1
|
Tsaban G, Aharon-Hananel G, Shalem S, Zelicha H, Yaskolka Meir A, Pachter D, Goldberg DT, Kamer O, Alufer L, Stampfer MJ, Wang DD, Qi L, Blüher M, Stumvoll M, Hu FB, Shai I, Tirosh A. The effect of Mankai plant consumption on postprandial glycaemic response among patients with type 2 diabetes: A randomized crossover trial. Diabetes Obes Metab 2024; 26:4713-4723. [PMID: 39134456 DOI: 10.1111/dom.15840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 09/19/2024]
Abstract
AIM To explore the effect of Mankai, a cultivated aquatic duckweed green plant, on postprandial glucose (PG) excursions in type 2 diabetes (T2D). METHODS In a 4-week, randomized crossover-controlled trial, we enrolled 45 adults with T2D (HbA1c range: 6.5%-8.5%) from two sites in Israel. Participants were randomized to drink Mankai (200 mL of raw-fresh-aquatic plant + 100 mL of water, 40 kcal, ~10 g of dry matter equivalent) or water (300 mL) following dinner, for 2 weeks each, with a 4-day washout interval, without dietary, physical activity or pharmacotherapy alterations. We used continuous glucose monitoring (CGM) devices. RESULTS Forty patients (adherence rate = 88.5%; 743 person-intervention-days, 68.9% men, age = 64 years, HbA1c = 6.8%) completed the study with a consistent diet and complete CGM reads. Only two-thirds of the individuals responded beneficially to Mankai. Overall, Mankai significantly lowered the PG peak by 19.3% (∆peak = 24.3 ± 16.8 vs. 30.1 ± 18.5 mg/dL; P < .001) and delayed the time-to-peak by 20.0% (112.5 [interquartile range: 75-135] vs. 90 [60-105] min; P < .001) compared with water. The PG incline and decline slopes were shallower following postdinner Mankai (incline slope: 16.8 vs. water: 29.9 mg/[dL h]; P < .001; decline slope: -6.1 vs. water: -7.9 mg/[dL h]; P < .01). Mean postprandial net incremental area-under-the-glucose-curve was lowered by 20.1% with Mankai compared with water (P = .03). Results were consistent across several sensitivity and subgroup analyses, including across antidiabetic pharmacotherapy treatment groups. Within 2 weeks, the triglycerides/high-density lipoprotein cholesterol ratio in the Mankai group (-0.5 ± 1.3) decreased versus water (+0.3 ± 1.5, P = .05). CONCLUSIONS Mankai consumption may mitigate the PG response in people with T2D with an ~20% improvement in glycaemic values. These findings provide case-study evidence for plant-based treatments in T2D to complement a healthy lifestyle and pharmacotherapy.
Collapse
Affiliation(s)
- Gal Tsaban
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
- Department of Cardiology, Soroka University Medical Centre, Beersheva, Israel
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Genya Aharon-Hananel
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Centre, Ramat Gan, Israel
| | - Shiran Shalem
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Centre, Ramat Gan, Israel
| | - Hila Zelicha
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
| | - Anat Yaskolka Meir
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Dafna Pachter
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
| | - Dana Tamar Goldberg
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
| | - Omer Kamer
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
| | - Liav Alufer
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
| | - Meir J Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dong D Wang
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Matthias Blüher
- Medical Department III-Endocrinology, Nephrology, Rheumatology and Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre Munich at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | | | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Iris Shai
- The Health and Nutrition Innovative International Research Centre, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Amir Tirosh
- Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Centre, Ramat Gan, Israel
| |
Collapse
|
2
|
Aizawa K, Hughes AD, Casanova F, Gooding KM, Gates PE, Mawson DM, Williams J, Goncalves I, Nilsson J, Khan F, Colhoun HM, Palombo C, Parker KH, Shore AC. Reservoir-excess pressure parameters are independently associated with NT-proBNP in older adults. ESC Heart Fail 2024; 11:3290-3298. [PMID: 38946623 PMCID: PMC11424359 DOI: 10.1002/ehf2.14926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024] Open
Abstract
AIMS Parameters derived from reservoir-excess pressure analysis have been demonstrated to predict cardiovascular events. Thus, altered reservoir-excess pressure parameters could have a detrimental effect on highly-perfused organs like the heart. We aimed to cross-sectionally determine whether reservoir-excess pressure parameters were associated with N-terminal pro-brain-type natriuretic peptide (NT-proBNP) in older adults. METHODS We studied 868 older adults with diverse cardiovascular risk. Reservoir-excess pressure parameters were obtained through radial artery tonometry including reservoir pressure integral, peak reservoir pressure, excess pressure integral (INTXSP), systolic rate constant (SRC) and diastolic rate constant (DRC). Plasma levels of NT-proBNP, as a biomarker of cardiac overload, were analysed by the Proximity Extension Assay technology. RESULTS Multivariable linear regression analyses revealed that all reservoir-excess pressure parameters studied were associated with NT-proBNP after adjusting for age and sex. After further adjustments for conventional cardiovascular risk factors, INTXSP [β = 0.191 (95% confidence interval, CI: 0.099, 0.283), P < 0.001], SRC [β = -0.080 (95% CI: -0.141, -0.019), P = 0.010] and DRC [β = 0.138 (95% CI: 0.073, 0.202), P < 0.001] remained associated with NT-proBNP. Sensitivity analysis found that there were occasions where the association between SRC and NT-proBNP was attenuated, but both INTXSP and DRC remained consistently associated with NT-proBNP. CONCLUSIONS The observed associations between reservoir-excess pressure parameters and NT-proBNP suggest that altered reservoir-excess pressure parameters may reflect an increased load inflicted on the left ventricular cardiomyocytes and could have a potential to be utilized in the clinical setting for cardiovascular risk stratification.
Collapse
Affiliation(s)
- Kunihiko Aizawa
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - Alun D. Hughes
- MRC unit for Lifelong Health and Ageing, Institute of Cardiovascular ScienceUniversity College LondonLondonUK
| | - Francesco Casanova
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - Kim M. Gooding
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - Phillip E. Gates
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - David M. Mawson
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - Jennifer Williams
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| | - Isabel Goncalves
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden
- Department of CardiologySkåne University HospitalMalmöSweden
| | - Jan Nilsson
- Department of Clinical Sciences MalmöLund UniversityMalmöSweden
| | - Faisel Khan
- Division of Systems MedicineUniversity of DundeeDundeeUK
| | - Helen M. Colhoun
- Centre for Genomic and Experimental MedicineUniversity of EdinburghEdinburghUK
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular and Critical Area PathologyUniversity of PisaPisaItaly
| | - Kim H. Parker
- Department of BioengineeringImperial CollegeLondonUK
| | - Angela C. Shore
- Vascular Research Centre, NIHR Exeter Clinical Research FacilityUniversity of Exeter Medical SchoolExeterUK
| |
Collapse
|
3
|
Al Hageh C, O'Sullivan S, Platt DE, Henschel A, Chacar S, Gauguier D, Abchee A, Alefishat E, Nader M, Zalloua PA. Coronary artery disease patients with rs7904519 (TCF7L2) are at a persistent risk of type 2 diabetes. Diabetes Res Clin Pract 2024; 207:111052. [PMID: 38072013 DOI: 10.1016/j.diabres.2023.111052] [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: 06/13/2023] [Revised: 10/04/2023] [Accepted: 12/07/2023] [Indexed: 02/10/2024]
Abstract
AIMS Type 2 diabetes (T2D) and coronary artery disease (CAD) often coexist and share genetic factors.This study aimed to investigate the common genetic factors underlying T2D and CAD in patients with CAD. METHODS A three-step association approach was conducted: a) a discovery step involving 943 CAD patients with T2D and 1,149 CAD patients without T2D; b) an eliminating step to exclude CAD or T2D specific variants; and c) a replication step using the UK Biobank data. RESULTS Ten genetic loci were associated with T2D in CAD patients. Three variants were specific to either CAD or T2D. Five variants lost significance after adjusting for covariates, while two SNPs remained associated with T2D in CAD patients (rs7904519*G: TCF7L2 and rs17608766*C: GOSR2). The T2D susceptibility rs7904519*G was associated with increased T2D risk, while the CAD susceptibility rs17608766*C was negatively associated with T2D in CAD patients. These associations were replicated in a UK Biobank data, confirming the results. CONCLUSIONS No significant common T2D and CAD susceptibility genetic association was demonstrated indicating distinct disease pathways. However, CAD patients carrying the T2D susceptibility gene TCF7L2 remain at higher risk for developing T2D emphasizing the need for frequent monitoring in this subgroup.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Siobhan O'Sullivan
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Daniel E Platt
- Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, NY, USA
| | - Andreas Henschel
- Department of Electrical Engineering and Computer, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stephanie Chacar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dominique Gauguier
- Université Paris Cité, INSERM UMR 1124, 45 rue des Saint-Pères, 75006 Paris, France; McGill University and Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada
| | | | - Eman Alefishat
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi UAE
| | - Moni Nader
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| | - Pierre A Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
4
|
Genovese F, Gonçalves I, Holm Nielsen S, Karsdal MA, Edsfeldt A, Nilsson J, Shore AC, Natali A, Khan F, Shami A. Plasma levels of PRO-C3, a type III collagen synthesis marker, are associated with arterial stiffness and increased risk of cardiovascular death. Atherosclerosis 2024; 388:117420. [PMID: 38128431 DOI: 10.1016/j.atherosclerosis.2023.117420] [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: 09/11/2023] [Revised: 11/17/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND AND AIMS The N-terminal propeptide of type III collagen (PRO-C3) assay measures a pro-peptide released during type III collagen synthesis, an important feature of arterial stiffening and atherogenesis. There is a clinical need for improved non-invasive, cheap and easily accessible methods for evaluating individuals at risk of cardiovascular disease (CVD). In this study, we investigate the potential of using circulating levels of PRO-C3 to mark the degree of vascular stenosis and risk of cardiovascular events. METHODS Baseline plasma levels of PRO-C3 were measured by ELISA in subjects belonging to the SUrrogate markers for Micro- and Macro-vascular hard endpoints for Innovative diabetes Tools (SUMMIT) cohort (N = 1354). Associations between PRO-C3 levels with vascular characteristics, namely stiffness and stenosis, and risk of future cardiovascular events were explored. Subjects were followed up after a median of 35 months (interquartile range 34-36 months), with recorded outcomes cardiovascular death and all-cause mortality. RESULTS We found a correlation between PRO-C3 levels and pulse wave velocity (rho 0.13, p = 0.000009), a measurement of arterial stiffness. Higher PRO-C3 levels were also associated with elevated blood pressure (rho 0.07, p = 0.014), as well as risk of cardiovascular mortality over a three-year follow-up period (OR 1.56, confidence interval 1.008-2.43, p = 0.046). CONCLUSIONS Elevated circulating PRO-C3 levels are associated with arterial stiffness and future cardiovascular death, in the SUMMIT cohort, suggesting a potential value of PRO-C3 as a novel marker for declining vascular health.
Collapse
Affiliation(s)
| | - Isabel Gonçalves
- Dept. of Clinical Sciences Malmö, Lund University, Clinical Research Center, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden; Dept. of Cardiology, Malmö, Skåne University Hospital, Lund University, Carl-Bertil Laurells Gata 9, 214 28, Malmö, Sweden
| | - Signe Holm Nielsen
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark; Biomedicine and Biotechnology, Technical University of Denmark, Søltofts Pl. 221, 2800, Kongens Lyngby, Denmark
| | - Morten A Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Andreas Edsfeldt
- Dept. of Clinical Sciences Malmö, Lund University, Clinical Research Center, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden; Dept. of Cardiology, Malmö, Skåne University Hospital, Lund University, Carl-Bertil Laurells Gata 9, 214 28, Malmö, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Klinikgatan 32, 221 84, Lund, Sweden
| | - Jan Nilsson
- Dept. of Clinical Sciences Malmö, Lund University, Clinical Research Center, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter, Medical School, National Institute for Health Research Exeter Clinical Research Facility, Barrack Road, Exeter, EX2 5AX, UK
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 8, 56100, Pisa, Italy
| | - Faisel Khan
- Division of Systems Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Annelie Shami
- Dept. of Clinical Sciences Malmö, Lund University, Clinical Research Center, Jan Waldenströms Gata 35, 214 28, Malmö, Sweden.
| |
Collapse
|
5
|
Aizawa K, Gates PE, Mawson DM, Casanova F, Gooding KM, Hope SV, Goncalves I, Nilsson J, Khan F, Colhoun HM, Natali A, Palombo C, Shore AC. Type 2 diabetes exacerbates changes in blood pressure-independent arterial stiffness: cross-sectional and longitudinal evidence from the SUMMIT study. J Appl Physiol (1985) 2024; 136:13-22. [PMID: 37969084 PMCID: PMC11208039 DOI: 10.1152/japplphysiol.00283.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/19/2023] [Accepted: 11/12/2023] [Indexed: 11/17/2023] Open
Abstract
Greater central artery stiffness is observed in people with type 2 diabetes (T2DM). Elevated blood pressure (BP) and altered arterial wall structure/composition in T2DM are generally considered as main drivers for this alteration. However, because conventional arterial stiffness measures are BP-dependent and as such an influence of BP remains in a measure, it is unclear if greater central artery stiffness is a function of greater BP, or due to changes in the structure and composition of the arterial wall. We aimed to measure BP-independent arterial stiffness (β0) cross-sectionally and longitudinally in T2DM. We studied 753 adults with T2DM (DM+) and 436 adults without (DM-) at baseline (Phase 1), and 310 DM+ and 210 DM- adults at 3-yr follow-up (Phase 2). We measured carotid-femoral pulse wave velocity and used it to calculate β0. In Phase 1, β0 was significantly greater in DM+ than DM- after adjusting for age and sex [27.5 (26.6-28.3) vs. 23.6 (22.4-24.8) au, P < 0.001]. Partial correlation analyses after controlling for age and sex showed that β0 was significantly associated with hemoglobin A1c (r = 0.15 P < 0.001) and heart rate [(HR): r = 0.23 P < 0.001)] in DM+. In Phase 2, percentage-change in β0 was significantly greater in DM+ than DM- [19.5 (14.9-24.0) vs. 5.0 (-0.6 to 10.6) %, P < 0.001] after adjusting for age, sex, and baseline β0. β0 was greater in DM+ than DM- and increased much more in DM+ than in DM- over 3 yr. This suggests that T2DM exacerbates BP-independent arterial stiffness and may have a complemental utility to existing arterial stiffness indices.NEW & NOTEWORTHY We demonstrate in this study a greater BP-independent arterial stiffness β0 in people with type 2 diabetes (T2DM) compared to those without, and also a greater change in β0 over 3 yr in people with T2DM than those without. These findings suggest that the intrinsic properties of the arterial wall may change in a different and more detrimental way in people with T2DM and likely represents accumulation of cardiovascular risk.
Collapse
Affiliation(s)
- Kunihiko Aizawa
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - Phillip E Gates
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - David M Mawson
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - Francesco Casanova
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - Kim M Gooding
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - Suzy V Hope
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Faisel Khan
- Division of Systems Medicine, University of Dundee, Dundee, United Kingdom
| | - Helen M Colhoun
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Angela C Shore
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Exeter, United Kingdom
| |
Collapse
|
6
|
Edsfeldt A, Gonçalves I, Vigren I, Jovanović A, Engström G, Shore AC, Natali A, Khan F, Nilsson J. Circulating soluble IL-6 receptor associates with plaque inflammation but not with atherosclerosis severity and cardiovascular risk. Vascul Pharmacol 2023; 152:107214. [PMID: 37634789 DOI: 10.1016/j.vph.2023.107214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/21/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND The residual cardiovascular risk in subjects receiving guideline-recommended therapy is related to persistent vascular inflammation and IL-6 represents a target for its treatment. IL-6 binds to receptors on leukocytes and hepatocytes and/or by forming complexes with soluble IL-6 receptors (sIL-6R) binding to gp130 which is present on all cells. Here we aimed to estimate the associations of these two pathways with risk of cardiovascular disease (CVD). METHODS IL-6 and sIL-6R were analyzed using the proximity extension assay. Baseline plasma samples were obtained from participants in the prospective Malmö Diet and Cancer (MDC) study (n = 4661), the SUMMIT VIP study (n = 1438) and the Carotid Plaque Imaging Project (CPIP, n = 285). Incident clinical events were obtained through national registers. Plaques removed at surgery were analyzed by immunohistochemistry and biochemical methods. RESULTS During 23.1 ± 7.0 years follow-up, 575 subjects in the MDC cohort suffered a first myocardial infarction. Subjects in the highest tertile of IL-6 had an increased risk compared to the lowest tertile (HR and 95% CI 2.60 [2.08-3.25]). High plasma IL-6 was also associated with more atherosclerosis, increased arterial stiffness, and impaired endothelial function in SUMMIT VIP, but IL-6 was only weakly associated with plaque inflammation in CPIP. sIL-6R showed no independent association with risk of myocardial infarction, atherosclerosis severity or vascular function, but was associated with plaque inflammation. CONCLUSIONS Our findings show that sIL-6R is a poor marker of CVD risk and associated vascular changes. However, the observation that sIL-6R reflects plaque inflammation highlights the complexity of the role of IL-6 in CVD.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Cardiology, Skåne University Hospital, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Sweden
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Cardiology, Skåne University Hospital, Sweden
| | - Isa Vigren
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Anja Jovanović
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Faisel Khan
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Sweden.
| |
Collapse
|
7
|
Aizawa K, Hughes AD, Casanova F, Gates PE, Mawson DM, Gooding KM, Gilchrist M, Goncalves I, Nilsson J, Khan F, Colhoun HM, Palombo C, Parker KH, Shore AC. Reservoir Pressure Integral Is Independently Associated With the Reduction in Renal Function in Older Adults. Hypertension 2022; 79:2364-2372. [PMID: 35993228 DOI: 10.1161/hypertensionaha.122.19483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Arterial hemodynamic parameters derived from reservoir-excess pressure analysis exhibit prognostic utility. Reservoir-excess pressure analysis may provide useful information about an influence of altered hemodynamics on target organ such as the kidneys. We determined whether the parameters derived from the reservoir-excess pressure analysis were associated with the reduction in estimated glomerular filtration rate in 542 older adults (69.4±7.9 years, 194 females) at baseline and after 3 years. METHODS Reservoir-excess pressure parameters, including reservoir pressure integral, excess pressure integral, systolic, and diastolic rate constants, were obtained by radial artery tonometry. RESULTS After 3 years, and in a group of 94 individuals (72.4±7.6 years, 26 females), there was an estimated glomerular filtration rate reduction of >5% per year (median reduction of 20.5% over 3 years). A multivariable logistic regression analysis revealed that higher baseline reservoir pressure integral was independently associated with a smaller reduction in estimated glomerular filtration rate after accounting for conventional cardiovascular risk factors and study centers (odds ratio: 0.660 [95% CIs, 0.494-0.883]; P=0.005). The association remained unchanged after further adjustments for potential confounders and baseline renal function (odds ratio: 0.528 [95% CIs, 0.351-0.794]; P=0.002). No other reservoir-excess pressure parameters exhibited associations with the reduction in renal function. CONCLUSIONS This study demonstrates that baseline reservoir pressure integral was associated with the decline in renal function in older adults at 3-year follow-up, independently of conventional cardiovascular risk factors. This suggests that reservoir pressure integral may play a role in the functional decline of the kidneys.
Collapse
Affiliation(s)
- Kunihiko Aizawa
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - Alun D Hughes
- MRC unit for Lifelong Health and Ageing, Institute of Cardiovascular Science, University College London, United Kingdom (A.D.H.)
| | - Francesco Casanova
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - Phillip E Gates
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - David M Mawson
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - Kim M Gooding
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - Mark Gilchrist
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| | - Isabel Goncalves
- Department of Clinical Sciences, Lund University, Malmö, Sweden (I.G., J.N.).,Department of Cardiology, Skåne University Hospital, Malmö, Sweden (I.G.)
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden (I.G., J.N.)
| | - Faisel Khan
- Division of Systems Medicine, University of Dundee, United Kingdom (F.K.)
| | - Helen M Colhoun
- Centre for Genomic and Experimental Medicine, University of Edinburgh, United Kingdom (H.M.C.)
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Italy (C.P.)
| | - Kim H Parker
- Department of Bioengineering, Imperial College, London, United Kingdom (K.H.P.)
| | - Angela C Shore
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., M.G., A.C.S.)
| |
Collapse
|
8
|
Khan F, Gonçalves I, Shore AC, Natali A, Palombo C, Colhoun HM, Östling G, Casanova F, Kennbäck C, Aizawa K, Persson M, Gooding KM, Strain D, Looker H, Dove F, Belch J, Pinnola S, Venturi E, Kozakova M, Nilsson J. Plaque characteristics and biomarkers predicting regression and progression of carotid atherosclerosis. Cell Rep Med 2022; 3:100676. [PMID: 35858591 PMCID: PMC9381367 DOI: 10.1016/j.xcrm.2022.100676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 04/10/2022] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
The factors that influence the atherosclerotic disease process in high-risk individuals remain poorly understood. Here, we used a combination of vascular imaging, risk factor assessment, and biomarkers to identify factors associated with 3-year change in carotid disease severity in a cohort of high-risk subjects treated with preventive therapy (n = 865). The results show that changes in intima-media thickness (IMT) are most pronounced in the carotid bulb. Progression of bulb IMT demonstrates independent associations with baseline bulb IMT, the plaque gray scale median (GSM), and the plasma level of platelet-derived growth factor (PDGF) (standardized β-coefficients and 95% confidence interval [CI] −0.14 [−0.06 to −0.02] p = 0.001, 0.15 [0.02–0.07] p = 0.001, and 0.20 [0.03–0.07] p < 0.001, respectively). Plasma PDGF correlates with the plaque GSM (0.23 [0.15–0.29] p < 0.001). These observations provide insight into the atherosclerotic process in high-risk subjects by showing that progression primarily occurs in fibrotic plaques and is associated with increased levels of PDGF. High age, male gender, and smoking increases risk of carotid disease progression Plaques that progress are more echogenic, indicating an increased degree of fibrosis Progression is associated with high plasma levels of pro-fibrotic growth factors Regression is most common in large, less fibrotic plaques
Collapse
Affiliation(s)
- Faisel Khan
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Gerd Östling
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Francesco Casanova
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Cecilia Kennbäck
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Kunihiko Aizawa
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | | | - Kim M Gooding
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - David Strain
- Diabetes and Vascular Medicine, University of Exeter Medical School and NIHR Exeter Clinical Research Facility, Exeter, UK
| | - Helen Looker
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Fiona Dove
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Jill Belch
- Division of Systems Medicine, University of Dundee, Dundee, UK
| | - Silvia Pinnola
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Venturi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy; Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Nagy EE, Puskás A, Kelemen P, Makó K, Brassai Z, Hársfalvi J, Frigy A. Elevated Serum Cystatin C and Decreased Cathepsin S/Cystatin C Ratio Are Associated with Severe Peripheral Arterial Disease and Polyvascular Involvement. Diagnostics (Basel) 2022; 12:diagnostics12040833. [PMID: 35453881 PMCID: PMC9029365 DOI: 10.3390/diagnostics12040833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral arterial disease (PAD) is frequently associated with atherosclerotic manifestations of the carotids and coronaries. Polyvascular involvement and low ankle−brachial index predict major cardiovascular events and high mortality. Cathepsin S (Cat S) promotes the inflammatory pathways of the arterial wall, while Cystatin C (Cys C) functions as its inhibitor; therefore, Cys C was proposed to be a biomarker of progression in PAD. In a single-center observational study, we investigated the correlations of serum Cys C and Cat S/Cys C ratio in a group of 90 PAD patients, predominantly with polyvascular involvement. Cys C and Cat S/Cys C were associated with ankle−brachial index (ABI) scores <0.4 in univariate and multiple regression models. Furthermore, both markers correlated positively with the plasma Von Willebrand Factor Antigen (VWF: Ag) and Von Willebrand Factor collagen-binding activity (VWF: CB). In addition, Cat S/Cys C was significantly decreased, whereas Cys C increased in subjects with three-bed atherosclerotic involvement. According to our results, high serum Cys C and low Cat S/Cys C ratios may indicate severe peripheral arterial disease and polyvascular atherosclerotic involvement.
Collapse
Affiliation(s)
- Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
- Correspondence: ; Tel.: +40-733-956-395
| | - Attila Puskás
- Angio-Center Vascular Medicine, 540074 Targu Mures, Romania;
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Piroska Kelemen
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Katalin Makó
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
- Hestia General Practioner Ltd., H-1188 Budapest, Hungary
| | - Zoltán Brassai
- Department of Internal Medicine II, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania; (P.K.); (K.M.); (Z.B.)
- II Clinic of Internal Medicine, Emergency Clinical County Hospital Targu Mures, 540142 Targu Mures, Romania
| | - Jolán Hársfalvi
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, H-1444 Budapest, Hungary;
| | - Attila Frigy
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Department of Cardiology, Clinical County Hospital Mures, 540072 Targu Mures, Romania
| |
Collapse
|
10
|
Guo XJ, Wu M, Pei SF, Xie P, Wu MY. Influence of Carotid Intima-Media Thickness Levels at Bifurcation on Short-Term Functional Outcomes Among Non-Cardiogenic Ischemic Stroke Patients with and without Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2022; 15:897-906. [PMID: 35356702 PMCID: PMC8958197 DOI: 10.2147/dmso.s351679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/10/2022] [Indexed: 04/13/2023] Open
Abstract
PURPOSE The intima-media thickness (IMT) is broadly reported to have relationships with non-cardiogenic ischemic stroke and with diabetes. But how does IMT affect the short-term prognosis of stroke seems unknown yet. We investigated the influence of the intima-media thickness at carotid bifurcation (IMTbif) on short-term functional outcomes among non-cardiogenic ischemic stroke patients with and without type 2 diabetes mellitus (T2DM). PATIENTS AND METHODS A total of 314 patients with non-cardiogenic ischemic stroke (122 with T2DM and 192 without diabetes) were included in this retrospective study. Poor functional outcome was defined as a modified Rankin Scale (mRS) > 2 at 3 months after stroke onset. Group comparisons were done in favorable and poor outcome groups. Linear regression analysis was utilized to verify the associations between IMTbif and mRS in subgroups with and without diabetes, respectively. RESULTS The median IMTbif of total patients was 1.40mm. Patients with poor outcomes were significantly older, had higher National Institutes of Health Stroke Scale (NIHSS) scores, lower haemoglobin, higher fasting glucose and higher systolic blood pressure values. Their IMTbif levels were also markedly higher. Among 122 included stroke patients with T2DM, IMTbif levels and NIHSS were independently associated with functional outcomes at 3 months, whereas there was no significant association between IMTbif levels and short-term functional outcomes among patients without diabetes. CONCLUSION The IMTbif levels were significantly associated with 3-month functional outcomes in non-cardiogenic ischemic stroke patients with T2DM. The ultrasound detection of the IMTbif therefore suggests a prognostic value among patients with stroke and T2DM.
Collapse
Affiliation(s)
- Xiao-Jing Guo
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Mian Wu
- Department of Endocrinology and Metabolism, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Shao-Fang Pei
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Ping Xie
- Department of Ultrasonography, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
| | - Min-Ya Wu
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, People’s Republic of China
- Correspondence: Min-Ya Wu, Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, People’s Republic of China, Tel +86 15151429862, Email
| |
Collapse
|
11
|
Carotid-femoral pulse wave velocity acquisition methods and their associations with cardiovascular risk factors and subclinical biomarkers of vascular health. J Hypertens 2021; 40:658-665. [PMID: 34879390 DOI: 10.1097/hjh.0000000000003055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Different methods to measure carotid-femoral pulse wave velocity (CFPWV) may affect the measurements obtained and influence the association between CFPWV, cardiovascular risk factors and biomarkers of subclinical vascular health. The estimation of distance between the carotid and femoral artery measurement sites (the arterial path length) is particularly problematic. METHOD We determined if CFPWV and equation-based estimates of CFPWV were influenced by arterial path length and if this affected the association of CFPWV with cardiovascular risk factors and subclinical vascular biomarkers. The CFPWV derived from the measurement of surface distance (CFPWV-D), arterial path length formula (CFPWV-F), and estimated CFPWV (ePWV) were obtained from 489 older adults (67.2 ± 8.8 years). Macrovascular [carotid artery: lumen diameter (LD), inter-adventitial diameter (IAD), intima-media thickness (IMT) and total plaque area (TPA)] and microvascular [reactive hyperaemia index and urinary albumin-creatinine ratio (UACR)] biomarkers were also measured. RESULTS CFPWV-D was significantly greater than CFPWV-F [9.6 (8.0-11.2) vs. 8.9 (7.6-10.5) m/s, P < 0.001], because of estimated path length being longer in CFPWV-D than CFPWV-F (495.4 ± 44.8 vs. 465.3 ± 20.6 mm, P < 0.001). ePWV was significantly greater than both CFPWV-F and CFPWV-D [11.0 (10.0-12.2) m/s, P < 0.001]. The three CFPWV methods were similarly associated with LD, IAD, IMT, TPA and UACR but not with cardiovascular risk factors. CONCLUSION Different methods to measure CFPWV affect the derived measurement values and the association with cardiovascular risk factors but not the association with subclinical biomarkers of vascular health. These hitherto unreported observations are important considerations in experimental design, data interpretation and of particular importance, comparison between studies where CFPWV is measured.
Collapse
|
12
|
Jaeger M, Stratmann B, Tschoepe D. Peripheral oscillometric arterial performance does not depict coronary status in patients with type 2 diabetes mellitus. Diab Vasc Dis Res 2021; 18:14791641211046522. [PMID: 34825586 PMCID: PMC8743959 DOI: 10.1177/14791641211046522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Arterial stiffness is associated with cardiovascular events. Matrix metalloproteases (MMPs), their tissue inhibitors (TIMPs) and galectin-3 are involved in the pathogenesis of end organ damage. This study aimed to evaluate the contribution of arterial stiffness, MMPs, TIMPs and galectin-3 with the current vascular status in type 2 diabetes mellitus (T2DM). METHODS 74 patients with T2DM, 36 with coronary heart disease (CHD) (T2DM + CHD) and 38 without CHD (T2DM - CHD) were included. Aortic pulse wave velocity (PWVao), aortic and brachial augmentation indices (AIx aortic and AIx brachial) and central-aortic blood pressure values were determined by non-invasive arteriography. MMPs, TIMPs and galectin-3 plasma concentrations were analysed by ELISA. RESULTS Patients with T2DM and CHD presented with significantly increased arterial stiffness determined as AIx and significantly elevated values for TIMP-4 and galectin-3. Heterogeneous peripheral vascular status regardless of the CHD status was observed, and increasing severity of CHD was associated with an increased arterial stiffness. TIMP-4 correlated significantly with an elevated PWVao in the whole cohort independently from CHD status. CONCLUSION Determination of arterial stiffness is an effective and, compared to laboratory markers, more reliable method for determining the peripheral vascular situation in patients with T2DM, but it does not clearly depict coronary situation.
Collapse
Affiliation(s)
- Magdalene Jaeger
- Herz- und Diabeteszentrum NRW, 39059Ruhr Universität Bochum, Bad Oeynhausen, Germany
| | - Bernd Stratmann
- Herz- und Diabeteszentrum NRW, 39059Ruhr Universität Bochum, Bad Oeynhausen, Germany
| | - Diethelm Tschoepe
- Herz- und Diabeteszentrum NRW, 39059Ruhr Universität Bochum, Bad Oeynhausen, Germany
- Stiftung DHD (Der herzkranke Diabetiker) Stiftung in der Deutschen Diabetes-Stiftung, Bad Oeynhausen, Germany
| |
Collapse
|
13
|
Shami A, Edsfeldt A, Bengtsson E, Nilsson J, Shore AC, Natali A, Khan F, Lutgens E, Gonçalves I. Soluble CD40 Levels in Plasma Are Associated with Cardiovascular Disease and in Carotid Plaques with a Vulnerable Phenotype. J Stroke 2021; 23:367-376. [PMID: 34649381 PMCID: PMC8521258 DOI: 10.5853/jos.2021.00178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/17/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE CD40 and CD40 ligand (CD40L) are costimulatory molecules of the tumor necrosis factor receptor superfamily and well known for their involvement in inflammatory diseases: atherosclerotic mouse models with disrupted CD40 signalling develop lesions of reduced size with a more stable plaque profile. This study investigated the potential of plasma and intraplaque levels of CD40 and CD40L as markers for cardiovascular disease (CVD) in humans and their association with plaque stability. METHODS Soluble CD40 and CD40L (sCD40L) were measured in plasma in 1,437 subjects from The SUrrogate markers for Micro- and Macro-vascular hard endpoints for Innovative diabetes Tools (SUMMIT) cohort. Intra-plaque levels of sCD40 and sCD40L were measured in atherosclerotic plaque homogenates from 199 subjects of the Carotid Plaque Imaging Project (CPIP) cohort. RESULTS Both plasma sCD40 and sCD40L levels were elevated in individuals with prevalent stroke, while sCD40 levels also were higher in individuals with a prior acute myocardial infarction. Plasma levels of sCD40 correlated with carotid intima-media thickness and total carotid plaque area and were associated with risk of cardiovascular events over a 3-year follow-up period. Intra-plaque levels of sCD40 and sCD40L were associated with plaque components characteristic for plaque vulnerability and extracellular matrix remodelling. CONCLUSIONS Higher plasma sCD40 and sCD40L levels are associated with prevalent CVD. Plasma sCD40 levels also correlate with the severity of carotid atherosclerosis and predict future cardiovascular events, while intra-plaque levels correlate with a vulnerable plaque phenotype. Our findings thus demonstrate that elevated levels of sCD40 and sCD40L are markers of CVD.
Collapse
Affiliation(s)
- Annelie Shami
- Department of Clinical Sciences Malmo, Clinical Research Center, Lund University, Malmo, Sweden
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmo, Clinical Research Center, Lund University, Malmo, Sweden.,Department of Cardiology, Skane University Hospital, Lund University, Malmo, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmo, Clinical Research Center, Lund University, Malmo, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmo, Clinical Research Center, Lund University, Malmo, Sweden
| | - Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, UK
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Faisel Khan
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Isabel Gonçalves
- Department of Clinical Sciences Malmo, Clinical Research Center, Lund University, Malmo, Sweden.,Department of Cardiology, Skane University Hospital, Lund University, Malmo, Sweden
| |
Collapse
|
14
|
Aizawa K, Casanova F, Gates PE, Mawson DM, Gooding KM, Strain WD, Östling G, Nilsson J, Khan F, Colhoun HM, Palombo C, Parker KH, Shore AC, Hughes AD. Reservoir-Excess Pressure Parameters Independently Predict Cardiovascular Events in Individuals With Type 2 Diabetes. Hypertension 2021; 78:40-50. [PMID: 34058850 DOI: 10.1161/hypertensionaha.121.17001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kunihiko Aizawa
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - Francesco Casanova
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - Phillip E Gates
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - David M Mawson
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - Kim M Gooding
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - W David Strain
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - Gerd Östling
- Department of Clinical Sciences, Lund University, Malmö, Sweden (G.O., J.N.)
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Malmö, Sweden (G.O., J.N.)
| | - Faisel Khan
- Division of Molecular & Clinical Medicine, University of Dundee, United Kingdom (F.K.)
| | - Helen M Colhoun
- Centre for Genomic and Experimental Medicine, University of Edinburgh, United Kingdom (H.M.C.)
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Italy (C.P.)
| | - Kim H Parker
- Department of Bioengineering, Imperial College, London, United Kingdom (K.H.P.)
| | - Angela C Shore
- Diabetes and Vascular Medicine Research Centre, NIHR Exeter Clinical Research Facility, University of Exeter Medical School, United Kingdom (K.A., F.C., P.E.G., D.M.M., K.M.G., W.D.S., A.C.S.)
| | - Alun D Hughes
- MRC Unit for Lifelong Health & Ageing, Institute of Cardiovascular Science, University College London, United Kingdom (A.D.H.)
| |
Collapse
|
15
|
Kärberg K, Lember M. Subclinical atherosclerosis in the carotid artery: can the ankle-brachial index predict it in type 2 diabetes patients? Scandinavian Journal of Clinical and Laboratory Investigation 2021; 81:237-243. [PMID: 33771060 DOI: 10.1080/00365513.2021.1904279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Atherosclerosis is a progressive asymmetrical systemic disease that progresses faster in patients with diabetes comorbidity. Therefore, type 2 diabetic (T2DM) patients who have a high risk of, or have already detected, early atherosclerosis should be treated aggressively to prevent premature mortality. We hypothesised that subclinical atherosclerosis is predictable with the ankle-brachial index (ABI). There are currently only a few studies to indicate which specific value of ABI can predict atherosclerosis in the carotid artery. Our study aimed to examine ABI ≤ 1.1 ability to predict ultrasound-visualised atherosclerosis in carotid arteries in patients with T2DM, who had not been previously diagnosed with atherosclerosis. A population-based cross-sectional multicentric study was performed in 216 participants (mean age 59 ± 8 years). Carotid artery intima-medial thickness (IMT) ≥1 mm ± plaque was defined as a marker for subclinical atherosclerosis and was compared with ABI. Mean duration of T2DM was 7.05 ± 6.0 years. Atherosclerosis in the carotid artery was found in 96 (44%) patients, with no significant differences between genders (47 vs 53%, p = .206). ABI ≤1.1 was associated with the carotid artery mean IMT ≥1 mm (p = .037), plaque (p = .027) and IMT ≥1 mm ± plaque (p = .037). The association between ABI ≤ 1.1 and IMT ≥ 1 mm ± plaque remained significant after adjustment for risk factors and age >50 years. Observations demonstrated that ABI ≤ 1.1 could be an indicator of subclinical atherosclerosis for T2DM male patients over 50 years old.
Collapse
Affiliation(s)
- Kati Kärberg
- Department of Internal Medicine, University of Tartu, Tartu, Estonia
| | - Margus Lember
- Department of Internal Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
16
|
Monjezi MR, Fouladseresht H, Farjadian S, Gharesi-Fard B, Khosropanah S, Doroudchi M. T Cell Proliferative Responses and IgG Antibodies to β2GPI in Patients with Diabetes and Atherosclerosis. Endocr Metab Immune Disord Drug Targets 2021; 21:495-503. [PMID: 32368987 DOI: 10.2174/1871530320666200505115850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/15/2020] [Accepted: 03/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Diabetes increases the risk of myocardial infarction (MI) by 2 to 3 folds. Tlymphocytes play a role in atherosclerosis, which is the main pathology behind MI. Cellular immune responses to beta-2 glycoprotein I (β2GPI) are shown in carotid atherosclerosis. OBJECTIVE To investigate the self-reactive, β2GPI-specific T-lymphocytes in patients with and without diabetes and atherosclerosis. METHODS Collectively, 164 subjects with and without diabetes that underwent coronary angiography were divided into four groups based on their diabetes status and coronary stenosis. Group I=Diabetic with ≥50% stenosis: A+D+ (n=66); Group II=Non-diabetic with ≥50% stenosis, A+D- (n=39); Group III=Diabetic with <50% stenosis: A-D+ (n=28); and Group IV=Non-diabetic with <50% stenosis: AD- (n=31). All groups were evaluated for anti-β2GPI IgG antibody by ELISA method. Then, PBMCs were isolated from 18 subjects and were stimulated with β2GPI-derived peptides to assess their proliferation in accordance with their HLA-DRB1 alleles. RESULTS Mean β2GPI IgG levels were higher in groups with ≥50% stenosis (A+) compared to those with <50% stenosis (A-), (P=0.02). The co-presence of diabetes in A+ individuals increased mean β2GPI-specific IgG. Auto-reactive β2GPI-specific T cells were detected in the repertoire of T-lymphocytes in all groups. β2GPI-peptides showed promiscuous restriction by various HLADRB1. CONCLUSION β2GPI is the target of cellular and humoral immune responses in patients with atherosclerosis. Since the T cell responses but not antibodies were detectable in A-D+ and A-D- groups, it is reasonable to assume that cellular responses preceded the humoral responses. Post-translation modifications of β2GPI under oxidative and glycemic stresses may have increased the IgG levels in patients with diabetes. Finally, identification of antigens that trigger immuno-pathogenesis in atherosclerosis and diabetes may help the development of immunomodulation methods to prevent or treat these debilitating diseases.
Collapse
Affiliation(s)
- Mohammad R Monjezi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Farjadian
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahdad Khosropanah
- Department of Cardiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Meakin PJ, Coull BM, Tuharska Z, McCaffery C, Akoumianakis I, Antoniades C, Brown J, Griffin KJ, Platt F, Ozber CH, Yuldasheva NY, Makava N, Skromna A, Prescott A, McNeilly AD, Siddiqui M, Palmer CN, Khan F, Ashford ML. Elevated circulating amyloid concentrations in obesity and diabetes promote vascular dysfunction. J Clin Invest 2021; 130:4104-4117. [PMID: 32407295 PMCID: PMC7410081 DOI: 10.1172/jci122237] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetes, obesity, and Alzheimer’s disease (AD) are associated with vascular complications and impaired nitric oxide (NO) production. Furthermore, increased β-site amyloid precursor protein–cleaving (APP-cleaving) enzyme 1 (BACE1), APP, and β-amyloid (Aβ) are linked with vascular disease development and increased BACE1 and Aβ accompany hyperglycemia and hyperlipidemia. However, the causal relationship between obesity and diabetes, increased Aβ, and vascular dysfunction is unclear. We report that diet-induced obesity (DIO) in mice increased plasma and vascular Aβ42 that correlated with decreased NO bioavailability, endothelial dysfunction, and increased blood pressure. Genetic or pharmacological reduction of BACE1 activity and Aβ42 prevented and reversed, respectively, these outcomes. In contrast, expression of human mutant APP in mice or Aβ42 infusion into control diet–fed mice to mimic obese levels impaired NO production, vascular relaxation, and raised blood pressure. In humans, increased plasma Aβ42 correlated with diabetes and endothelial dysfunction. Mechanistically, higher Aβ42 reduced endothelial NO synthase (eNOS), cyclic GMP (cGMP), and protein kinase G (PKG) activity independently of diet, whereas endothelin-1 was increased by diet and Aβ42. Lowering Aβ42 reversed the DIO deficit in the eNOS/cGMP/PKG pathway and decreased endothelin-1. Our findings suggest that BACE1 inhibitors may have therapeutic value in the treatment of vascular disease associated with diabetes.
Collapse
Affiliation(s)
- Paul J Meakin
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom.,Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Bethany M Coull
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Zofia Tuharska
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Christopher McCaffery
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Level 6 West Wing, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Charalambos Antoniades
- Cardiovascular Medicine Division, Level 6 West Wing, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jane Brown
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Kathryn J Griffin
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Fiona Platt
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Claire H Ozber
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Nadira Y Yuldasheva
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Natallia Makava
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Anna Skromna
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Alan Prescott
- School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Alison D McNeilly
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Moneeza Siddiqui
- Division of Population Health & Genomics, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Colin Na Palmer
- Division of Population Health & Genomics, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Michael Lj Ashford
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
18
|
Ott MV, Sumin AN, Kovalenko AV. [Possibilities of application of cardio-ankle vascular index in patients with cerebrovascular diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:37-44. [PMID: 33016675 DOI: 10.17116/jnevro202012008237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This publication focuses on the feasibility of using the cardio-ankle vascular index (CAVI) in patients with cerebrovascular diseases. The authors consider the pathological conditions and risk factors of stroke associated with increased arterial stiffness, methods for its assessment, the advantages of using CAVI, the experience of using CAVI in patients with cardiovascular diseases, in particular, in neurological patients. The complexity of the application of CAVI in the Russian population, promising directions for determining the index in neurology as well as the importance of CAVI borderline indicators are shown. It is emphasized that stroke patients should be screened with a mandatory study of CAVI.
Collapse
Affiliation(s)
- M V Ott
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A N Sumin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A V Kovalenko
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia.,Kemerovo State Medical University, Kemerovo, Russia
| |
Collapse
|
19
|
Nesti L, Mengozzi A, Natali A. Statins, LDL Cholesterol Control, Cardiovascular Disease Prevention, and Atherosclerosis Progression: A Clinical Perspective. Am J Cardiovasc Drugs 2020; 20:405-412. [PMID: 31840213 DOI: 10.1007/s40256-019-00391-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Despite the longstanding and widespread use of statins, they are used quite inefficiently in everyday clinical practice. This might be because of a lack of robust evidence or the wide variety of different guidelines that are frequently changed. Using data from clinical trials and some simple mathematical modeling, we sought to expand upon the relation between low-density lipoprotein cholesterol (LDL-C) control and cardiovascular risk to offer a firm basis for independent decision making in everyday clinical practice. Analysis of the dose-response curves of different statins indicated that doubling the dose will provide a < 5% extra LDL-C gradient and that the relationship among different statin dose equipotencies is fourfold in the lower range and threefold in the higher range. Thus, the use of potent statins at very low doses might overcome patient statin reluctance. Moreover, whereas statins lower LDL-C percentwise, the prevention of atherosclerosis-related cardiovascular events (ARCVEs) depends on the absolute LDL-C gradient produced and the level of risk. Consequently, and counterintuitively, the lower the baseline LDL-C and/or ARCVE risk, the higher the statin therapy strength required, and approach that is also cost effective. We discuss the issue of threshold versus gradient in terms of clinical trials on plaque regression and speculate on the relationship between LDL-C and atherosclerosis.
Collapse
Affiliation(s)
- Lorenzo Nesti
- Laboratory of Metabolism, Nutrition and Atherosclerosis, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 8, 56100, Pisa, Italy.
| | - Alessandro Mengozzi
- Laboratory of Metabolism, Nutrition and Atherosclerosis, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 8, 56100, Pisa, Italy
| | - Andrea Natali
- Laboratory of Metabolism, Nutrition and Atherosclerosis, Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 8, 56100, Pisa, Italy
| |
Collapse
|
20
|
Lunde NN, Gregersen I, Ueland T, Shetelig C, Holm S, Kong XY, Michelsen AE, Otterdal K, Yndestad A, Broch K, Gullestad L, Nyman TA, Bendz B, Eritsland J, Hoffmann P, Skagen K, Gonçalves I, Nilsson J, Grenegård M, Poreba M, Drag M, Seljeflot I, Sporsheim B, Espevik T, Skjelland M, Johansen HT, Solberg R, Aukrust P, Björkbacka H, Andersen GØ, Halvorsen B. Legumain is upregulated in acute cardiovascular events and associated with improved outcome - potentially related to anti-inflammatory effects on macrophages. Atherosclerosis 2019; 296:74-82. [PMID: 31870625 DOI: 10.1016/j.atherosclerosis.2019.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS We have previously found increased levels of the cysteine protease legumain in plasma and plaques from patients with carotid atherosclerosis. This study further investigated legumain during acute cardiovascular events. METHODS Circulating levels of legumain from patients and legumain released from platelets were assessed by enzyme-linked-immunosorbent assay. Quantitative PCR and immunoblotting were used to study expression, while localization was visualized by immunohistochemistry. RESULTS In the SUMMIT Malmö cohort (n = 339 with or without type 2 diabetes and/or cardiovascular disease [CVD], and 64 healthy controls), the levels of circulating legumain were associated with the presence of CVD in non-diabetics, with no relation to outcome. In symptomatic carotid plaques and in samples from both coronary and intracerebral thrombi obtained during acute cardiovascular events, legumain was co-localized with macrophages in the same regions as platelets. In vitro, legumain was shown to be present in and released from platelets upon activation. In addition, THP-1 macrophages exposed to releasate from activated platelets showed increased legumain expression. Interestingly, primary peripheral blood mononuclear cells stimulated with recombinant legumain promoted anti-inflammatory responses. Finally, in a STEMI population (POSTEMI; n = 272), patients had significantly higher circulating legumain before and immediately after percutaneous coronary intervention compared with healthy controls (n = 67), and high levels were associated with improved outcome. CONCLUSIONS Our data demonstrate for the first time that legumain is upregulated during acute cardiovascular events and is associated with improved outcome.
Collapse
Affiliation(s)
- Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway.
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Christian Shetelig
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Tuula A Nyman
- Proteomics Core Facility, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Rikshospitalet Oslo, Oslo, Norway
| | - Bjørn Bendz
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jan Eritsland
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pavel Hoffmann
- Section of Interventional Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Karolina Skagen
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Isabel Gonçalves
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden; Department of Cardiology, Skåne University Hospital, Sweden
| | - Jan Nilsson
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden
| | | | - Marcin Poreba
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Marcin Drag
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bjørnar Sporsheim
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona Skjelland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Harald Thidemann Johansen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Harry Björkbacka
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden
| | - Geir Øystein Andersen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
Rattik S, Engelbertsen D, Wigren M, Ljungcrantz I, Östling G, Persson M, Nordin Fredrikson G, Bengtsson E, Nilsson J, Björkbacka H. Elevated circulating effector memory T cells but similar levels of regulatory T cells in patients with type 2 diabetes mellitus and cardiovascular disease. Diab Vasc Dis Res 2019; 16:270-280. [PMID: 30574794 DOI: 10.1177/1479164118817942] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type 2 diabetes mellitus is associated with an elevated risk of cardiovascular disease, but the mechanism through which diabetes contributes to cardiovascular disease development remains incompletely understood. In this study, we compared the association of circulating regulatory T cells, naïve T cells, effector memory T cells or central memory T cells with cardiovascular disease in patients with and without type 2 diabetes mellitus. Percentage of circulating T cell subsets was analysed by flow cytometry in type 2 diabetes mellitus subjects with and without prevalent cardiovascular disease as well as in non-diabetic subjects with and without prevalent cardiovascular disease from the Malmö SUMMIT cohort. Subjects with type 2 diabetes mellitus had elevated percentages of effector memory T cells (CD4+CD45RO+CD62L-; 21.8% ± 11.2% vs 17.0% ± 9.2% in non-type 2 diabetes mellitus, p < 0.01) and central memory T cells (CD4+CD45RO+CD62L+; 38.0% ± 10.7% vs 36.0% ± 9.5% in non-type 2 diabetes mellitus, p < 0.01). In contrast, the frequency of naïve T cells was reduced (CD4+CD45RO-CD62L+, 35.0% ± 16.5% vs 42.9% ± 14.4% in non-type 2 diabetes mellitus, p < 0.001). The proportion of effector memory T cells was increased in type 2 diabetes mellitus subjects with cardiovascular disease as compared to those without (26.4% ± 11.5% vs 18.4% ± 10.2%, p < 0.05), while no difference in regulatory T cells was observed between these two patient groups. This study identifies effector memory T cells as a potential cellular biomarker for cardiovascular disease among subjects with type 2 diabetes mellitus, suggesting a state of exacerbated immune activation in type 2 diabetes mellitus patients with cardiovascular disease.
Collapse
Affiliation(s)
- Sara Rattik
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Daniel Engelbertsen
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Maria Wigren
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Gerd Östling
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Margaretha Persson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Eva Bengtsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
22
|
Scuteri A, Rovella V, Alunni Fegatelli D, Tesauro M, Gabriele M, Di Daniele N. An operational definition of SHATS (Systemic Hemodynamic Atherosclerotic Syndrome): Role of arterial stiffness and blood pressure variability in elderly hypertensive subjects. Int J Cardiol 2018; 263:132-137. [PMID: 29754908 DOI: 10.1016/j.ijcard.2018.03.117] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/20/2018] [Accepted: 03/26/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND CV risk exponentially increases as the number of damaged organs increases The Systemic Hemodynamic Atherosclerotic Syndrome (SHATS) represents a novel conceptualization of the CV continuum focusing on simultaneous multi-organ alteration. This is the first study operationally defining SHATS and aimed at identifying its determinants. METHODS Left Ventricular Hypertrophy (echocardiography), Common Carotid Artery plaque and increased thickness (ultrasound), and Chronic Kidney Disease (estimated Glomerular Filtration Rate) indexed selective target organ damage. SHATS was operationally defined as their simultaneous presence in a patient. PWV was measured by Sphygmocor® and BP variability by 24 h ABPM. RESULTS SHATS affected 19.9% of the 367 studied subjects. Subjects with SHATS had a similar prevalence in diabetes mellitus, but a greater prevalence of very stiff artery (84.9 vs 64.3%, p < 0.01) and use of antihypertensive medications. In the presence of similar office BP, SHATS was associated with higher 24 h SBP and lower 24 h DBP (a greater pulsatile pressure!), reduced nighttime SBP fall, and a twofold greater prevalence of reverse dipper status (48.2 vs 20.2%, p < 0.001). BMI (positive correlation) and DBP (negative correlation) were the only traditional CV risk factors significantly associated with the odds of having SHATS. Very stiff artery and BP variability were significant independent determinants of SHATS, with highly predictive accuracy. CONCLUSION SHATS, the simultaneous damage of multiple target organs, may easily operationally defined. Very stiff artery and BP variability represent key factors for SHATS. The present results support the hypothesis of SHATS as a systemic condition, needing further characterization.
Collapse
Affiliation(s)
- Angelo Scuteri
- Department of Medical, Surgical, and Experimental Medicine, University of Sassari, Sassari, Italy.
| | - Valentina Rovella
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata -, Rome, Italy
| | | | - Manfredi Tesauro
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata -, Rome, Italy
| | - Marco Gabriele
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata -, Rome, Italy
| | - Nicola Di Daniele
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata -, Rome, Italy
| |
Collapse
|
23
|
Catov JM, Snyder GG, Bullen BL, Barinas-Mitchell EJM, Holzman C. Women with Preterm Birth Have Evidence of Subclinical Atherosclerosis a Decade After Delivery. J Womens Health (Larchmt) 2018; 28:621-627. [PMID: 30388049 DOI: 10.1089/jwh.2018.7148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Women with preterm birth (PTB) have excess risk of cardiovascular disease (CVD) and metabolic dysregulation after delivery, but vascular mechanisms are poorly understood. We considered that women with PTB may have evidence of subclinical atherosclerosis after delivery, perhaps related to cardiometabolic risk factors. Materials and Methods: The Pregnancy Outcomes and Community Health Moms (POUCHmoms) study followed women from pregnancy through 7 to 15 years after delivery (n = 678). Women underwent B-mode ultrasound to measure the average intima-media thickness (IMT) across the common carotid, bulb, and internal carotid artery segments at follow-up (n = 605). Linear regression estimated the overall and segment-specific difference in IMT between women with preterm and term births. Results: Women were, on average, 38 years old (SD 5.7) at the follow-up visit. Those with a prior preterm versus term birth had thicker mean IMT (average of eight segments, 0.592 mm vs. 0.575, p = 0.04). Differences persisted after accounting for age, race, smoking, and body mass index (difference = +0.018 mm, p = 0.019) and were attenuated after adjustment for blood pressure, medication use, and total cholesterol (difference = +0.014, p = 0.052). Thicker mean bulb IMT in women with PTB was robust to cardiovascular risk factor adjustments (fully adjusted difference = +0.033, p = 0.029). Excluding cases of prepregnancy hypertension or preeclampsia did not change results. Conclusions: Mechanisms leading to subclinical atherosclerosis may link PTB with future CVD. PTB differences in maternal vessel remodeling in the carotid bulb, an arterial segment more prone to early development of atherosclerosis, were independent of traditional risk factors suggesting that novel processes may be involved.
Collapse
Affiliation(s)
- Janet M Catov
- 1 Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,2 Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Gabrielle G Snyder
- 2 Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Bertha L Bullen
- 3 Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan
| | - Emma J M Barinas-Mitchell
- 2 Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Claudia Holzman
- 3 Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan
| |
Collapse
|
24
|
Shore AC, Colhoun HM, Natali A, Palombo C, Khan F, Östling G, Aizawa K, Kennbäck C, Casanova F, Persson M, Gooding K, Gates PE, Looker H, Dove F, Belch J, Pinnola S, Venturi E, Kozakova M, Goncalves I, Kravic J, Björkbacka H, Nilsson J. Use of Vascular Assessments and Novel Biomarkers to Predict Cardiovascular Events in Type 2 Diabetes: The SUMMIT VIP Study. Diabetes Care 2018; 41:2212-2219. [PMID: 30061319 DOI: 10.2337/dc18-0185] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/06/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Cardiovascular disease (CVD) risk prediction represents an increasing clinical challenge in the treatment of diabetes. We used a panel of vascular imaging, functional assessments, and biomarkers reflecting different disease mechanisms to identify clinically useful markers of risk for cardiovascular (CV) events in subjects with type 2 diabetes (T2D) with or without manifest CVD. RESEARCH DESIGN AND METHODS The study cohort consisted of 936 subjects with T2D recruited at four European centers. Carotid intima-media thickness and plaque area, ankle-brachial pressure index, arterial stiffness, endothelial function, and circulating biomarkers were analyzed at baseline, and CV events were monitored during a 3-year follow-up period. RESULTS The CV event rate in subjects with T2D was higher in those with (n = 440) than in those without (n = 496) manifest CVD at baseline (5.53 vs. 2.15/100 life-years, P < 0.0001). New CV events in subjects with T2D with manifest CVD were associated with higher baseline levels of inflammatory biomarkers (interleukin 6, chemokine ligand 3, pentraxin 3, and hs-CRP) and endothelial mitogens (hepatocyte growth factor and vascular endothelial growth factor A), whereas CV events in subjects with T2D without manifest CVD were associated with more severe baseline atherosclerosis (median carotid plaque area 30.4 mm2 [16.1-92.2] vs. 19.5 mm2 [9.5-40.5], P = 0.01). Conventional risk factors, as well as measurements of arterial stiffness and endothelial reactivity, were not associated with CV events. CONCLUSIONS Our observations demonstrate that markers of inflammation and endothelial stress reflect CV risk in subjects with T2D with manifest CVD, whereas the risk for CV events in subjects with T2D without manifest CVD is primarily related to the severity of atherosclerosis.
Collapse
Affiliation(s)
- Angela C Shore
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, U.K
| | - Helen M Colhoun
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, U.K
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Faisel Khan
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Gerd Östling
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Kunihiko Aizawa
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, U.K
| | - Cecilia Kennbäck
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Francesco Casanova
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, U.K
| | | | - Kim Gooding
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, U.K
| | - Phillip E Gates
- Diabetes and Vascular Medicine, University of Exeter Medical School, National Institute for Health Research Exeter Clinical Research Facility, Exeter, U.K
| | - Helen Looker
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Fiona Dove
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Jill Belch
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, U.K
| | - Silvia Pinnola
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Venturi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Surgical, Medical, Molecular, and Critical Area Pathology, University of Pisa, Pisa, Italy
| | - Isabel Goncalves
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Jasmina Kravic
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Harry Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | | |
Collapse
|
25
|
Strawbridge RJ, van Zuydam NR. Shared Genetic Contribution of Type 2 Diabetes and Cardiovascular Disease: Implications for Prognosis and Treatment. Curr Diab Rep 2018; 18:59. [PMID: 29938349 PMCID: PMC6015804 DOI: 10.1007/s11892-018-1021-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE OF REVIEW The increased cardiovascular disease (CVD) risk in subjects with type 2 diabetes (T2D) is well established. This review collates the available evidence and assesses the shared genetic background between T2D and CVD: the causal contribution of common risk factors to T2D and CVD and how genetics can be used to improve drug development and clinical outcomes. RECENT FINDINGS Large-scale genome-wide association studies (GWAS) of T2D and CVD support a shared genetic background but minimal individual locus overlap. Mendelian randomisation (MR) analyses show that T2D is causal for CVD, but GWAS of CVD, T2D and their common risk factors provided limited evidence for individual locus overlap. Distinct but functionally related pathways were enriched for CVD and T2D genetic associations reflecting the lack of locus overlap and providing some explanation for the variable associations of common risk factors with CVD and T2D from MR analyses.
Collapse
Affiliation(s)
- Rona J. Strawbridge
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Room 113, 1 Lilybank Gardens, Glasgow, G12 8RZ UK
- Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Natalie R. van Zuydam
- Wellcome Centre Human Genetics, University of Oxford, Roosevelt Drive, Headington, Oxford, Oxfordshire, OX3 7BN UK
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Headington, Oxford, Oxfordshire, OX3 7LE UK
| |
Collapse
|
26
|
Weir-McCall JR, Brown L, Summersgill J, Talarczyk P, Bonnici-Mallia M, Chin SC, Khan F, Struthers AD, Sullivan F, Colhoun HM, Shore AC, Aizawa K, Groop L, Nilsson J, Cockcroft JR, McEniery CM, Wilkinson IB, Ben-Shlomo Y, Houston JG. Development and Validation of a Path Length Calculation for Carotid-Femoral Pulse Wave Velocity Measurement: A TASCFORCE, SUMMIT, and Caerphilly Collaborative Venture. Hypertension 2018; 71:937-945. [PMID: 29555666 PMCID: PMC5902134 DOI: 10.1161/hypertensionaha.117.10620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 12/28/2017] [Accepted: 01/30/2018] [Indexed: 11/19/2022]
Abstract
Supplemental Digital Content is available in the text. Current distance measurement techniques for pulse wave velocity (PWV) calculation are susceptible to intercenter variability. The aim of this study was to derive and validate a formula for this distance measurement. Based on carotid femoral distance in 1183 whole-body magnetic resonance angiograms, a formula was derived for calculating distance. This was compared with distance measurements in 128 whole-body magnetic resonance angiograms from a second study. The effects of recalculation of PWV using the new formula on association with risk factors, disease discrimination, and prediction of major adverse cardiovascular events were examined within 1242 participants from the multicenter SUMMIT study (Surrogate Markers of Micro- and Macrovascular Hard End-Points for Innovative Diabetes Tools) and 825 participants from the Caerphilly Prospective Study. The distance formula yielded a mean error of 7.8 mm (limits of agreement =−41.1 to 56.7 mm; P<0.001) compared with the second whole-body magnetic resonance angiogram group. Compared with an external distance measurement, the distance formula did not change associations between PWV and age, blood pressure, or creatinine (P<0.01) but did remove significant associations between PWV and body mass index (BMI). After accounting for differences in age, sex, and mean arterial pressure, intercenter differences in PWV persisted using the external distance measurement (F=4.6; P=0.004), whereas there was a loss of between center difference using the distance formula (F=1.4; P=0.24). PWV odds ratios for cardiovascular mortality remained the same using both the external distance measurement (1.14; 95% confidence interval, 1.06–1.24; P=0.001) and the distance formula (1.17; 95% confidence interval, 1.08–1.28; P<0.001). A population-derived automatic distance calculation for PWV obtained from routinely collected clinical information is accurate and removes intercenter measurement variability without impacting the diagnostic utility of carotid–femoral PWV.
Collapse
Affiliation(s)
- Jonathan R Weir-McCall
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Liam Brown
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Jennifer Summersgill
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Piotr Talarczyk
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Michael Bonnici-Mallia
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Sook C Chin
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Faisel Khan
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Allan D Struthers
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Frank Sullivan
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Helen M Colhoun
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Angela C Shore
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Kunihiko Aizawa
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Leif Groop
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Jan Nilsson
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - John R Cockcroft
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Carmel M McEniery
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Ian B Wilkinson
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - Yoav Ben-Shlomo
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.)
| | - J Graeme Houston
- From the Division of Molecular and Clinical Medicine, College of Medicine, University of Dundee, United Kingdom (J.R.W.-M., L.B., J.S., F.K., A.D.S., J.G.H.); NHS Tayside Clinical Radiology, Ninewells Hospital, Dundee, United Kingdom (P.T., M.B.-M., S.C.C.); Department of Research and Innovation, North York General Hospital, University of Toronto, Canada (F.S.); Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, United Kingdom (H.M.C.); NIHR Exeter Clinical Research Facility, Royal Devon and Exeter Hospital and University of Exeter Medical School, United Kingdom (A.C.S., K.A.); Lund University Diabetes Centre, Lund University, Malmö, Sweden (L.G.); Department of Clinical Sciences Malmö, Lund University, Sweden (J.N.); Department of Cardiology, Wales Heart Research Institute, Cardiff, United Kingdom (J.R.C.); Division of Experimental Medicine & Immunotherapeutics, University of Cambridge, United Kingdom (C.M.M., I.B.W.); and Population Health Sciences, University of Bristol, United Kingdom (Y.B.-S.).
| |
Collapse
|
27
|
Epicardial adipose tissue is related to arterial stiffness and inflammation in patients with cardiovascular disease and type 2 diabetes. BMC Cardiovasc Disord 2018; 18:31. [PMID: 29433433 PMCID: PMC5809843 DOI: 10.1186/s12872-018-0770-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/02/2018] [Indexed: 11/10/2022] Open
Abstract
Background Epicardial adipose tissue (EAT) is an emerging cardio-metabolic risk factor and has been shown to correlate with adverse cardiovascular (CV) outcome; however the underlying pathophysiology of this link is not well understood. The aim of this study was to evaluate the relationship between EAT and a comprehensive panel of cardiovascular risk biomarkers and pulse wave velocity (PWV) and indexed left ventricular mass (LVMI) in a cohort of patients with cardiovascular disease (CVD) and diabetes compared to controls. Methods One hundred forty-five participants (mean age 63.9 ± 8.1 years; 61% male) were evaluated. All patients underwent cardiovascular magnetic resonance (CMR) examination and PWV. EAT measurements from CMR were performed on the 4-chamber view. Blood samples were taken and a range of CV biomarkers was evaluated. Results EAT measurements were significantly higher in the groups with CVD, with or without T2DM compared to patients without CVD or T2DM (group 1 EAT 15.9 ± 5.5 cm2 vs. group 4 EAT 11.8 ± 4.1 cm2, p = 0.001; group 3 EAT 15.1 ± 4.3 cm2 vs. group 4 EAT 11.8 ± 4.1 cm2, p = 0.024). EAT was independently associated with IL-6 (beta 0.2, p = 0.019). When added to clinical variables, both EAT (beta 0.16, p = 0.035) and IL-6 (beta 0.26, p = 0.003) were independently associated with PWV. EAT was significantly associated with LVMI in a univariable analysis but not when added to significant clinical variables. Conclusions In patients with cardio-metabolic disease, EAT was independently associated with PWV. EAT may be associated with CVD risk due to an increase in systemic vascular inflammation. Whether targeting EAT may reduce inflammation and/or cardiovascular risk should be evaluated in prospective studies. Electronic supplementary material The online version of this article (10.1186/s12872-018-0770-z) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Casanova F, Adingupu DD, Adams F, Gooding KM, Looker HC, Aizawa K, Dove F, Elyas S, Belch JJF, Gates PE, Littleford RC, Gilchrist M, Colhoun HM, Shore AC, Khan F, Strain WD. The impact of cardiovascular co-morbidities and duration of diabetes on the association between microvascular function and glycaemic control. Cardiovasc Diabetol 2017; 16:114. [PMID: 28915818 PMCID: PMC5603035 DOI: 10.1186/s12933-017-0594-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/04/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Good glycaemic control in type 2 diabetes (T2DM) protects the microcirculation. Current guidelines suggest glycaemic targets be relaxed in advanced diabetes. We explored whether disease duration or pre-existing macrovascular complications attenuated the association between hyperglycaemia and microvascular function. METHODS 743 participants with T2DM (n = 222), cardiovascular disease (CVD = 183), both (n = 177) or neither (controls = 161) from two centres in the UK, underwent standard clinical measures and endothelial dependent (ACh) and independent (SNP) microvascular function assessment using laser Doppler imaging. RESULTS People with T2DM and CVD had attenuated ACh and SNP responses compared to controls. This was additive in those with both (ANOVA p < 0.001). In regression models, cardiovascular risk factors accounted for attenuated ACh and SNP responses in CVD, whereas HbA1c accounted for the effects of T2DM. HbA1c was associated with ACh and SNP response after adjustment for cardiovascular risk factors (adjusted standardised beta (β) -0.096, p = <0.008 and -0.135, p < 0.001, respectively). Pre-existing CVD did not modify this association (β -0.099; p = 0.006 and -0.138; p < 0.001, respectively). Duration of diabetes accounted for the association between HbA1c and ACh (β -0.043; p = 0.3), but not between HbA1c and SNP (β -0.105; p = 0.02). CONCLUSIONS In those with T2DM and CVD, good glycaemic control is still associated with better microvascular function, whereas in those with prolonged disease this association is lost. This suggests duration of diabetes may be a better surrogate for "advanced disease" than concomitant CVD, although this requires prospective validation.
Collapse
Affiliation(s)
- F Casanova
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - D D Adingupu
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - F Adams
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - K M Gooding
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - H C Looker
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - K Aizawa
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - F Dove
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - S Elyas
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - J J F Belch
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - P E Gates
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - R C Littleford
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - M Gilchrist
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - H M Colhoun
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - A C Shore
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK
| | - F Khan
- Vascular and Inflammatory Diseases Research Unit, Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| | - W D Strain
- Diabetes and Vascular Medicine Research Centre, Institute of Biomedical and Clinical Science and NIHR Exeter Clinical Research Facility, University of Exeter Medical School, Barrack Rd, Exeter, EX2 5AX, UK.
| |
Collapse
|
29
|
Rani J, Mittal I, Pramanik A, Singh N, Dube N, Sharma S, Puniya BL, Raghunandanan MV, Mobeen A, Ramachandran S. T2DiACoD: A Gene Atlas of Type 2 Diabetes Mellitus Associated Complex Disorders. Sci Rep 2017; 7:6892. [PMID: 28761062 PMCID: PMC5537262 DOI: 10.1038/s41598-017-07238-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 06/28/2017] [Indexed: 12/11/2022] Open
Abstract
We performed integrative analysis of genes associated with type 2 Diabetes Mellitus (T2DM) associated complications by automated text mining with manual curation and also gene expression analysis from Gene Expression Omnibus. They were analysed for pathogenic or protective role, trends, interaction with risk factors, Gene Ontology enrichment and tissue wise differential expression. The database T2DiACoD houses 650 genes, and 34 microRNAs associated with T2DM complications. Seven genes AGER, TNFRSF11B, CRK, PON1, ADIPOQ, CRP and NOS3 are associated with all 5 complications. Several genes are studied in multiple years in all complications with high proportion in cardiovascular (75.8%) and atherosclerosis (51.3%). T2DM Patients' skeletal muscle tissues showed high fold change in differentially expressed genes. Among the differentially expressed genes, VEGFA is associated with several complications of T2DM. A few genes ACE2, ADCYAP1, HDAC4, NCF1, NFE2L2, OSM, SMAD1, TGFB1, BDNF, SYVN1, TXNIP, CD36, CYP2J2, NLRP3 with details of protective role are catalogued. Obesity is clearly a dominant risk factor interacting with the genes of T2DM complications followed by inflammation, diet and stress to variable extents. This information emerging from the integrative approach used in this work could benefit further therapeutic approaches. The T2DiACoD is available at www.http://t2diacod.igib.res.in/ .
Collapse
Affiliation(s)
- Jyoti Rani
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Inna Mittal
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Atreyi Pramanik
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Namita Singh
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Namita Dube
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Smriti Sharma
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Bhanwar Lal Puniya
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Muthukurussi Varieth Raghunandanan
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
| | - Ahmed Mobeen
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India
- Academy of Scientific and Innovative Research, CSIR-IGIB South Campus, New Delhi, 110025, India
| | - Srinivasan Ramachandran
- G N Ramachandran Knowledge of Centre, Council of Scientific and Industrial Research - Institute of Genomics and Integrative Biology (CSIR-IGIB), Room No. 130, Mathura Road, New Delhi, 110025, India.
- Academy of Scientific and Innovative Research, CSIR-IGIB South Campus, New Delhi, 110025, India.
| |
Collapse
|
30
|
Noh M, Kwon H, Jung CH, Kwon SU, Kim MS, Lee WJ, Park JY, Han Y, Kim H, Kwon TW, Cho YP. Impact of diabetes duration and degree of carotid artery stenosis on major adverse cardiovascular events: a single-center, retrospective, observational cohort study. Cardiovasc Diabetol 2017; 16:74. [PMID: 28587650 PMCID: PMC5461631 DOI: 10.1186/s12933-017-0556-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/29/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We aimed to investigate the impact of diabetes duration and carotid artery stenosis (CAS) on the occurrence of major adverse cardiovascular events (MACE) in patients with type 2 diabetes mellitus (T2DM) without clinical cardiovascular disease. METHODS A total of 2006 patients with T2DM, without clinical cardiovascular disease, aged >50 years, and who underwent baseline carotid Doppler ultrasound screening with regular follow-ups at the outpatient clinic of our diabetes center, were stratified into four subgroups according to diabetes duration and CAS degree. The primary outcomes included the occurrence of MACE, defined as fatal or nonfatal stroke and myocardial infarction, and all-cause mortality. RESULTS The difference in the MACE incidence was significantly greater in patients with a longer diabetes duration (≥10 years) and significant CAS (50-69% luminal narrowing) (p < 0.001). Analysis of individual MACE components indicated a trend towards an increased incidence of stroke (p < 0.001), parallel to a longer diabetes duration and significant CAS. In contrast, the risk of myocardial infarction was significantly higher in patients with a diabetes duration <10 years and significant CAS (p = 0.039). Multivariate regression analysis showed that patients with both a longer diabetes duration and significant CAS demonstrated additive and very high risks of MACE (hazard ratio [HR], 2.07; 95% confidence interval [CI] 1.17-3.66; p = 0.012) and stroke (HR, 3.38; 95% CI 1.54-7.44; p = 0.002). CONCLUSIONS The risk of MACE is significantly greater in patients with T2DM, without clinical cardiovascular disease, who have both a longer diabetes duration and significant CAS, compared with those who have a shorter duration and/or nonsignificant CAS.
Collapse
Affiliation(s)
- Minsu Noh
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Hyunwook Kwon
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Chang Hee Jung
- Department of Internal Medicine, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Sun U. Kwon
- Department of Neurology, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Min Seon Kim
- Department of Internal Medicine, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Woo Je Lee
- Department of Internal Medicine, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Joong Yeol Park
- Department of Internal Medicine, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Youngjin Han
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Hyangkyoung Kim
- Department of Surgery, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul, 06973 South Korea
| | - Tae-Won Kwon
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| | - Yong-Pil Cho
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Asanbyeongwon-gil 86, Songpa-gu, Seoul, 05505 South Korea
| |
Collapse
|
31
|
Weir-McCall JR, Khan F, Cassidy DB, Thakur A, Summersgill J, Matthew SZ, Adams F, Dove F, Gandy SJ, Colhoun HM, Belch JJF, Houston JG. Effects of inaccuracies in arterial path length measurement on differences in MRI and tonometry measured pulse wave velocity. BMC Cardiovasc Disord 2017; 17:118. [PMID: 28486936 PMCID: PMC5424356 DOI: 10.1186/s12872-017-0546-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/28/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Carotid-femoral pulse wave velocity (cf-PWV) and aortic PWV measured using MRI (MRI-PWV) show good correlation, but with a significant and consistent bias across studies. The aim of the current study was to evaluate whether the differences between cf.-PWV and MRI-PWV can be accounted for by inaccuracies of currently used distance measurements. METHODS One hundred fourteen study participants were recruited into one of 4 groups: Type 2 diabetes melltus (T2DM) with cardiovascular disease (CVD) (n = 23), T2DM without CVD (n = 41), CVD without T2DM (n = 25) and a control group (n = 25). All participants underwent cf.-PWV, cardiac MRI and whole body MR angiography(WB-MRA). 90 study participants also underwent aortic PWV using MRI. cf.-PWVEXT was performed using a SphygmoCor device (Atcor Medical, West Ryde, Australia). The true intra-arterial pathlength was measured using the WB-MRA and then used to recalculate the cf.-PWVEXT to give a cf.-PWVMRA. RESULTS Distance measurements were significantly lower on WB-MRA than on external tape measure (mean diff = -85.4 ± 54.0 mm,p < 0.001). MRI-PWV was significantly lower than cf.-PWVEXT (MRI-PWV = 8.1 ± 2.9 vs. cf.-PWVEXT = 10.9 ± 2.7 ms-1,p < 0.001). When cf.-PWV was recalculated using the inter-arterial distance from WB-MRA, this difference was significantly reduced but not lost (MRI-PWV = 8.1 ± 2.9 ms-1 vs. cf.-PWVMRA 9.1 ± 2.1 ms-1, mean diff = -0.96 ± 2.52 ms-1,p = 0.001). Recalculation of the PWV increased correlation with age and pulse pressure. CONCLUSION Differences in cf.-PWV and MRI PWV can be predominantly but not entirely explained by inaccuracies introduced by the use of simple surface measurements to represent the convoluted arterial path between the carotid and femoral arteries.
Collapse
Affiliation(s)
- Jonathan R Weir-McCall
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Faisel Khan
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Deirdre B Cassidy
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Arsh Thakur
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Jennifer Summersgill
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Shona Z Matthew
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Fiona Adams
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Fiona Dove
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - Stephen J Gandy
- NHS Tayside Medical Physics, Ninewells Hospital, Dundee, Scotland, UK
| | - Helen M Colhoun
- Centre for Genomic and Experimental Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU Scotland, UK
| | - Jill JF Belch
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| | - J Graeme Houston
- Division of Molecular and Clinical Medicine, Medical Research Institute, University of Dundee, Dundee, DD1 9SY UK
| |
Collapse
|
32
|
Casanova MA, Medeiros F, Trindade M, Cohen C, Oigman W, Neves MF. Omega-3 fatty acids supplementation improves endothelial function and arterial stiffness in hypertensive patients with hypertriglyceridemia and high cardiovascular risk. ACTA ACUST UNITED AC 2016; 11:10-19. [PMID: 27876342 DOI: 10.1016/j.jash.2016.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/23/2016] [Accepted: 10/18/2016] [Indexed: 01/07/2023]
Abstract
Association between hypertriglyceridemia and cardiovascular (CV) disease is still controversial. The purpose of this study was to compare omega-3 and ciprofibrate effects on the vascular structure and function in low and high CV risk hypertensive patients with hypertriglyceridemia. Twenty-nine adults with triglycerides 150-499 mg/dL were divided into low (<7.5%) and high (≥7.5%) CV risk, randomized to receive omega-3 fatty acids 1800 mg/d or ciprofibrate 100 mg/d for 12 weeks. Treatment was switched after 8-week washout. Clinical evaluation and vascular tests were assessed at baseline and after intervention. Peripheral (131 ± 3 to 125 ± 3 mm Hg, P < .05) and aortic (124 ± 3 to 118 ± 2 mg/dL, P < .05) systolic blood pressure were decreased by ciprofibrate in low-risk patients. In high-risk patients, pulse wave velocity was reduced (10.4 ± 0.4 to 9.4 ± 0.3 m/s, P < .05) and flow-mediated dilation was increased (11.1 ± 1.6 to 13.5 ± 1.2%, P < .05) by omega-3. In conclusion, omega-3 improved arterial stiffness and endothelial function, pointing out the beneficial effect of this therapy on vascular aging, in high-risk patients.
Collapse
Affiliation(s)
- Marcela A Casanova
- Department of Clinical Medicine, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Fernanda Medeiros
- Department of Applied Nutrition, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle Trindade
- Department of Clinical Medicine, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Célia Cohen
- Department of Clinical Medicine, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wille Oigman
- Department of Clinical Medicine, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mario Fritsch Neves
- Department of Clinical Medicine, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Gonçalves I, Edsfeldt A, Colhoun HM, Shore AC, Palombo C, Natali A, Fredrikson GN, Björkbacka H, Wigren M, Bengtsson E, Östling G, Aizawa K, Casanova F, Persson M, Gooding K, Gates P, Khan F, Looker HC, Adams F, Belch J, Pinnola S, Venturi E, Kozakova M, Gan LM, Schnecke V, Nilsson J. Association between renin and atherosclerotic burden in subjects with and without type 2 diabetes. BMC Cardiovasc Disord 2016; 16:171. [PMID: 27596252 PMCID: PMC5011869 DOI: 10.1186/s12872-016-0346-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/11/2016] [Indexed: 01/01/2023] Open
Abstract
Background Activation of the renin-angiotensin-aldosterone-system (RAAS) has been proposed to contribute to development of vascular complications in type 2 diabetes (T2D). The aim of the present study was to determine if plasma renin levels are associated with the severity of vascular changes in subjects with and without T2D. Methods Renin was analyzed by the Proximity Extension Assay in subjects with (n = 985) and without (n = 515) T2D participating in the SUMMIT (SUrrogate markers for Micro- and Macro-vascular hard endpoints for Innovative diabetes Tools) study and in 205 carotid endarterectomy patients. Vascular changes were assessed by determining ankle-brachial pressure index (ABPI), carotid intima-media thickness (IMT), carotid plaque area, pulse wave velocity (PWV) and the reactivity hyperemia index (RHI). Results Plasma renin was elevated in subjects with T2D and demonstrated risk factor-independent association with prevalent cardiovascular disease both in subjects with and without T2D. Renin levels increased with age, body mass index, HbA1c and correlated inversely with HDL. Subjects with T2D had more severe carotid disease, increased arterial stiffness, and impaired endothelial function. Risk factor-independent associations between renin and APBI, bulb IMT, carotid plaque area were observed in both T2D and non-T2D subjects. These associations were independent of treatment with RAAS inhibitors. Only weak associations existed between plasma renin and the expression of pro-inflammatory and fibrous components in plaques from 205 endarterectomy patients. Conclusions Our findings provide clinical evidence for associations between systemic RAAS activation and atherosclerotic burden and suggest that this association is of particular importance in T2D.
Collapse
Affiliation(s)
- Isabel Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Andreas Edsfeldt
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Helen M Colhoun
- Medical Research Institute, University of Dundee, Dundee, UK
| | - Angela C Shore
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular Pathology, and Critical Area Medicine, Pisa, Italy
| | - Andrea Natali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Harry Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Maria Wigren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Gerd Östling
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Kunihiko Aizawa
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK
| | - Francesco Casanova
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK
| | | | - Kim Gooding
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK
| | - Phil Gates
- Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK
| | - Faisel Khan
- Medical Research Institute, University of Dundee, Dundee, UK
| | - Helen C Looker
- Medical Research Institute, University of Dundee, Dundee, UK
| | - Fiona Adams
- Medical Research Institute, University of Dundee, Dundee, UK
| | - Jill Belch
- Medical Research Institute, University of Dundee, Dundee, UK
| | - Silvia Pinnola
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Venturi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Li-Ming Gan
- AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden
| | - Volker Schnecke
- AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.
| | | |
Collapse
|
34
|
Spigoni V, Aldigeri R, Picconi A, Derlindati E, Franzini L, Haddoub S, Prampolini G, Vigna GB, Zavaroni I, Bonadonna RC, Dei Cas A. Telomere length is independently associated with subclinical atherosclerosis in subjects with type 2 diabetes: a cross-sectional study. Acta Diabetol 2016; 53:661-7. [PMID: 27020053 DOI: 10.1007/s00592-016-0857-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/12/2016] [Indexed: 12/12/2022]
Abstract
AIMS Individuals with type 2 diabetes show shorter leukocyte telomere length (LTL) compared to people without diabetes. Reduced LTL is associated with increased carotid intima-media thickness (IMT) in healthy subjects. The aim of the study is to assess whether LTL also correlates with IMT in patients with diabetes. METHODS In a cohort of 104 subjects with type 2 diabetes and atherogenic dyslipidemia, we assessed anthropometric, hemodynamic and metabolic parameters. Common carotid IMT was expressed as the maximum IMT. LTL was assessed by a specific real-time PCR reaction. RESULTS At univariate analysis, IMT values were positively correlated with age (p < 0.001), previous history of cardiovascular events (p < 0.005), fasting plasma glucose (p < 0.01), HbA1c (p < 0.05) and negatively correlated with LTL (p < 0.05). In a multivariate model, age (p < 0.001) and LTL (p < 0.05) were the only independent predictors of maximum IMT, with an adjusted R (2) of 0.22. CONCLUSIONS LTL is an independent predictor of subclinical atherosclerosis pointing to a role of LTL as an early marker of vascular burden and cardiovascular disease also in type 2 diabetes.
Collapse
Affiliation(s)
- Valentina Spigoni
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Raffaella Aldigeri
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Angela Picconi
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Eleonora Derlindati
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Laura Franzini
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Silvia Haddoub
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Giorgia Prampolini
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Giovanni B Vigna
- Internal Medicine Unit, Department of Medicine, Azienda Ospedaliero-Universitaria of Ferrara, Ferrara, Italy
| | - Ivana Zavaroni
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Riccardo C Bonadonna
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy
| | - Alessandra Dei Cas
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci, 14, 43126, Parma, Italy.
| |
Collapse
|
35
|
Li MF, Zhao CC, Li TT, Tu YF, Lu JX, Zhang R, Chen MY, Bao YQ, Li LX, Jia WP. The coexistence of carotid and lower extremity atherosclerosis further increases cardio-cerebrovascular risk in type 2 diabetes. Cardiovasc Diabetol 2016; 15:43. [PMID: 26944724 PMCID: PMC4779218 DOI: 10.1186/s12933-016-0360-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 02/25/2016] [Indexed: 12/27/2022] Open
Abstract
Background Both carotid and lower limb atherosclerosis are associated with increased cardiovascular and cerebrovascular risks. However, it is still unclear whether the concomitant presence of carotid and lower extremity atherosclerosis further increases the cardiovascular and cerebrovascular risks. Therefore, our aim is to investigate whether the coexistence of carotid and lower extremity atherosclerosis was associated with higher cardiovascular and cerebrovascular risks in patients with type 2 diabetes. Methods This cross-sectional study was performed in 2830 hospitalized patients with type 2 diabetes. Based on carotid and lower limb Doppler ultrasound results, the patients were divided into three groups including 711 subjects without atherosclerosis, 999 subjects with either carotid or lower limb atherosclerosis, and 1120 subjects with both carotid and lower limb atherosclerosis. And we compared the clinical characteristics and prevalence of both cardio-cerebrovascular events (CCBVEs) and self-reported cardio- cerebrovascular diseases (CCBVDs) among the three groups. Results After adjusting for age, sex, and duration of diabetes, there were significant increases in the prevalence of both CCBVEs (3.8 vs. 11.8 vs. 26.4 %, p < 0.001 for trend) and self-reported CCBVDs (6.9 vs. 19.9 vs. 36.5 %, p < 0.001 for trend) across the three groups (diabetics without atherosclerosis, diabetics with either carotid or lower limb atherosclerosis, and diabetics with both carotid and lower extremity atherosclerosis). A fully adjusted logistic regression analysis also revealed that compared with those without atherosclerosis, those with either carotid or lower limb atherosclerosis had higher risk of CCBVEs (OR 1.724, 95 % CI 1.001–2.966) and self-reported CCBVDs (OR 1.705, 95 % CI 1.115–2.605), and those with concomitant presence of carotid and lower extremity atherosclerosis had the highest risk of CCBVEs (OR 2.869, 95 % CI 1.660–4.960) and self-reported CCBVDs (2.147, 95 % CI 1.388–3.320)(p < 0.001 for trend in CCBVEs and p = 0.002 for trend in CCBVDs, respectively). Conclusions Either carotid or lower limb atherosclerosis was obviously related to increased cardio-cerebrovascular risk in type 2 diabetes. The concomitant presence of carotid and lower extremity atherosclerosis further increased cardio-cerebrovascular risk in patients with type 2 diabetes. The combined application of carotid and lower extremity ultrasonography may help identify type 2 diabetics with higher cardio-cerebrovascular risk.
Collapse
Affiliation(s)
- Mei-Fang Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China. .,Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Cui-Chun Zhao
- Department of VIP, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Ting-Ting Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Yin-Fang Tu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Jun-Xi Lu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Rong Zhang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Ming-Yun Chen
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Yu-Qian Bao
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Lian-Xi Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Wei-Ping Jia
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Clinical Center for Metabolic Diseases, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
36
|
Wu CF, Liu PY, Wu TJ, Hung Y, Yang SP, Lin GM. Therapeutic modification of arterial stiffness: An update and comprehensive review. World J Cardiol 2015; 7:742-753. [PMID: 26635922 PMCID: PMC4660469 DOI: 10.4330/wjc.v7.i11.742] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/30/2015] [Accepted: 09/28/2015] [Indexed: 02/06/2023] Open
Abstract
Arterial stiffness has been recognized as a marker of cardiovascular disease and associated with long-term worse clinical outcomes in several populations. Age, hypertension, smoking, and dyslipidemia, known as traditional vascular risk factors, as well as diabetes, obesity, and systemic inflammation lead to both atherosclerosis and arterial stiffness. Targeting multiple modifiable risk factors has become the main therapeutic strategy to improve arterial stiffness in patients at high cardiovascular risk. Additionally to life style modifications, long-term ω-3 fatty acids (fish oil) supplementation in diet may improve arterial stiffness in the population with hypertension or metabolic syndrome. Pharmacological treatment such as renin-angiotensin-aldosterone system antagonists, metformin, and 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors were useful in individuals with hypertension and diabetes. In obese population with obstructive sleep apnea, weight reduction, aerobic exercise, and continuous positive airway pressure treatment may also improve arterial stiffness. In the populations with chronic inflammatory disease such as rheumatoid arthritis, a use of antibodies against tumor necrosis factor-alpha could work effectively. Other therapeutic options such as renal sympathetic nerve denervation for patients with resistant hypertension are investigated in many ongoing clinical trials. Therefore our comprehensive review provides knowledge in detail regarding many aspects of pathogenesis, measurement, and management of arterial stiffness in several populations, which would be helpful for physicians to make clinical decision.
Collapse
|
37
|
Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J 2015; 36:2288-96. [PMID: 26063450 DOI: 10.1093/eurheartj/ehv239] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/16/2015] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by multiple pathophysiologic abnormalities. With time, multiple glucose-lowering medications are commonly required to reduce and maintain plasma glucose concentrations within the normal range. Type 2 diabetes mellitus individuals also are at a very high risk for microvascular complications and the incidence of heart attack and stroke is increased two- to three-fold compared with non-diabetic individuals. Therefore, when selecting medications to normalize glucose levels in T2DM patients, it is important that the agent not aggravate, and ideally even improve, cardiovascular risk factors (CVRFs) and reduce cardiovascular morbidity and mortality. In this review, we examine the effect of oral (metformin, sulfonylureas, meglitinides, thiazolidinediones, DPP4 inhibitors, SGLT2 inhibitors, and α-glucosidase inhibitors) and injectable (glucagon-like peptide-1 receptor agonists and insulin) glucose-lowering drugs on established CVRFs and long-term studies of cardiovascular outcomes. Firm evidence that in T2DM cardiovascular disease can be reversed or prevented by improving glycaemic control is still incomplete and must await large, long-term clinical trials in patients at low risk using modern treatment strategies, i.e., drug combinations designed to maximize HbA1c reduction while minimizing hypoglycaemia and excessive weight gain.
Collapse
Affiliation(s)
- Ele Ferrannini
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
38
|
Goncalves I, Bengtsson E, Colhoun HM, Shore AC, Palombo C, Natali A, Edsfeldt A, Dunér P, Fredrikson GN, Björkbacka H, Östling G, Aizawa K, Casanova F, Persson M, Gooding K, Strain D, Khan F, Looker HC, Adams F, Belch J, Pinnoli S, Venturi E, Kozakova M, Gan LM, Schnecke V, Nilsson J. Elevated Plasma Levels of MMP-12 Are Associated With Atherosclerotic Burden and Symptomatic Cardiovascular Disease in Subjects With Type 2 Diabetes. Arterioscler Thromb Vasc Biol 2015; 35:1723-31. [PMID: 25953645 DOI: 10.1161/atvbaha.115.305631] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Matrix metalloproteinases (MMPs) degrade extracellular matrix proteins and play important roles in development and tissue repair. They have also been shown to have both protective and pathogenic effects in atherosclerosis, and experimental studies have suggested that MMP-12 contributes to plaque growth and destabilization. The objective of this study was to investigate the associations between circulating MMPs, atherosclerosis burden, and incidence of cardiovascular disease with a particular focus on type 2 diabetes mellitus. APPROACH AND RESULTS Plasma levels of MMP-1, -3, -7, -10, and -12 were analyzed by the Proximity Extension Assay technology in 1500 subjects participating in the SUMMIT (surrogate markers for micro- and macrovascular hard end points for innovative diabetes tools) study, 384 incident coronary cases, and 409 matched controls in the Malmö Diet and Cancer study and in 205 carotid endarterectomy patients. Plasma MMP-7 and -12 were higher in subjects with type 2 diabetes mellitus, increased with age and impaired renal function, and was independently associated with prevalent cardiovascular disease, atherosclerotic burden (as assessed by carotid intima-media thickness and ankle-brachial pressure index), arterial stiffness, and plaque inflammation. Baseline MMP-7 and -12 levels were increased in Malmö Diet and Cancer subjects who had a coronary event during follow-up. CONCLUSIONS The plasma level of MMP-7 and -12 are elevated in type 2 diabetes mellitus, associated with more severe atherosclerosis and an increased incidence of coronary events. These observations provide clinical support to previous experimental studies, demonstrating a role for these MMPs in plaque development, and suggest that they are potential biomarkers of atherosclerosis burden and cardiovascular disease risk.
Collapse
Affiliation(s)
- Isabel Goncalves
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Eva Bengtsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen M Colhoun
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Angela C Shore
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Carlo Palombo
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andrea Natali
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andreas Edsfeldt
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Pontus Dunér
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gunilla Nordin Fredrikson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Harry Björkbacka
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gerd Östling
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kunihiko Aizawa
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Francesco Casanova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Margaretha Persson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kim Gooding
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - David Strain
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Faisel Khan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen C Looker
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Fiona Adams
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jill Belch
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Silvia Pinnoli
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Elena Venturi
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Michaela Kozakova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Li-Ming Gan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Volker Schnecke
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.).
| | | |
Collapse
|