101
|
Halon DA, Lavi I, Barnett-Griness O, Rubinshtein R, Zafrir B, Azencot M, Lewis BS. Plaque Morphology as Predictor of Late Plaque Events in Patients With Asymptomatic Type 2 Diabetes. JACC Cardiovasc Imaging 2019; 12:1353-1363. [DOI: 10.1016/j.jcmg.2018.02.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 02/23/2018] [Indexed: 01/09/2023]
|
102
|
Sharma AN, Deyell JS, Sharma SN, Barseghian A. Role of and Recent Evidence for Antiplatelet Therapy in Prevention of Cardiovascular Disease in Diabetes. Curr Cardiol Rep 2019; 21:78. [PMID: 31254105 DOI: 10.1007/s11886-019-1168-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW When treating patients with diabetes mellitus (DM), the benefits of antiplatelet therapy in preventing cardiovascular disease must be weighed against an increased risk of bleeding. Recent trials have sought to determine both the optimal anti-platelet regimen for patients with DM, and who specifically requires medication among the DM population. This paper will review recent trials and evidence recommending the use of antiplatelet therapy in the prevention of cardiovascular disease in patients with diabetes. RECENT FINDINGS Seven notable trials assessed the effectiveness of antiplatelet therapy in the DM population. The ASCEND trial concluded 100 mg aspirin/day reduced rates of serious vascular events (OR 0.88, p < 0.01) but also increased rates of major bleeding events (OR 1.29, p < 0.01). The DAPT study revealed a longer dual antiplatelet regimen (30 months vs. 18 months) after coronary stent placement was more effective in reducing rates of stent thrombosis (0.5% vs. 1.1%, p = 0.06) and rates of myocardial infarction (3.5% vs. 4.8%, p = 0.06). DECLARE DIABETES showed that adding cilostazol to dual antiplatelet therapy after a coronary stent procedure reduced rates of in-stent and in-segment late loss and increased rates of revascularization (p < 0.04). In PEGASUS-TIMI, daily ticagrelor demonstrated reduced rates of major adverse cardiovascular and cerebrovascular events (OR 0.84, p < 0.04). The DAVID trial compared daily picotamide with daily aspirin therapy, finding reduced mortality rates in the picotamide group (OR 0.55, p < 0.05). Lastly, ACUITY found bivalirudin monotherapy resulted in lower rates of major bleeding events when compared to a glycoprotein IIb/IIa inhibitor and heparin or bivalirudin combination regimen (p < 0.01). Dual antiplatelet therapy guidelines still typically revolve around aspirin, but an increasing number of studies have demonstrated other drugs that may have a role in preventing atherosclerotic cardiovascular disease while decreasing the risk of major bleeding. Overall, it is wise to weigh the cardiovascular risk of a DM patient before prescribing antiplatelet medication. More research is necessary to determine a universal drug or combination of drugs that is safe and effective for DM patients.
Collapse
Affiliation(s)
- Ajay Nair Sharma
- School of Medicine, University of California Irvine, Irvine, CA, USA.,Division of Cardiology, UC Irvine, 333 City Blvd West. Ste 400, Orange, CA, 92868, USA
| | - Jacob S Deyell
- School of Medicine, University of California Irvine, Irvine, CA, USA.,Division of Cardiology, UC Irvine, 333 City Blvd West. Ste 400, Orange, CA, 92868, USA
| | | | - Ailin Barseghian
- Division of Cardiology, UC Irvine, 333 City Blvd West. Ste 400, Orange, CA, 92868, USA.
| |
Collapse
|
103
|
MacLean M, Derk J, Ruiz HH, Juranek JK, Ramasamy R, Schmidt AM. The Receptor for Advanced Glycation End Products (RAGE) and DIAPH1: Implications for vascular and neuroinflammatory dysfunction in disorders of the central nervous system. Neurochem Int 2019; 126:154-164. [PMID: 30902646 PMCID: PMC10976457 DOI: 10.1016/j.neuint.2019.03.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/11/2022]
Abstract
The Receptor for Advanced Glycation End Products (RAGE) is expressed by multiple cell types in the brain and spinal cord that are linked to the pathogenesis of neurovascular and neurodegenerative disorders, including neurons, glia (microglia and astrocytes) and vascular cells (endothelial cells, smooth muscle cells and pericytes). Mounting structural and functional evidence implicates the interaction of the RAGE cytoplasmic domain with the formin, Diaphanous1 (DIAPH1), as the key cytoplasmic hub for RAGE ligand-mediated activation of cellular signaling. In aging and diabetes, the ligands of the receptor abound, both in the central nervous system (CNS) and in the periphery. Such accumulation of RAGE ligands triggers multiple downstream events, including upregulation of RAGE itself. Once set in motion, cell intrinsic and cell-cell communication mechanisms, at least in part via RAGE, trigger dysfunction in the CNS. A key outcome of endothelial dysfunction is reduction in cerebral blood flow and increased permeability of the blood brain barrier, conditions that facilitate entry of activated leukocytes into the CNS, thereby amplifying primary nodes of CNS cellular stress. This contribution details a review of the ligands of RAGE, the mechanisms and consequences of RAGE signal transduction, and cites multiple examples of published work in which RAGE contributes to the pathogenesis of neurovascular perturbation. Insights into potential therapeutic modalities targeting the RAGE signal transduction axis for disorders of CNS vascular dysfunction and neurodegeneration are also discussed.
Collapse
Affiliation(s)
- Michael MacLean
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Julia Derk
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Henry H Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Judyta K Juranek
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
104
|
Grauen Larsen H, Yndigegn T, Marinkovic G, Grufman H, Mares R, Nilsson J, Goncalves I, Schiopu A. The soluble receptor for advanced glycation end-products (sRAGE) has a dual phase-dependent association with residual cardiovascular risk after an acute coronary event. Atherosclerosis 2019; 287:16-23. [PMID: 31181415 DOI: 10.1016/j.atherosclerosis.2019.05.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS The pro-inflammatory alarmin S100A12 (EN-RAGE) and the soluble form of its receptor, the receptor for advanced glycation endproducts (sRAGE), have diverging roles in cardiovascular disease. In experimental studies, S100A12 promoted atherosclerosis while sRAGE treatment was anti-atherogenic and reduced myocardial infarction size by scavenging RAGE ligands. Here, we aimed to explore the links between S100A12, sRAGE, and long-term prognosis after an acute coronary syndrome (ACS). METHODS We measured S100A12 and sRAGE in 524 patients within 24 h after an ACS, and again 6 weeks later in a subgroup of 114 patients. This subgroup also completed a follow-up echocardiography after 1 year. The median follow-up time for recurrent major adverse cardiovascular events (MACE), defined as recurrent ACS or cardiovascular death, was 25.7 ± 12.6 months. RESULTS In Cox proportional hazard analyses, baseline S100A12 and sRAGE were positively associated with the risk of MACE, independently of traditional cardiovascular risk factors. The association between sRAGE and MACE remained significant after additional adjustment for troponin T, NT-proBNP and hsCRP [HR 95%CI for highest versus lowest tertile 3.2 (1.5-6.5), p = 0.002]. High sRAGE was also associated with deteriorating left ventricular function and an increased rate of heart failure hospitalization post-discharge. In contrast, patients with increasing sRAGE at 6 weeks compared to baseline had lower incidence of recurrent ACS. CONCLUSIONS Our data suggest that sRAGE has a dual, phase-dependent association with residual cardiovascular risk after ACS. These findings are important for the design and interpretation of future studies on sRAGE as biomarker and potential treatment in ACS patients.
Collapse
Affiliation(s)
- Helena Grauen Larsen
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden
| | - Troels Yndigegn
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Goran Marinkovic
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Helena Grufman
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Razvan Mares
- University of Medicine, Pharmacy, Sciences and Technology of Tirgu-Mures, 38 Gh. Marinescu Str., Tirgu-Mures, Romania
| | - Jan Nilsson
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden
| | - Isabel Goncalves
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden
| | - Alexandru Schiopu
- Experimental Cardiovascular Research Unit, Department of Clinical Sciences Malmö, Lund University, CRC 91:12 Jan Waldenströms gata 35, 214 28, Malmö, Sweden; Department of Cardiology, Skane University Hospital Malmö, Sweden; University of Medicine, Pharmacy, Sciences and Technology of Tirgu-Mures, 38 Gh. Marinescu Str., Tirgu-Mures, Romania.
| |
Collapse
|
105
|
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
|
106
|
Jang EJ, Baek SE, Kim EJ, Park SY, Kim CD. HMGB1 enhances AGE-mediated VSMC proliferation via an increase in 5-LO-linked RAGE expression. Vascul Pharmacol 2019; 118-119:106559. [PMID: 30954689 DOI: 10.1016/j.vph.2019.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/28/2019] [Accepted: 04/02/2019] [Indexed: 01/11/2023]
Abstract
Receptors for advanced glycation end-product (RAGE) play a pivotal role in the progression of proliferative vascular diseases. However, the precise mechanisms regulating RAGE expression in vascular smooth muscle cells (VSMCs) of the injured vasculatures is unclear. Given the potential importance of 5-lipoxygenase (5-LO) derived mediators in cellular responses mediated by RAGE, this study aimed to evaluate in VSMCs treated with high mobility group box 1 (HMGB1): 1) the RAGE expression; 2) the AGE-induced VSMC proliferation; 3) the role of 5-LO signaling in HMGB1-induced RAGE expression. In cultured human VSMCs stimulated with HMGB1 (100 ng/ml), RAGE mRNA and protein expression were markedly increased along with an increase in AGE-mediated VSMC proliferation. Both of these effects were markedly attenuated in cells pretreated with zileuton (1-10 μM), a 5-LO inhibitor, as well as in cells transfected with 5-LO siRNA, suggesting a potential involvement of 5-LO signaling in HMGB1-mediated RAGE expression in VSMCs. Moreover, 5-LO expression, accompanied by production of leukotrienes was markedly increased in HMGB1-stimulated VSMCs, which was attenuated in cells deficient of TLR2 or RAGE. Taken together, our results suggest that HMGB1-induced increase in 5-LO expression enhances RAGE expression in VSMCs, which stimulates AGE-mediated VSMC proliferation. Thus, the 5-LO-RAGE signaling axis in VSMCs might serve as a potential therapeutic target for vascular remodeling in the injured vasculature.
Collapse
Affiliation(s)
- Eun Jeong Jang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Seung Eun Baek
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Eun Jung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|
107
|
Wang J, Jia L, Li X, Jin S, Li X, Liu F, Shan C, Zhang Y, Yang Y. New Insights into the Association between Fibrinogen and Coronary Atherosclerotic Plaque Vulnerability: An Intravascular Optical Coherence Tomography Study. Cardiovasc Ther 2019; 2019:8563717. [PMID: 31772619 PMCID: PMC6740041 DOI: 10.1155/2019/8563717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 01/31/2019] [Accepted: 02/24/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fibrinogen levels have been associated with coronary plaque vulnerability in experimental studies. However, it has yet to be determined if serum fibrinogen levels are independently associated with coronary plaque vulnerability as detected by optical coherence tomography (OCT) in patients with coronary heart disease. METHODS Patients with coronary heart disease (CHD) who underwent coronary angiography and OCT in our department from January 2015 to August 2018 were included in this study. Coronary lesions were categorized as ruptured plaque, nonruptured with thin-cap fibroatheroma (TCFA), and nonruptured and non-TCFA. Presence of ruptured plaque and nonruptured with TCFA was considered to be vulnerable lesions. Determinants of coronary vulnerability were evaluated by multivariable logistic regression analyses. RESULTS A total of 154 patients were included in this study; 17 patients had ruptured plaques, 15 had nonruptured plaques with TCFA, and 122 had nonruptured plaques with non-TCFA. Results of univariate analyses showed that being male, diabetes, current smoking, high body mass index (BMI), and clinical diagnosis of acute coronary syndrome (ACS) were associated with coronary vulnerability. No significant differences were detected in patient characteristics, coronary angiographic findings, and OCT results between patients with higher and normal fibrinogen. Results of multivariate logistic analyses showed that diabetes and ACS were associated with TCFA, while diabetes, higher BMI, and ACS were associated with plaque rupture. CONCLUSIONS Diabetes, higher BMI, and ACS are independently associated with coronary vulnerability as detected by OCT. Serum fibrinogen was not associated with coronary vulnerability in our cohort.
Collapse
Affiliation(s)
- Jun Wang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Lu Jia
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xing Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Siyu Jin
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Xiaomei Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Fen Liu
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Chunfang Shan
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Yu Zhang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| | - Yining Yang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China
| |
Collapse
|
108
|
Schmidt AM. Diabetes Mellitus and Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:558-568. [PMID: 30786741 PMCID: PMC6532416 DOI: 10.1161/atvbaha.119.310961] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease remains a leading cause of morbidity and mortality in people with types 1 or 2 diabetes mellitus. Although beneficial roles for strict control of hyperglycemia have been suggested, such a strategy is not without liabilities. Specifically, the risk of hypoglycemia and its consequences remain an omnipresent threat with such approaches. The advent of the CVOT (Cardiovascular Outcomes Trials) for new antidiabetes mellitus treatments has uncovered unexpected benefits of cardiovascular protection in some of the new classes of agents, such as the GLP-1 RAs (glucagon-like peptide-1 receptor agonists) and the SGLT-2 (sodium-glucose cotransporter-2) inhibitors. Further, state-of-the-art approaches, such as antibodies to PCKSK9 (proprotein convertase subtilisin-kexin type 9); RNA therapeutics; agents targeting distinct components of the immune/inflammatory response; and novel small molecules that block the actions of RAGE (receptor for advanced glycation end products) signaling, also hold potential as new therapies for diabetes mellitus and cardiovascular disease. Finally, interventions such as weight loss, through bariatric surgery, may hold promise for benefit in diabetes and cardiovascular disease. In this Brief Review, some of the novel approaches and emerging targets for the treatment of diabetes mellitus and cardiovascular disease are discussed. Ultimately, identification of the optimal timing and combinations of such interventions, especially in the context of personalized approaches, together with emerging disease-modifying agents, holds great promise to reduce the burden that diabetes poses to the cardiovascular system.
Collapse
Affiliation(s)
- Ann Marie Schmidt
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York
| |
Collapse
|
109
|
Unraveling the Cardiovascular PROSPECTs of Patients With Prediabetes. JACC Cardiovasc Imaging 2019; 12:742-744. [DOI: 10.1016/j.jcmg.2017.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/09/2017] [Accepted: 10/12/2017] [Indexed: 11/19/2022]
|
110
|
Su G, Zhang T, Yang HX, Dai WL, Wang T, Tian L, Mi SH. Association of Isoprostanes-Related Oxidative Stress with Vulnerability of Culprit Lesions in Diabetic Patients with Acute Coronary Syndrome. Int Heart J 2019; 60:271-279. [PMID: 30745536 DOI: 10.1536/ihj.18-233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Urinary excretion of 8-iso-prostaglandin F2α (8-iso-PGF2α), a reliable biomarker for enhanced oxidant stress in vivo, has been described in association with diabetes and coronary heart disease. The aim of this study was to evaluate the relationship between urinary 8-iso-PGF2α levels and the characteristics of coronary culprit lesion in diabetic patients with acute coronary syndrome (ACS). A total of 79 diabetic patients with ACS were included. iMAP intravascular ultrasound (iMAP-IVUS) was performed to evaluate the characteristics of culprit plaques. Fasting urinary 8-iso-PGF2α level was measured and corrected by creatinine clearance. iMAP-IVUS data showed culprit plaques in high urinary 8-iso-PGF2α level patients had a greater percentage of necrotic core and less fibrous components. High urinary 8-iso-PGF2α levels were correlated with increased necrotic plaque components (r = 0.325, P = 0.003). Meanwhile, the presence of thin-capped fibroatheroma (50.0% versus 11.5%, P = 0.003), ruptured plaques (30.8% versus 7.7%, P = 0.035), and thrombus (38.5% versus 7.7%, P = 0.008) were significantly more frequent in the upper tertile of urinary 8-iso-PGF2α levels than in the low tertile. Multivariate analysis showed high levels of urinary 8-iso-PGF2α (OR 4.240, P = 0.007) was independently associated with the presence of vulnerable culprit plaque in diabetic ACS patients. Urinary 8-iso-PGF2α also displayed a significant value in predicting vulnerable plaques in diabetic patients with ACS by constructing the receiver-operating characteristic (ROC) curve (Area under the ROC curve: 0.713, P = 0.001). Urinary 8-iso-PGF2α levels are associated with the vulnerability of the coronary culprit lesion in diabetic patients with ACS and may provide additional information for risk assessment in suspected vulnerable patients.
Collapse
Affiliation(s)
- Gong Su
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| | - Tao Zhang
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| | - Hong-Xia Yang
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| | - Wen-Long Dai
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| | - Tao Wang
- Department of Thoracic Surgery, People Liberation Army General Hospital
| | - Lei Tian
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| | - Shu-Hua Mi
- Center of Cardiology, Beijing An Zhen Hospital, Capital Medical University
| |
Collapse
|
111
|
Bjornstad P, Wiromrat P, Johnson RJ, Sippl R, Cherney DZI, Wong R, Rewers MJ, Snell-Bergeon JK. Serum Uromodulin Predicts Less Coronary Artery Calcification and Diabetic Kidney Disease Over 12 Years in Adults With Type 1 Diabetes: The CACTI Study. Diabetes Care 2019; 42:297-302. [PMID: 30482755 PMCID: PMC6341281 DOI: 10.2337/dc18-1527] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/23/2018] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Novel biomarkers are needed to better predict coronary artery calcification (CAC), a marker of subclinical atherosclerosis, and diabetic kidney disease (DKD) in type 1 diabetes. We evaluated the associations between serum uromodulin (SUMOD [a biomarker associated with anti-inflammatory and renal protective properties]), CAC progression, and DKD development over 12 years. RESEARCH DESIGN AND METHODS Participants (n = 527, 53% females) in the Coronary Artery Calcification in Type 1 Diabetes (CACTI) study were examined during 2002-2004, at a mean age of 39.6 ± 9.0 years and a median duration of diabetes of 24.8 years. Urine albumin-to-creatinine ratio (ACR) and estimated glomerular filtration rate (eGFR) determined by the CKD-EPI (Chronic Kidney Disease Epidemiology Collaboration) creatinine equation were measured at baseline and after a mean follow-up period of 12.1 ± 1.5 years. Elevated albumin excretion was defined as ACR ≥30 mg/g, rapid GFR decline (>3 mL/min/1.73 m2/year), and impaired GFR as eGFR <60 mL/min/1.73 m2. SUMOD was measured on stored baseline plasma samples (Meso Scale Discovery). CAC was measured using electron beam computed tomography. CAC progression was defined as a change in the square root-transformed CAC volume of ≥2.5. RESULTS Higher baseline SUMOD level conferred lower odds of CAC progression (odds ratio 0.68; 95% CI 0.48-0.97), incident elevated albumin excretion (0.37; 0.16-0.86), rapid GFR decline (0.56; 0.35-0.91), and impaired GFR (0.44; 0.24-0.83) per 1 SD increase in SUMOD (68.44 ng/mL) after adjustment for baseline age, sex, systolic blood pressure, LDL cholesterol, and albuminuria/GFR. The addition of SUMOD to models with traditional risk factors also significantly improved the prediction performance for CAC progression and incident DKD. CONCLUSIONS Higher baseline SUMOD level predicted lower odds of both CAC progression and incident DKD over 12 years in adults with type 1 diabetes.
Collapse
Affiliation(s)
- Petter Bjornstad
- Section of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO .,Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| | - Pattara Wiromrat
- Section of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, CO
| | - Richard J Johnson
- Division of Renal Disease and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Rachel Sippl
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, and Department of Physiology, University of Toronto, Ontario, Canada
| | - Randy Wong
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| | - Marian J Rewers
- Barbara Davis Center for Diabetes, University of Colorado Denver, Aurora, CO
| | | |
Collapse
|
112
|
Lovshin JA, Bjornstad P, Lovblom LE, Bai JW, Lytvyn Y, Boulet G, Farooqi MA, Santiago S, Orszag A, Scarr D, Weisman A, Keenan HA, Brent MH, Paul N, Bril V, Perkins BA, Cherney DZI. Atherosclerosis and Microvascular Complications: Results From the Canadian Study of Longevity in Type 1 Diabetes. Diabetes Care 2018; 41:2570-2578. [PMID: 30275283 PMCID: PMC6245210 DOI: 10.2337/dc18-1236] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Type 1 diabetes carries a significant risk for cardiovascular mortality, but it is unclear how atherosclerosis associates with microvascular complications. We aimed to determine the relationships between atherosclerotic burden and neuropathy, retinopathy, and diabetic kidney disease (DKD) in adults with a ≥50-year history of type 1 diabetes. RESEARCH DESIGN AND METHODS Adults with type 1 diabetes (n = 69) underwent coronary artery calcification (CAC) volume scoring by wide-volume computerized tomography. Microvascular complications were graded as follows: neuropathy by clinical assessment, electrophysiology, vibration and cooling detection thresholds, heart rate variability, and corneal confocal microscopy; retinopathy by ultra-wide-field retinal imaging; and DKD by renal hemodynamic function measured by inulin and para-aminohippurate clearance at baseline and after intravenous infusion of angiotensin II. The cohort was dichotomized to high (≥300 Agatston units [AU]) or low (<300 AU) CAC and was stratified by diabetes status. A comparator group without diabetes (n = 73) matched for age and sex also underwent all study procedures except for retinal imaging. RESULTS CAC scores were higher in participants with type 1 diabetes (median Agatston score 1,000 [interquartile range = 222, 2,373] AU vs. 1 [0.75] AU in comparators, P < 0.001). In participants with type 1 diabetes, high CAC scores associated with markers of neuropathy and retinopathy, but not with DKD, or renal hemodynamic function at baseline or in response to angiotensin II. CONCLUSIONS The presence of high CAC in adults with longstanding type 1 diabetes was associated with large nerve fiber neuropathy and retinopathy but not with renal hemodynamic function, suggesting that neuropathy, retinopathy, and macrovascular calcification share common risk factors.
Collapse
Affiliation(s)
- Julie A Lovshin
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada .,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Petter Bjornstad
- Division of Endocrinology, Department of Pediatrics, University of Colorado, Aurora, CO.,Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Leif E Lovblom
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Johnny-Wei Bai
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Yuliya Lytvyn
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Geneviève Boulet
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mohammed A Farooqi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Sam Santiago
- Joint Department of Medical Imaging, Division of Cardiothoracic Radiology, University Health Network, Toronto, Ontario, Canada
| | - Andrej Orszag
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Daniel Scarr
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Alanna Weisman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | - Michael H Brent
- Department of Ophthalmology and Vision Sciences, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Narinder Paul
- Joint Department of Medical Imaging, Division of Cardiothoracic Radiology, University Health Network, Toronto, Ontario, Canada
| | - Vera Bril
- The Ellen and Martin Prosserman Centre for Neuromuscular Diseases, Krembil Neuroscience Centre, Division of Neurology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Bruce A Perkins
- Division of Endocrinology and Metabolism, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - David Z I Cherney
- Division of Nephrology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
113
|
Miyoshi A, Koyama S, Sasagawa-Monden M, Kadoya M, Konishi K, Shoji T, Inaba M, Yamamoto Y, Koyama H. JNK and ATF4 as two important platforms for tumor necrosis factor-α-stimulated shedding of receptor for advanced glycation end products. FASEB J 2018; 33:3575-3589. [PMID: 30452882 DOI: 10.1096/fj.201701553rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Soluble receptor for advanced glycation end products (sRAGE), shed from cell surfaces, is found in human circulation and has been implicated in cardiovascular disease. Its pathophysiological regulation and underlying mechanisms are scarcely understood. In endothelium-specific human RAGE transgenic mice, human sRAGE was detected in circulation, whereas its level was markedly increased after LPS treatment. That increase was preceded by a rapid rise in TNF-α level. Treatment with TNF-α also significantly increased serum sRAGE. In human microvascular endothelial cells or human umbilical vein endothelial cells with RAGE overexpression, TNF-α markedly induced RAGE shedding, which was dependent on MMP9 and ADAM10. TNF-α-stimulated MMP9 expression was completely dependent on JNK activation, with its inhibition partially effective in suppressing TNF-α-induced RAGE shedding. In contrast, TNF-α transiently induced activation transcription factor (ATF)4, a major component in unfolded protein response (UPR), whereas knockdown of ATF4 abrogated TNF-α-stimulated RAGE shedding. Protein levels of the pro and activated forms of ADAM10 were also decreased by ATF4 knockdown, whereas inhibition of other components of UPR, including XBP1 and ATF6, failed to block TNF-α-stimulated RAGE shedding. Although the endoplasmic reticulum stressors thapsigargin and tunicamycin induced markedly and sustained expression of ATF4 and XBP-1, they did not induce RAGE shedding to the same level as TNF-α, suggesting that ATF4 is necessary but not sufficient alone for TNF-α-mediated RAGE shedding. ATF4 inhibition did not affect TNF-α-stimulated MMP9 expression, whereas inhibition of JNK activity did not influence ADAM10 activation. Thus, inflammatory cascades including TNF-α induced RAGE shedding in endothelial cells in vivo and in vitro. JNK and ATF4 may be 2 platforms for regulation of TNF-α-stimulated RAGE shedding.-Miyoshi, A., Koyama, S., Sasagawa-Monden, M., Kadoya, M., Konishi, K., Shoji, T., Inaba, M., Yamamoto, Y., Koyama, H. JNK and ATF4 as two important platforms for tumor necrosis factor-α-stimulated shedding of receptor for advanced glycation end products.
Collapse
Affiliation(s)
- Akio Miyoshi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Sachie Koyama
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masayo Sasagawa-Monden
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan.,Department of Endocrinology, Metabolism, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan; and
| | - Manabu Kadoya
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Kosuke Konishi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takuhito Shoji
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masaaki Inaba
- Department of Endocrinology, Metabolism, and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan; and
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hidenori Koyama
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
114
|
Ramuš SM, Petrovič D. Genetic Variations and Subclinical Markers of Carotid Atherosclerosis in Patients with Type 2 Diabetes Mellitus. Curr Vasc Pharmacol 2018; 17:16-24. [DOI: 10.2174/1570161116666180206112635] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
Abstract
Atherosclerosis and its cardiovascular complications are the main cause of death in diabetic
patients. Patients with diabetes mellitus have a greater than 10-fold risk of cardiovascular disease in
their lifetime. The carotid Intima-Media Thickness (cIMT), a surrogate marker for the presence and
progression of atherosclerosis, predicts future cardiovascular events in asymptomatic subjects with Type
2 Diabetes Mellitus (T2DM). This review focuses on genetic variants that contribute to the pathobiology
of subclinical atherosclerosis in the setting of T2DM. Specifically, we devoted our attention to wellstudied
genes selected for their relevance for atherosclerosis. These include: The Renin-Angiotensin-
Aldosterone System (RAAS), Apolipoprotein E (ApoE), Methylenetetrahydrofolate Reductase (MTHFR)
and pro-inflammatory genes.
</P><P>
The ever-growing availability of advanced genotyping technologies has made Genome-Wide Association
Studies (GWAS) possible. Although several bioinformatics tools have been developed to manage
and interpret the huge amounts of data produced, there has been limited success in the many attempts to
uncover the biological meaning of the novel susceptibility loci for atherosclerosis.
Collapse
Affiliation(s)
- Sara Mankoč Ramuš
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Daniel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
115
|
Chellan B, Sutton NR, Hofmann Bowman MA. S100/RAGE-Mediated Inflammation and Modified Cholesterol Lipoproteins as Mediators of Osteoblastic Differentiation of Vascular Smooth Muscle Cells. Front Cardiovasc Med 2018; 5:163. [PMID: 30467547 PMCID: PMC6235906 DOI: 10.3389/fcvm.2018.00163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/19/2018] [Indexed: 01/01/2023] Open
Abstract
Arterial calcification is a feature of atherosclerosis and shares many risk factors including diabetes, dyslipidemia, chronic kidney disease, hypertension, and age. Although there is overlap in risk factors, anti-atherosclerotic therapies, including statins, fail to reduce arterial, and aortic valve calcifications. This suggests that low density lipoprotein (LDL) may not be the main driver for aortic valve disease and arterial calcification. This review focuses on modified LDLs and their role in mediating foam cell formation in smooth muscle cells (SMCs), with special emphasis on enzyme modified non-oxidized LDL (ELDL). In vivo, ELDL represents one of the many forms of modified LDLs present in the atherosclerotic vessel. Phenotypic changes of macrophages and SMCs brought about by the uptake of modified LDLs overlap significantly in an atherosclerotic milieu, making it practically impossible to differentiate between the effects from oxidized LDL, ELDL, and other LDL modification. By studying in vitro-generated modifications of LDL, we were able to demonstrate marked differences in the transcriptome of human coronary artery SMCs (HCASMCs) upon uptake of ELDL, OxLDL, and native LDL, indicating that specific modifications of LDL in atherosclerotic plaques may determine the biology and functional consequences in vasculature. Enzyme-modified non-oxidized LDL (ELDL) induces calcification of SMCs and this is associated with reduced mRNA levels for genes protective for calcification (ENPP1, MGP) and upregulation of osteoblastic genes. A second focus of this review is on the synergy between hyperlipidemia and accelerated calcification In vivo in a mouse models with transgenic expression of human S100A12. We summarize mechanisms of S100A12/RAGE mediated vascular inflammation promoting vascular and valve calcification in vivo.
Collapse
Affiliation(s)
- Bijoy Chellan
- Department of Medicine, University of Illinois, Chicago, IL, United States
| | - Nadia R Sutton
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | | |
Collapse
|
116
|
Bajpai A, Tilley DG. The Role of Leukocytes in Diabetic Cardiomyopathy. Front Physiol 2018; 9:1547. [PMID: 30443223 PMCID: PMC6221939 DOI: 10.3389/fphys.2018.01547] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetes is predominant risk factor for cardiovascular diseases such as myocardial infarction and heart failure. Recently, leukocytes, particularly neutrophils, macrophages, and lymphocytes, have become targets of investigation for their potential role in a number of chronic inflammatory diseases such as diabetes and heart failure. While leukocytes contribute significantly to the progression of diabetes and heart failure individually, understanding their participation in the pathogenesis of diabetic heart failure is much less understood. The present review summarizes the role of leukocytes in the complex interplay between diabetes and heart failure, which is critical to the discovery of new targeted therapies for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Anamika Bajpai
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
117
|
Abstract
PURPOSE OF REVIEW Insulin resistance (IR) is recognized to play an important role in the pathogenesis of dyslipidemia. This review summarizes the complex interplay between IR and dyslipidemia in people with and without diabetes. RECENT FINDINGS IR impacts the metabolism of triglycerides, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and very low-density lipoprotein cholesterol (VLDL-C) by several mechanisms. Trials with insulin sensitizing therapies, including biguanides and thiazolidinediones, have provided inconsistent results on lipid lowering in people with and without diabetes. In this review, we focus on the pathophysiological interplay between IR and dyslipidemia and recapitulate lipid and lipoprotein data from insulin-sensitizing trials. Further research elucidating the reciprocal relationship between IR and dyslipidemia is needed to better target these important risk factors for cardiovascular disease.
Collapse
Affiliation(s)
- Petter Bjornstad
- Department of Pediatrics, Division of Endocrinology, University of Colorado School of Medicine, 13123 East 16th Ave, Box B26, Aurora, CO, 80045, USA.
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Robert H Eckel
- Department of Medicine, Division of Endocrinology and Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
118
|
Chang HY, Yazdani A, Li X, Douglas KAA, Mantzoros CS, Karniadakis GE. Quantifying Platelet Margination in Diabetic Blood Flow. Biophys J 2018; 115:1371-1382. [PMID: 30224049 PMCID: PMC6170725 DOI: 10.1016/j.bpj.2018.08.031] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/23/2018] [Accepted: 08/24/2018] [Indexed: 12/23/2022] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) develop thrombotic abnormalities strongly associated with cardiovascular diseases. In addition to the changes of numerous coagulation factors such as elevated levels of thrombin and fibrinogen, the abnormal rheological effects of red blood cells (RBCs) and platelets flowing in blood are crucial in platelet adhesion and thrombus formation in T2DM. An important process contributing to the latter is the platelet margination. We employ the dissipative particle dynamics method to seamlessly model cells, plasma, and vessel walls. We perform a systematic study on RBC and platelet transport in cylindrical vessels by considering different cell shapes, sizes, and RBC deformabilities in healthy and T2DM blood, as well as variable flowrates and hematocrit. In particular, we use cellular-level RBC and platelet models with parameters derived from patient-specific data and present a sensitivity study. We find T2DM RBCs, which are less deformable compared to normal RBCs, lower the transport of platelets toward the vessel walls, whereas platelets with higher mean volume (often observed in T2DM) lead to enhanced margination. Furthermore, increasing the flowrate or hematocrit enhances platelet margination. We also investigated the effect of platelet shape and observed a nonmonotonic variation with the highest near-wall concentration corresponding to platelets with a moderate aspect ratio of 0.38. We examine the role of white blood cells (WBCs), whose count is increased notably in T2DM patients. We find that WBC rolling or WBC adhesion tends to decrease platelet margination due to hydrodynamic effects. To the best of our knowledge, such simulations of blood including all blood cells have not been performed before, and our quantitative findings can help separate the effects of hydrodynamic interactions from adhesive interactions and potentially shed light on the associated pathological processes in T2DM such as increased inflammatory response, platelet activation and adhesion, and ultimately thrombus formation.
Collapse
Affiliation(s)
- Hung-Yu Chang
- Division of Applied Mathematics, Brown University, Providence, Rhode Island
| | - Alireza Yazdani
- Division of Applied Mathematics, Brown University, Providence, Rhode Island
| | - Xuejin Li
- Division of Applied Mathematics, Brown University, Providence, Rhode Island
| | - Konstantinos A A Douglas
- S. Lepida Biomedical Laboratory, Athens, Greece; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
119
|
Dattani N, Sayers RD, Bown MJ. Diabetes mellitus and abdominal aortic aneurysms: A review of the mechanisms underlying the negative relationship. Diab Vasc Dis Res 2018; 15:367-374. [PMID: 29874945 DOI: 10.1177/1479164118780799] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Diabetes mellitus appears to be negatively associated with abdominal aortic aneurysm; however, the mechanisms underlying this relationship remain poorly understood. The aim of this article is to provide a comprehensive review of the currently understood biological pathways underlying this relationship. METHODS A review of the literature ('diabetes' OR 'hyperglycaemia' AND 'aneurysm') was performed and relevant studies grouped into biological pathways. RESULTS This review identified a number of biological pathways through which diabetes mellitus may limit the presence, growth and rupture of abdominal aortic aneurysms. These include those influencing extracellular matrix volume, extracellular matrix glycation, the formation of advanced glycation end-products, inflammation, oxidative stress and intraluminal thrombus biology. In addition, there is an increasing evidence to suggest that the medications used to treat diabetes can also limit the development and progression of abdominal aortic aneurysms. CONCLUSION The negative association between diabetes and abdominal aortic aneurysm is robust. Future studies should attempt to target the pathways identified in this review to develop novel therapeutic agents aimed at slowing or even halting aneurysm progression.
Collapse
Affiliation(s)
- Nikesh Dattani
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Unit and British Heart Foundation Cardiovascular Research Centre, University of Leicester, Leicester, UK
| | - Robert D Sayers
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Unit and British Heart Foundation Cardiovascular Research Centre, University of Leicester, Leicester, UK
| | - Matthew J Bown
- Department of Cardiovascular Sciences, NIHR Leicester Cardiovascular Biomedical Research Unit and British Heart Foundation Cardiovascular Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
120
|
La réduction de l’hyperglycémie entraîne-t-elle un bénéfice cardiovasculaire ? Presse Med 2018; 47:764-768. [DOI: 10.1016/j.lpm.2018.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 11/21/2022] Open
|
121
|
RAGE and TLRs as Key Targets for Antiatherosclerotic Therapy. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7675286. [PMID: 30225265 PMCID: PMC6129363 DOI: 10.1155/2018/7675286] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/08/2018] [Indexed: 02/08/2023]
Abstract
Receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs) are the key factors indicating a danger to the organism. They recognize the microbial origin pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). The primary response induced by PAMPs or DAMPs is inflammation. Excessive stimulation of the innate immune system occurs in arterial wall with the participation of effector cells. Persistent adaptive responses can also cause tissue damage and disease. However, inflammation mediated by the molecules innate responses is an important way in which the adaptive immune system protects us from infection. The specific detection of PAMPs and DAMPs by host receptors drives a cascade of signaling that converges at nuclear factor-κB (NF-κB) and interferon regulatory factors (IRFs) and induces the secretion of proinflammatory cytokines, type I interferon (IFN), and chemokines, which promote direct killing of the pathogen. Therefore, signaling of these receptors' pathways also appear to present new avenue for the modulation of inflammatory responses and to serve as potential novel therapeutic targets for antiatherosclerotic therapy.
Collapse
|
122
|
Sugiyama T, Yamamoto E, Bryniarski K, Xing L, Fracassi F, Lee H, Jang IK. Coronary Plaque Characteristics in Patients With Diabetes Mellitus Who Presented With Acute Coronary Syndromes. J Am Heart Assoc 2018; 7:e009245. [PMID: 30006490 PMCID: PMC6064844 DOI: 10.1161/jaha.118.009245] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 06/12/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Diabetes mellitus (DM) is a major risk factor for cardiovascular events. We aimed to investigate the coronary plaque phenotype of diabetic patients who presented with acute coronary syndromes by optical coherence tomography. METHODS AND RESULTS A total of 322 patients with acute coronary syndromes who underwent preintervention optical coherence tomography imaging of the culprit lesion were included. Culprit plaque characteristics were compared between patients with DM (n=95) and those without DM (n=227). In the subgroup of 250 patients in whom sufficient length of nonculprit region in the culprit vessel was imaged by optical coherence tomography, the characteristics of nonculprit plaques were also evaluated. Patients with DM had a higher prevalence of lipid-rich plaque (58.9% versus 44.9%, P=0.030) and macrophage accumulation (60.0% versus 44.9%, P=0.019) in the culprit lesion compared with patients without DM. The prevalence of plaque rupture (33.7% versus 30.4%, P=0.896) and plaque erosion (21.1% versus 22.0%, P=0.458) was similar. In the nonculprit lesions, the DM group had greater maximal lipid arc (248.9°±83.9° versus 179.9°±58.3°, P=0.006), thinner fibrous cap thickness (103.3±56.2 μm versus 140.7±70.0 μm, P=0.013), and a higher prevalence of thin-cap fibroatheroma (17.2% versus 6.3%, P=0.031), compared with the non-DM group. CONCLUSIONS Compared with patients without DM, those with DM had more vulnerable features in both culprit and nonculprit lesions, thus indicating a higher level of panvascular instability. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01110538.
Collapse
Affiliation(s)
- Tomoyo Sugiyama
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Erika Yamamoto
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Krzysztof Bryniarski
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Lei Xing
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Francesco Fracassi
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital Harvard Medical School, Boston, MA
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital Harvard Medical School, Boston, MA
- Division of Cardiology, Kyung Hee University Hospital, Seoul, South Korea
| |
Collapse
|
123
|
Kang J, Park KW, Lee MS, Zheng C, Han JK, Yang HM, Kang HJ, Koo BK, Kim HS. The natural course of nonculprit coronary artery lesions; analysis by serial quantitative coronary angiography. BMC Cardiovasc Disord 2018; 18:130. [PMID: 29954346 PMCID: PMC6027760 DOI: 10.1186/s12872-018-0870-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/21/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Nonculprit lesions are the major cause of future cardiovascular events. However, the natural course of nonculprit lesions and angiographic predictors of plaque progression are not well-studied. The purpose of our study was to observe the natural course of nonculprit lesions, and to identify predictors of unanticipated future events and angiographic progression in nonculprit lesions. METHODS We analyzed 640 nonculprit lesions with a length of ≥2 mm and luminal narrowing ≥30% from 320 patients who had two serial angiographic follow-ups; 9 to 13 months post-PCI and 24 months post-PCI. The study endpoints were nonculprit-ischemia driven revascularization (IDR) and the rate of diameter stenosis (DS) progression. Those with progression of DS > 12%/year were defined as 'rapid progressors'. RESULTS During the median follow-up period of 737 days, 20 lesions in 20 patients (6.3%) required nonculprit-IDR. Independent predictors of nonculprit-IDR were diabetes (hazard ratio [HR] 2.93, 95% confidence interval [CI] 1.072-8.007, p = 0.036) and lesion type B2/C (HR 4.017, 95% CI 1.614-9.997, p = 0.003). The presence of one or both of the two major risk factors was associated with significant DS progression (3.0 ± 6.8% vs. 3.5 ± 6.1% vs. 6.8 ± 9.9% for lesions with 0, 1 and both risk factors, p < 0.001). Among the 640 lesions, 38 lesions (5.9%) in 33 patients were rapid progressors, while risk factors of rapid progressors included lesion type B2/C as a lesion-related risk factor (HR 1.998, 95% CI 1.006-3.791, p = 0.048) and diabetes mellitus as a patient-related risk factor (HR 3.725, 95% CI 1.937-7.538, p < 0.001). Lesions with both risk factors (type B2/C lesions in diabetic patients) were at the highest risk of rapid progression (odds ratio 3.250, 95% CI 1.451-7.282), compared to type A/B1 lesions in non-diabetic patients. CONCLUSION Nonculprit-IDR was not uncommon during the 2-year follow up period in our population. The major risk factors of nonculprit lesion progression were diabetes and lesion type B2/C. TRIAL REGISTRATION Retrospectively registered and approved by the institutional review board of Seoul National University Hospital (No.: 1801-138-918) on February 2nd, 2018.
Collapse
Affiliation(s)
- Jeehoon Kang
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Kyung Woo Park
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea.
| | - Michael S Lee
- Division of Cardiology, University of California Los Angeles Medical Center, Los Angeles, CA, USA
| | - Chengbin Zheng
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Jung-Kyu Han
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Han-Mo Yang
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Hyun-Jae Kang
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Bon-Kwon Koo
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine and Cardiovascular Center, Seoul National University Hospital, 101 Daehakro, Jongno Gu, Seoul, 110-744, South Korea
| |
Collapse
|
124
|
Zhang S, Dai J, Jia H, Hu S, Du H, Li N, Zou Y, Zou Y, Jing S, Wang Y, Sun R, Yu B. Non-culprit plaque characteristics in acute coronary syndrome patients with raised hemoglobinA1c: an intravascular optical coherence tomography study. Cardiovasc Diabetol 2018; 17:90. [PMID: 29907160 PMCID: PMC6002985 DOI: 10.1186/s12933-018-0729-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/06/2018] [Indexed: 02/03/2023] Open
Abstract
Background Raised hemoglobinA1c (HbA1c) is an indicator of pre-diabetes, which is associated with increased risk of coronary artery disease. However, the detailed morphological characteristics of non-culprit plaques in acute coronary syndrome (ACS) patients remain largely unknown. Methods A total of 305 non-culprit plaques from 216 ACS patients were analyzed by intravascular optical coherence tomography. These patients were divided into three groups according to the serum glycosylated hemoglobin level: normal HbA1c (< 5.7%), pre-diabetes with raised HbA1c (5.7–6.4%) and diabetes mellitus (DM). Results Plaques in patients with raised HbA1c had a longer lipid length (17.0 ± 8.3 mm vs. 13.9 ± 7.2 mm, P = 0.004) and greater lipid index (2775.0 ± 1694.0 mm° vs. 1592.1 ± 981.2 mm°, P = 0.001) than those with normal HbA1c but were similar to DM. The prevalence of calcification in patients with raised HbA1c was significantly higher (38.7% vs. 26.3%, P = 0.048) than normal HbA1c but was similar to DM. The percentage of macrophage infiltration in the DM group was higher than that in the normal HbA1c group (20.5% vs. 7.4%, P = 0.005). Conclusions Compared to patients with normal HbA1c, the non-culprit plaques in ACS patients with raised HbA1c had more typical vulnerable features but were similar to DM.
Collapse
Affiliation(s)
- Shaotao Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Jiannan Dai
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Haibo Jia
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Sining Hu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Hongwei Du
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Ning Li
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yongpeng Zou
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yanan Zou
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Shenhong Jing
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Rong Sun
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China.
| |
Collapse
|
125
|
Kim U, Leipsic JA, Sellers SL, Shao M, Blanke P, Hadamitzky M, Kim YJ, Conte E, Andreini D, Pontone G, Budoff MJ, Gottlieb I, Lee BK, Chun EJ, Cademartiri F, Maffei E, Marques H, Shin S, Choi JH, Virmani R, Samady H, Stone PH, Berman DS, Narula J, Shaw LJ, Bax JJ, Min JK, Chang HJ. Natural History of Diabetic Coronary Atherosclerosis by Quantitative Measurement of Serial Coronary Computed Tomographic Angiography: Results of the PARADIGM Study. JACC Cardiovasc Imaging 2018; 11:1461-1471. [PMID: 29778853 DOI: 10.1016/j.jcmg.2018.04.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES This study aimed to determine the rate and extent of plaque progression (PP), changes in plaque features, and clinical predictors of PP in patients with diabetes mellitus (DM). BACKGROUND The natural history of coronary PP in patients with DM is not well established. METHODS A total of 1,602 patients (age 61.3 ± 9.0 years; 60.3% men; median scan interval 3.8 years) who underwent serial coronary computed tomography angiography over a period of at least 24 months were enrolled and analyzed from the PARADIGM (Progression of Atherosclerotic Plaque Determined by Computed Tomographic Angiography Imaging) trial. Study endpoints were changes in plaque features in diabetics with PP and risk factors for PP by serial coronary computed tomography angiography between patients with and without DM. PP was defined if plaque volume at follow-up minus plaque volume at baseline was >0. RESULTS DM was an independent risk factor for PP (84.6%; 276 of 326 patients with PP) in multivariate analysis (odds ratio [OR]: 1.526; 95% confidence interval [CI]: 1.100 to 2.118; p = 0.011). Independent risk factors for PP in patients with DM were male sex (OR: 1.485; 95% CI: 1.003 to 2.199; p = 0.048) and mean plaque burden at baseline ≥75% (OR: 3.121; 95% CI: 1.701 to 5.725; p ≤0.001). After propensity matching, percent changes in overall plaque volume (30.3 ± 36.9% in patients without DM and 36.0 ± 29.7% in those with DM; p = 0.032) and necrotic core volume (-7.0 ± 35.8% in patients without DM and 21.5 ± 90.5% in those with DM; p = 0.007) were significantly greater in those with DM. The frequency of spotty calcification, positive remodeling, and burden of low-attenuation plaque were significantly greater in patients with DM. CONCLUSIONS People with DM experience greater PP, particularly significantly greater progression in adverse plaque, than those without DM. Male sex and mean plaque burden >75% at baseline were identified as independent risk factors for PP.
Collapse
Affiliation(s)
- Ung Kim
- Department of Radiology, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada; Yeungnam University Medical Center, Daegu, South Korea
| | - Jonathon A Leipsic
- Department of Radiology, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, British Columbia, Canada.
| | - Stephanie L Sellers
- Department of Radiology, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada; Centre for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Michael Shao
- Department of Radiology, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Philipp Blanke
- Department of Radiology, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Hadamitzky
- Department of Radiology and Nuclear Medicine, German Heart Center Munich, Munich, Germany
| | - Yong-Jin Kim
- Department of Radiology and Nuclear Medicine, German Heart Center Munich, Munich, Germany; Seoul National University Hospital, Seoul, South Korea
| | - Edoardo Conte
- Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Daniele Andreini
- Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Gianluca Pontone
- Centro Cardiologico Monzino, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Matthew J Budoff
- Department of Medicine, Harbor UCLA Medical Center, Los Angeles, California
| | - Ilan Gottlieb
- Department of Radiology, Casa de Saude São Jose, Rio de Janeiro, Brazil
| | - Byoung Kwon Lee
- Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Ju Chun
- Seoul National University Bundang Hospital, South Korea
| | | | - Erica Maffei
- Department of Radiology, Area Vasta 1/Azienda Sanitaria Unica Regionale (ASUR) Marche, Urbino, Italy
| | | | - Sanghoon Shin
- National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | | | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Habib Samady
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Peter H Stone
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel S Berman
- Department of Imaging, Cedars-Sinai Medical Center, Cedars-Sinai Heart Institute, Los Angeles, California
| | - Jagat Narula
- Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, Zena and Michael A. Wiener Cardiovascular Institute, and Marie-Josee and Henry R. Kravis Center for Cardiovascular Health, New York, New York
| | - Leslee J Shaw
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Jeroen J Bax
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands
| | - James K Min
- Dalio Institute of Cardiovascular Imaging, New York-Presbyterian Hospital and Weill Cornell Medical College, New York, New York
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Integrative Cardiovascular Imaging Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
126
|
Guo M, Xiao J, Sheng X, Zhang X, Tie Y, Wang L, Zhao L, Ji X. Ginsenoside Rg3 Mitigates Atherosclerosis Progression in Diabetic apoE-/- Mice by Skewing Macrophages to the M2 Phenotype. Front Pharmacol 2018; 9:464. [PMID: 29867472 PMCID: PMC5954105 DOI: 10.3389/fphar.2018.00464] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/20/2018] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis (AS) in diabetic patients is often associated with low stability, which might be largely attributed to unfavorable macrophage polarization and increased inflammatory response induced by hyperglycaemia. Ginsenoside Rg3 is one of the main active principles of Panax Ginseng, which has been reported to be a natural ligand of peroxisome proliferator-activated receptor-gamma (PPARγ), a key nuclear transcriptional factor involved in inflammation and macrophage differentiation. However, it remains unclear if Rg3 could exert protective effects on plaque stability in diabetes. In this study, we investigated the role of ginsenoside 20(S)-Rg3 in macrophage polarization and AS plaque stability using advanced glycation end products-treated macrophages and diabetic AS mice models. In vitro, advanced glycation end products (AGEs) treatment promoted the expression of proinflammatory molecules and M1 surface markers, whereas 20(S)-Rg3 could reverse the M1 polarization to the M2 phenotype. In vivo, the administration of 20(S)-Rg3 promoted AS lesion stability and reduced the plaque burden, accompanied by increased M2 macrophages and reduced M1 macrophages. In addition, PPARγ antagonist GW9662 co-administration mostly blocked these effects, suggesting the important role of PPARγ pathways in mediating 20(S)-Rg3 effects in macrophage polarization and atherosclerosis progression. Together, these results demonstrated an immunomodulatory role of ginsenoside 20(S)-Rg3 in promoting macrophages to a profile of the M2 type through PPARγ-dependent mechanisms, and indicated a potential role of 20(S)-Rg3 in the prevention and treatment of diabetic atherosclerosis.
Collapse
Affiliation(s)
- Mengqi Guo
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Xiao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xi Sheng
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyu Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Tie
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lang Zhao
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoping Ji
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
127
|
Ofstad AP, Atar D, Gullestad L, Langslet G, Johansen OE. The heart failure burden of type 2 diabetes mellitus-a review of pathophysiology and interventions. Heart Fail Rev 2018; 23:303-323. [PMID: 29516230 PMCID: PMC5937871 DOI: 10.1007/s10741-018-9685-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diabetes and heart failure (HF) are both global epidemics with tremendous costs on society with increased rates of HF hospitalizations and worsened prognosis when co-existing, making it a significant "deadly duo." The evidence for pharmacological treatment of HF in patients with type 2 diabetes mellitus (T2DM) stems typically from either subgroup analyses of patients that were recruited to randomized controlled trials of HF interventions, usually in patients with reduced ejection fraction (EF), or from subgroup analyses of HF patients recruited to cardiovascular (CV) outcome trials (CVOT) of glucose lowering agents involving patients with T2DM. Studies in patients with HF with preserved EF are sparse. This review summarizes the literature on pathophysiology and interventions aiming to reduce the HF burden in T2DM and includes HF trials of ACEi, digoxin, β-blocker, ARB, If-blocker, MRA, and ARNI involving 38,600 patients, with or without prevalent diabetes, and CV outcome trials in T2DM involving 74,351 patients, with or without prevalent HF. In all HF trials, HF outcomes by prevalent diabetes were reported with an incremental risk of HF and death confessed by prevalent diabetes and a treatment effect similar to those without diabetes. All T2DM CVOTs reported on HF outcomes with heterogeneity between trials with two reporting benefits (empagliflozin and canagliflozin) and two reporting increased risk (saxagliptin, pioglitazone). In vulnerable T2DM patients with concomitant HF, guideline-recommended HF drugs are effective. When choosing glucose-lowering therapy, outcomes from available CVOTs should be considered.
Collapse
Affiliation(s)
- Anne Pernille Ofstad
- Bærum Hospital, Vestre Viken HF, Rud, Norway.
- Medical Department, Boehringer Ingelheim, Asker, Norway.
| | - Dan Atar
- Department of Cardiology B, Oslo University Hospital, Ullevål, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Gisle Langslet
- Rikshospitalet, Lipid Clinic, Oslo University Hospital, Oslo, Norway
| | - Odd Erik Johansen
- Bærum Hospital, Vestre Viken HF, Rud, Norway
- Medical Department, Boehringer Ingelheim, Asker, Norway
| |
Collapse
|
128
|
Nilsson J. Atherosclerotic plaque vulnerability in the statin era. Eur Heart J 2018; 38:1638-1644. [PMID: 28387815 DOI: 10.1093/eurheartj/ehx143] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 03/07/2017] [Indexed: 12/24/2022] Open
Abstract
Life style changes and improved medical therapy have decreased cardiovascular mortality in many countries over the last decades. This has been accompanied by changes in disease characteristics including more non-ST segment elevation myocardial infraction and less vulnerable plaques as assessed by histological analysis of surgical specimens. However, many patients with established disease still suffer from recurrent cardiovascular events in spite of treatment with state-of-the-art-therapy including statins. It is likely that this reflects a state of the disease in which statins control the pro-inflammatory effects of lipids allowing other statin-unresponsive disease mechanisms to become increasingly important. If this assumption is correct it means that patients with established disease with time will get insuffient protection by current therapies alone. Against this background it is critical to reach a better understanding of alternative mechanisms for plaque vulnerability. Examples of such mechanisms include altered patterns of blood flow caused by plaque stenosis resulting in down-regulation of the anti-inflammatory and anti-thrombotic signals in the endothelium, impaired vascular repair associated with diabetes and plaque inflammation driven by cholesterol crystals, infectious pathogens as well as autoimmune responses against modified plaque components. Novel biomarkers and other diagnostics are needed to establish the clinical importance of these mechanisms as well as to determine how they are affected by current treatments. Consequently, there will also be a need for development of new treatments targeting these mechanisms and that can act in concert with current therapies.
Collapse
Affiliation(s)
- Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, 20502 Malmö, Sweden
| |
Collapse
|
129
|
Benenati S, De Maria GL, Scarsini R, Porto I, Banning AP. Invasive “in the cath-lab” assessment of myocardial ischemia in patients with coronary artery disease: When does the gold standard not apply? CARDIOVASCULAR REVASCULARIZATION MEDICINE 2018; 19:362-372. [DOI: 10.1016/j.carrev.2018.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 01/16/2018] [Indexed: 02/08/2023]
|
130
|
Severino P, D'Amato A, Netti L, Pucci M, De Marchis M, Palmirotta R, Volterrani M, Mancone M, Fedele F. Diabetes Mellitus and Ischemic Heart Disease: The Role of Ion Channels. Int J Mol Sci 2018; 19:802. [PMID: 29534462 PMCID: PMC5877663 DOI: 10.3390/ijms19030802] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus is one the strongest risk factors for cardiovascular disease and, in particular, for ischemic heart disease (IHD). The pathophysiology of myocardial ischemia in diabetic patients is complex and not fully understood: some diabetic patients have mainly coronary stenosis obstructing blood flow to the myocardium; others present with coronary microvascular disease with an absence of plaques in the epicardial vessels. Ion channels acting in the cross-talk between the myocardial energy state and coronary blood flow may play a role in the pathophysiology of IHD in diabetic patients. In particular, some genetic variants for ATP-dependent potassium channels seem to be involved in the determinism of IHD.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Andrea D'Amato
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Lucrezia Netti
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Mariateresa Pucci
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marialaura De Marchis
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Clinical Oncology Oncogenomic Research Center, 'Aldo Moro' University of Bari, 70124 Bari, Italy.
| | - Maurizio Volterrani
- Department of Cardiac Rehabilitation, IRCCS San Raffaele, 00163 Rome, Italy.
| | - Massimo Mancone
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| | - Francesco Fedele
- Department of Cardiovascular, Respiratory, Nephrology, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
131
|
Subramanian S, Hirsch IB. Intensive Diabetes Treatment and Cardiovascular Outcomes in Type 1 Diabetes Mellitus: Implications of the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Study 30-Year Follow-up. Endocrinol Metab Clin North Am 2018; 47:65-79. [PMID: 29407057 DOI: 10.1016/j.ecl.2017.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Type 1 diabetes mellitus, an autoimmune disorder characterized by beta-cell destruction and absolute insulin deficiency, is associated with significantly increased cardiovascular disease risk but the mechanisms underlying this enhanced risk are unclear. Results of the pivotal Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications study have shown that compared to conventional therapy, intensive glycemic control results in decreased cardiovascular morbidity and mortality. Evidence from this study also revealed contributions of blood pressure, renal disease, body weight, and lipids to cardiovascular disease in type 1 diabetes mellitus. Extrapolating from existing evidence, this article addresses clinical strategies to mitigate cardiovascular risks.
Collapse
Affiliation(s)
- Savitha Subramanian
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, 4245 Roosevelt Way Northeast, Box 354691, Seattle, WA 98105, USA.
| | - Irl B Hirsch
- Division of Metabolism, Endocrinology and Nutrition, University of Washington, 4245 Roosevelt Way Northeast, Box 354691, Seattle, WA 98105, USA
| |
Collapse
|
132
|
Beneit N, Martín-Ventura JL, Rubio-Longás C, Escribano Ó, García-Gómez G, Fernández S, Sesti G, Hribal ML, Egido J, Gómez-Hernández A, Benito M. Potential role of insulin receptor isoforms and IGF receptors in plaque instability of human and experimental atherosclerosis. Cardiovasc Diabetol 2018; 17:31. [PMID: 29463262 PMCID: PMC5819698 DOI: 10.1186/s12933-018-0675-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Clinical complications associated with atherosclerotic plaques arise from luminal obstruction due to plaque growth or destabilization leading to rupture. We previously demonstrated that overexpression of insulin receptor isoform A (IRA) and insulin-like growth factor-I receptor (IGF-IR) confers a proliferative and migratory advantage to vascular smooth muscle cells (VSMCs) promoting plaque growth in early stages of atherosclerosis. However, the role of insulin receptor (IR) isoforms, IGF-IR or insulin-like growth factor-II receptor (IGF-IIR) in VSMCs apoptosis during advanced atherosclerosis remains unclear. METHODS We evaluated IR isoforms expression in human carotid atherosclerotic plaques by consecutive immunoprecipitations of insulin receptor isoform B (IRB) and IRA. Western blot analysis was performed to measure IGF-IR, IGF-IIR, and α-smooth muscle actin (α-SMA) expression in human plaques. The expression of those proteins, as well as the presence of apoptotic cells, was analyzed by immunohistochemistry in experimental atherosclerosis using BATIRKO; ApoE-/- mice, a model showing more aggravated vascular damage than ApoE-/- mice. Finally, apoptosis of VSMCs bearing IR (IRLoxP+/+ VSMCs), or not (IR-/- VSMCs), expressing IRA (IRA VSMCs) or expressing IRB (IRB VSMCs), was assessed by Western blot against cleaved caspase 3. RESULTS We observed a significant decrease of IRA/IRB ratio in human complicated plaques as compared to non-complicated regions. Moreover, complicated plaques showed a reduced IGF-IR expression, an increased IGF-IIR expression, and lower levels of α-SMA indicating a loss of VSMCs. In experimental atherosclerosis, we found a significant decrease of IRA with an increased IRB expression in aorta from 24-week-old BATIRKO; ApoE-/- mice. Furthermore, atherosclerotic plaques from BATIRKO; ApoE-/- mice had less VSMCs content and higher number of apoptotic cells. In vitro experiments showed that IGF-IR inhibition by picropodophyllin induced apoptosis in VSMCs. Apoptosis induced by thapsigargin was lower in IR-/- VSMCs expressing higher IGF-IR levels as compared to IRLoxP+/+ VSMCs. Finally, IRB VSMCs are more prone to thapsigargin-induced apoptosis than IRA or IRLoxP+/+ VSMCs. CONCLUSIONS In advanced human atherosclerosis, a reduction of IRA/IRB ratio, decreased IGF-IR expression, or increased IGF-IIR may contribute to VSMCs apoptosis, promoting plaque instability and increasing the risk of plaque rupture and its clinical consequences.
Collapse
Affiliation(s)
- Nuria Beneit
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - José Luis Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Díaz-Autonoma University, Madrid, Spain.,CIBER of Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Carlota Rubio-Longás
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Óscar Escribano
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Gema García-Gómez
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Silvia Fernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Marta Letizia Hribal
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Jesús Egido
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain.,Vascular Research Lab, IIS-Fundación Jiménez Díaz-Autonoma University, Madrid, Spain.,CIBER of Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - Almudena Gómez-Hernández
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain. .,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain. .,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain.
| | - Manuel Benito
- Biochemistry and Molecular Biology II Department, School of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.,Health Research Institute of San Carlos Clinic Hospital (IdISSC), Madrid, Spain.,CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| |
Collapse
|
133
|
Mori H, Torii S, Kutyna M, Sakamoto A, Finn AV, Virmani R. Coronary Artery Calcification and its Progression. JACC Cardiovasc Imaging 2018; 11:127-142. [DOI: 10.1016/j.jcmg.2017.10.012] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/29/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022]
|
134
|
Kovarnik T, Chen Z, Mintz GS, Wahle A, Bayerova K, Kral A, Chval M, Kopriva K, Lopez J, Sonka M, Linhart A. Plaque volume and plaque risk profile in diabetic vs. non-diabetic patients undergoing lipid-lowering therapy: a study based on 3D intravascular ultrasound and virtual histology. Cardiovasc Diabetol 2017; 16:156. [PMID: 29212544 PMCID: PMC5719721 DOI: 10.1186/s12933-017-0637-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022] Open
Abstract
Background Coronary atherosclerosis progresses faster in patients with diabetes mellitus (DM) and causes higher morbidity and mortality in such patients compared to non-diabetics ones (non-DM). We quantify changes in plaque volume and plaque phenotype during lipid-lowering therapy in DM versus non-DM patients using advanced intracoronary imaging. Methods We analyzed data from 61 patients with stable angina pectoris included to the PREDICT trial searching for prediction of plaque changes during intensive lipid-lowering therapy (40 mg rosuvastatin daily). Geometrically correct, fully 3-D representation of the vascular wall surfaces and intravascular ultrasound virtual histology (IVUS-VH) defined tissue characterization was obtained via fusion of two-plane angiography and IVUS-VH. Frame-based indices of plaque morphology and virtual histology analyses were computed and averaged in 5 mm long baseline/follow-up registered vessel segments covering the entire length of the two sequential pullbacks (baseline, 1-year). We analyzed 698 5-mm-long segments and calculated the Liverpool active plaque score (LAPS). Results Despite reaching similar levels of LDL cholesterol (DM 2.12 ± 0.91 mmol/l, non-DM 1.8 ± 0.66 mmol/l, p = 0.21), DM patients experienced, compared to non-DM ones, higher progression of mean plaque area (0.47 ± 1.15 mm2 vs. 0.21 ± 0.97, p = 0.001), percent atheroma volume (0.7 ± 2.8% vs. − 1.4 ± 2.5%, p = 0.007), increase of LAPS (0.23 ± 1.66 vs. 0.13 ± 1.79, p = 0.018), and exhibited more locations with TCFA (Thin-Cap Fibro-Atheroma) plaque phenotype in 5 mm vessel segments (20.3% vs. 12.5%, p = 0.01). However, only non-DM patients reached significant decrease of LDL cholesterol. Plaque changes were more pronounced in PIT (pathologic intimal thickening) compared to TCFA with increased plaque area in both phenotypes in DM patients. Conclusion Based on detailed 3D analysis, we found advanced plaque phenotype and further atherosclerosis progression in DM patients despite the same reached levels of LDLc as in non-DM patients. Trial registration ClinicalTrials.gov identifier: NCT01773512
Collapse
Affiliation(s)
- Tomas Kovarnik
- 2nd Department of Medicine-Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, II. interni klinika VFN a 1. LF UK, U nemocnice 2, 128 08, Praha 2, Czech Republic.
| | - Zhi Chen
- Department of Electrical & Computer Engineering and Iowa Institute for Biomedical Imaging, The University of Iowa, Iowa City, IA, USA
| | - Gary S Mintz
- Cardiovascular Research Foundation, New York, USA
| | - Andreas Wahle
- Department of Electrical & Computer Engineering and Iowa Institute for Biomedical Imaging, The University of Iowa, Iowa City, IA, USA
| | - Kristyna Bayerova
- 2nd Department of Medicine-Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, II. interni klinika VFN a 1. LF UK, U nemocnice 2, 128 08, Praha 2, Czech Republic
| | - Ales Kral
- 2nd Department of Medicine-Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, II. interni klinika VFN a 1. LF UK, U nemocnice 2, 128 08, Praha 2, Czech Republic
| | - Martin Chval
- Institute for Research and Development of Education, Faculty of Education, Charles University in Prague, Prague, Czech Republic
| | - Karel Kopriva
- Cardiology Department, Na Homolce Hospital, Prague, Czech Republic
| | - John Lopez
- Loyola University Stritch School of Medicine, Maywood, IL, USA
| | - Milan Sonka
- Department of Electrical & Computer Engineering and Iowa Institute for Biomedical Imaging, The University of Iowa, Iowa City, IA, USA
| | - Ales Linhart
- 2nd Department of Medicine-Department of Cardiovascular Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, II. interni klinika VFN a 1. LF UK, U nemocnice 2, 128 08, Praha 2, Czech Republic
| |
Collapse
|
135
|
Mrgan M, Funck KL, Gaur S, Øvrehus KA, Dey D, Kusk MW, Nørgaard BL, Gram JB, Olsen MH, Gram J, Sand NPR. High burden of coronary atherosclerosis in patients with a new diagnosis of type 2 diabetes. Diab Vasc Dis Res 2017; 14:468-476. [PMID: 28866908 DOI: 10.1177/1479164117728014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The purposes of this study were to compare the presence, extent and composition of coronary plaques in asymptomatic patients with newly diagnosed type 2 diabetes to age- and sex-matched controls. METHODS Patients with newly diagnosed (<1 year) type 2 diabetes ( n = 44) and controls ( n = 44) underwent contrast-enhanced coronary computed tomography angiography. Advanced plaque analysis including total plaque volume and volumes of plaque components (calcified plaque and non-calcified plaque, including low-attenuation [low-density non-calcified plaque]) was performed using validated semi-automated software. RESULTS Coronary artery calcification was more often seen in patients with type 2 diabetes (66%) versus controls (48%), p < 0.05. Both the absolute volume (median; interquartile range) of low-density non-calcified plaque (7.9 mm3; 0-50.5 mm3 vs 0; 0-34.3 mm3, p < 0.05) and the increase in low-density non-calcified plaque ratio in relation to total plaque volume ( τ = 0.5, p < 0.001) were significantly higher in patients with type 2 diabetes. More patients with type 2 diabetes had spotty calcification (31% vs 0%, p < 0.05). By multivariate analysis, the presence of any low-density non-calcified plaque was higher in males (odds ratio: 4.06, p < 0.05), who also demonstrated a larger low-density non-calcified plaque volume ( p < 0.001). The presence and extent of low-density non-calcified plaque increased with age, smoking, hypertension and hyperglycaemia, all p < 0.05. CONCLUSION Asymptomatic patients with newly diagnosed type 2 diabetes had plaque features associated with increased vulnerability as compared with age- and sex-matched controls.
Collapse
Affiliation(s)
- Monija Mrgan
- 1 Department of Cardiology, Hospital of Southwest Denmark, Esbjerg, Denmark
| | - Kristian L Funck
- 2 Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Sara Gaur
- 3 Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Damini Dey
- 5 Biomedical Imaging Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Martin W Kusk
- 6 Department of Radiology, Hospital of Southwest Denmark, Esbjerg, Denmark
| | - Bjarne L Nørgaard
- 3 Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen B Gram
- 7 Unit for Thrombosis Research, University of Southern Denmark, Esbjerg, Denmark
- 8 Department of Clinical Biochemistry, Hospital of Southwest Denmark, Esbjerg, Denmark
| | - Michael H Olsen
- 9 Cardiology Section, Department of Internal Medicine, Holbæk Hospital, Holbæk, Denmark
- 10 Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Jeppe Gram
- 11 Department of Endocrinology, Hospital of Southwest Denmark, Esbjerg, Denmark
| | - Niels Peter R Sand
- 1 Department of Cardiology, Hospital of Southwest Denmark, Esbjerg, Denmark
- 12 Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
136
|
Taguchi Y, Itoh T, Oda H, Uchimura Y, Kaneko K, Sakamoto T, Goto I, Sakuma M, Ishida M, Terashita D, Otake H, Morino Y, Shinke T. Coronary risk factors associated with OCT macrophage images and their response after CoCr everolimus-eluting stent implantation in patients with stable coronary artery disease. Atherosclerosis 2017; 265:117-123. [DOI: 10.1016/j.atherosclerosis.2017.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/27/2017] [Accepted: 08/16/2017] [Indexed: 02/02/2023]
|
137
|
Sun B, Li X, Liu X, Ge X, Lu Q, Zhao X, Pu J, Xu J, Zhao H. Association between carotid plaque characteristics and acute cerebral infarction determined by MRI in patients with type 2 diabetes mellitus. Cardiovasc Diabetol 2017; 16:111. [PMID: 28893252 PMCID: PMC5594451 DOI: 10.1186/s12933-017-0592-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/28/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) might aggravate the carotid plaque vulnerability, and increase the risk for ischemic stroke. Few studies reported the acute stroke subtype with carotid plaque characteristics in T2DM patients. This study aimed to investigate the association between carotid plaque characteristics and acute cerebral infarct (ACI) lesion features determined by MRI in T2DM patients. METHODS Patients with acute cerebrovascular syndrome in internal carotid artery territory were recruited. All patients were stratified into T2DM and non-T2DM groups and underwent both carotid and brain MRI scans. Ipsilateral carotid plaque morphological and compositional characteristics, intracranial and extracranial carotid artery stenosis were also determined. Stroke subtype based on the Trial of ORG 10172 in Acute Stroke Treatment classification and ACI lesion patterns were evaluated. RESULTS Of the recruited 140 patients, 68 (48.6%) patients had T2DM (mean age 64.16 ± 11.38 years, 40 males). T2DM patients exhibited higher prevalence of carotid type IV-VI lesions, larger plaque burden as well as larger lipid-rich necrotic core (LRNC) compared with non-T2DM patients. Among the patients with carotid LRNC on symptomatic side, more concomitant large perforating artery infarct patterns and larger ACI size in the internal carotid artery territory were found in T2DM group than those in non-T2DM group. Carotid plaque with LRNC% > 22.0% was identified as an independent risk factor for the presence of ACI lesions confined to the carotid territory in T2DM patients, regardless of other risk factors. CONCLUSIONS This study shows that more concomitant large perforating artery infarct patterns and larger ACI size in the internal carotid artery territory were found in the T2DM patients with ipsilateral carotid LRNC plaque than those in non-T2DM patients. Quantification of the carotid plaque characteristics, particularly the LRNC% by MRI has the potential usefulness for stroke risk stratification.
Collapse
Affiliation(s)
- Beibei Sun
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Xiao Li
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Xiaosheng Liu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| | - Xiaoqian Ge
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Qing Lu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Xihai Zhao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| | - Jianrong Xu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China
| | - Huilin Zhao
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, China.
| |
Collapse
|
138
|
Jiang W, Wang Z, Hu Z, Wu H, Hu R, Hu X, Ren Z, Huang J. Blocking the ERK1/2 signal pathway can inhibit S100A12 induced human aortic smooth muscle cells damage. Cell Biol Int 2017; 41:1307-1315. [PMID: 28816402 DOI: 10.1002/cbin.10840] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/12/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Wanli Jiang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Hongbing Wu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Rui Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Jizhen Huang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| |
Collapse
|
139
|
Chamaria S, Johnson KW, Vengrenyuk Y, Baber U, Shameer K, Divaraniya AA, Glicksberg BS, Li L, Bhatheja S, Moreno P, Maehara A, Mehran R, Dudley JT, Narula J, Sharma SK, Kini AS. Intracoronary Imaging, Cholesterol Efflux, and Transcriptomics after Intensive Statin Treatment in Diabetes. Sci Rep 2017; 7:7001. [PMID: 28765529 PMCID: PMC5539108 DOI: 10.1038/s41598-017-07029-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
Residual atherothrombotic risk remains higher in patients with versus without diabetes mellitus (DM) despite statin therapy. The underlying mechanisms are unclear. This is a retrospective post-hoc analysis of the YELLOW II trial, comparing patients with and without DM (non-DM) who received rosuvastatin 40 mg for 8–12 weeks and underwent intracoronary multimodality imaging of an obstructive nonculprit lesion, before and after therapy. In addition, blood samples were drawn to assess cholesterol efflux capacity (CEC) and changes in gene expression in peripheral blood mononuclear cells (PBMC). There was a significant reduction in low density lipoprotein-cholesterol (LDL-C), an increase in CEC and beneficial changes in plaque morphology including increase in fibrous cap thickness and decrease in the prevalence of thin cap fibro-atheroma by optical coherence tomography in DM and non-DM patients. While differential gene expression analysis did not demonstrate differences in PBMC transcriptome between the two groups on the single-gene level, weighted gene coexpression network analysis revealed two modules of coexpressed genes associated with DM, Collagen Module and Platelet Module, related to collagen catabolism and platelet function respectively. Bayesian network analysis revealed key driver genes within these modules. These transcriptomic findings might provide potential mechanisms responsible for the higher cardiovascular risk in DM patients.
Collapse
Affiliation(s)
| | - Kipp W Johnson
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA
| | | | - Usman Baber
- Mount Sinai Heart, Mount Sinai Hospital, New York, USA
| | - Khader Shameer
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA
| | - Aparna A Divaraniya
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA
| | - Benjamin S Glicksberg
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA
| | - Li Li
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA
| | | | - Pedro Moreno
- Mount Sinai Heart, Mount Sinai Hospital, New York, USA
| | | | - Roxana Mehran
- Mount Sinai Heart, Mount Sinai Hospital, New York, USA
| | - Joel T Dudley
- Institute for Next Generation Healthcare, Icahn School of Medicine at Mount Sinai, New York, USA.,Department of Genetics and Genomic Sciences, Icahn Institute for Genetics and Genomic Sciences, Icahn School of Medicine, New York, USA.,Department of Population Health and Health Policy, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jagat Narula
- Mount Sinai Heart, Mount Sinai Hospital, New York, USA
| | | | | |
Collapse
|
140
|
Vascular Damage and Kidney Transplant Outcomes: An Unfriendly and Harmful Link. Am J Med Sci 2017; 354:7-16. [DOI: 10.1016/j.amjms.2017.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
|
141
|
Matsuura Y, Yamashita A, Zhao Y, Iwakiri T, Yamasaki K, Sugita C, Koshimoto C, Kitamura K, Kawai K, Tamaki N, Zhao S, Kuge Y, Asada Y. Altered glucose metabolism and hypoxic response in alloxan-induced diabetic atherosclerosis in rabbits. PLoS One 2017; 12:e0175976. [PMID: 28410399 PMCID: PMC5391952 DOI: 10.1371/journal.pone.0175976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/03/2017] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus accelerates atherosclerosis that causes most cardiovascular events. Several metabolic pathways are considered to contribute to the development of atherosclerosis, but comprehensive metabolic alterations to atherosclerotic arterial cells remain unknown. The present study investigated metabolic changes and their relationship to vascular histopathological changes in the atherosclerotic arteries of rabbits with alloxan-induced diabetes. Diabetic atherosclerosis was induced in rabbit ilio-femoral arteries by injecting alloxan (100 mg/kg), injuring the arteries using a balloon, and feeding with a 0.5% cholesterol diet. We histologically assessed the atherosclerotic lesion development, cellular content, pimonidazole positive-hypoxic area, the nuclear localization of hypoxia-inducible factor-1α, and apoptosis. We evaluated comprehensive arterial metabolism by performing metabolomic analyses using capillary electrophoresis-time of flight mass spectrometry. We evaluated glucose uptake and its relationship to vascular hypoxia using 18F-fluorodeoxyglucose and pimonidazole. Plaque burden, macrophage content, and hypoxic areas were more prevalent in arteries with diabetic, than non-diabetic atherosclerosis. Metabolomic analyses highlighted 12 metabolites that were significantly altered between diabetic and non-diabetic atherosclerosis. A half of them were associated with glycolysis metabolites, and their levels were decreased in diabetic atherosclerosis. The uptake of glucose evaluated as 18F-fluorodeoxyglucose in atherosclerotic lesions increased according to increased macrophage content or hypoxic areas in non-diabetic, but not diabetic rabbits. Despite profound hypoxic areas, the nuclear localization of hypoxia-inducible factor-1α decreased and the number of apoptotic cells increased in diabetic atherosclerotic lesions. Altered glycolysis metabolism and an impaired response to hypoxia in atherosclerotic lesions under conditions of insulin-dependent diabetes might be involved in the development of diabetic atherosclerosis.
Collapse
Affiliation(s)
- Yunosuke Matsuura
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Atsushi Yamashita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- * E-mail:
| | - Yan Zhao
- Department of Tracer Kinetics and Bioanalysis, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Iwakiri
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuaki Yamasaki
- Department of Tracer Kinetics and Bioanalysis, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Chihiro Sugita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Kyusyu University of Health and Welfare, Nobeoka, Japan
| | - Chihiro Koshimoto
- Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| | - Kazuo Kitamura
- Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Keiichi Kawai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Songji Zhao
- Department of Tracer Kinetics and Bioanalysis, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Kuge
- Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
142
|
Schmidt AM. 22016 ATVB Plenary Lecture: Receptor for Advanced Glycation Endproducts and Implications for the Pathogenesis and Treatment of Cardiometabolic Disorders: Spotlight on the Macrophage. Arterioscler Thromb Vasc Biol 2017; 37:613-621. [PMID: 28183700 PMCID: PMC5364055 DOI: 10.1161/atvbaha.117.307263] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/30/2017] [Indexed: 12/23/2022]
Abstract
The receptor for advanced glycation endproducts (RAGE) interacts with a unique repertoire of ligands that form and collect in the tissues and circulation in diabetes mellitus, aging, inflammation, renal failure, and obesity. RAGE is expressed on multiple cell types linked to tissue perturbation in these settings. This brief review focuses on the role of RAGE in monocytes/macrophages and how RAGE ligand engagement on these cells mediates seminal changes in monocyte/macrophage migration, oxidative stress, cholesterol efflux, and pro- versus anti-inflammatory cues that signal to tissue damage. Studies using mice devoid of Ager (gene encoding RAGE) or pharmacological antagonists of RAGE are protective in animal models of diabetes mellitus, atherosclerosis, and high-fat diet-induced obesity, in least in part through key roles in monocytes/macrophages. RAGE signal transduction requires the interaction of RAGE cytoplasmic domain with the formin, DIAPH1 (diaphanous 1) and novel antagonists of this interaction show significant promise in attenuation of the maladaptive effects of RAGE ligands in cellular and in vivo models. Finally, this brief review discusses evidence for RAGE axis perturbation in human monocytes/macrophages and how tracing RAGE activity in these cells may identify target engagement biomarkers of RAGE antagonism for future clinical trials.
Collapse
Affiliation(s)
- Ann Marie Schmidt
- From the Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York.
| |
Collapse
|
143
|
Buyukterzi Z, Can U, Alpaydin S, Guzelant A, Karaarslan S, Kocyigit D, Gurses KM. Enhanced S100A9 and S100A12 expression in acute coronary syndrome. Biomark Med 2017; 11:229-237. [PMID: 28157385 DOI: 10.2217/bmm-2016-0253] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AIMS In this study, we aimed to investigate whether serum S100A8, S100A9 and S100A12 levels were markers of acute coronary syndrome (ACS). MATERIALS & METHODS Patients who underwent coronary angiography and/or percutaneous coronary interventions between June 2015-October 2015 were consecutively recruited in this study and categorized three groups each containing 30 patients (normal coronary arteries, stable coronary artery disease, and acute coronary syndrome). Baseline characteristics, including co- morbidities and medications, were recorded and serum S100A8, S100A9, S100A12, and C- reactive protein levels were measured besides routine laboratory tests. RESULTS A total of 90 patients (63.00 [56.00-73.00] years, 62.89% male) have been included. None of the groups differed from each other regarding baseline characteristics (p > 0.05). S100A9 levels were elevated in ACS when compared with the normal coronary arteries (p = 0.033) and S100A12 levels were found to be elevated in ACS when compared with both patients with normal coronary arteries and stable coronary artery disease (p = 0.001). S100A12 was identified as an independent associate of ACS (p = 0.002). CONCLUSION These results suggest that S100A12 may serve as a marker of coronary plaque instability, and may have a therapeutic implication in ACS treatment.
Collapse
Affiliation(s)
- Zafer Buyukterzi
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Ummugulsum Can
- Department of Biochemistry, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sertac Alpaydin
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Asuman Guzelant
- Department of Microbiology & Infectious Diseases, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| | - Sukru Karaarslan
- Department of Cardiology, Ufuk University Faculty of Medicine, 06520 Balgat Ankara, Turkey
| | - Duygu Kocyigit
- Department of Cardiology, Hacettepe University Faculty of Medicine, 06100 Sihhiye Ankara, Turkey
| | - Kadri Murat Gurses
- Department of Cardiology, Konya Training & Research Hospital, University of Health Sciences, 42090 Meram Konya, Turkey
| |
Collapse
|
144
|
van Haelst ST, Haitjema S, de Vries JPP, Moll FL, Pasterkamp G, den Ruijter HM, de Borst GJ. Patients with diabetes differ in atherosclerotic plaque characteristics and have worse clinical outcome after iliofemoral endarterectomy compared with patients without diabetes. J Vasc Surg 2017; 65:414-421.e5. [DOI: 10.1016/j.jvs.2016.06.110] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/25/2016] [Indexed: 01/16/2023]
|
145
|
van Diepen JA, Thiem K, Stienstra R, Riksen NP, Tack CJ, Netea MG. Diabetes propels the risk for cardiovascular disease: sweet monocytes becoming aggressive? Cell Mol Life Sci 2016; 73:4675-4684. [PMID: 27469259 PMCID: PMC5097107 DOI: 10.1007/s00018-016-2316-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/27/2022]
Abstract
Diabetes strongly predisposes to cardiovascular disease (CVD), the leading cause of mortality in these patients, as well as in the entire population. Hyperglycemia is an important cardiovascular risk factor as shown by the observation that even transient periods of hyperglycemia, despite return to normoglycemia during follow-up, increase the risk for CVD, a phenomenon termed 'hyperglycemic memory'. The molecular mechanisms underlying this phenomenon remain largely unknown. As inflammation plays an important role in the pathogenesis of atherosclerosis, we propose that long-term functional reprogramming of monocytes and macrophages, induced by hyperglycemia, plays an important role in the phenomenon of hyperglycemic memory leading to cardiovascular complications in patients with diabetes. In this review, we discuss recent insights showing that innate immune cells possess the capacity to reprogram their function through epigenetically mediated rewiring of gene transcription, a process termed 'trained immunity'. The long-term reprogramming of monocytes can be induced by microbial as well as metabolic products, and involves a shift in cellular metabolism from oxidative phosphorylation to aerobic glycolysis. We hypothesize that hyperglycemia in diabetes patients induces long-term activation of monocytes and macrophages through similar mechanisms, thereby contributing to plaque development and subsequent macrovascular complications.
Collapse
Affiliation(s)
- Janna A van Diepen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Kathrin Thiem
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, 6703 HA, Wageningen, The Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen Medical Center, Radboudumc, (463), P. O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Nijmegen Medical Center, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
146
|
Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani R. Pathology of Human Coronary and Carotid Artery Atherosclerosis and Vascular Calcification in Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2016; 37:191-204. [PMID: 27908890 DOI: 10.1161/atvbaha.116.306256] [Citation(s) in RCA: 367] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022]
Abstract
The continuing increase in the prevalence of diabetes mellitus in the general population is predicted to result in a higher incidence of cardiovascular disease. Although the mechanisms of diabetes mellitus-associated progression of atherosclerosis are not fully understood, at clinical and pathological levels, there is an appreciation of increased disease burden and higher levels of arterial calcification in these subjects. Plaques within the coronary arteries of patients with diabetes mellitus generally exhibit larger necrotic cores and significantly greater inflammation consisting mainly of macrophages and T lymphocytes relative to patients without diabetes mellitus. Moreover, there is a higher incidence of healed plaque ruptures and positive remodeling in hearts from subjects with type 1 diabetes mellitus and type 2 diabetes mellitus, suggesting a more active atherogenic process. Lesion calcification in the coronary, carotid, and other arterial beds is also more extensive. Although the role of coronary artery calcification in identifying cardiovascular disease and predicting its outcome is undeniable, our understanding of how key hormonal and physiological alterations associated with diabetes mellitus such as insulin resistance and hyperglycemia influence the process of vascular calcification continues to grow. Important drivers of atherosclerotic calcification in diabetes mellitus include oxidative stress, endothelial dysfunction, alterations in mineral metabolism, increased inflammatory cytokine production, and release of osteoprogenitor cells from the marrow into the circulation. Our review will focus on the pathophysiology of type 1 diabetes mellitus- and type 2 diabetes mellitus-associated vascular disease with particular focus on coronary and carotid atherosclerotic calcification.
Collapse
Affiliation(s)
- Kazuyuki Yahagi
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Frank D Kolodgie
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Christoph Lutter
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Hiroyoshi Mori
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Maria E Romero
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Aloke V Finn
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.)
| | - Renu Virmani
- From the CVPath Institute, Inc, Gaithersburg, MD (K.Y., F.D.K., C.L., H.M., M.E.R., A.V.F., R.V.); and University of Maryland, School of Medicine, Baltimore (A.V.F.).
| |
Collapse
|
147
|
Samarghandian S, Azimi-Nezhad M, Farkhondeh T. Crocin attenuate Tumor Necrosis Factor-alpha (TNF-α) and interleukin-6 (IL-6) in streptozotocin-induced diabetic rat aorta. Cytokine 2016; 88:20-28. [PMID: 27529541 DOI: 10.1016/j.cyto.2016.08.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022]
|
148
|
Nakanishi R, Ceponiene I, Osawa K, Luo Y, Kanisawa M, Megowan N, Nezarat N, Rahmani S, Broersen A, Kitslaar PH, Dailing C, Budoff MJ. Plaque progression assessed by a novel semi-automated quantitative plaque software on coronary computed tomography angiography between diabetes and non-diabetes patients: A propensity-score matching study. Atherosclerosis 2016; 255:73-79. [PMID: 27835741 DOI: 10.1016/j.atherosclerosis.2016.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/26/2016] [Accepted: 11/02/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS We aimed at investigating whether diabetes is associated with progression in coronary plaque components. METHODS We identified 142 study subjects undergoing serial coronary computed tomography angiography. The resulting propensity score was applied 1:1 to match diabetic patients to non-diabetic patients for clinical risk factors, prior coronary stenting, coronary artery calcium (CAC) score and the serial scan interval, resulting in the 71 diabetes and 71 non-diabetes patients. Coronary plaque (total, calcified, non-calcified including fibrous, fibrous-fatty and low attenuation plaque [LAP]) volume normalized by total coronary artery length was measured using semi-automated plaque software and its change overtime between diabetic and non-diabetic patients was evaluated. RESULTS The matching was successful without significant differences between the two groups in all matched variables. The baseline volumes in each plaque also did not differ. During a mean scan interval of 3.4 ± 1.8 years, diabetic patients showed a 2-fold greater progression in normalized total plaque volume (TPV) than non-diabetes patients (52.8 mm3vs. 118.3 mm3, p = 0.005). Multivariable linear regression model revealed that diabetes was associated with normalized TPV progression (β 72.3, 95%CI 24.3-120.3). A similar trend was observed for the non-calcified components, but not calcified plaque (β 3.8, 95%CI -27.0-34.7). Higher baseline CAC score was found to be associated with total, non-calcified and calcified plaque progression. However, baseline non-calcified volume but not CAC score was associated with LAP progression. CONCLUSIONS The current study among matched patients indicates diabetes is associated with a greater plaque progression. Our results show the need for strict adherence of diabetic patients to the current preventive guidelines.
Collapse
Affiliation(s)
- Rine Nakanishi
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.
| | - Indre Ceponiene
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Kazuhiro Osawa
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Yanting Luo
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Mitsuru Kanisawa
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Nichelle Megowan
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Negin Nezarat
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Sina Rahmani
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | | | - Pieter H Kitslaar
- Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands; Medis Medical Systems bv, Leiden, 2316 XG, The Netherlands
| | - Christopher Dailing
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Matthew J Budoff
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| |
Collapse
|
149
|
Perales-Torres AL, Castillo-Ruíz O, Castañeda Licón MT, Alemán-Castillo SE, Jiménez Andrade JM. [Diabetes and type of diet as determinant factor in the progression of atherosclerosis]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2016; 86:326-334. [PMID: 26775035 DOI: 10.1016/j.acmx.2015.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 12/02/2015] [Accepted: 12/02/2015] [Indexed: 11/30/2022] Open
Abstract
The purpose of this review is to analyze the biochemical progression of atherosclerotic plaque and its association with diet and diabetes. This study shows the scientific evidence of demonstrating that diabetic patients present high levels of fatty acids like palmitic acid and linoleic acid in their atheroma plaques in comparison with non-diabetic patients. This study also establishes how patients with diabetes mellitus have a higher prevalence of atherosclerotic heart diseases in the form of Coronary Thrombosis and have different anatomopathological appearance like higher necrotic core and thin fibrotic layer than the general population. Furthermore this review describes the different anatomopathological appearance and cellular changes involved in the formation of these plaques and how diet can affect the development of these plaques.
Collapse
Affiliation(s)
| | - Octelina Castillo-Ruíz
- Departamento de Nutrición y Alimentos, Universidad Autónoma de Tamaulipas, Reynosa, Tamaulipas, México.
| | | | | | | |
Collapse
|
150
|
Immunomodulatory and antioxidant effects of saffron aqueous extract (Crocus sativus L.) on streptozotocin-induced diabetes in rats. Indian Heart J 2016; 69:151-159. [PMID: 28460761 PMCID: PMC5414951 DOI: 10.1016/j.ihj.2016.09.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/13/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
Introduction Crocus sativus L. (saffron) has many biological effects such as antioxidant property. The present study investigated the immunomodulatory effects of the aqueous saffron extract on streptozotocin (STZ)-induced diabetic rats. Materials and methods In this study, the rats were divided into the following groups of 9 animals each: control, untreated diabetic, three saffron extract-treated diabetic groups. Diabetes was induced by STZ in rats. Saffron was administered 3 days after STZ administration; these injections were continued to the end of the study (4 weeks). At the end of the 4-week period, blood was drawn for biochemical assays and the abdominal aorta was removed for detecting the inflammatory cytokines expression. Results We found that saffron decreased blood glucose, malondialdehyde, nitric oxide, total lipids, triglycerides, cholesterol levels significantly (p < 0.01) and increased glutathione level, catalase, and superoxide dismutase activities in the saffron-treated diabetic groups compared with the untreated groups, in a dose dependent manner (p < 0.05, p < 0.01, p < 0.001). On the other hand, saffron-treated diabetic rats inhibited the expression of inflammatory cytokines in the abdominal aorta versus the untreated diabetic rats. Conclusion Our results validate the use of saffron as a treatment against diabetes mellitus and its vascular complications.
Collapse
|