1
|
Antza C, Sitmalidou M, Belančić A, Katsiki N, Kotsis V. Clinic, Ambulatory and Home Blood Pressure Monitoring for Metabolic Syndrome: Time to Change the Definition? MEDICINA (KAUNAS, LITHUANIA) 2025; 61:434. [PMID: 40142246 PMCID: PMC11944059 DOI: 10.3390/medicina61030434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Metabolic syndrome (MetS) is considered a global epidemic, and its diagnosis is crucial, allowing early intervention and management. The main aim of this study was to examine any possible blood pressure (BP) differences based on office and out-of-office measurements in patients with and without MetS, and to investigate if any of these measurements correlated better with MetS. The secondary aim was to investigate any possible cardiovascular risk differences. Materials and Methods: The study population consisted of individuals attending the outpatient hypertension clinic. Office and out-of-office BP measurements were recorded in all of the patients, as well as different cardiovascular risk scores and echocardiography. MetS was defined according to ACC/AHA criteria. Results: A total of 282 (39.9% men) individuals (56.8 ± 15.8 years) were analyzed; 60.8% of them had MetS. The patients with MetS had a significantly higher systolic BP (SBP) in all of the BP measurements, higher ASCVD risk (22% vs. 12%), Framingham risk scores (11.8% vs. 6.9%), a significantly higher prevalence of LVH (49.2% vs. 22.7%) and early vascular aging (54.8% vs. 27.4%) compared with the patients without MetS (p < 0.05 for all). In a univariate analysis, MetS was significantly correlated with the average 24h SBP, daytime and nighttime ambulatory SBP, office SBP, and home SBP in the morning (p < 0.05). No significant differences were observed for any of the DBP measurements. Finally, 50.5% of the MetS patients had sustained hypertension, 15.2% masked hypertension, and 11.5% white-coat hypertension based on ABPM, and these values were 45.1%, 19.3%, and 13.6%, respectively, based on HBPM. Furthermore, most of the MetS patients had non-dipping hypertension (56.4%). Conclusions: The present findings highlight the importance of out-of-office BP measurements in the diagnosis of MetS, since both a high office and out-of-office SBP were significant features of the syndrome (whereas this was not the case with DBP). This is further supported by the increased prevalence of different hypertension phenotypes observed in the MetS patients. Higher ASCVD risk scores and LVH and EVA prevalence were also related to MetS, thus strongly supporting the necessity for early detection and treatment.
Collapse
Affiliation(s)
- Christina Antza
- 3rd Department of Internal Medicine, Aristotle University, Hypertension, Hypertension-24h Ambulatory Blood Pressure Monitoring Center, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (C.A.); (M.S.)
| | - Maria Sitmalidou
- 3rd Department of Internal Medicine, Aristotle University, Hypertension, Hypertension-24h Ambulatory Blood Pressure Monitoring Center, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (C.A.); (M.S.)
| | - Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece;
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus
| | - Vasilios Kotsis
- 3rd Department of Internal Medicine, Aristotle University, Hypertension, Hypertension-24h Ambulatory Blood Pressure Monitoring Center, Papageorgiou Hospital, 56429 Thessaloniki, Greece; (C.A.); (M.S.)
| |
Collapse
|
2
|
Costa Hoffmeister M, Hammel Lovison V, Priesnitz Friedrich E, da Costa Rodrigues T. Ambulatory blood pressure monitoring and vascular complications in patients with type 1 diabetes mellitus - Systematic review and meta-analysis of observational studies. Diabetes Res Clin Pract 2024; 217:111873. [PMID: 39343143 DOI: 10.1016/j.diabres.2024.111873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
AIMS This study aimed to evaluate the role of the 24-Hour Ambulatory Blood Pressure Monitoring (ABPM) as a possible predictor of vascular outcomes in office normotensive people with type 1 diabetes mellitus (T1DM). METHODS This is a systematic review including cohort studies from the Embase, PubMed/Medline, and Web of Science databases on people with T1DM undergoing ABPM and subsequent evaluation of vascular complications. Measurements of difference (MD) were obtained using random effect model meta-analysis. RESULTS We found 364 articles and 49 duplicates. Seven studies were included, comprising 635 participants aged 25.8 ± 6.2 years. Most (57.5 %) were men, mean duration of diabetes was 11.8 ± 5.3 years, mean glycated hemoglobin level among participants was 8.5 % ± 1.6 %, and mean follow-up time was 4.2 years. Lower night systolic blood pressure MD - 4.37 mmHg (p = 0.0009) and night diastolic blood pressure MD - 3.97 mmHg (p < 0.0001) were associated with lower incidence of albuminuria. People withT1DM who presented no beginning or progression of retinopathy were those with lower night diastolic blood pressure MD - 3.62 mmHg (p = 0.042), diurnal diastolic blood pressure MD - 2.69 mmHg (p = 0.0138), and 24-hour diastolic blood pressure MD - 3.65 mmHg (p = 0.037). CONCLUSION Small mean differences in blood pressure parameters, as measured by ABPM, between people with T1DM are associated with a lower incidence or risk of progression of nephropathy and retinopathy.
Collapse
Affiliation(s)
- Mariana Costa Hoffmeister
- Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Vinicius Hammel Lovison
- Acadêmico da Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Priesnitz Friedrich
- Acadêmico da Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ticiana da Costa Rodrigues
- Serviço de Endocrinologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Departamento de Medicina Interna, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
3
|
Khurana I, Kaipananickal H, Maxwell S, Birkelund S, Syreeni A, Forsblom C, Okabe J, Ziemann M, Kaspi A, Rafehi H, Jørgensen A, Al-Hasani K, Thomas MC, Jiang G, Luk AO, Lee HM, Huang Y, Thewjitcharoen Y, Nakasatien S, Himathongkam T, Fogarty C, Njeim R, Eid A, Hansen TW, Tofte N, Ottesen EC, Ma RC, Chan JC, Cooper ME, Rossing P, Groop PH, El-Osta A. Reduced methylation correlates with diabetic nephropathy risk in type 1 diabetes. J Clin Invest 2023; 133:160959. [PMID: 36633903 PMCID: PMC9927943 DOI: 10.1172/jci160959] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023] Open
Abstract
Diabetic nephropathy (DN) is a polygenic disorder with few risk variants showing robust replication in large-scale genome-wide association studies. To understand the role of DNA methylation, it is important to have the prevailing genomic view to distinguish key sequence elements that influence gene expression. This is particularly challenging for DN because genome-wide methylation patterns are poorly defined. While methylation is known to alter gene expression, the importance of this causal relationship is obscured by array-based technologies since coverage outside promoter regions is low. To overcome these challenges, we performed methylation sequencing using leukocytes derived from participants of the Finnish Diabetic Nephropathy (FinnDiane) type 1 diabetes (T1D) study (n = 39) that was subsequently replicated in a larger validation cohort (n = 296). Gene body-related regions made up more than 60% of the methylation differences and emphasized the importance of methylation sequencing. We observed differentially methylated genes associated with DN in 3 independent T1D registries originating from Denmark (n = 445), Hong Kong (n = 107), and Thailand (n = 130). Reduced DNA methylation at CTCF and Pol2B sites was tightly connected with DN pathways that include insulin signaling, lipid metabolism, and fibrosis. To define the pathophysiological significance of these population findings, methylation indices were assessed in human renal cells such as podocytes and proximal convoluted tubule cells. The expression of core genes was associated with reduced methylation, elevated CTCF and Pol2B binding, and the activation of insulin-signaling phosphoproteins in hyperglycemic cells. These experimental observations also closely parallel methylation-mediated regulation in human macrophages and vascular endothelial cells.
Collapse
Affiliation(s)
- Ishant Khurana
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Harikrishnan Kaipananickal
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Maxwell
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Sørine Birkelund
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,University College Copenhagen, Faculty of Health, Department of Technology, Biomedical Laboratory Science, Copenhagen, Denmark
| | - Anna Syreeni
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jun Okabe
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Antony Kaspi
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Haloom Rafehi
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anne Jørgensen
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Keith Al-Hasani
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Merlin C. Thomas
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | | | - Andrea O.Y. Luk
- Department of Medicine and Therapeutics,,Hong Kong Institute of Diabetes and Obesity,,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Heung Man Lee
- Department of Medicine and Therapeutics,,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | | | | | | | - Christopher Fogarty
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rachel Njeim
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Nete Tofte
- Steno Diabetes Center Copenhagen, Herlev, Denmark
| | | | - Ronald C.W. Ma
- Department of Medicine and Therapeutics,,Hong Kong Institute of Diabetes and Obesity,,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Juliana C.N. Chan
- Department of Medicine and Therapeutics,,Hong Kong Institute of Diabetes and Obesity,,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Mark E. Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Per-Henrik Groop
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Laboratory and,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia.,University College Copenhagen, Faculty of Health, Department of Technology, Biomedical Laboratory Science, Copenhagen, Denmark.,Hong Kong Institute of Diabetes and Obesity,,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Morić BV, Šamija I, Sabolić LLG, Stipančić G. Is there a characteristic pattern of ambulatory blood pressure profile in type 1 diabetic children and adolescents? Ann Pediatr Endocrinol Metab 2022; 27:300-307. [PMID: 36200311 PMCID: PMC9816465 DOI: 10.6065/apem.2244022.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/28/2022] [Indexed: 01/13/2023] Open
Abstract
PURPOSE To examine the characteristics of ambulatory blood pressure (ABP) including blood pressure variability (BPV) and its association with albuminuria in type 1 diabetic (T1D) children and to identify potential predictors of high-normal albuminuria and microalbuminuria. METHODS ABP monitoring was performed in 201 T1D children and adolescents (mean age, 14.7±3.8 years) with T1D duration over 1 year. The level of albuminuria was assessed as the albumin/creatinine ratio (ACR) and patients were further classified as low-normal, high-normal or microalbuminuria. RESULTS Fifteen T1D children (7.5%) were hypertensive using office blood pressure (BP) and 10 (5%) according to ABP. T1D subjects had elevated 24-hour systolic BP (SBP) and diastolic BP (DBP) (+0.2 and + 0.3 standard deviation score [SDS]) and nighttime SBP and DBP (+0.6 and +0.8 SDS) compared to reference values. Patients with microalbuminuria had significantly higher 24-hour, daytime and nighttime DBP compared to normoalbuminuric subjects. There was a high percentage of nondippers (74.1%). Nighttime diastolic BPV was significantly higher in subjects with high-normal compared to low-normal albuminuria (p=0.01). A weak correlation was found between ACR and daytime DBP SDS (r=0.29, p<0.001 and nighttime DBP SDS (r=0.21, p=0.003). Age and nighttime diastolic BPV were predictors of high-normal albuminuria while nighttime DBP was a strong predictor for microalbuminuria. CONCLUSION T1D children have impaired BP regulation although most of them do not fulfill the criteria for sustained hypertension. There is an association between diastolic ABP and diastolic BPV with rising levels of albuminuria pointing to a clear connection between BP and incipient diabetic nephropathy.
Collapse
Affiliation(s)
- Bernardica Valent Morić
- Department of Pediatrics, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia,Address for correspondence: Bernardica Valent Morić Department of Pediatrics, Sestre milosrdnice University Hospital Center, Vinogradska 29, 10000 Zagreb, Croatia
| | - Ivan Šamija
- Department of Oncology and Nuclear Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | | | - Gordana Stipančić
- Department of Pediatrics, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| |
Collapse
|
5
|
Lee JS, Lee YJ, Lee YA, Shin CH. Effects of circadian blood pressure patterns on development of microvascular complications in pediatric patients with type 1 diabetes mellitus. Ann Pediatr Endocrinol Metab 2022; 27:44-51. [PMID: 35038838 PMCID: PMC8984744 DOI: 10.6065/apem.2142084.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The effects of circadian blood pressure (BP) alterations on the development and progression of microvascular complications in type 1 diabetes mellitus (T1DM) patients are unknown. We evaluated the effects of circadian BP alterations with development of microvascular complications during follow-up with patients with childhood-onset T1DM. METHODS We investigated the medical records of 81 pediatric patients with T1DM who underwent 24-hour ambulatory BP monitoring (ABPM) between January 2009 and February 2010. RESULTS Mean age at diagnosis and ABPM evaluation was 8.0±3.9 and 15.6±2.4 years, respectively. Hypertension (daytime, nighttime, and 24-hour mean hypertension) data were available in 42 patients. During the 8 years of follow-up after ABPM, microvascular complications occurred in 8 patients (diabetic retinopathy [DR] alone in 5, microalbuminuria alone in 2, and both in 1), of whom 7 had nondipper BP. Nighttime diastolic BP, nighttime mean arterial pressure, and glycated hemoglobin A (HbA1c) level were higher in patients with DR than in those without DR (P<0.05 for all). Daytime or nighttime BP and presence of dipper BP were not related to microvascular complications, but diabetic microvascular complications were more likely to occur in patients with an older age at diagnosis and higher HbA1c level. The proportion of patients with DR was higher in those with nondipper hypertension (83.3%) compared with dipper and nondipper normotension (0% and 16.7%, respectively; P=0.021). CONCLUSION As a predictor of microvascular complications, nondipper hypertension was not significant. Glycemic control rather than nondipper hypertension is the predominant factor determining DR in T1DM patients.
Collapse
Affiliation(s)
- Jeong-Seon Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Jeong Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Young Ah Lee
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Choong Ho Shin
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea,Address for correspondence: Choong Ho Shin Department of Pediatrics, Seoul National University Children’s Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| |
Collapse
|
6
|
Gaebe K, White CA, Mahmud FH, Scholey JW, Elia YT, Sochett EB, Cherney DZ. Evaluation of novel glomerular filtration rate estimation equations in adolescents and young adults with type 1 diabetes. J Diabetes Complications 2022; 36:108081. [PMID: 34756765 DOI: 10.1016/j.jdiacomp.2021.108081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/25/2022]
Abstract
AIMS Individuals with type 1 diabetes (T1D) are at an increased risk of chronic kidney disease making estimation of glomerular filtration rate (eGFR) an important component of diabetes care. Which eGFR equation is most appropriate to use in patients with T1D during the transition to adult care is unclear. We, therefore, sought to evaluate the performance of five eGFR equations in adolescents and young adults with T1D. METHODS Measured iohexol-based glomerular filtration rate was compared to the Chronic Kidney Disease and Epidemiology Collaboration (CKD-EPI) eGFR, Chronic Kidney Disease in Children (CKiD) eGFR, and three recently developed age-adjusted versions of these in 53 patients with T1D and preserved GFR using bias, precision, and accuracy. RESULTS The best performance was found in the sex-dependent CKiD equation (bias: -0.8, accuracy: 11.8 ml/min/1.73 m2). Bias and accuracy (26.4 and 26.8 ml/min/1.73 m2) were worst in the CKD-EPI equation. Age-dependent adjustment improved performance for this equation (bias: 5.3, accuracy: 13.4 ml/min/1.73 m2), but not for the CKiD equation (bias: 15.5, accuracy: 18.8 ml/min/1.73 m2). CONCLUSION Age-adjustment improved performance for the CKD-EPI equation, but not for the CKiD equation. The sex-adjusted CKiD equation performed best out of all equations.
Collapse
Affiliation(s)
- Karolina Gaebe
- Division of Endocrinology and Metabolism, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| | - Christine A White
- Division of Nephrology, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Farid H Mahmud
- Division of Endocrinology and Metabolism, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - James W Scholey
- Division of Nephrology, Department of Medicine, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Yesmino T Elia
- Division of Endocrinology and Metabolism, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Etienne B Sochett
- Division of Endocrinology and Metabolism, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - David Z Cherney
- Division of Nephrology, Department of Medicine, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Koca SB, Akdogan M, Koca S. Evaluation of early retinal vascular changes by optical coherence tomography angiography in children with type 1 diabetes mellitus without diabetic retinopathy. Int Ophthalmol 2021; 42:423-433. [PMID: 34625889 DOI: 10.1007/s10792-021-02059-7] [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: 04/24/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE To evaluate macular and peripapillary vascular changes by optical coherence tomography angiography (OCTA) in children with type 1 diabetes mellitus (T1DM) without diabetic retinopathy (DR). METHODS This study included 46 patients with T1DM and 46 age-sex matched healthy subjects. All participants were evaluated in terms of macular and optic disk parameters by using AngioVue. Foveal avascular zone (FAZ) area, macular and optic disk vessel density (VD) were analyzed. The correlation of these parameters with metabolic factors such as disease duration, mean hemoglobin A1c (HbA1c), insulin-like growth factor 1 (IGF-1) standard deviation score (SDS), homocysteine (Hcy) level, body mass index (BMI) SDS and daily insulin dose was also investigated in T1DM group. RESULTS No significant difference was found in FAZ area and optic disk radial peripapillary capillary (RPC) VD comparing diabetic and control groups. In all macular regions, VD was significantly lower in T1DM versus control group both in superficial capillary plexus (SCP) and deep capillary plexus (DCP). None of the metabolic parameters was correlated with FAZ area and optic disk RPC-VD. Vascular density in SCP was negatively correlated with mean HbA1c and positively correlated with IGF-1 SDS. Homocysteine level was negatively correlated with DCP-VD in all areas. CONCLUSION In children with T1DM without clinically apparent DR, VD in SCP and DCP was decreased and OCTA is a valuable imaging technique for detecting early vascular changes. The metabolic parameters such as mean HbA1c, IGF-1 SDS and Hcy affect the macular VD in diabetic children. TRIAL REGISTRATION NUMBER 2011-KAEK-2, 2021/4, Trial registration date: 02.04.2021.
Collapse
Affiliation(s)
- Serkan Bilge Koca
- Faculty of Medicine, Department of Pediatrics, Division of Pediatric Endocrinology, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Muberra Akdogan
- Faculty of Medicine, Department of Ophthalmology, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Semra Koca
- Faculty of Medicine, Department of Ophthalmology, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey. .,, 1444. Sokak Kandilli Konakları D Blok 2/8, Afyonkarahisar, Türkiye.
| |
Collapse
|
8
|
Guo X, Liang S, Wang W, Zheng Y, Zhang C, Chen X, Cai G. Lowest nocturnal systolic blood pressure is related to heavy proteinuria and outcomes in elderly patients with chronic kidney disease. Sci Rep 2021; 11:5846. [PMID: 33712668 PMCID: PMC7955052 DOI: 10.1038/s41598-021-85071-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022] Open
Abstract
Ambulatory blood pressure monitoring (ABPM) can produce many variables, of which the lowest nocturnal systolic blood pressure (LNSBP) currently used in calculating morning surge is occasionally overlooked in recent kidney studies compared with other ABPM parameters. We explored the clinical effects of LNSBP in elderly patients with chronic kidney disease (CKD) in a multicenter, observational cohort study. A total of 356 elderly patients with CKD from 19 clinics were included in this analysis. We used multiple logistic regression and survival analyses to assess the associations between the lowest nocturnal systolic blood pressure and heavy proteinuria and kidney disease outcomes, respectively. The median age was 66 years, and 66.6% were men. The median eGFR was 49.2 ml/min/1.73 m2. Multivariate logistic regression analysis demonstrated that LNSBP (OR 1.24; 95% CI 1.10–1.39; P < 0.001; per 10 mmHg) was associated with heavy proteinuria. During the median follow-up of 23 months, 70 patients (19.7%) had a composite outcome; of these, 25 initiated dialysis, 25 had 40% eGFR loss, and 20 died. Cox analysis showed that the renal risk of LNSBP for CKD outcomes remained significant even after adjusting for background factors, including age, sex, medical history of hypertension and diabetes, smoking status, eGFR, 24-h proteinuria, and etiology of CKD (HR 1.18; 95% CI 1.06–1.32; P = 0.002; per 10 mmHg). Concentrating on LNSBP could be valuable in guiding antihypertensive treatment to control heavy proteinuria and improve renal prognosis in elderly CKD patients.
Collapse
Affiliation(s)
- Xinru Guo
- Medical School of Chinese PLA, Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Shuang Liang
- Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China
| | - Wenling Wang
- Department of Nephrology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ying Zheng
- Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China
| | - Chun Zhang
- Department of Nephrology, Xinjiang Armed Police Crops Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xiangmei Chen
- Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, The First Medical Centre, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China.
| |
Collapse
|
9
|
Mamilly L, Mastrandrea LD, Mosquera Vasquez C, Klamer B, Kallash M, Aldughiem A. Evidence of Early Diabetic Nephropathy in Pediatric Type 1 Diabetes. Front Endocrinol (Lausanne) 2021; 12:669954. [PMID: 33995287 PMCID: PMC8113955 DOI: 10.3389/fendo.2021.669954] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/10/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most common microvascular complications in type 1 diabetes Mellitus (T1D). Urinary markers of renal damage or oxidative stress may signal early stages of DN. The association of these markers with blood pressure (BP) patterns and glycemic variability (GV) in children is yet to be explored. METHODS Subjects between the ages of 10 and 21 years with T1D were enrolled. Continuous glucose monitoring (CGM) and ambulatory blood pressure monitoring (ABPM) were performed on each subject. Urine samples were collected and analyzed for albumin, creatinine, neutrophil gelatinase-associated lipocalin (NGAL) and pentosidine. RESULTS The study included 21 subjects (62% female) with median age of 16.8 (IQR: 14.5, 18.9). Median HbA1C was 8.4 (IQR: 7.5, 9.3). While microalbuminuria was negative in all but one case (4.8%), urinary NGAL/Cr and pentosidine/Cr ratios were significantly elevated (P<0.001) in diabetic patients despite having normal microalbuminuria, and they correlated significantly with level of microalbumin/Cr (r=0.56 [CI: 0.17, 0.8] and r=0.79 [CI: 0.54, 0.91], respectively). Using ABPM, none had hypertension, however, poor nocturnal systolic BP dipping was found in 48% of cases (95% CI: 28-68%). Urinary NGAL/Cr negatively correlated with nocturnal SBP dipping (r=-0.47, CI: -0.76, -0.03). Urine NGAL/Cr also showed a significant negative correlation with HbA1c measurements, mean blood glucose, and high blood glucose index (r=-0.51 [CI: -0.78, -0.09], r=-0.45 [CI: -0.74, -0.03], and r=-0.51 [CI: -0.77, -0.1], respectively). Median urinary NGAL/Cr and pentosidine/Cr ratios were higher in the high GV group but were not significantly different. DISCUSSION This pilot study explores the role of ABPM and urinary markers of tubular health and oxidative stress in early detection of diabetic nephropathy. GV may play a role in the process of this diabetic complication.
Collapse
Affiliation(s)
- Leena Mamilly
- Section of Pediatric Endocrinology, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
- *Correspondence: Leena Mamilly,
| | - Lucy D. Mastrandrea
- Division of Endocrinology/Diabetes, UBMD Pediatrics and University at Buffalo/Oishei Children’s Hospital of Buffalo, NY, United States
| | - Claudia Mosquera Vasquez
- Section of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Brett Klamer
- Biostatistics Resource at Nationwide Children’s Hospital, Nationwide Children’s Hospital, Columbus, OH, United States
- Section of Pediatric Nephrology, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Mahmoud Kallash
- Section of Pediatric Nephrology, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Ahmad Aldughiem
- Section of Pediatric Nephrology, Dayton Children’s Hospital, Dayton, OH, United States
| |
Collapse
|
10
|
Chrysaidou K, Chainoglou A, Karava V, Dotis J, Printza N, Stabouli S. Secondary Hypertension in Children and Adolescents: Novel Insights. Curr Hypertens Rev 2020; 16:37-44. [PMID: 31038068 DOI: 10.2174/1573402115666190416152820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 12/25/2022]
Abstract
Hypertension is a significant risk factor for cardiovascular morbidity and mortality, not only in adults, but in youths also, as it is associated with long-term negative health effects. The predominant type of hypertension in children is the secondary hypertension, with the chronic kidney disease being the most common cause, however, nowadays, there is a rising incidence of primary hypertension due to the rising incidence of obesity in children. Although office blood pressure has guided patient management for many years, ambulatory blood pressure monitoring provides useful information, facilitates the diagnosis and management of hypertension in children and adolescents, by monitoring treatment and evaluation for secondary causes or specific phenotypes of hypertension. In the field of secondary hypertension, there are numerous studies, which have reported a strong association between different determinants of 24-hour blood pressure profile and the underlying cause. In addition, in children with secondary hypertension, ambulatory blood pressure monitoring parameters offer the unique advantage to identify pediatric low- and high-risk children for target organ damage. Novel insights in the pathogenesis of hypertension, including the role of perinatal factors or new cardiovascular biomarkers, such as fibroblast growth factor 23, need to be further evaluated in the near future.
Collapse
Affiliation(s)
- Katerina Chrysaidou
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| | - Athanasia Chainoglou
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| | - Vasiliki Karava
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| | - John Dotis
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| | - Nikoleta Printza
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| | - Stella Stabouli
- 1st Department of Pediatrics, Aristotle University Thessaloniki, Hippokratio General Hospital, Thessaloniki, Greece
| |
Collapse
|
11
|
Mejia-Vilet JM, López-Hernández YJ, Trujeque-Matos M, Santander-Velez JI, Cano-Verduzco ML, Cruz C, Morales-Buenrostro LE. High frequency of nocturnal hypertension in lupus nephritis: should ABPM be implemented in usual practice? Clin Rheumatol 2019; 39:1147-1155. [PMID: 31838636 DOI: 10.1007/s10067-019-04830-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/08/2019] [Accepted: 10/23/2019] [Indexed: 01/01/2023]
Abstract
INTRODUCTION/OBJECTIVES Hypertension management in lupus nephritis (LN) is guided by in-office blood pressure (BP); however, recent studies demonstrate that lupus patients frequently have nocturnal hypertension and reduced BP dipping. The aim of the study was to evaluate 24-h blood pressure in patients with active LN and after response to treatment. METHODS Seventy active LN patients were evaluated during a LN flare by ambulatory blood pressure monitoring (ABPM). Later, 10 patients with complete response were re-evaluated after 12 months along with 20 matched controls. Overall, daytime and nightime BP, day-to-night dipping, BP load and variability, and the incidence of abnormal BP patterns were assessed. Blood pressure levels were correlated with clinical and histologic parameters and independent associations evaluated by linear regression. RESULTS Overall systolic hypertension occurred in 25 (36%) patients and diastolic hypertension in 28 (40%). Nighttime systolic and diastolic hypertension occurred in 35 (50%) and 44 (63%) of patients, respectively. Nocturnal systolic day-to-night BP decrease was abnormal in 59 (84%) patients. Only 18 (26%) were diagnosed with HT by in-office evaluation while 29 (41%) had masked hypertension (MH)/masked uncontrolled hypertension (MUCH), and 3 (4%) had white coat hypertension. Patients with MH had lower eGFR, complement C3, hemoglobin, and higher systolic variability compared with patients with normal BP. Systolic and diastolic BP levels were associated with the years under corticosteroid treatment, activity biomarkers (proteinuria, complement C3), and the degree of interstitial inflammation in the kidney biopsy. A re-evaluation at 12 months showed that although 9 out of 10 patients had normal in-office BP and BP loads improved, still 5 patients remained with MH due to nocturnal hypertension, and 7 remained with abnormal day-to-night dipping. These numbers were higher than those of matched controls. CONCLUSIONS Due to the high frequency of nocturnal hypertension and abnormal day-to-night dipping, office BP measurements alone may not be sufficient to guide hypertension management in patients with LN.Key Points• Nocturnal hypertension and abnormal BP patterns are frequent and not detectable by the standard in-office BP evaluation in LN patients.• BP abnormalities may not be fully corrected after a complete clinical response to treatment in lupus nephritis and are only detectable by ABPM.• The degree of interstitial inflammation in the kidney biopsy in LN patients is associated to BP levels. This supports the hypotheses underlining the role of interstitial inflammation in salt sensitivity and hypertension.
Collapse
Affiliation(s)
- Juan M Mejia-Vilet
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Yesser J López-Hernández
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Mariedel Trujeque-Matos
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - J Iván Santander-Velez
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Mayra L Cano-Verduzco
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Cristino Cruz
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico
| | - Luis E Morales-Buenrostro
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga No. 15, Sección XVI, Tlalpan, 14080, Mexico City, Mexico.
| |
Collapse
|
12
|
Maksoud AAA, Sharara SM, Nanda A, Khouzam RN. The renal resistive index as a new complementary tool to predict microvascular diabetic complications in children and adolescents: a groundbreaking finding. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:422. [PMID: 31660321 PMCID: PMC6787385 DOI: 10.21037/atm.2019.08.65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/12/2019] [Indexed: 11/06/2022]
Abstract
The increasing prevalence of type 1 diabetes mellitus (DM) has made it necessary to have new markers for early detection of diabetic nephropathy. Renal resistive index (RI) by using renal Doppler can be a helpful tool in detecting functional alterations in renal hemodynamics. This study was conducted on 100 children and adolescents with type 1 DM. They were further subdivided into two equal subgroups: group 1 with type 1 DM and normo-albuminuria [urinary albumin excretion (UAE) <30 mg/24 hours], and group 2 with type 1 DM and hyper-albuminuria (increased UAE >30 mg/24 hours). There were 37 males (37%) and 63 females (63%); their mean ages were 13.6±2.53 (range, 10-19) years and mean disease duration was 8.867±2.260 (range, 5-13) years. Progressive increase in RI was significantly associated with increased disease duration more than 10 years, elevated serum HbA1c more than 7.5% and early pubertal stages. While not significantly related to sex, weight, height, blood pressure or serum lipid profile, diabetic micro-vascular complications (nephropathy and sensory neuropathy) were more prevalent among patients with RI more than 0.58. Renal RI could be a useful complementary test for the evaluation of functional alterations in renal hemodynamics in the early stages of diabetic nephropathy.
Collapse
Affiliation(s)
| | | | - Amit Nanda
- Department of Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rami N. Khouzam
- Department of Medicine, Division of Cardiovascular Diseases, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
13
|
Marcovecchio ML, Dalton RN, Daneman D, Deanfield J, Jones TW, Neil HAW, Dunger DB. A new strategy for vascular complications in young people with type 1 diabetes mellitus. Nat Rev Endocrinol 2019; 15:429-435. [PMID: 30996294 DOI: 10.1038/s41574-019-0198-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes vascular complications, including cardiovascular disease, diabetic nephropathy and retinopathy, have a negative effect on the long-term prognosis of young people with type 1 diabetes mellitus (T1DM). Poor glycaemic control and consequent increased HbA1c levels are major risk factors for the development of vascular complications. HbA1c levels are the main focus of current management strategies; however, the recommended target is rarely achieved in adolescents. Thus, a clear need exists for improved biomarkers to identify high-risk young people early and to develop new intervention strategies. Evidence is accumulating that early increases in urinary albumin excretion could be predictive of adolescents with T1DM who are at an increased risk of developing vascular complications, independent of HbA1c levels. These findings present an opportunity to move towards the personalized care of adolescents with T1DM, which takes into consideration changes in albumin excretion and other risk factors in addition to HbA1c levels.
Collapse
Affiliation(s)
| | - R Neil Dalton
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Denis Daneman
- Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - John Deanfield
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Timothy W Jones
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - H Andrew W Neil
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW This review considers the relationship between abnormal blood pressure (BP) variability and autonomic dysfunction through an attempt to answer questions about its clinical relevance and pertinence to diabetes and cardiovascular autonomic neuropathy (CAN) and which therapeutic measures can lessen its cardiovascular impact. RECENT FINDINGS Office, ambulatory, and home BP monitoring identify posture-related, circadian, short-term, and long-term BP variabilities. Abnormal BP variability is a risk marker for organ damage, mortality, and cardiovascular events. Moreover, BP variability changes are common in diabetes and associated with CAN and possibly exacerbated by comorbidities like nephropathy, obstructive sleep apnoea syndrome, and chronic pain. The prognostic role of nondipping and reverse dipping is well documented in diabetes. Some findings suggest the possibility of restoring dipping with the dosage time of antihypertensive agents. Diabetes is a favorable scenario for altered BP variability, which might mediate the harmful effects of CAN. Preliminary data suggest the protective effect of targeting BP variability. However, further longitudinal outcome studies are needed. In the meantime, BP variability measures and practical expedients in antihypertensive treatment should be implemented in diabetes.
Collapse
Affiliation(s)
- Vincenza Spallone
- Endocrinology, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133, Rome, Italy.
| |
Collapse
|
15
|
Donaghue KC, Marcovecchio ML, Wadwa RP, Chew EY, Wong TY, Calliari LE, Zabeen B, Salem MA, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2018: Microvascular and macrovascular complications in children and adolescents. Pediatr Diabetes 2018; 19 Suppl 27:262-274. [PMID: 30079595 PMCID: PMC8559793 DOI: 10.1111/pedi.12742] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/27/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Kim C Donaghue
- The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Camperdown, Australia
| | | | - R P Wadwa
- University of Colorado School of Medicine, Denver, Colorado
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, the National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Tien Y Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | | | - Bedowra Zabeen
- Department of Paediatrics and Changing Diabetes in Children Program, Bangladesh Institute of Research and Rehabilitation in Diabetes, Endocrine and Metabolic Disorders, Dhaka, Bangladesh
| | - Mona A Salem
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maria E Craig
- The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, University of Sydney, Camperdown, Australia
- School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
16
|
Lu L, Marcovecchio ML, Dalton RN, Dunger D. Cardiovascular autonomic dysfunction predicts increasing albumin excretion in type 1 diabetes. Pediatr Diabetes 2018; 19:464-469. [PMID: 29171134 DOI: 10.1111/pedi.12614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 11/01/2017] [Accepted: 11/01/2017] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To determine the potential role of cardiovascular autonomic dysfunction in the development of renal complications in young people with type 1 diabetes (T1D). METHODS In this prospective study, 199 children and adolescents recruited to the Oxford Regional Prospective Study underwent assessment of autonomic function ~5 years after diagnosis, and were subsequently followed with longitudinal assessments of HbA1c and urine albumin-creatinine ratio (ACR) over 8.6 ± 3.4 years. Autonomic function was assessed with 4 standardized tests of cardiovascular reflexes: heart rate (HR) response to (1) Valsalva Maneuver, (2) deep breathing, (3) standing, and (4) blood pressure (BP) response to standing. Linear mixed models were used to assess the association between autonomic parameters and future changes in ACR. RESULTS Independent of HbA1c , each SD increase in HR response to Valsalva Maneuver predicted an ACR increase of 2.16% [95% CI: 0.08; 4.28] per year (P = .04), while each SD increase in diastolic BP response to standing predicted an ACR increase of 2.55% [95% CI: 0.37; 4.77] per year (P = .02). The effect of HR response to standing on ACR reached borderline significance (-2.07% [95% CI: -4.11; 0.01] per year per SD increase, P = .051). CONCLUSIONS In this cohort of young people with T1D, enhanced cardiovascular reflexes at baseline predicted future increases in ACR. These results support a potential role for autonomic dysfunction in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Liangjian Lu
- Department of Paediatrics, MRL Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, UK.,Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
| | - M Loredana Marcovecchio
- Department of Paediatrics, MRL Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - R Neil Dalton
- WellChild Laboratory, Evelina London Children's Hospital, London, UK
| | - David Dunger
- Department of Paediatrics, MRL Wellcome Trust-MRC Institute of Metabolic Science, NIHR Cambridge Comprehensive Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Mei ZB, Xiao Y. ACE Inhibitors and Statins in Adolescents with Type 1 Diabetes. N Engl J Med 2018; 378:579. [PMID: 29419270 DOI: 10.1056/nejmc1715763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Zu-Bing Mei
- Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Xiao
- Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
18
|
Marcovecchio ML, Tossavainen PH, Owen K, Fullah C, Benitez-Aguirre P, Masi S, Ong K, Nguyen H, Chiesa ST, Dalton RN, Deanfield J, Dunger DB. Clustering of cardio-metabolic risk factors in parents of adolescents with type 1 diabetes and microalbuminuria. Pediatr Diabetes 2017; 18:947-954. [PMID: 28271589 PMCID: PMC6186416 DOI: 10.1111/pedi.12515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/12/2017] [Accepted: 02/06/2017] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE To evaluate the association between a clustering of cardio-metabolic risk factors in parents and the development of microalbuminuria (MA) in their offspring with childhood-onset type 1 diabetes (T1D). METHODS The study population comprised 53 parents (mean age [±SD]: 56.7±6.2 years) of 35 T1D young people with MA (MA+) and 86 parents (age: 56.1±6.3 years) of 50 matched offspring with normoalbuminuria (MA-), who underwent clinical, biochemical and cardiovascular imaging assessments. The primary study endpoint was the difference between parents from the MA+ and MA- groups in a cardio-metabolic risk score, calculated as the average value of the standardized measures (z-scores) for waist circumference, blood pressure, fasting glucose, insulin, HDL-cholesterol and triglycerides levels. Cardiovascular parameters, including carotid intima-media thickness (cIMT), flow-mediated dilatation (FMD) and pulse wave velocity (PWV), were also assessed. A DXA scan was performed to assess body composition. RESULTS The cardio-metabolic risk score was significantly higher in parents of MA+ compared to parents of MA- offspring (mean [95% CI]: 1.066[0.076; 2.056] vs -0.268[-0.997; 0.460], P = .03). Parents of MA+ offspring had slightly higher values of waist circumference, lipids, insulin and blood pressure, although only diastolic blood pressure was statistically different between the 2 groups (P = .0085). FMD, cIMT, PWV (all P > .3), and DXA parameters (all P > .2) were not significantly different between the 2 groups. CONCLUSIONS Parents of young offspring with childhood-onset T1D and MA showed an abnormal metabolic profile, reflected by a calculated risk score. The finding supports the role of a familial predisposition to risk of developing diabetic nephropathy.
Collapse
Affiliation(s)
| | - Päivi H Tossavainen
- Department of Paediatrics, PEDEGO Research Unit and Medical Research Centre Oulu, Oulu, University Hospital and University of Oulu, Oulu, Finland
| | - Katharine Owen
- Oxford Centre for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
| | - Catherine Fullah
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Paul Benitez-Aguirre
- Institute of Endocrinology and Diabetes, University of Sydney, Sydney, Australia
| | - Stefano Masi
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Ken Ong
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Helen Nguyen
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - Scott T Chiesa
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - R Neil Dalton
- WellChild Laboratory, King's College London, Evelina Children's Hospital, London, UK
| | - John Deanfield
- Vascular Physiology Unit, Institute of Cardiovascular Science, University College London, London, UK
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
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: 84] [Impact Index Per Article: 10.5] [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
|
20
|
Rica I, Mingorance A, Gómez-Gila AL, Clemente M, González I, Caimari M, García-Cuartero B, Barrio R. Achievement of metabolic control among children and adolescents with type 1 diabetes in Spain. Acta Diabetol 2017; 54:677-683. [PMID: 28421336 DOI: 10.1007/s00592-017-0991-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/02/2017] [Indexed: 12/27/2022]
Abstract
AIMS To assess metabolic control in a paediatric T1D population in Spain and analyse the rate of severe acute decompensations and chronic complications. METHODS Data from patients treated at eight paediatric diabetes units with experienced diabetes teams between June and December 2014 were analysed in an observational prospective study. Variables included: age, sex, diabetes duration, number of follow-up visits/year, anthropometrical data, insulin treatment modalities, mean annual HbA1c and the prevalence of acute and chronic complications. SPSS statistics 21.0 was used. RESULTS A total of 853 patients (49.7% female) with a mean age of 12.1 ± 3.7 years were included. Anthropometric data were normal. Mean diabetes duration was 8 ± 3.4 years. Mean outpatient follow-up was 4.7 ± 0.04 visits/year. Twenty-five per cent were on continuous subcutaneous insulin infusion (CSII). Mean HbA1c was 7.3 ± 1% (56 ± 8 mmol/mol) and 66.6% had HbA1c < 7.5% (58 mmol/mol). HbA1c value correlated negatively with age at onset and positively with years of diabetes, number of visits/year and current age (F = 7.06; p = 0.01). Patients on CSII (n = 213) were younger, attended the outpatient clinic more frequently, were diagnosed earlier, had better metabolic control and had presented more severe hypoglycaemic episodes the previous year. The rate of severe decompensation (episodes/100 patients/year) was ketoacidosis 1.5 and severe hypoglycaemia 4.5. The prevalence of chronic complications was very low. CONCLUSIONS Our data describe the good compliance of paediatric T1D patients treated at eight paediatric units in Spain following international standards of metabolic control.
Collapse
Affiliation(s)
- Itxaso Rica
- Paediatric Endocrinology, Hospital Universitario de Cruces, CIBERDEM, CIBERER, BIOCRUCES, Plaza Cruces s/n, 48903, Barakaldo, Bizkaia, Spain
| | - Andrés Mingorance
- Paediatric Endocrinology, Hospital General Universitario de Alicante, Pintor Baeza 11, 03010, Alicante, Spain
| | - Ana Lucía Gómez-Gila
- Paediatric Endocrinology Unit. Paediatric Service, Hospital Infantil, HHUU Virgen Rocío, Avda Marques Luca de Tena s/n, Seville, Spain
| | - María Clemente
- Paediatric Endocrinology Unit, Paediatric Service, Growth and Development Research Unit, Vall d'Hebron Research Institute (VHIR), Hospital Vall d'Hebron, CIBERER, Instituto de Salud Carlos III, Ps. Vall d´Hebron 119-129, 08035, Barcelona, Spain.
| | - Isabel González
- Paediatric Endocrinology, Hospital Infantil La Paz, Pº Castellana 261, 28046, Madrid, Spain
| | - María Caimari
- Paediatric Endocrinology, Hospital Universitario Son Espases, Carretera Valldemosa, 79, 07010, Palma de Mallorca, Spain
| | - Beatriz García-Cuartero
- Paediatric Endocrinology Unit, Severo Ochoa University Hospital, 28911, Leganés, Madrid, Spain
| | - Raquel Barrio
- Paediatric Endocrinology and Diabetes Unit, Ramón y Cajal University Hospital, Cta. Colmenar Viejo 9, 100, 28034, Madrid, Spain
| |
Collapse
|
21
|
Dunger DB. Banting Memorial Lecture 2016 Reducing lifetime risk of complications in adolescents with Type 1 diabetes. Diabet Med 2017; 34:460-466. [PMID: 27973749 DOI: 10.1111/dme.13299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2016] [Indexed: 12/19/2022]
Abstract
Adolescence is a challenging period of life for any young person, and for those with Type 1 diabetes, physiological and psychological factors can result in a deterioration in glycaemic control. In young people with Type 1 diabetes, puberty may be an additional risk factor impacting on the lifetime risk for renal and cardiovascular complications. Our longitudinal studies have identified that increases in urinary albumin excretion through childhood are associated with the development of microalbuminuria and a generalized endotheliopathy linked to cardiovascular risk. Screening of participants recruited to the Adolescent type 1 Diabetes cardio-renal Intervention Trial (AdDIT) confirms that these early changes in albumin excretion are related to both diabetic nephropathy and cardiovascular risk; in part, independent of glycaemic control. Thus, as well as current attempts to improve glycaemic control through enhanced targeted insulin delivery, pumps, sensors and closed loop, we have explored the role of angiotensin-converting enzyme inhibitors and statins in providing cardio-renal protection during adolescence.
Collapse
Affiliation(s)
- D B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| |
Collapse
|
22
|
Sandholm N, Van Zuydam N, Ahlqvist E, Juliusdottir T, Deshmukh HA, Rayner NW, Di Camillo B, Forsblom C, Fadista J, Ziemek D, Salem RM, Hiraki LT, Pezzolesi M, Trégouët D, Dahlström E, Valo E, Oskolkov N, Ladenvall C, Marcovecchio ML, Cooper J, Sambo F, Malovini A, Manfrini M, McKnight AJ, Lajer M, Harjutsalo V, Gordin D, Parkkonen M, Tuomilehto J, Lyssenko V, McKeigue PM, Rich SS, Brosnan MJ, Fauman E, Bellazzi R, Rossing P, Hadjadj S, Krolewski A, Paterson AD, Florez JC, Hirschhorn JN, Maxwell AP, Dunger D, Cobelli C, Colhoun HM, Groop L, McCarthy MI, Groop PH. The Genetic Landscape of Renal Complications in Type 1 Diabetes. J Am Soc Nephrol 2016; 28:557-574. [PMID: 27647854 DOI: 10.1681/asn.2016020231] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022] Open
Abstract
Diabetes is the leading cause of ESRD. Despite evidence for a substantial heritability of diabetic kidney disease, efforts to identify genetic susceptibility variants have had limited success. We extended previous efforts in three dimensions, examining a more comprehensive set of genetic variants in larger numbers of subjects with type 1 diabetes characterized for a wider range of cross-sectional diabetic kidney disease phenotypes. In 2843 subjects, we estimated that the heritability of diabetic kidney disease was 35% (P=6.4×10-3). Genome-wide association analysis and replication in 12,540 individuals identified no single variants reaching stringent levels of significance and, despite excellent power, provided little independent confirmation of previously published associated variants. Whole-exome sequencing in 997 subjects failed to identify any large-effect coding alleles of lower frequency influencing the risk of diabetic kidney disease. However, sets of alleles increasing body mass index (P=2.2×10-5) and the risk of type 2 diabetes (P=6.1×10-4) associated with the risk of diabetic kidney disease. We also found genome-wide genetic correlation between diabetic kidney disease and failure at smoking cessation (P=1.1×10-4). Pathway analysis implicated ascorbate and aldarate metabolism (P=9.0×10-6), and pentose and glucuronate interconversions (P=3.0×10-6) in pathogenesis of diabetic kidney disease. These data provide further evidence for the role of genetic factors influencing diabetic kidney disease in those with type 1 diabetes and highlight some key pathways that may be responsible. Altogether these results reveal important biology behind the major cause of kidney disease.
Collapse
Affiliation(s)
- Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Natalie Van Zuydam
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Medical Research Institute
| | - Emma Ahlqvist
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Harshal A Deshmukh
- Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - N William Rayner
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Human Genetics, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Barbara Di Camillo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Joao Fadista
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Daniel Ziemek
- Computational Sciences, Pfizer Worldwide Research and Development, Berlin, Germany
| | - Rany M Salem
- Departments of Genetics,Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Divisions of Endocrinology and Genetics, Boston Children's Hospital, Boston, Massachusetts
| | - Linda T Hiraki
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Marcus Pezzolesi
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, Massachusetts
| | - David Trégouët
- Sorbonne Universities, Pierre et Marie Curie University (UPMC) and National Institute for Health and Medical Research, Mixed Research Unit in Health (UMR_S) 1166, Paris, France.,Institute for Cardiometabolism and Nutrition, Genomics and pathophysiology of Cardiovascular diseases, Paris, France
| | - Emma Dahlström
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Erkka Valo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Nikolay Oskolkov
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Claes Ladenvall
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Jason Cooper
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Francesco Sambo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alberto Malovini
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.,Laboratory of Informatics and Systems Engineering for Clinical Research, Scientific Institute for Research, Hospitalization and Health Care, IRCCS (Instituto di Ricovero e Cura a Carattere Scientifico); Salvatore Maugeri Foundation, Pavia, Italy
| | - Marco Manfrini
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Amy Jayne McKnight
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, United Kingdom
| | - Maria Lajer
- Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Daniel Gordin
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | - Maija Parkkonen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
| | | | - Jaakko Tuomilehto
- The Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland.,Centre for Vascular Prevention, Danube University Krems, Krems, Austria
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden.,Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark
| | - Paul M McKeigue
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | | | - Eric Fauman
- Computational Sciences, Pfizer Worldwide Research and Development, Cambridge, Massachusetts
| | - Riccardo Bellazzi
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Peter Rossing
- Diabetic Complications, Steno Diabetes Center, Gentofte, Denmark.,Department of Health, Aarhus University, Aarhus, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Samy Hadjadj
- Functional Research Unit of Medicine and Pharmacy, University of Poitiers, Poitiers, France.,Department of Endocrinology-Diabetology and Center of Clinical Investigation, Poitiers University Hospital, Poitiers, France.,Institute National pour la Santé et la Recherche Médicale, National Institute for Health and Medical Research, Center of Clinical Investigation 1402 and Unit 1082, Poitiers, France
| | - Andrzej Krolewski
- Section on Genetics and Epidemiology, Joslin Diabetes Center, Boston, Massachusetts
| | - Andrew D Paterson
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts
| | - Joel N Hirschhorn
- Departments of Genetics,Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts.,Divisions of Endocrinology and Genetics, Boston Children's Hospital, Boston, Massachusetts
| | - Alexander P Maxwell
- Nephrology Research, Centre for Public Health, Queen's University of Belfast, Belfast, United Kingdom.,Regional Nephrology Unit, Belfast City Hospital, Belfast, United Kingdom; and
| | | | - David Dunger
- Department of Paediatrics, Institute of Metabolic Science, and
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Helen M Colhoun
- Division of Population Health Sciences, University of Dundee, Dundee, United Kingdom
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics,Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland.,Baker IDI (International Diabetes Institute) Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | |
Collapse
|
23
|
Parati G, Ochoa JE, Bilo G, Agarwal R, Covic A, Dekker FW, Fliser D, Heine GH, Jager KJ, Gargani L, Kanbay M, Mallamaci F, Massy Z, Ortiz A, Picano E, Rossignol P, Sarafidis P, Sicari R, Vanholder R, Wiecek A, London G, Zoccali C. Hypertension in Chronic Kidney Disease Part 2. Hypertension 2016; 67:1102-10. [DOI: 10.1161/hypertensionaha.115.06896] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gianfranco Parati
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Juan Eugenio Ochoa
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Grzegorz Bilo
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Rajiv Agarwal
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Adrian Covic
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Friedo W. Dekker
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Danilo Fliser
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Gunnar H. Heine
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Kitty J. Jager
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Luna Gargani
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Mehmet Kanbay
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Francesca Mallamaci
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Ziad Massy
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Alberto Ortiz
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Eugenio Picano
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Patrick Rossignol
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Pantelis Sarafidis
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Rosa Sicari
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Raymond Vanholder
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Andrzej Wiecek
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Gerard London
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| | - Carmine Zoccali
- From the Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (G.P., J.E.O.); Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy (G.P., J.E.O., G.B.); Indiana University and VAMC, Indianapolis (R.A.); Clinic of Nephrology, C. I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.C.); Department of Clinical Epidemiology, Leiden University Medical Center,
| |
Collapse
|
24
|
Bradley TJ, Slorach C, Mahmud FH, Dunger DB, Deanfield J, Deda L, Elia Y, Har RLH, Hui W, Moineddin R, Reich HN, Scholey JW, Mertens L, Sochett E, Cherney DZI. Early changes in cardiovascular structure and function in adolescents with type 1 diabetes. Cardiovasc Diabetol 2016; 15:31. [PMID: 26879273 PMCID: PMC4754808 DOI: 10.1186/s12933-016-0351-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/05/2016] [Indexed: 01/24/2023] Open
Abstract
Background Children with type 1 diabetes (T1D) are at higher risk of early adult-onset cardiovascular disease. We assessed cardiovascular structure and function in adolescents with T1D compared with healthy controls and the relationships between peripheral vascular function and myocardial parameters. Methods and results 199 T1D [14.4 ± 1.6 years, diabetes duration 6.2 (2.0–12.8) years] and 178 controls (14.4 ± 2.1 years) completed endothelial function by flow mediated vasodilatation (FMD), arterial stiffness using pulse wave velocity (PWV) along with M-mode, pulse wave and tissue Doppler, and myocardial deformation echocardiographic imaging. Systolic (113 ± 10 vs. 110 ± 9 mmHg; p = 0.0005) and diastolic (62 ± 7 vs. 58 ± 7 mmHg; p < 0.0001) blood pressures, carotid femoral PWV and endothelial dysfunction measurements were increased in T1D compared with controls. Systolic and diastolic left ventricular dimensions and function by M-mode and pulse wave Doppler assessment were not significantly different. Mitral valve lateral e’ (17.6 ± 2.6 vs. 18.6 ± 2.6 cm/s; p < 0.001) and a’ (5.4 ± 1.1 vs. 5.9 ± 1.1 cm/s; p < 0.001) myocardial velocities were decreased and E/e’ (7.3 ± 1.2 vs. 6.7 ± 1.3; p = 0.0003) increased in T1D. Left ventricular mid circumferential strain (−20.4 ± 2.3 vs. −19.5 ± 1.7 %; p < 0.001) was higher, whereas global longitudinal strain was lower (−19.0 ± 1.9 vs. −19.8 ± 1.5 % p < 0.001) in T1D. Conclusions Adolescents with T1D exhibit early changes in blood pressure, peripheral vascular function and left ventricular myocardial deformation indices with a shift from longitudinal to circumferential shortening. Longitudinal follow-up of these changes in ongoing prospective trials may allow detection of those most at risk for cardiovascular abnormalities including hypertension that could preferentially benefit from early therapeutic interventions.
Collapse
Affiliation(s)
- Timothy J Bradley
- Department of Paediatrics, Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Cameron Slorach
- Department of Paediatrics, Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Farid H Mahmud
- Department of Paediatrics, Division of Endocrinology, JDRF-Canadian Clinical Trial Network (JDRF-CCTN) Sick Kids Multicenter Clinical Trial Center, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - David B Dunger
- Department of Pediatrics, University of Cambridge, Cambridge, UK.
| | - John Deanfield
- University College Hospital, London, UK. .,Heart Hospital and Great Ormond Street Hospital, London, UK.
| | - Livia Deda
- Department of Paediatrics, Division of Endocrinology, JDRF-Canadian Clinical Trial Network (JDRF-CCTN) Sick Kids Multicenter Clinical Trial Center, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Yesmino Elia
- Department of Paediatrics, Division of Endocrinology, JDRF-Canadian Clinical Trial Network (JDRF-CCTN) Sick Kids Multicenter Clinical Trial Center, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Ronnie L H Har
- Department of Paediatrics, Division of Endocrinology, JDRF-Canadian Clinical Trial Network (JDRF-CCTN) Sick Kids Multicenter Clinical Trial Center, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Wei Hui
- Department of Paediatrics, Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Rahim Moineddin
- Department of Family and Community Medicine, University of Toronto, Toronto, Canada.
| | - Heather N Reich
- Department of Medicine, Division of Nephrology, University Health Network, Toronto General Hospital, University of Toronto, 585 University Avenue, 8 N-845, Toronto, ON, M5G 2N2, Canada.
| | - James W Scholey
- Department of Medicine, Division of Nephrology, University Health Network, Toronto General Hospital, University of Toronto, 585 University Avenue, 8 N-845, Toronto, ON, M5G 2N2, Canada.
| | - Luc Mertens
- Department of Paediatrics, Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - Etienne Sochett
- Department of Paediatrics, Division of Endocrinology, JDRF-Canadian Clinical Trial Network (JDRF-CCTN) Sick Kids Multicenter Clinical Trial Center, The Hospital for Sick Children, University of Toronto, Toronto, Canada.
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, University Health Network, Toronto General Hospital, University of Toronto, 585 University Avenue, 8 N-845, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
25
|
Cho YH, Craig ME, Davis EA, Cotterill AM, Couper JJ, Cameron FJ, Benitez-Aguirre PZ, Dalton RN, Dunger DB, Jones TW, Donaghue KC. Cardiac autonomic dysfunction is associated with high-risk albumin-to-creatinine ratio in young adolescents with type 1 diabetes in AdDIT (adolescent type 1 diabetes cardio-renal interventional trial). Diabetes Care 2015; 38:676-81. [PMID: 25573882 DOI: 10.2337/dc14-1848] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study examined the association between cardiac autonomic dysfunction and high albumin-to-creatinine ratio (ACR) in adolescents with type 1 diabetes. RESEARCH DESIGN AND METHODS Adolescents recruited as part of a multicenter screening study (n = 445, 49% female, aged 10-17 years, mean duration 6.9 years; mean HbA1c 8.4%, 68 mmol/mol) underwent a 10-min continuous electrocardiogram recording for heart rate variability analysis. Time-domain heart rate variability measures included baseline heart rate, SD of the R-R interval (SDNN), and root mean squared difference of successive R-R intervals (RMSSD). Spectral analysis included sympathetic (low-frequency) and parasympathetic (high-frequency) components. Standardized ACR were calculated from six early morning urine collections using an established algorithm, reflecting age, sex, and duration, and stratified into ACR tertiles, where the upper tertile reflects higher nephropathy risk. RESULTS The upper-tertile ACR group had a faster heart rate (76 vs. 73 bpm; P < 0.01) and less heart rate variability (SDNN 68 vs. 76 ms, P = 0.02; RMSSD 63 vs. 71 ms, P = 0.04). HbA1c was 8.5% (69 mmol/mmol) in the upper tertile vs. 8.3% (67 mmol/mol) in the lower tertiles (P = 0.07). In multivariable analysis, upper-tertile ACR was associated with faster heart rate (β = 2.5, 95% CI 0.2-4.8, P = 0.03) and lower RMSSD (β = -9.5, 95% CI -18.2 to -0.8, P = 0.03), independent of age and HbA1c. CONCLUSIONS Adolescents at potentially higher risk for nephropathy show an adverse cardiac autonomic profile, indicating sympathetic overdrive, compared with the lower-risk group. Longitudinal follow-up of this cohort will further characterize the relationship between autonomic and renal dysfunction and the effect of interventions in this population.
Collapse
Affiliation(s)
- Yoon Hi Cho
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, New South Wales, Australia Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Maria E Craig
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, New South Wales, Australia Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia School of Women's and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Elizabeth A Davis
- Department of Endocrinology and Diabetes, Princess Margaret Hospital for Children, Perth, Western Australia, Australia Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Andrew M Cotterill
- Department of Paediatric Endocrinology, Mater Children's Hospital, Brisbane, Queensland, Australia
| | - Jennifer J Couper
- Endocrinology and Diabetes Centre, Women's and Children's Hospital, and Robinson Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Melbourne, Victoria, Australia Murdoch Children's Research Institute, Melbourne, Victoria, Australia The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Z Benitez-Aguirre
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, New South Wales, Australia Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| | - R Neil Dalton
- WellChild Laboratory, St Thomas' Hospital, London, U.K
| | - David B Dunger
- University Department of Paediatrics, Addenbrooke's Hospital, Cambridge, U.K
| | - Timothy W Jones
- Department of Endocrinology and Diabetes, Princess Margaret Hospital for Children, Perth, Western Australia, Australia Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Kim C Donaghue
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, New South Wales, Australia Discipline of Paediatrics and Child Health, The University of Sydney, Sydney, New South Wales, Australia
| | | |
Collapse
|
26
|
Engelen L, Soedamah-Muthu SS, Geleijnse JM, Toeller M, Chaturvedi N, Fuller JH, Schalkwijk CG, Stehouwer CDA. Higher dietary salt intake is associated with microalbuminuria, but not with retinopathy in individuals with type 1 diabetes: the EURODIAB Prospective Complications Study. Diabetologia 2014; 57:2315-23. [PMID: 25172228 PMCID: PMC4181505 DOI: 10.1007/s00125-014-3367-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/05/2014] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS High dietary salt intake has been associated with elevated BP and may also have a deleterious effect on microvascular complications. We studied the cross-sectional associations between dietary salt intake (estimated from 24 h urinary sodium excretion) and urinary potassium excretion on the one hand, and the prevalence of microvascular complications on the other, in individuals with type 1 diabetes. METHODS We measured sodium and potassium concentrations in two 24 h urine samples in 1,212 individuals with type 1 diabetes (40 ± 10 years old, 51% men) who participated in the EURODIAB Prospective Complications Study. We used multiple logistic regression analyses to investigate associations between dietary salt intake and microvascular complications adjusted for age and sex, and additionally for BMI, smoking, urinary potassium excretion, antihypertensive medication and physical activity, and total energy, protein, alcohol, saturated fat and fibre intake. RESULTS After full adjustment, 1 g/day higher dietary salt intake was positively associated with the presence of microalbuminuria (OR 1.06 [95% CI 1.01, 1.10]), but not macroalbuminuria (OR 0.99 [95% CI 0.94, 1.05]), non-proliferative retinopathy (OR 1.00 (95% CI 0.96, 1.04]) or proliferative retinopathy (OR 1.02 (95% CI 0.95, 1.08]). After excluding individuals with cardiovascular disease and/or antihypertensive medication (n = 418), we found a non-significant association with microalbuminuria (OR 1.04 [95% CI 0.99, 1.10]) and macroalbuminuria (OR 1.05 [95% CI 0.96, 1.16]). The association between dietary salt intake and microalbuminuria was stronger in individuals with a BMI above 25 kg/m(2) (OR 1.11 [95% CI 1.04, 1.18]) than in those with BMI below 25 kg/m(2) (OR 1.03 [95% CI 0.97, 1.09]). No significant associations were found between urinary potassium excretion and microvascular complications. CONCLUSIONS/INTERPRETATION In individuals with type 1 diabetes, higher dietary salt intake, as determined by 24 h urinary sodium excretion, may be positively associated with microalbuminuria, particularly in overweight individuals.
Collapse
Affiliation(s)
- Lian Engelen
- Department of Internal Medicine, Maastricht University Medical Centre, Universiteitssingel 50, 6200 MD, Maastricht, the Netherlands,
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Yang GK, Har RLH, Lytvyn Y, Yip P, Cherney DZI. Renal hyperfiltration is associated with glucose-dependent changes in fractional excretion of sodium in patients with uncomplicated type 1 diabetes. Diabetes Care 2014; 37:2774-81. [PMID: 25011944 DOI: 10.2337/dc14-0798] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Renal hyperfiltration is a common abnormality associated with diabetic nephropathy in patients with type 1 diabetes (T1D). In animal models, increased proximal tubular sodium reabsorption results in decreased distal sodium delivery, tubuloglomerular feedback activation, afferent vasodilatation, and hyperfiltration. The role of tubular factors is less well understood in humans. The aim of the current study was therefore to compare the fractional sodium excretion (FENa) in hyperfiltering (T1D-H) versus normofiltering (T1D-N) patients and healthy control (HC) subjects, as well as the role of ambient hyperglycemia on FENa. RESEARCH DESIGN AND METHODS Blood pressure, renal function (inulin for glomerular filtration rate [GFR], and paraaminohippurate for effective renal plasma flow), FENa, and circulating neurohormones were measured in T1D-H (n = 28, GFR ≥135 mL/min/1.73 m(2)), T1D-N (n = 30), and HC (n = 35) subjects during clamped euglycemia. Studies were repeated in a subset of patients during clamped hyperglycemia. RESULTS During clamped euglycemia, T1D-H exhibited lower FENa than T1D-N and HC subjects (0.64 ± 0.06% vs. 0.91 ± 0.12% and 0.90 ± 0.10%, P < 0.05). During clamped hyperglycemia, FENa increased (Δ + 0.88 ± 0.22% vs. Δ + 0.02 ± 0.21%; between-group effect, P = 0.01) significantly in T1D-H, whereas FENa did not change in T1D-N. When treated as continuous variables, elevated GFR values were associated with hyperglycemia-induced increases in FENa (R(2) = 0.20, P = 0.007). CONCLUSIONS Patients with uncomplicated T1D-H exhibit lower FENa under euglycemic conditions, which may help to identify patients with hyperfiltration outside of a controlled laboratory setting. Increased FENa in T1D-H but not T1D-N under clamped hyperglycemic conditions suggests that the mechanisms responsible for increased sodium reabsorption leading to hyperfiltration can be saturated.
Collapse
Affiliation(s)
- Gary K Yang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ronnie L H Har
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Yuliya Lytvyn
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Paul Yip
- University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David Z I Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Donaghue KC, Wadwa RP, Dimeglio LA, Wong TY, Chiarelli F, Marcovecchio ML, Salem M, Raza J, Hofman PL, Craig ME. ISPAD Clinical Practice Consensus Guidelines 2014. Microvascular and macrovascular complications in children and adolescents. Pediatr Diabetes 2014; 15 Suppl 20:257-69. [PMID: 25182318 DOI: 10.1111/pedi.12180] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 06/13/2014] [Indexed: 01/21/2023] Open
Affiliation(s)
- Kim C Donaghue
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Paediatrics and Child Health, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Rollo A, Salardi S, Ciavarella A, Forlani G, Scipione M, Maltoni G, Balsamo C, Martini AL, Zucchini S. Transition from pediatric to adult care. eight years after the transition from pediatric to adult diabetes care: metabolic control, complications and associated diseases. J Endocrinol Invest 2014; 37:653-9. [PMID: 24852416 DOI: 10.1007/s40618-014-0090-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 04/30/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Transition from pediatric to adult care is a critical process in the life of patients with diabetes. AIM Primary aim of the study was to compare the metabolic control between pediatric care and adult care at least 5 years in a group of patients with type 1 diabetes mellitus (T1DM). Secondary aim was to evaluate the presence of complications, associated diseases and psychological-psychiatric disorders. SUBJECTS AND METHODS We obtained data from 73 % (69/94) patients (current mean age 34 years) transferred to local adult centers between 1985 and 2005 at a mean age of 23.8 years. Data were collected for HbA1c, diabetic complications and associated diseases. RESULTS Mean HbA1c did not change during the pediatric, transition and adult period [8.4 ± 1.8 % (68 ± 18 mmol/mol), 8.3 ± 1.4 % (67 ± 15 mmol/mol) and 8.4 ± 1.3 % (68 ± 14 mmol/mol), respectively]. 13 patients dropped out, after 2-12 years since transition, and their HbA1c mean value at transition was 10.4 %. After a mean of 25.9 years of disease, 35/69 patients (50.7 %) showed retinopathy, and 12/69 patients (17.3 %) nephropathy. Thyroid diseases were the most frequent associated diseases (18.3 %), followed by depression (11.2 %) and benign neoplasms (9.8 %). Drug or alcohol addictions were present in four cases (5.6 %). CONCLUSIONS After a mean follow-up of 8 years metabolic control after transition did not change significantly in patients constantly attending to adult care centre. Patients with diabetes onset between 20 and 40 years ago were free from complications in 50 % of cases when considering retinopathy and in more than 80 % considering nephropathy. Thyroid problems were the most common associated diseases. Poor metabolic control at transition is associated with higher risk of drop-out and psychosocial morbidity.
Collapse
Affiliation(s)
- Alessandra Rollo
- Department of Pediatrics, S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 11, 40100, Bologna, Italy,
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Sandholm N, Forsblom C, Mäkinen VP, McKnight AJ, Osterholm AM, He B, Harjutsalo V, Lithovius R, Gordin D, Parkkonen M, Saraheimo M, Thorn LM, Tolonen N, Wadén J, Tuomilehto J, Lajer M, Ahlqvist E, Möllsten A, Marcovecchio ML, Cooper J, Dunger D, Paterson AD, Zerbini G, Groop L, Tarnow L, Maxwell AP, Tryggvason K, Groop PH. Genome-wide association study of urinary albumin excretion rate in patients with type 1 diabetes. Diabetologia 2014; 57:1143-53. [PMID: 24595857 DOI: 10.1007/s00125-014-3202-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/04/2014] [Indexed: 01/15/2023]
Abstract
AIMS/HYPOTHESIS An abnormal urinary albumin excretion rate (AER) is often the first clinically detectable manifestation of diabetic nephropathy. Our aim was to estimate the heritability and to detect genetic variation associated with elevated AER in patients with type 1 diabetes. METHODS The discovery phase genome-wide association study (GWAS) included 1,925 patients with type 1 diabetes and with data on 24 h AER. AER was analysed as a continuous trait and the analysis was stratified by the use of antihypertensive medication. Signals with a p value <10(-4) were followed up in 3,750 additional patients with type 1 diabetes from seven studies. RESULTS The narrow-sense heritability, captured with our genotyping platform, was estimated to explain 27.3% of the total AER variability, and 37.6% after adjustment for covariates. In the discovery stage, five single nucleotide polymorphisms in the GLRA3 gene were strongly associated with albuminuria (p < 5 × 10(-8)). In the replication group, a nominally significant association (p = 0.035) was observed between albuminuria and rs1564939 in GLRA3, but this was in the opposite direction. Sequencing of the surrounding genetic region in 48 Finnish and 48 UK individuals supported the possibility that population-specific rare variants contribute to the synthetic association observed at the common variants in GLRA3. The strongest replication (p = 0.026) was obtained for rs2410601 between the PSD3 and SH2D4A genes. Pathway analysis highlighted natural killer cell mediated immunity processes. CONCLUSIONS/INTERPRETATION This study suggests novel pathways and molecular mechanisms for the pathogenesis of albuminuria in type 1 diabetes.
Collapse
Affiliation(s)
- Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Downes K, Marcovecchio ML, Clarke P, Cooper JD, Ferreira RC, Howson JMM, Jolley J, Nutland S, Stevens HE, Walker NM, Wallace C, Dunger DB, Todd JA. Plasma concentrations of soluble IL-2 receptor α (CD25) are increased in type 1 diabetes and associated with reduced C-peptide levels in young patients. Diabetologia 2014; 57:366-72. [PMID: 24264051 PMCID: PMC3890035 DOI: 10.1007/s00125-013-3113-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 10/30/2013] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is a common autoimmune disease that has genetic and environmental determinants. Variations within the IL2 and IL2RA (also known as CD25) gene regions are associated with disease risk, and variation in expression or function of these proteins is likely to be causal. We aimed to investigate if circulating concentrations of the soluble form of CD25, sCD25, an established marker of immune activation and inflammation, were increased in individuals with type 1 diabetes and if this was associated with the concentration of C-peptide, a measure of insulin production that reflects the degree of autoimmune destruction of the insulin-producing beta cells. METHODS We used immunoassays to measure sCD25 and C-peptide in peripheral blood plasma from patient and control samples. RESULTS We identified that sCD25 was increased in patients with type 1 diabetes compared with controls and replicated this result in an independent set of 86 adult patient and 80 age-matched control samples (p = 1.17 × 10(-3)). In 230 patients under 20 years of age, with median duration-of-disease of 6.1 years, concentrations of sCD25 were negatively associated with C-peptide concentrations (p = 4.8 × 10(-3)). CONCLUSIONS/INTERPRETATION The 25% increase in sCD25 in patients, alongside the inverse association between sCD25 and C-peptide, probably reflect the adverse effects of an on-going, actively autoimmune and inflammatory immune system on beta cell function in patients.
Collapse
Affiliation(s)
- Kate Downes
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
- Present Address: Department of Haematology, NHS Blood and Transplant, University of Cambridge, Long Road, Cambridge, UK
| | - M. Loredana Marcovecchio
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
- Present Address: Department of Paediatrics, University of Chieti, Chieti, Italy
| | - Pamela Clarke
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - Jason D. Cooper
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
- Present Address: Department of Chemical Engineering and Biotechnology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Ricardo C. Ferreira
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - Joanna M. M. Howson
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
- Present Address: Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Jennifer Jolley
- Department of Haematology, NHS Blood and Transplant, University of Cambridge, Long Road, Cambridge, UK
| | - Sarah Nutland
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - Helen E. Stevens
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - Neil M. Walker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - Chris Wallace
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| | - David B. Dunger
- Department of Paediatrics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - John A. Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Addenbrooke’s Hospital, Cambridge, CB2 0XY UK
| |
Collapse
|
32
|
Marcovecchio ML, Woodside J, Jones T, Daneman D, Neil A, Prevost T, Dalton RN, Deanfield J, Dunger DB. Adolescent Type 1 Diabetes Cardio-Renal Intervention Trial (AdDIT): urinary screening and baseline biochemical and cardiovascular assessments. Diabetes Care 2013; 37:805-13. [PMID: 24198300 DOI: 10.2337/dc13-1634] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We assessed the association between early increases in albumin excretion and cardiovascular (CV) and renal markers in a large cohort of young people with type 1 diabetes. RESEARCH DESIGN AND METHODS As part of preliminary screening for a multicenter, randomized controlled trial of statins/ACE inhibitors, we measured albumin-creatinine ratio (ACR) in six early morning urine samples from 3,353 adolescents (10-16 years of age) and calculated tertiles based on an established algorithm. From those subjects deemed to be at higher risk (upper ACR tertile), we recruited 400 into the intervention study (trial cohort). From those subjects deemed to be at lower risk (middle-lower ACR tertiles), we recruited 329 to the observation cohort. At baseline, vascular measurements (carotid intima-media thickness, pulse wave velocity [PWV], flow-mediated dilatation, digital pulse amplitude tonometry), renal markers (symmetric dimethylarginine, cystatin C, creatinine), and CV disease markers (lipids and apolipoproteins [Apo] A-1 and B, C-reactive protein, asymmetric dimethylarginine) were assessed. RESULTS Age- and sex-adjusted PWV was higher in the trial than in the observational cohort (5.00 ± 0.84 vs. 4.86 ± 0.70 m/s; P = 0.021). Similarly, non-HDL cholesterol (2.95 ± 0.83 vs. 2.81 ± 0.78 mmol/L; P = 0.02) and ApoB-ApoA-1 ratio (0.50 ± 0.14 vs. 0.47 ± 0.11; P = 0.04) were higher in the trial cohort. Cystatin C and creatinine were decreased (0.88 ± 0.13 vs. 0.90 ± 0.13 mg/L, P = 0.04; 51.81 ± 10.45 vs. 55.35 ± 11.05 μmol/L, P < 0.001; respectively) and estimated glomerular filtration rate (137.05 ± 23.89 vs. 129.31 ± 22.41 mL/min/1.73 m(2); P < 0.001) increased in the trial compared with the observational cohort. CONCLUSIONS Our data demonstrate that in adolescents with type 1 diabetes, the group with the highest tertile of albumin excretion showed more evidence of early renal and CV disease than those in the lower tertiles.
Collapse
|
33
|
Yang GK, Maahs DM, Perkins BA, Cherney DZI. Renal hyperfiltration and systemic blood pressure in patients with uncomplicated type 1 diabetes mellitus. PLoS One 2013; 8:e68908. [PMID: 23861950 PMCID: PMC3701674 DOI: 10.1371/journal.pone.0068908] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/07/2013] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Patients with type 1 diabetes mellitus (DM) and renal hyperfiltration also exhibit systemic microvascular abnormalities, including endothelial dysfunction. The effect of renal hyperfiltration on systemic blood pressure (BP) is less clear. We therefore measured BP, renal hemodynamic function and circulating renin angiotensin aldosterone system (RAAS) mediators in type 1 DM patients with hyperfiltration (n = 36, DM-H, GFR≥135 ml/min/1.73 m(2)) or normofiltration (n = 40, DM-N), and 56 healthy controls (HC). Since renal hyperfiltration represents a state of intrarenal RAAS activation, we hypothesized that hyperfiltration would be associated with higher BP and elevated levels of circulating RAAS mediators. METHODS BP, glomerular filtration rate (GFR - inulin), effective renal plasma flow (paraaminohippurate) and circulating RAAS components were measured in DM-H, DM-N and HC during clamped euglycemia (4-6 mmol/L). Studies were repeated in DM-H and DM-N during clamped hyperglycemia (9-11 mmol/L). RESULTS Baseline GFR was elevated in DM-H vs. DM-N and HC (167±6 vs. 115±2 and 115±2 ml/min/1.73 m(2), p<0.0001). Baseline systolic BP (SBP, 117±2 vs. 111±2 vs. 109±1, p = 0.004) and heart rate (76±1 vs. 67±1 vs. 61±1, p<0.0001) were higher in DM-H vs. DM-N and HC. Despite higher SBP in DM-H, plasma aldosterone was lower in DM-H vs. DM-N and HC (42±5 vs. 86±14 vs. 276±41 ng/dl, p = 0.01). GFR (p<0.0001) and SBP (p<0.0001) increased during hyperglycemia in DM-N but not in DM-H. CONCLUSIONS DM-H was associated with higher heart rate and SBP values and an exaggerated suppression of systemic aldosterone. Future work should focus on the mechanisms that explain this paradox in diabetes of renal hyperfiltration coupled with systemic RAAS suppression.
Collapse
Affiliation(s)
- Gary K. Yang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David M. Maahs
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Bruce A. Perkins
- Department of Medicine, Division of Endocrinology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - David Z. I. Cherney
- Department of Medicine, Division of Nephrology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
34
|
Blood pressure variability, cardiovascular risk, and risk for renal disease progression. Curr Hypertens Rep 2013; 14:421-31. [PMID: 22903810 DOI: 10.1007/s11906-012-0290-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The adverse cardiovascular consequences of high blood pressure (BP) not only depend on absolute BP values, but also on BP variability (BPV). Evidence has been provided that independently of mean BP levels, BP variations in the short- and long-term are associated with the development, progression and severity of cardiac, vascular and renal organ damage, and with an increased risk of CV events and mortality. Alterations in BPV have also been shown to be predictive of the development and progression of renal damage, which is of relevance if considering that impaired renal function in a hypertensive patient constitutes a very potent predictor of future CV events and mortality even in treated subjects. This review will address whether antihypertensive treatment should target alterations in BPV, in addition to reducing absolute BP levels, in order to achieve the highest CV and renal protection in hypertensive and renal patients.
Collapse
|
35
|
Marcovecchio ML, Tossavainen PH, Heywood JJ, Dalton RN, Dunger DB. An independent effect of parental lipids on the offspring lipid levels in a cohort of adolescents with type 1 diabetes. Pediatr Diabetes 2012; 13:463-9. [PMID: 22369206 DOI: 10.1111/j.1399-5448.2012.00860.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/01/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Genetic factors modulate lipid levels and an intrafamilial aggregation of abnormal lipid profiles has been reported in the general population. As dyslipidemia is common among people with diabetes and has been related to diabetic nephropathy, we investigated whether parental lipid levels were related to lipids and albumin excretion in young offspring with childhood-onset type 1 diabetes (T1D). METHODS Non-fasting blood samples were collected from 895 offspring, 808 mothers and 582 fathers. Total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C) and non-HDL-C were measured. Three early morning urinary albumin-creatinine ratios (ACR), hemoglobin A1C (HbA1c) and anthropometric parameters were also assessed. RESULTS The offspring's mean age (±SD) was 14.5 ± 2.2 yr, mean diabetes duration 5.5 ± 3.7 yr; the fathers' age was 45.7 ± 6.1 yr and the mothers' age was 42.8 ± 5.5 yr. After adjusting for the offspring age, gender, body mass index, HbA1c, maternal (TC: β = 0.242; TG: β = 0.152; HDL-C: β = 0.285; LDL-C: β = 0.278; non-HDL-C: β = 0.253; all p < 0.001) and paternal lipid levels (TC: β = 0.188; TG: β = 0.108; HDL-C: β = 0.253; LDL-C: β = 0.187; non-HDL-C: β = 0.173; all p < 0.001) were significantly associated with the offspring's lipid parameters. In contrast, no significant association was found between parental lipid levels and the offspring's ACR. CONCLUSIONS In the present study, parental lipid levels were independently associated with the same traits in the offspring, suggesting a role of genetic influences and/or shared environmental factors in modulating the metabolic profile of adolescents with T1D. In contrast, there was no significant association between parental lipid levels and the offspring's albumin excretion.
Collapse
|
36
|
Kanaoka T, Tamura K, Ohsawa M, Yanagi M, Haku S, Wakui H, Maeda A, Dejima T, Azushima K, Mitsuhashi H, Okano Y, Fujikawa T, Toya Y, Mizushima S, Tochikubo O, Umemura S. Relationship of ambulatory blood pressure and the heart rate profile with renal function parameters in hypertensive patients with chronic kidney disease. Clin Exp Hypertens 2012; 34:264-9. [PMID: 22578052 DOI: 10.3109/10641963.2012.681082] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Strict blood pressure (BP) control is reportedly important for the management of hypertensive patients with chronic kidney disease (CKD). The purpose of this cross-sectional study was to examine whether the variables of ambulatory BP and the heart rate (HR) profile, central hemodynamics, and arterial stiffness were closely related to the renal function parameters (urine albumin excretion rate [UACR] and estimated glomerular filtration rate [eGFR]) observed in 25 consecutive hospitalized hypertensive patients with CKD. There were significant positive relationships between UACR and 24-hour, daytime, and nighttime ambulatory systolic BP. In addition, there were significant negative relationships between UACR and 24-hour and daytime HR variability. The circulating B-type natriuretic peptide level and hemoglobin A1c were also positively related to UACR. With respect to eGFR, although the 24-hour and nighttime HR variability were positively associated with eGFR, the circulating pentosidine and nighttime HR had a negative relationship with eGFR. On the other hand, central hemodynamics and arterial stiffness did not exhibit any significant association with renal function parameters. These results indicate that ambulatory BP and the HR profile are closely modulated by renal function deterioration. Further studies are needed to investigate the causal relationship between ambulatory BP and the HR profile and renal function parameters in hypertensive patients with CKD.
Collapse
Affiliation(s)
- Tomohiko Kanaoka
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rosengård-Bärlund M, Bernardi L, Sandelin A, Forsblom C, Groop PH. Baroreflex sensitivity and its response to deep breathing predict increase in blood pressure in type 1 diabetes in a 5-year follow-up. Diabetes Care 2011; 34:2424-30. [PMID: 21953796 PMCID: PMC3198281 DOI: 10.2337/dc11-0629] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We have recently demonstrated that early autonomic dysfunction, defined as low baroreflex sensitivity (BRS), could be functional and reversible. However, potential temporal changes in BRS have not yet been addressed by longitudinal studies in type 1 diabetes. Moreover, it is not known whether low BRS predisposes to hypertension or other nonfatal diabetes complications. RESEARCH DESIGN AND METHODS We conducted a 5-year prospective study including 80 patients with type 1 diabetes. We measured ambulatory blood pressure and autonomic function tests. BRS was assessed by six different methods during spontaneous, controlled, and slow deep breathing at baseline and follow-up. RESULTS Spontaneous BRS declined over time (BRS(average) 16.2 ± 0.8 vs. 13.2 ± 0.8 ms/mmHg; P < 0.01), but the change was not significant when adjusted for time of follow-up. Low BRS at baseline did not progress to cardiac autonomic neuropathy but predicted an increase in the nighttime systolic blood pressure (BRS(average) r = -0.37; P < 0.05). Additionally, BRS response to deep breathing at baseline predicted an increase in 24-h ambulatory blood pressure (BRS-αLF r = 0.323-0.346; P < 0.05). CONCLUSIONS The decline in spontaneous BRS over time in patients with type 1 diabetes seems to be due to normal aging, which supports a functional etiology behind early autonomic derangements. Decreased resting BRS and the magnitude of improvement by deep breathing may be due to sympathovagal imbalance, a well-known mechanism in the development of hypertension. Early interventions aiming to reduce sympathetic overactivity in patients with low BRS might delay the development of hypertension.
Collapse
Affiliation(s)
- Milla Rosengård-Bärlund
- Division of Nephrology, Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
38
|
Marcovecchio ML, Dalton RN, Chiarelli F, Dunger DB. A1C variability as an independent risk factor for microalbuminuria in young people with type 1 diabetes. Diabetes Care 2011; 34:1011-3. [PMID: 21335371 PMCID: PMC3064014 DOI: 10.2337/dc10-2028] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the potential association between A1C variability (A1C-SD) and microalbuminuria in young people with type 1 diabetes. RESEARCH DESIGN AND METHODS Serially collected samples for A1C measurement were available for 1,232 subjects with childhood-onset type 1 diabetes recruited to the Oxford Regional Prospective Study and the Nephropathy Family Study. RESULTS The median (range) number of A1C assessments was 4 (2-16). Mean intrapersonal A1C was 9.5% and A1C-SD was 0.91. Mean A1C and A1C-SD values were higher in subjects with microalbuminuria (n = 227) than in those with normoalbuminuria (10.3 vs. 9.4%; 1.12 vs. 0.86, P < 0.001). In a Cox regression model, A1C-SD was independently associated with microalbuminuria (hazard ratio 1.31 [95% CI 1.01-1.35]). CONCLUSIONS In the current study, A1C variability was an independent variable that added to the effect of A1C on the risk for microalbuminuria in youth with type 1 diabetes, a population highly vulnerable to vascular complications.
Collapse
|
39
|
Marcovecchio ML, Chiarelli F. Microvascular disease in children and adolescents with type 1 diabetes and obesity. Pediatr Nephrol 2011; 26:365-75. [PMID: 20721674 DOI: 10.1007/s00467-010-1624-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 07/13/2010] [Accepted: 07/14/2010] [Indexed: 12/16/2022]
Abstract
The incidence of type 1 diabetes (T1D) is increasing worldwide and is associated with a significant burden, mainly related to the development of vascular complications. Over the last decades, concomitant with the epidemic of childhood obesity, there has been an increasing number of cases of type 2 diabetes (T2D) among children and adolescents. Microvascular complications of diabetes, which include nephropathy, retinopathy and neuropathy, are characterized by damage to the microvasculature of the kidney, retina and neurons. Although clinically evident microvascular complications are rarely seen among children and adolescents with diabetes, there is clear evidence that their pathogenesis and early signs develop during childhood and accelerate during puberty. Diabetic vascular complications are often asymptomatic during their early stages, and once symptoms develop, there is little to be done to cure them. Therefore, screening needs to be started early during adolescence and, in the case of T2D, already at diagnosis. Identification of risk factors and subclinical signs of complications is essential for the early implementation of preventive and therapeutic strategies, which could change the course of vascular complications and improve the prognosis of children, adolescents and young adults with diabetes.
Collapse
|
40
|
Lemley KV. When to initiate ACEI/ARB therapy in patients with type 1 and 2 diabetes. Pediatr Nephrol 2010; 25:2021-34. [PMID: 20352458 DOI: 10.1007/s00467-010-1498-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 02/08/2010] [Accepted: 02/16/2010] [Indexed: 10/19/2022]
Abstract
Angiotensin converting enzyme inhibitors (ACEI) and angiotensin 2 receptor blockers (ARB) have become a mainstay of adjunctive therapy for the prevention and amelioration of diabetic nephropathy. Although ACEI were shown over 20 years ago to slow the rate of loss of renal function in diabetic subjects with decreased renal function, the question of how early in the course of diabetes to introduce them remains unresolved. Recent studies suggest that very early initiation of ACEI/ARB therapy may not have demonstrable beneficial effects even over a period of years.
Collapse
Affiliation(s)
- Kevin V Lemley
- Division of Nephrology, MS#40, Childrens Hospital Los Angeles, 4650 Sunset Boulevard, Los Angeles, CA 90027, USA.
| |
Collapse
|
41
|
Marcovecchio ML, Tossavainen PH, Acerini CL, Barrett TG, Edge J, Neil A, Shield J, Widmer B, Dalton RN, Dunger DB. Maternal but not paternal association of ambulatory blood pressure with albumin excretion in young offspring with type 1 diabetes. Diabetes Care 2010; 33:366-71. [PMID: 19918004 PMCID: PMC2809284 DOI: 10.2337/dc09-1152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Familial predisposition to hypertension has been associated with the development of diabetic nephropathy in adults, but there are limited data in adolescents. Our aim was to assess whether parental ambulatory blood pressure (ABP) was associated with ABP and albumin excretion in young offspring with type 1 diabetes. RESEARCH DESIGN AND METHODS Twenty-four-hour ABP monitoring was performed in 509 young offspring (mean +/- SD age 15.8 +/- 2.3 years) with type 1 diabetes, 311 fathers, and 444 mothers. Systolic (SBP) and diastolic blood pressure (DBP) measurements during 24 h, daytime, and nighttime were calculated. Three early morning urinary albumin-to-creatinine ratios (ACRs), A1C, and anthropometric parameters were available for the offspring. RESULTS All paternal ABP parameters, except for nighttime SBP, were independently related to the offspring's ABP (24-h SBP beta = 0.18, 24-h DBP beta = 0.22, daytime SBP beta = 0.25, daytime DBP beta = 0.23, and nighttime DBP beta = 0.18; all P < 0.01). Maternal 24-h DBP (beta = 0.19, P = 0.004), daytime DBP (beta = 0.09, P = 0.04), and nighttime SBP (beta = 0.24 P = 0.001) were related to the corresponding ABP parameter in the offspring. Significant associations were found between the offspring's logACR and maternal ABP. The association with 24-h DBP (beta = 0.16, P = 0.02), daytime DBP (beta = 0.16 P = 0.02), and nighttime DBP (beta = 0.15 P = 0.03) persisted even after adjustment for the offspring's ABP. Mothers of offspring with microalbuminuria had higher ABP than mothers of offspring without microalbuminuria (all P < 0.05). CONCLUSIONS In this cohort, parental ABP significantly influenced offspring blood pressure, therefore confirming familial influences on this trait. In addition, maternal ABP, particularly DBP, was closely related to ACR in the offspring, suggesting a dominant effect of maternal genes or an effect of the intrauterine environment on microalbuminuria risk.
Collapse
|
42
|
Marcovecchio ML, Tossavainen PH, Dunger DB. Prevention and treatment of microvascular disease in childhood type 1 diabetes. Br Med Bull 2010; 94:145-64. [PMID: 20053672 DOI: 10.1093/bmb/ldp053] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The incidence of type 1 diabetes (T1D) is increasing worldwide, particularly in children, and is associated with a significant burden, mainly related to the development of vascular complications. The prevention and treatment of microvascular complications, which include nephropathy, retinopathy and neuropathy, are of paramount importance to decrease the associated mortality and morbidity. SOURCES OF DATA A literature search was performed on Medline and articles on microvascular complications, with particular emphasis on the increasing incidence of childhood T1D and its implications on prevention and treatment of complications, were selected. AREAS OF AGREEMENT The incidence of childhood T1D is increasing. Early identification of subjects at risk for long-term complications and early implementation of preventive and therapeutic strategies are fundamental in order to reduce the burden associated with microvascular complications of diabetes. Improving glycaemic control is the principle way of preventing and treating T1D complications. AREAS OF CONTROVERSY In adults with T1D and microvascular complications, treatment with anti-hypertensive drugs and statins is increasingly common, whereas there are no definitive indications for treatment with these drugs in children and adolescents with early signs of complications. GROWING POINTS There is growing interest in the development of new preventive and therapeutic strategies targeting specific pathways implicated in the pathogenesis of microvascular complications. AREAS TIMELY FOR DEVELOPING RESEARCH Investigations to clarify genetic and environmental factors implicated in the pathogenesis of microvascular complications could lead to the identification of biochemical markers with high predictive values, to be used as a guide for screening and intervention programmes.
Collapse
Affiliation(s)
- M L Marcovecchio
- Department of Paediatrics and Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
43
|
Abstract
BACKGROUND The prognosis for young people diagnosed with diabetes during childhood remains poor and this is mainly related to the long-term risk of developing vascular complications.Microalbuminuria identifies subjects at risk for diabetic nephropathy (DN) and cardiovascular disease (CVD). It is often detected in adolescence but is rarely treated before the age of 18 years, as at the end of puberty albumin excretion may decline and in some subjects will return into the normal range. However, evidence indicates that subjects with both transient and persistent microalbuminuria have experienced renal damage during puberty and thus reno-protection to prevent long-term complications is warranted. In adults with diabetes and microalbuminuria, the use of angiotensin converting enzyme inhibitors (ACEI) and Statins is increasing, and in order to determine whether these agents are of value in the adolescent population a large randomized controlled clinical trial is needed. METHODS/DESIGN The Adolescent type 1 Diabetes cardio-renal Intervention Trial (AdDIT) is a multi-center, randomized, double-blind, placebo-controlled trial of ACEI and Statin therapy in adolescents with type 1 diabetes. 500 high-risk adolescents, defined on the basis of their albumin excretion, are randomized to receive either ACEI (Quinapril) or Statins (Atorvastatin) or combination therapy or placebo for 3-4 years. There will also be a parallel open observational study, based on the follow-up of 400 low-risk non-randomized adolescents. The major endpoint of the study is the change in albumin excretion; secondary endpoints include markers of CVD, renal function, retinopathy, quality of life combined with assessment of compliance and potential health economic benefits. DISCUSSION AdDIT will provide important data on the potential renal and cardiovascular protective effects of ACEI and Statins in high-risk adolescents. Long-term follow-up of the randomized subjects will provide direct evidence of disease outcomes, in addition to the data on early surrogate measures of DN and CVD. Follow-up of non-randomized low-risk subjects will determine the potential impact of intervention on DN and CVD. AdDIT will help to determine whether, in addition to encouraging young people to achieve good glycaemic control, pharmacological cardio-renal protection should also be implemented. EUDRACT NUMBER: 2007-001039-72 TRIAL REGISTRATION NUMBER ISRCTN91419926.
Collapse
|
44
|
Current literature in diabetes. Diabetes Metab Res Rev 2009; 25:i-x. [PMID: 19790194 DOI: 10.1002/dmrr.1037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|