2) age ≥18 years; (3) creatinine-based CKD-EPI eGFR as index test; (4) measured GFR using the clearance/plasma disappearance of inulin, iohexol, iothalamate, diethylenetriamine-pentaacetic acid (DTPA) or chromium labelled ethylenediaminetetraacetic acid (Cr-EDTA) as reference test; (5) report of the diagnostic accuracy of the index test. Exclusion criteria will be participants with renal transplant, chronic use of corticosteroids, chronic inflammatory diseases, pregnancy, non-diabetes related kidney disease, thalassaemia, heart failure, pregnancy and potential kidney donors as well as critically ill patients. Screening, eligibility check, risk of bias assessment and data extraction will be carried out by two independent reviewers. Any discrepancies will be discussed, and third-party opinion will be sought. The risk of bias will be assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 tool. A quantitative synthesis of the aggregated-data will be used if the included studies are homogenous. ETHICS AND DISSEMINATION No ethics approval is required. The outcome will be published in a peer-reviewed journal. The results will help researchers and clinicians evaluate the diagnostic accuracy of the creatinine-based CKD-EPI eGFR in adults with diabetes. PROSPERO REGISTRATION NUMBER CRD42018108776.
Collapse
Affiliation(s)
- Neda Zafari
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Leonid Churilov
- Florey Institute of Neuroscience and Mental Health, Heidelberg,Melbourne, Victoria, Australia
| | - Richard J MacIsaac
- Department of Endocrinology & Diabetes, St Vincent's Hospital, The University of Melbourne, Fitzroy, Melbourne, Victoria, Australia
| | - Niloufar Torkamani
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | - Helen Baxter
- Austin Health Sciences Library, Austin Health, Heidelberg, Victoria, Australia
| | - Katerina V Kiburg
- Department of Endocrinology & Diabetes, St Vincent's Hospital, The University of Melbourne, Fitzroy, Melbourne, Victoria, Australia
| | - Elif Ekinci
- Department of Medicine, The University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Korolenko TA, Shintyapina AB, Pupyshev AB, Akopyan AA, Russkikh GS, Dikovskaya MA, Vavilin VA, Zavjalov EL, Tikhonova MA, Amstislavskaya TG. The regulatory role of cystatin C in autophagy and neurodegeneration. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autophagy is a dynamic cellular process involved in the turnover of proteins, protein complexes, and organelles through lysosomal degradation. It is particularly important in neurons, which do not have a proliferative option for cellular repair. Autophagy has been shown to be suppressed in the striatum of a transgenic mouse model of Parkinson’s disease. Cystatin C is one of the potent regulators of autophagy. Changes in the expression and secretion of cystatin C in the brain have been shown in amyotrophic lateral sclerosis, Alzheimer’s and Parkinson’s diseases, and in some animal models of neurodegeneration, thus proving a protective function of cystatin C. It has been suggested that cystatin C plays the primary role in amyloidogenesis and shows promise as a therapeutic agent for neurodegenerative diseases (Alzheimer’s and Parkinson’s diseases). Cystatin C colocalizes with the amyloid β-protein in the brain during Alzheimer’s disease. Controlled expression of a cystatin C peptide has been proposed as a new approach to therapy for Alzheimer’s disease. In Parkinson’s disease, serum cystatin C levels can predict disease severity and cognitive dysfunction, although the exact involvement of cystatin C remains unclear. The aim: to study the role of cystatin C in neurodegeneration and evaluate the results in relation to the mechanism of autophagy. In our study on humans, a higher concentration of cystatin C was noted in cerebrospinal fluid than in serum; much lower concentrations were observed in other biological fluids (intraocular fluid, bile, and sweat). In elderly persons (61–80 years old compared to practically healthy people at 40–60 years of age), we revealed increased cystatin C levels both in serum and intraocular fluid. In an experiment on C57Bl/6J mice, cystatin C concentration was significantly higher in brain tissue than in the liver and spleen: an indication of an important function of this cysteine protease inhibitor in the brain. Using a transgenic mouse model of Parkinson’s disease (5 months old), we demonstrated a significant increase in osmotic susceptibility of brain lysosomes, depending on autophagy, while in a murine model of Alzheimer’s disease, this parameter did not differ from that in the appropriate control.
Collapse
Affiliation(s)
- T. A. Korolenko
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. B. Shintyapina
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine
| | - A. B. Pupyshev
- Scientific Research Institute of Physiology and Basic Medicine
| | - A. A. Akopyan
- Scientific Research Institute of Physiology and Basic Medicine
| | - G. S. Russkikh
- Scientific Research Institute of Biochemistry, Federal Research Center for Basic and Translational Medicine
| | - M. A. Dikovskaya
- Scientific Research Institute of Physiology and Basic Medicine; S.N. Fedorov NMRC “MNTK “Eye Microsurgery”, Novosibirsk Branch
| | - V. A. Vavilin
- Scientific Research Institute of Molecular Biology and Biophysics, Federal Research Center for Basic and Translational Medicine; Institute of Cytology and Genetics, SB RAS
| | | | - M. A. Tikhonova
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| | - T. G. Amstislavskaya
- Scientific Research Institute of Physiology and Basic Medicine; Novosibirsk State University
| |
Collapse
|
21
|
Scarr D, Bjornstad P, Lovblom LE, Lovshin JA, Boulet G, Lytvyn Y, Farooqi MA, Lai V, Orszag A, Weisman A, Keenan HA, Brent MH, Paul N, Bril V, Cherney DZ, Perkins BA. Estimating GFR by Serum Creatinine, Cystatin C, and β2-Microglobulin in Older Adults: Results From the Canadian Study of Longevity in Type 1 Diabetes. Kidney Int Rep 2019; 4:786-796. [PMID: 31194091 PMCID: PMC6551543 DOI: 10.1016/j.ekir.2019.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Glomerular filtration rate (GFR) is routinely used for clinical assessment of kidney function. However, the accuracy of estimating equations in older adults is uncertain. METHODS In 66 adults with ≥50 years type 1 diabetes (T1D) duration and 73 nondiabetic controls from age/sex-matched subgroups (65 ± 8 years old and 77[55%] were women) we evaluated the performance of estimated GFR (eGFR) by creatinine (Modification of Diet and Renal Disease [MDRD], Chronic Kidney Disease-Epidemiology [CKD-EPI]cr), cystatin C (CKD-EPIcys, CKD-EPIcr-cys), and β2-microglobulin (β2M) compared with measured GFR by inulin clearance (mGFR). Performance was evaluated using metrics of bias (mean difference), precision (SD), and accuracy (proportion of eGFR that differed by >20% of mGFR). RESULTS Mean mGFR was 104 ± 18 ml/min per 1.73 m2 (range: 70-154 ml/min per 1.73 m2) and was not different between T1D and controls (103 ± 17 vs. 105 ± 19 ml/min per 1.73 m2, P = 0.39). All equations significantly underestimated mGFR (bias: -15 to -30 ml/min per 1.73 m2, P < 0.001 for all comparisons) except for β2M, which had bias of 1.9 ml/min per 1.73 m2 (P = 0.61). Bias was greatest in cystatin C-based equations. Precision was lowest for β2M (SD: 43.5 ml/min per 1.73 m2, P < 0.001 for each comparison). Accuracy was lowest for CKD-EPIcysC (69.1%, P < 0.001 for each comparison). Cystatin C-based equations demonstrated greater bias and lower accuracy in older age subgroups (<60, 60-69, ≥70 years). All equations demonstrated greater bias across higher ranges of mGFR (60-89, 90-119, ≥120 ml/min per 1.73 m2). Results were similar between T1D and controls except that β2M had lower performance in T1D. CONCLUSION Better estimates of GFR in older adults are needed for research and clinical practice, as this subgroup of the population has an amplified risk for the development of chronic kidney disease (CKD) that requires accurate GFR estimation methods.
Collapse
Affiliation(s)
- Daniel Scarr
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Petter Bjornstad
- Division of Endocrinology, Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
- Division of Nephrology, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Leif E. Lovblom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Julie A. Lovshin
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Genevieve Boulet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yuliya Lytvyn
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mohammed A. Farooqi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Vesta Lai
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Andrej Orszag
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Alanna Weisman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hillary A. Keenan
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Michael H. Brent
- Department of Ophthalmology and Vision Sciences, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Narinder Paul
- Joint Department of Medical Imaging, Division of Cardiothoracic Radiology, University Health Network, Toronto, Ontario, Canada
| | - Vera Bril
- The Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Krembil Neuroscience Centre, Division of Neurology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Z.I. Cherney
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bruce A. Perkins
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Sartore G, Chilelli NC, Seraglia R, Ragazzi E, Marin R, Roverso M, Cosma C, Vaccaro O, Burlina S, Lapolla A. Long-term effect of pioglitazone vs glimepiride on lipoprotein oxidation in patients with type 2 diabetes: a prospective randomized study. Acta Diabetol 2019; 56:505-513. [PMID: 30740640 DOI: 10.1007/s00592-018-01278-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/14/2018] [Indexed: 01/07/2023]
Abstract
AIMS Type 2 diabetes (DM2) is associated to oxidative modifications of high-density lipoproteins (HDL), which can interfere with their function. Pioglitazone has proved effective in raising HDL cholesterol (HDL-C) and lowering small dense low-density lipoprotein (LDL), but no clinical studies have examined its effect on lipoprotein oxidation in patients with DM2. METHODS We assessed the effect of pioglitazone vs glimepiride after 1 year on HDL oxidation, expressed as relative abundance of peptides containing Met112O in ApoA-I (oxApoA-I) estimated by mass spectrometry (MALDI/TOF/TOF), in 95 patients with DM2. The oxLDL and AGE were quantified by ELISA. RESULTS Patients receiving pioglitazone showed a significant increase in the concentration of ApoA-I (Δ = 7.2 ± 14.8 mg/dL, p < 0.02) and a reduction in oxApoA-I (Δ = - 1.0 ± 2.6%, p < 0.02); this reduction was not significantly different from glimepiride. oxLDL showed a slight, but not significant increase in both treatment groups. Regression analysis showed a correlation between ΔoxApoA-I and ΔAGE (r = 0.30; p = 0.007) in all patients, while both of these parameters were unrelated to changes in HbA1c, HDL-C, duration of illness, or use of statins. CONCLUSIONS Long-term treatment with pioglitazone was effective in reducing the oxidation of HDL, but not LDL in patients with DM2, while glimepiride didn't. This finding seems to be associated to the change of glyco-oxidation status, not to any improvement in glycemic control or lipid profile. TRIAL REGISTRATION NCT00700856, ClinicalTrials.gov Registered June 18, 2008.
Collapse
Affiliation(s)
- Giovanni Sartore
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Nino Cristiano Chilelli
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy.
| | - Roberta Seraglia
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Raffaella Marin
- Lipid Laboratory, Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - Marco Roverso
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University of Padova, Padua, Italy
| | - Olga Vaccaro
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Silvia Burlina
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Annunziata Lapolla
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| |
Collapse
|
23
|
Sauder KA, Stafford JM, Mayer-Davis EJ, Jensen ET, Saydah S, Mottl A, Dolan LM, Hamman RF, Lawrence JM, Pihoker C, Marcovina S, D'Agostino RB, Dabelea D. Co-occurrence of early diabetes-related complications in adolescents and young adults with type 1 diabetes: an observational cohort study. THE LANCET. CHILD & ADOLESCENT HEALTH 2019; 3:35-43. [PMID: 30409691 PMCID: PMC6295346 DOI: 10.1016/s2352-4642(18)30309-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/14/2018] [Accepted: 09/17/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND One in three adolescents and young adults with type 1 diabetes have at least one early diabetes-related complication or comorbidity. We aimed to examine the prevalence and pattern of co-occurring complications in this population, as well as the related risk factors. METHODS This observational cohort study includes data from individuals diagnosed with type 1 diabetes before age 20 years who participated in the SEARCH for Diabetes in Youth Study across five sites in the USA. We assessed sociodemographic and metabolic risk factors at baseline and at follow-up, and diabetes complications at follow-up. A frequency analysis was done to examine the difference in observed versus expected prevalence (calculated using a contingency table assuming independence across cells) of co-occurring complications or comorbidities. A cluster analysis was done to identify unique clusters of participants based on demographic characteristics and metabolic risk factors. FINDINGS 1327 participants who completed the follow-up visit were included in the frequency analysis. The mean age was 10·1 (SD 3·9) years at the time of type 1 diabetes diagnosis and 18·0 (4·1) years at follow-up. At a mean diabetes duration of 7·8 [SD 1·9] years, co-occurrence of any two or more complications was observed in 78 (5·9%) participants, more frequently than expected by chance alone (58 [4·4%], p=0·015). Specifically, the complications that co-occurred more frequently than expected were retinopathy and diabetic kidney disease (11 [0·8%] vs three [0·2%]; p=0·0007), retinopathy and arterial stiffness (13 [1·0%] vs four [0·3%]; p=0·0016), and arterial stiffness and cardiovascular autonomic neuropathy (24 [1·8%] vs 13 [1·0%]; p=0·015). We identified four unique clusters characterised by progressively worsening metabolic risk factor profiles (longer duration of diabetes and higher glycated haemoglobin, non-HDL cholesterol, and waist-to-height ratio). The prevalence of at least two complications increased across the clusters (six [2·3%] of 261 in the low-risk cluster, 32 [6·3%] of 509 in the moderate-risk cluster, 28 [8%] of 348 in the high-risk cluster, and five [20·8%] of 24 in the highest-risk cluster). Compared with the low-risk and moderate-risk clusters, the high-risk and highest-risk clusters were characterised by a lower proportion of participants who were non-Hispanic white, and a higher proportion of participants who had a household income below US$50 000 and did not have private health insurance. INTERPRETATION Early complications co-occur in adolescents and young adults with type 1 diabetes more frequently than expected. Identification of individuals with adverse risk factors could enable targeted behavioural or medical interventions that reduce the likelihood of early development of lifelong diabetes-related morbidity. FUNDING US Centers for Disease Control and Prevention, US National Institutes of Health.
Collapse
Affiliation(s)
- Katherine A Sauder
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Jeanette M Stafford
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sharon Saydah
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Amy Mottl
- Division of Nephrology and Hypertension, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Lawrence M Dolan
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Richard F Hamman
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Jean M Lawrence
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Catherine Pihoker
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Santica Marcovina
- Northwest Lipid Research Laboratory, University of Washington, Seattle, WA, USA
| | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dana Dabelea
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA; Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| |
Collapse
|
24
|
Marcovecchio ML, Chiesa ST, Armitage J, Daneman D, Donaghue KC, Jones TW, Mahmud FH, Marshall SM, Neil HAW, Dalton RN, Deanfield J, Dunger DB, Acerini C, Ackland F, Anand B, Barrett T, Birrell V, Campbell F, Charakida M, Cheetham T, Chiesa S, Cooper C, Doughty I, Dutta A, Edge J, Gray A, Hamilton-Shield J, Mann N, Marcovecchio ML, Rayman G, Robinson JM, Russell-Taylor M, Sankar V, Smith A, Thalange N, Yaliwal C, Benitez-Aguirre P, Cameron F, Cotterill A, Couper J, Craig M, Davis E, Donaghue K, Jones TW, Verge C, Bergman P, Rodda C, Clarson C, Curtis J, Daneman D, Mahmud F, Sochett E, Marshall S, Armitage J, Bingley P, Van’t Hoff W, Dunger D, Dalton N, Daneman D, Neil A, Deanfield J, Jones T, Donaghue K, Baigent C, Emberson J, Flather M, Bilous R. Renal and Cardiovascular Risk According to Tertiles of Urinary Albumin-to-Creatinine Ratio: The Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT). Diabetes Care 2018; 41:1963-1969. [PMID: 30026334 DOI: 10.2337/dc18-1125] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/17/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Baseline data from the Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT) indicated that tertiles of urinary albumin-to-creatinine ratios (ACRs) in the normal range at age 10-16 years are associated with risk markers for diabetic nephropathy (DN) and cardiovascular disease (CVD). We aimed to determine whether the top ACR tertile remained associated with DN and CVD risk over the 2-4-year AdDIT study. RESEARCH DESIGN AND METHODS One hundred fifty adolescents (mean age 14.1 years [SD 1.6]) with baseline ACR in the upper tertile (high-ACR group) recruited to the AdDIT trial, who remained untreated, and 396 (age 14.3 years [1.6]) with ACR in the middle and lower tertiles (low-ACR group), who completed the parallel AdDIT observational study, were evaluated prospectively with assessments of ACR and renal and CVD markers, combined with carotid intima-media thickness (cIMT) at baseline and end of study. RESULTS After a median follow-up of 3.9 years, the cumulative incidence of microalbuminuria was 16.3% in the high-ACR versus 5.5% in the low-ACR group (log-rank P < 0.001). Cox models showed independent contributions of the high-ACR group (hazard ratio 4.29 [95% CI 2.08-8.85]) and HbA1c (1.37 [1.10-1.72]) to microalbuminuria risk. cIMT change from baseline was significantly greater in the high- versus low-ACR group (mean difference 0.010 mm [0.079], P = 0.006). Changes in estimated glomerular filtration rate, systolic blood pressure, and hs-CRP were also significantly greater in the high-ACR group (P < 0.05). CONCLUSIONS ACR at the higher end of the normal range at the age of 10-16 years is associated with an increased risk of progression to microalbuminuria and future CVD risk, independently of HbA1c.
Collapse
Affiliation(s)
| | - Scott T. Chiesa
- National Centre for Cardiovascular Prevention and Outcomes, University College London, London, U.K
| | - Jane Armitage
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Denis Daneman
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kim C. Donaghue
- Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead, University of Sydney, Camperdown, New South Wales, Australia
| | - Timothy W. Jones
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Farid H. Mahmud
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Sally M. Marshall
- Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - H. Andrew W. Neil
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, U.K
| | - R. Neil Dalton
- Guy’s and St Thomas’ National Health Service Foundation Trust, London, U.K
| | - John Deanfield
- National Centre for Cardiovascular Prevention and Outcomes, University College London, London, U.K
| | - David B. Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, U.K
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, U.K
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Edmonds DJ, Kung DW, Kalgutkar AS, Filipski KJ, Ebner DC, Cabral S, Smith AC, Aspnes GE, Bhattacharya SK, Borzilleri KA, Brown JA, Calabrese MF, Caspers NL, Cokorinos EC, Conn EL, Dowling MS, Eng H, Feng B, Fernando DP, Genung NE, Herr M, Kurumbail RG, Lavergne SY, Lee ECY, Li Q, Mathialagan S, Miller RA, Panteleev J, Polivkova J, Rajamohan F, Reyes AR, Salatto CT, Shavnya A, Thuma BA, Tu M, Ward J, Withka JM, Xiao J, Cameron KO. Optimization of Metabolic and Renal Clearance in a Series of Indole Acid Direct Activators of 5′-Adenosine Monophosphate-Activated Protein Kinase (AMPK). J Med Chem 2018; 61:2372-2383. [DOI: 10.1021/acs.jmedchem.7b01641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- David J. Edmonds
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Daniel W. Kung
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S. Kalgutkar
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Kevin J. Filipski
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - David C. Ebner
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Shawn Cabral
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Aaron C. Smith
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gary E. Aspnes
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Samit K. Bhattacharya
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Kris A. Borzilleri
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Janice A. Brown
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew F. Calabrese
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nicole L. Caspers
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Emily C. Cokorinos
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Edward L. Conn
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S. Dowling
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Heather Eng
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Bo Feng
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Dilinie P. Fernando
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Nathan E. Genung
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael Herr
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ravi G. Kurumbail
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sophie Y. Lavergne
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Esther C.-Y. Lee
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Qifang Li
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sumathy Mathialagan
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Russell A. Miller
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jane Panteleev
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jana Polivkova
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Francis Rajamohan
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Allan R. Reyes
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Christopher T. Salatto
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Andre Shavnya
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Benjamin A. Thuma
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Meihua Tu
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jessica Ward
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Jane M. Withka
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jun Xiao
- Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kimberly O. Cameron
- Pfizer Worldwide Research and Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
26
|
Jakhotia S, Sivaprasad M, Shalini T, Reddy PY, Viswanath K, Jakhotia K, Sahay R, Sahay M, Reddy GB. Circulating levels of Hsp27 in microvascular complications of diabetes: Prospects as a biomarker of diabetic nephropathy. J Diabetes Complications 2018; 32:221-225. [PMID: 29175119 DOI: 10.1016/j.jdiacomp.2017.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/26/2017] [Accepted: 10/04/2017] [Indexed: 10/18/2022]
Abstract
AIM Heat shock protein 27 (Hsp27) is a small heat shock protein known to protect the cells from apoptosis under stress. In the present study, we determined the plasma Hsp27 levels in type 2 diabetes subjects without and with microvascular complications- diabetic retinopathy (DRe), diabetic nephropathy (DNe), and diabetic neuropathy (DNu) to understand if it could serve as a marker for these complications. METHODS This is a hospital-based case-control study with 754 subjects including 247 controls, 195 subjects with diabetes, 123 with DRe, 80 with DNe and 109 with DNu. Plasma Hsp27 levels were measured by ELISA. RESULTS The mean plasma Hsp27 was higher in the DNe group (631.5±355.2) compared to the control (496.55±308.54), diabetes (523.41±371.01), DRe (494.60±391.48) and DNu (455.21±319.74) groups with a p-value of 0.018. Receiver operating characteristic (ROC) curve analysis of Hsp27 in DNe group showed an area under the curve (AUC) of 0.617. Spearman correlation analysis shows a positive correlation of plasma Hsp27 with serum creatinine (p=0.053, r-value 0.083). Gender, age and BMI did not affect the plasma Hsp27 levels. CONCLUSION The plasma Hsp27 levels in the DNe group are higher compared to the control and other complications, thereby it could be explored to be used as a potential biomarker of DNe.
Collapse
Affiliation(s)
- Sneha Jakhotia
- Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Tattari Shalini
- Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | | | | | - Rakesh Sahay
- Osmania Medical College and General Hospital, Hyderabad, India
| | - Manisha Sahay
- Osmania Medical College and General Hospital, Hyderabad, India
| | | |
Collapse
|
27
|
Lovshin JA, Boulet G, Lytvyn Y, Lovblom LE, Bjornstad P, Farooqi MA, Lai V, Cham L, Tse J, Orszag A, Scarr D, Weisman A, Keenan HA, Brent MH, Paul N, Bril V, Perkins BA, Cherney DZ. Renin-angiotensin-aldosterone system activation in long-standing type 1 diabetes. JCI Insight 2018; 3:96968. [PMID: 29321380 PMCID: PMC5821172 DOI: 10.1172/jci.insight.96968] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/28/2017] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND In type 1 diabetes (T1D), adjuvant treatment with inhibitors of the renin-angiotensin-aldosterone system (RAAS), which dilate the efferent arteriole, is associated with prevention of progressive albuminuria and renal dysfunction. Uncertainty still exists as to why some individuals with long-standing T1D develop diabetic kidney disease (DKD) while others do not (DKD resistors). We hypothesized that those with DKD would be distinguished from DKD resistors by the presence of RAAS activation. METHODS Renal and systemic hemodynamic function was measured before and after exogenous RAAS stimulation by intravenous infusion of angiotensin II (ANGII) in 75 patients with prolonged T1D durations and in equal numbers of nondiabetic controls. The primary outcome was change in renal vascular resistance (RVR) in response to RAAS stimulation, a measure of endogenous RAAS activation. RESULTS Those with DKD had less change in RVR following exogenous RAAS stimulation compared with DKD resistors or controls (19%, 29%, 31%, P = 0.008, DKD vs. DKD resistors), reflecting exaggerated endogenous renal RAAS activation. All T1D participants had similar changes in renal efferent arteroilar resistance (9% vs. 13%, P = 0.37) irrespective of DKD status, which reflected less change versus controls (20%, P = 0.03). In contrast, those with DKD exhibited comparatively less change in afferent arteriolar vascular resistance compared with DKD resistors or controls (33%, 48%, 48%, P = 0.031, DKD vs. DKD resistors), indicating higher endogenous RAAS activity. CONCLUSION In long-standing T1D, the intrarenal RAAS is exaggerated in DKD, which unexpectedly predominates at the afferent rather than the efferent arteriole, stimulating vasoconstriction. FUNDING JDRF operating grant 17-2013-312.
Collapse
Affiliation(s)
- Julie A. Lovshin
- Division of Endocrinology and Metabolism and
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Geneviève Boulet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yuliya Lytvyn
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Leif E. Lovblom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Petter Bjornstad
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Research Division, Barbara Davis Center for Diabetes, Aurora, Colorado, USA
| | - Mohammed A. Farooqi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Vesta Lai
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Leslie Cham
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Josephine Tse
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Andrej Orszag
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Daniel Scarr
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Alanna Weisman
- Division of Endocrinology and Metabolism and
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Hillary A. Keenan
- Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Michael H. Brent
- Department of Ophthalmology and Vision Sciences, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Narinder Paul
- Joint Department of Medical Imaging, Division of Cardiothoracic Radiology, University Health Network, Toronto, Ontario, Canada
| | - Vera Bril
- Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Krembil Neuroscience Centre, Division of Neurology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Bruce A. Perkins
- Division of Endocrinology and Metabolism and
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - David Z.I. Cherney
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Benitez-Aguirre PZ, Januszewski AS, Cho YH, Craig ME, Jenkins AJ, Donaghue KC. Early changes of arterial elasticity in Type 1 diabetes with microvascular complications - A cross-sectional study from childhood to adulthood. J Diabetes Complications 2017; 31:1674-1680. [PMID: 28941950 DOI: 10.1016/j.jdiacomp.2017.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 01/01/2023]
Abstract
AIM To examine the trajectory of small artery elasticity (SAE) and pulse pressure (PP) in people with Type 1 diabetes and non-diabetic controls across the lifespan, and explore associations with microvascular complications (CX+). METHODS This cross-sectional study included 477 Type 1 diabetes patients (188 with CX+, 289 without CX-) and 515 controls. Relationships between SAE and PP and age were evaluated using segmented linear regression. Logistic regression was used to assess the associations between microvascular complications (retinopathy and/or nephropathy) and SAE and PP. RESULTS SAE peaked significantly later among controls than diabetic patients CX- vs. CX+ (21.2 vs. 20.4 vs. 17.6 years respectively, p < 0.001). In adults, mean SAE was significantly lower in CX+ vs. CX- vs. controls (6.8 vs. 7.8 vs. 8.0 ml/mm Hg × 10; p < 0.0001), and mean PP was significantly higher in CX+ vs CX- and controls (60 vs. 55 vs. 53 mm Hg; p < 0.0001). CONCLUSION Type 1 diabetes CX+ subjects have an earlier peak and decline in SAE relative to CX- and controls, who did not differ. Lower SAE and higher PP were associated with increased odds of Type 1 diabetes complications in adults. These clinically applicable techniques demonstrate an association between accelerated vascular aging and vascular complications in diabetes.
Collapse
Affiliation(s)
- P Z Benitez-Aguirre
- Discipline of Paediatrics and Child Health, University of Sydney, Australia; Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia
| | - A S Januszewski
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Department of Medicine, University of Melbourne, Australia
| | - Y H Cho
- Discipline of Paediatrics and Child Health, University of Sydney, Australia; Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia
| | - M E Craig
- Discipline of Paediatrics and Child Health, University of Sydney, Australia; Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Australia
| | - A J Jenkins
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Department of Medicine, University of Melbourne, Australia
| | - K C Donaghue
- Discipline of Paediatrics and Child Health, University of Sydney, Australia; Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia.
| |
Collapse
|
29
|
Marcovecchio ML, Chiesa ST, Bond S, Daneman D, Dawson S, Donaghue KC, Jones TW, Mahmud FH, Marshall SM, Neil HAW, Dalton RN, Deanfield J, Dunger DB. ACE Inhibitors and Statins in Adolescents with Type 1 Diabetes. N Engl J Med 2017; 377:1733-1745. [PMID: 29091568 DOI: 10.1056/nejmoa1703518] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Among adolescents with type 1 diabetes, rapid increases in albumin excretion during puberty precede the development of microalbuminuria and macroalbuminuria, long-term risk factors for renal and cardiovascular disease. We hypothesized that adolescents with high levels of albumin excretion might benefit from angiotensin-converting-enzyme (ACE) inhibitors and statins, drugs that have not been fully evaluated in adolescents. METHODS We screened 4407 adolescents with type 1 diabetes between the ages of 10 and 16 years of age and identified 1287 with values in the upper third of the albumin-to-creatinine ratios; 443 were randomly assigned in a placebo-controlled trial of an ACE inhibitor and a statin with the use of a 2-by-2 factorial design minimizing differences in baseline characteristics such as age, sex, and duration of diabetes. The primary outcome for both interventions was the change in albumin excretion, assessed according to the albumin-to-creatinine ratio calculated from three early-morning urine samples obtained every 6 months over 2 to 4 years, and expressed as the area under the curve. Key secondary outcomes included the development of microalbuminuria, progression of retinopathy, changes in the glomerular filtration rate, lipid levels, and measures of cardiovascular risk (carotid intima-media thickness and levels of high-sensitivity C-reactive protein and asymmetric dimethylarginine). RESULTS The primary outcome was not affected by ACE inhibitor therapy, statin therapy, or the combination of the two. The use of an ACE inhibitor was associated with a lower incidence of microalbuminuria than the use of placebo; in the context of negative findings for the primary outcome and statistical analysis plan, this lower incidence was not considered significant (hazard ratio, 0.57; 95% confidence interval, 0.35 to 0.94). Statin use resulted in significant reductions in total, low-density lipoprotein, and non-high-density lipoprotein cholesterol levels, in triglyceride levels, and in the ratio of apolipoprotein B to apolipoprotein A1, whereas neither drug had significant effects on carotid intima-media thickness, other cardiovascular markers, the glomerular filtration rate, or progression of retinopathy. Overall adherence to the drug regimen was 75%, and serious adverse events were similar across the groups. CONCLUSIONS The use of an ACE inhibitor and a statin did not change the albumin-to-creatinine ratio over time. (Funded by the Juvenile Diabetes Research Foundation and others; AdDIT ClinicalTrials.gov number, NCT01581476 .).
Collapse
Affiliation(s)
- M Loredana Marcovecchio
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Scott T Chiesa
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Simon Bond
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Denis Daneman
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Sarah Dawson
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Kim C Donaghue
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Timothy W Jones
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Farid H Mahmud
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - Sally M Marshall
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - H Andrew W Neil
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - R Neil Dalton
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - John Deanfield
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| | - David B Dunger
- From the Department of Paediatrics (M.L.M., D.B.D.) and the Wellcome Trust-Medical Research Council Institute of Metabolic Science (D.B.D.), University of Cambridge, and the Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital (S.B., S.D.), Cambridge, the National Centre for Cardiovascular Prevention and Outcomes, University College London (S.T.C., J.D.), and the WellChild Laboratory, Evelina London Children's Hospital, St. Thomas' Hospital (R.N.D.), London, the Institute of Cellular Medicine (Diabetes), Faculty of Clinical Medical Sciences, Newcastle University, Newcastle upon Tyne (S.M.M.), and the Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford (H.A.W.N.) - all in the United Kingdom; the Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto (D.D., F.H.M.); and the Institute of Endocrinology and Diabetes, Children's Hospital at Westmead and University of Sydney, Sydney (K.C.D.), and the Telethon Kids Institute, University of Western Australia, Perth (T.W.J.) - both in Australia
| |
Collapse
|
30
|
Affiliation(s)
- Petter Bjornstad
- Department of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, Colorado
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, Colorado; and
- Department of Medicine, Division of Nephrology and
- Department of Physiology, University of Toronto, Ontario, Canada
| | - David Z.I. Cherney
- Department of Medicine, Division of Nephrology and
- Department of Physiology, University of Toronto, Ontario, Canada
| |
Collapse
|
31
|
Bevc S, Hojs N, Hojs R, Ekart R, Gorenjak M, Puklavec L. Estimation of Glomerular Filtration Rate in Elderly Chronic Kidney Disease Patients: Comparison of Three Novel Sophisticated Equations and Simple Cystatin C Equation. Ther Apher Dial 2017; 21:126-132. [PMID: 28296256 DOI: 10.1111/1744-9987.12523] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/07/2016] [Accepted: 12/06/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine; University Clinical Centre Maribor; Maribor Slovenia
- Faculty of Medicine; University of Maribor; Maribor Slovenia
| | - Nina Hojs
- Department of Nephrology, Clinic for Internal Medicine; University Clinical Centre Maribor; Maribor Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine; University Clinical Centre Maribor; Maribor Slovenia
- Faculty of Medicine; University of Maribor; Maribor Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine; University Clinical Centre Maribor; Maribor Slovenia
- Faculty of Medicine; University of Maribor; Maribor Slovenia
| | - Maksimiljan Gorenjak
- Department of Nuclear Medicine, Clinic for Internal Medicine; University Clinical Centre Maribor; Maribor Slovenia
| | - Ludvik Puklavec
- Department of Clinical Chemistry; University Clinical Centre Maribor; Maribor Slovenia
- Faculty of Medicine; University of Maribor; Maribor Slovenia
| |
Collapse
|
32
|
Dabelea D, Stafford JM, Mayer-Davis EJ, D'Agostino R, Dolan L, Imperatore G, Linder B, Lawrence JM, Marcovina SM, Mottl AK, Black MH, Pop-Busui R, Saydah S, Hamman RF, Pihoker C. Association of Type 1 Diabetes vs Type 2 Diabetes Diagnosed During Childhood and Adolescence With Complications During Teenage Years and Young Adulthood. JAMA 2017; 317:825-835. [PMID: 28245334 PMCID: PMC5483855 DOI: 10.1001/jama.2017.0686] [Citation(s) in RCA: 419] [Impact Index Per Article: 59.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance The burden and determinants of complications and comorbidities in contemporary youth-onset diabetes are unknown. Objective To determine the prevalence of and risk factors for complications related to type 1 diabetes vs type 2 diabetes among teenagers and young adults who had been diagnosed with diabetes during childhood and adolescence. Design, Setting, and Participants Observational study from 2002 to 2015 in 5 US locations, including 2018 participants with type 1 and type 2 diabetes diagnosed at younger than 20 years, with single outcome measures between 2011 and 2015. Exposures Type 1 and type 2 diabetes and established risk factors (hemoglobin A1c level, body mass index, waist-height ratio, and mean arterial blood pressure). Main Outcomes and Measures Diabetic kidney disease, retinopathy, peripheral neuropathy, cardiovascular autonomic neuropathy, arterial stiffness, and hypertension. Results Of 2018 participants, 1746 had type 1 diabetes (mean age, 17.9 years [SD, 4.1]; 1327 non-Hispanic white [76.0%]; 867 female patients [49.7%]), and 272 had type 2 (mean age, 22.1 years [SD, 3.5]; 72 non-Hispanic white [26.5%]; 181 female patients [66.5%]). Mean diabetes duration was 7.9 years (both groups). Patients with type 2 diabetes vs those with type 1 had higher age-adjusted prevalence of diabetic kidney disease (19.9% vs 5.8%; absolute difference [AD], 14.0%; 95% CI, 9.1%-19.9%; P < .001), retinopathy (9.1% vs 5.6%; AD, 3.5%; 95% CI, 0.4%-7.7%; P = .02), peripheral neuropathy (17.7% vs 8.5%; AD, 9.2%; 95% CI, 4.8%-14.4%; P < .001), arterial stiffness (47.4% vs 11.6%; AD, 35.9%; 95% CI, 29%-42.9%; P < .001), and hypertension (21.6% vs 10.1%; AD, 11.5%; 95% CI, 6.8%-16.9%; P < .001), but not cardiovascular autonomic neuropathy (15.7% vs 14.4%; AD, 1.2%; 95% CI, -3.1% to 6.5; P = .62). After adjustment for established risk factors measured over time, participants with type 2 diabetes vs those with type 1 had significantly higher odds of diabetic kidney disease (odds ratio [OR], 2.58; 95% CI, 1.39-4.81; P=.003), retinopathy (OR, 2.24; 95% CI, 1.11-4.50; P = .02), and peripheral neuropathy (OR, 2.52; 95% CI, 1.43-4.43; P = .001), but no significant difference in the odds of arterial stiffness (OR, 1.07; 95% CI, 0.63-1.84; P = .80) and hypertension (OR, 0.85; 95% CI, 0.50-1.45; P = .55). Conclusions and Relevance Among teenagers and young adults who had been diagnosed with diabetes during childhood or adolescence, the prevalence of complications and comorbidities was higher among those with type 2 diabetes compared with type 1, but frequent in both groups. These findings support early monitoring of youth with diabetes for development of complications.
Collapse
Affiliation(s)
- Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, Aurora
| | - Jeanette M Stafford
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | | | - Ralph D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Lawrence Dolan
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Giuseppina Imperatore
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Barbara Linder
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Jean M Lawrence
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | | | - Amy K Mottl
- UNC Division of Nephrology and Hypertension, University of North Carolina School of Medicine, Chapel Hill
| | - Mary Helen Black
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena
| | - Rodica Pop-Busui
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor
| | - Sharon Saydah
- Division of Diabetes Translation, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Richard F Hamman
- Department of Epidemiology, Colorado School of Public Health, Aurora
| | | |
Collapse
|
33
|
Mathews PM, Levy E. Cystatin C in aging and in Alzheimer's disease. Ageing Res Rev 2016; 32:38-50. [PMID: 27333827 DOI: 10.1016/j.arr.2016.06.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 12/13/2022]
Abstract
Under normal conditions, the function of catalytically active proteases is regulated, in part, by their endogenous inhibitors, and any change in the synthesis and/or function of a protease or its endogenous inhibitors may result in inappropriate protease activity. Altered proteolysis as a result of an imbalance between active proteases and their endogenous inhibitors can occur during normal aging, and such changes have also been associated with multiple neuronal diseases, including Amyotrophic Lateral Sclerosis (ALS), rare heritable neurodegenerative disorders, ischemia, some forms of epilepsy, and Alzheimer's disease (AD). One of the most extensively studied endogenous inhibitor is the cysteine-protease inhibitor cystatin C (CysC). Changes in the expression and secretion of CysC in the brain have been described in various neurological disorders and in animal models of neurodegeneration, underscoring a role for CysC in these conditions. In the brain, multiple in vitro and in vivo findings have demonstrated that CysC plays protective roles via pathways that depend upon the inhibition of endosomal-lysosomal pathway cysteine proteases, such as cathepsin B (Cat B), via the induction of cellular autophagy, via the induction of cell proliferation, or via the inhibition of amyloid-β (Aβ) aggregation. We review the data demonstrating the protective roles of CysC under conditions of neuronal challenge and the protective pathways induced by CysC under various conditions. Beyond highlighting the essential role that balanced proteolytic activity plays in supporting normal brain aging, these findings suggest that CysC is a therapeutic candidate that can potentially prevent brain damage and neurodegeneration.
Collapse
Affiliation(s)
- Paul M Mathews
- Departments of Psychiatry, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Efrat Levy
- Departments of Psychiatry, New York University School of Medicine, USA; Biochemistry and Molecular Pharmacology, New York University School of Medicine, USA; Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA.
| |
Collapse
|
34
|
Bjornstad P, Škrtić M, Lytvyn Y, Maahs DM, Johnson RJ, Cherney DZI. The Gomez' equations and renal hemodynamic function in kidney disease research. Am J Physiol Renal Physiol 2016; 311:F967-F975. [PMID: 27605583 PMCID: PMC6347069 DOI: 10.1152/ajprenal.00415.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/02/2016] [Indexed: 12/16/2022] Open
Abstract
Diabetic kidney disease (DKD) remains the leading cause of end-stage renal disease. A major challenge in preventing DKD is the difficulty in identifying high-risk patients at an early, pre-clinical stage. Albuminuria and eGFR as measures of renal function in DKD research and clinical practice are limited by regression of one-third of patients with microalbuminuria to normoalbuminuria and eGFR is biased and imprecise in the normal-elevated range. Moreover, existing methods that are used to assess renal function do not give detailed insight into the location of the renal hemodynamic effects of pharmacological agents at the segmental level. To gain additional information about the intrarenal circulation in-vivo in humans, mathematical equations were developed by Gomez et al in the 1950s. These equations used measurements of GFR, renal blood flow (RBF), effective renal plasma flow (ERPF), renal vascular resistance (RVR), hematocrit and serum protein to calculate afferent and efferent arteriolar resistances, glomerular hydrostatic pressure and filtration pressure. Although indirect and based on physiological assumptions, these techniques have the potential to improve researchers' ability to identify early pre-clinical changes in renal hemodynamic function in patients with a variety of conditions including DKD, thereby offering tremendous potential in mechanistic human research studies. In this review, we focus on the application of Gomez' equations and summarize the potential and limitations of this technique in DKD research. We also summarize illustrative data derived from Gomez' equations in patients with type 1 (T1D) and type 2 diabetes (T2D) and hypertension.
Collapse
|
35
|
Pan Y, Jiang S, Qiu D, Shi J, Zhou M, An Y, Ge Y, Xie H, Liu Z. Comparing the GFR estimation equations using both creatinine and cystatin c to predict the long-term renal outcome in type 2 diabetic nephropathy patients. J Diabetes Complications 2016; 30:1478-1487. [PMID: 27593903 DOI: 10.1016/j.jdiacomp.2016.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 11/26/2022]
Abstract
AIMS This study aimed to determine whether eGFRcre-cys and its slope could improve the prediction of the long-term renal outcome in patients with type 2 diabetic nephropathy (DN). METHODS The cross-sectional and longitudinal analyses included 501 type 2DN patients from 2003 to 2009. GFR was estimated using either eGFRcre-cys or the serum creatinine-based equation (eGFRcre) or the cystatin C-based equation (eGFRcys), and was classified into 3 categories (≥90, 60-90, ≤60ml/min per 1.73m2). The proportion of patients was evaluated in each creatinine-calculated eGFR category for which the category was reclassified based on either cystatin C or the combined measurement. Long-term changes in eGFRcre-cys, eGFRcys and eGFRcre were estimated using linear mixed effect models. The receiver operating characteristic (ROC) curves was applied to study the sensitivity and specificity of different eGFR slopes for predicting the renal endpoint. RESULTS In the cross-sectional analyses, eGFRcre was overestimated compared to eGFRcre-cys [median bias -8.5 (95% CI: -25.01, 1.21)]. The reclassification of eGFRcre to a higher value was associated with an increased risk of ESRD [OR: 4.01 (95% CI: 2.36 to 6.82)]. In the longitudinal analyses for predicting end-stage renal disease (ERSD), the ROC curves for eGFRcre-cys (AUC=0.86±0.03) over 24months were increased compared with the ROC curves for eGFRcre and eGFRcys (p<0.05). CONCLUSIONS The study suggests that the eGFRcre-cys equation may be more precise and sensitive for predicting the renal outcome in T2DN patients. Tracking renal decline using eGFRcre-cys may be used as a surrogate for determining the renal endpoint in a clinical setting.
Collapse
Affiliation(s)
- Yu Pan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Song Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Dandan Qiu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Minlin Zhou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Yu An
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Yongchun Ge
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Honglang Xie
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine.
| |
Collapse
|
36
|
Zhang H, Wang H, Yan M, Zhao T, Lu X, Zhu B, Gong Y, Li P. A recombinant TGF-β1 vaccine ameliorates diabetic nephropathy in OLETF rats. Immunotherapy 2016; 8:1045-57. [PMID: 27485077 DOI: 10.2217/imt-2015-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The aim of this study was to investigate the potential of a recombinant vaccine encoding TGF-β1 in OLETF rats with diabetic nephropathy (DN). METHODS OLETF rats were treated with vehicle or TGF-β1 vaccine. LETO rats were used as normal controls. At 42 weeks after immunization with vaccine, samples from blood, urine and kidney were collected for biochemical, histologic, immunohistochemical and molecular analyses. RESULTS OLETF rats treated with the vaccine reduced blood glucose levels, improved renal pathological changes, and inhibited overexpression of TGF-β1 and p-Smad3, as well as MCP-1, TNF-α and IL-1β. CONCLUSION TGF-β1 vaccine attenuated diabetic nephropathy in OLETF rats through reduction of inflammation, improvement of kidney fibrosis and partial correction of glucose metabolism.
Collapse
Affiliation(s)
- Haojun Zhang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Hua Wang
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Meihua Yan
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Tingting Zhao
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Xiaoguang Lu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Zhu
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Yuewen Gong
- College of Pharmacy, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|