1
|
Aras T, Tayeh M, Aswad A, Sharkawy M, Almuzakki Z, Dorweiler B, Majd P. The BG Study Part 1 (Bergisch Gladbach): Development of a Prototype Coronary Artery Disease Risk Score Incorporating Peripheral Vascular Parameters-Preliminary Insights for Future CAD Risk Prediction Models in Vascular Patients. J Clin Med 2025; 14:1297. [PMID: 40004827 PMCID: PMC11856496 DOI: 10.3390/jcm14041297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Peripheral vascular parameters may provide valuable insights into coronary artery disease (CAD) risk stratification. This study aimed to develop a CAD risk score by integrating carotid duplex velocities, ankle-brachial index (ABI), and clinical history into a point-based model. Methods: We analyzed data from 902 cardiology patients, of whom 592 (65.6%) had confirmed CAD based on coronary angiography. Peripheral vascular assessments included carotid duplex ultrasonography and ABI measurements. Predictors were identified through multivariate logistic regression, addressing multicollinearity and interaction effects. A point-based scoring system was developed using statistically significant variables and evaluated via receiver operating characteristic (ROC) analysis. Results: Key predictors included external carotid artery velocities, ABI, carotid stenosis, chronic kidney disease (CKD) stage, smoking history, diabetes, hypertension, and age. The scoring system demonstrated moderate discriminative ability (AUC: 0.683) and high sensitivity (97%) for detecting CAD-positive cases but lower specificity (11%) for CAD-negative cases. Patients were stratified into risk categories, with an optimal threshold of ≥7 points maximizing the F1 score. Conclusions: This novel scoring system highlights the clinical relevance of integrating peripheral vascular assessments into CAD risk models. While its high sensitivity ensures robust detection of CAD-positive patients, future multicenter studies are needed to improve specificity and validate its broader clinical utility.
Collapse
Affiliation(s)
- Tuna Aras
- Department of Vascular Surgery, EVK Bergisch Gladbach, Ferrenbergstraße 24, 51465 Bergisch Gladbach, Germany; (M.T.); (P.M.)
| | - Mahmoud Tayeh
- Department of Vascular Surgery, EVK Bergisch Gladbach, Ferrenbergstraße 24, 51465 Bergisch Gladbach, Germany; (M.T.); (P.M.)
| | - Adel Aswad
- Al-Qassimi Teaching Hospital and Cardiac Centre, University of Sharjah, Sharjah P.O. Box 3500, United Arab Emirates; (A.A.); (Z.A.)
| | - Mohamed Sharkawy
- Department of Vascular Surgery, Faculty of Medicine, Cairo University Hospital, Cairo 4240310, Egypt;
| | - Zaki Almuzakki
- Al-Qassimi Teaching Hospital and Cardiac Centre, University of Sharjah, Sharjah P.O. Box 3500, United Arab Emirates; (A.A.); (Z.A.)
| | - Bernhard Dorweiler
- Department of Vascular and Endovascular Surgery, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937 Cologne, Germany;
| | - Payman Majd
- Department of Vascular Surgery, EVK Bergisch Gladbach, Ferrenbergstraße 24, 51465 Bergisch Gladbach, Germany; (M.T.); (P.M.)
| |
Collapse
|
2
|
Wei X, Wang M, Yu S, Han Z, Li C, Zhong Y, Zhang M, Yang T. Mapping the knowledge of omics in myocardial infarction: A scientometric analysis in R Studio, VOSviewer, Citespace, and SciMAT. Medicine (Baltimore) 2025; 104:e41368. [PMID: 39960900 PMCID: PMC11835070 DOI: 10.1097/md.0000000000041368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Many researchers nowadays choose multi-omics techniques for myocardial infarction studies. However, there's yet to be a review article integrating myocardial infarction multi-omics. Hence, this study adopts the popular bibliometrics. Based on its principles, we use software like R Studio, Vosviewer, Citespace, and SciMAT to analyze literature data of myocardial infarction omics research (1991-2022) from Web of Science. By extracting key information and calculating weights, we conduct analyses from 4 aspects: Collaboration Network Analysis, Co-word Analysis, Citing and Cited Journal Analysis, and Co-citation and Clustering Analysis, aiming to understand the field's cooperation, research topic evolution, and knowledge flow. The results show that myocardial infarction omics research is still in its early stage with limited international cooperation. In terms of knowledge flow, there's no significant difference within the discipline, but non-biomedical disciplines have joined, indicating an interdisciplinary integration trend. In the overall research field, genomics remains the main topic with many breakthroughs identifying susceptibility sites. Meanwhile, other omics fields like lipidomics and proteomics are also progressing, clarifying the pathogenesis. The cooperation details in this article enable researchers to connect with others, facilitating their research. The evolution trend of subject terms helps them set goals and directions, quickly grasp the development context, and read relevant literature. Journal analysis offers submission suggestions, and the analysis of research base and frontier provides references for the research's future development.
Collapse
Affiliation(s)
- Xuan Wei
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Min Wang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Shengnan Yu
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Zhengqi Han
- Institute for Digital Technology and Law (IDTL), China University of Political Science and Law, Beijing, China
- CUPL Scientometrics and Evaluation Center of Rule of Law, China University of Political Science and Law, Beijing, China
| | - Chang Li
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Yue Zhong
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Mengzhou Zhang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| | - Tiantong Yang
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, China
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, China
| |
Collapse
|
3
|
Burtscher J, Millet GP, Fresa M, Lanzi S, Mazzolai L, Pellegrin M. The link between impaired oxygen supply and cognitive decline in peripheral artery disease. Prog Cardiovasc Dis 2024; 85:63-73. [PMID: 38061613 DOI: 10.1016/j.pcad.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023]
Abstract
Although peripheral artery disease (PAD) primarily affects large arteries outside the brain, PAD is also associated with elevated cerebral vulnerabilities, including greater risks for brain injury (such as stroke), cognitive decline and dementia. In the present review, we aim to evaluate recent literature and extract information on potential mechanisms linking PAD and consequences on the brain. Furthermore, we suggest novel therapeutic avenues to mitigate cognitive decline and reduce risk of brain injury in patients with PAD. Various interventions, notably exercise, directly or indirectly improve systemic blood flow and oxygen supply and are effective strategies in patients with PAD or cognitive decline. Moreover, triggering protective cellular and systemic mechanisms by modulating inspired oxygen concentrations are emerging as potential novel treatment strategies. While several genetic and pharmacological approaches to modulate adaptations to hypoxia showed promising results in preclinical models of PAD, no clear benefits have yet been clinically demonstrated. We argue that genetic/pharmacological regulation of the involved adaptive systems remains challenging but that therapeutic variation of inspired oxygen levels (e.g., hypoxia conditioning) are promising future interventions to mitigate associated cognitive decline in patients with PAD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Marco Fresa
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Stefano Lanzi
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Lucia Mazzolai
- Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Maxime Pellegrin
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Angiology Department, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
4
|
Omiye JA, Ghanzouri I, Lopez I, Wang F, Cabot J, Amal S, Ye J, Lopez NG, Adebayo-Tijani F, Ross EG. Clinical use of polygenic risk scores for detection of peripheral artery disease and cardiovascular events. PLoS One 2024; 19:e0303610. [PMID: 38758931 PMCID: PMC11101066 DOI: 10.1371/journal.pone.0303610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 04/26/2024] [Indexed: 05/19/2024] Open
Abstract
We have previously shown that polygenic risk scores (PRS) can improve risk stratification of peripheral artery disease (PAD) in a large, retrospective cohort. Here, we evaluate the potential of PRS in improving the detection of PAD and prediction of major adverse cardiovascular and cerebrovascular events (MACCE) and adverse events (AE) in an institutional patient cohort. We created a cohort of 278 patients (52 cases and 226 controls) and fit a PAD-specific PRS based on the weighted sum of risk alleles. We built traditional clinical risk models and machine learning (ML) models using clinical and genetic variables to detect PAD, MACCE, and AE. The models' performances were measured using the area under the curve (AUC), net reclassification index (NRI), integrated discrimination improvement (IDI), and Brier score. We also evaluated the clinical utility of our PAD model using decision curve analysis (DCA). We found a modest, but not statistically significant improvement in the PAD detection model's performance with the inclusion of PRS from 0.902 (95% CI: 0.846-0.957) (clinical variables only) to 0.909 (95% CI: 0.856-0.961) (clinical variables with PRS). The PRS inclusion significantly improved risk re-classification of PAD with an NRI of 0.07 (95% CI: 0.002-0.137), p = 0.04. For our ML model predicting MACCE, the addition of PRS did not significantly improve the AUC, however, NRI analysis demonstrated significant improvement in risk re-classification (p = 2e-05). Decision curve analysis showed higher net benefit of our combined PRS-clinical model across all thresholds of PAD detection. Including PRS to a clinical PAD-risk model was associated with improvement in risk stratification and clinical utility, although we did not see a significant change in AUC. This result underscores the potential clinical utility of incorporating PRS data into clinical risk models for prevalent PAD and the need for use of evaluation metrics that can discern the clinical impact of using new biomarkers in smaller populations.
Collapse
Affiliation(s)
- Jesutofunmi A. Omiye
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ilies Ghanzouri
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ivan Lopez
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Fudi Wang
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - John Cabot
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Saeed Amal
- Department of Bioengineering, The Roux Institute at Northeastern University, Portland, Maine, United States of America
| | - Jianqin Ye
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicolas Gabriel Lopez
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Faatihat Adebayo-Tijani
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Elsie Gyang Ross
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Vascular Surgery, Department of Surgery, UC San Diego School of Medicine, La Jolla, California, United States of America
| |
Collapse
|
5
|
Chang WT, Huang PS, Su LW, Liao CT, Siong Toh H, Chen YC, Chung‑Han H, Chen ZC, Hsu PC, Hong CS. Utility of the ACD-GENE-CLI Score in Asian Patients with Critical Limb Ischemia Undergoing Endovascular Interventions. J Atheroscler Thromb 2024; 31:572-586. [PMID: 38092392 PMCID: PMC11079481 DOI: 10.5551/jat.64326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/19/2023] [Indexed: 05/03/2024] Open
Abstract
AIMS Critical limb ischemia (CLI) is an emerging public health threat and lacks a reliable score for predicting the outcomes. The Age, Body Mass Index, Chronic Kidney Disease, Diabetes, and Genotyping (ABCD-GENE) risk score helps identify patients with coronary artery disease who have cytochrome P450 2C19 (CYP2C19) polymorphism-related drug resistance and are at risk for cardiovascular adverse events. However, its application to CLI remains unknown. In this study, we aim to validate a modified ACD-GENE-CLI score to improve the prediction of major adverse limb events (MALEs) in patients with CLI receiving clopidogrel. METHODS Patients with CLI receiving clopidogrel post-endovascular intervention were enrolled prospectively in two medical centers. Amputation and revascularization as MALEs were regarded as the outcomes. RESULTS A total of 473 patients were recruited, with a mean follow-up duration of 25 months. Except for obesity, old age, diabetes, chronic kidney disease (CKD), and CYP2C19 polymorphisms were significantly associated with MALEs. Using bootstrap regression analysis, we established a modified risk score (ACD-GENE-CLI) that included old age (≥ 65 years), diabetes, CKD, and CYP2C19 polymorphisms. At a cutoff value of 8, the ACD-GENE-CLI score was superior to the CYP2C19 deficiency only, and the conventional ABCD-GENE score in predicting MALEs (area under the curve: 0.69 vs. 0.59 vs. 0.67, p=0.01). The diagnostic ability of the ACD-GENE-CLI score was consistent in the external validation. Also, Kaplan-Meier curves showed that in CYP2C19 deficiency, the ABCD-GENE and ACD-GENE-CLI scores could all differentiate patients with CLI who are free from MALEs. CONCLUSIONS The modified ACD-GENE-CLI score could differentiate patients with CLI receiving clopidogrel who are at risk of MALEs. Further studies are required to generalize the utility of the score.
Collapse
Affiliation(s)
- Wei-Ting Chang
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Po-Sen Huang
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Li-Wei Su
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chia-Te Liao
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - Han Siong Toh
- Department of Intensive Care Medicine, Chi Mei Medical Centre, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yi-Chen Chen
- Department of Medical Research, Chi-Mei Medical Center, Tainan
| | - Ho Chung‑Han
- Department of Medical Research, Chi-Mei Medical Center, Tainan
- Department of Information Management, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Po-Chao Hsu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Internal Medicine, Faculty of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chon-Seng Hong
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
6
|
Via M, Pera G, Forés R, Costa-Garrido A, Heras A, Baena-Díez JM, Pedrosa E, Clemente IC, Lamonja-Vicente N, Mataró M, Torán-Montserrat P, Alzamora MT. Genetic Variants at the 9p21.3 Locus Are Associated with Risk for Non-Compressible Artery Disease: Results from the ARTPER Study. Genes (Basel) 2023; 15:2. [PMID: 38275585 PMCID: PMC10815029 DOI: 10.3390/genes15010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Peripheral artery disease (PAD) and non-compressible artery disease (NCAD) constitute predictors of subclinical atherosclerosis easily assessed through the ankle brachial index (ABI). Although both diseases show substantial genetic influences, few genetic association studies have focused on the ABI and PAD, and none have focused on NCAD. To overcome these limitations, we assessed the role of several candidate genes on the ABI, both in its continuous distribution and in the clinical manifestations associated to its extreme values: PAD and NCAD. We examined 13 candidate genomic regions in 1606 participants from the ARTPER study, a prospective population-based cohort, with the ABI assessed through ultrasonography. Association analyses were conducted independently for individuals with PAD (ABI < 0.9) or with NCAD (ABI > 1.4) vs. healthy participants. After including potential covariates and correction for multiple testing, minor alleles in the genetic markers rs10757278 and rs1333049, both in the 9p21.3 region, were significantly associated with a decreased risk of NCAD. Associations with the ABI showed limited support to these results. No significant associations were detected for PAD. The locus 9p21.3 constitutes the first genetic locus associated with NCAD, an assessment of subclinical atherosclerosis feasible for implementation in primary healthcare settings that has been systematically neglected from genetic studies.
Collapse
Affiliation(s)
- Marc Via
- Brainlab-Grup de Recerca en Neurociència Cognitiva, Departament de Psicologia Clínica i Psicobiologia, Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain;
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
| | - Guillem Pera
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
| | - Rosa Forés
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
| | - Anna Costa-Garrido
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
| | - Antonio Heras
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
- Centre d’Atenció Primària Riu Nord-Riu Sud Santa Coloma de Gramenet, Direcció d’Atenció Primària Barcelonés Nord i Maresme, Institut Català de la Salut, 08921 Santa Coloma de Gramenet, Spain
| | - José Miguel Baena-Díez
- Centre d’Atenció Primària la Marina, Direcció d’Atenció Primària Barcelona Ciutat, Institut Català de la Salut, 08038 Barcelona, Spain;
- Institut Universitari d’Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), 08007 Barcelona, Spain
| | - Edurne Pedrosa
- IGTP-HUGTP Biobank, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain;
| | - Inmaculada C. Clemente
- Brainlab-Grup de Recerca en Neurociència Cognitiva, Departament de Psicologia Clínica i Psicobiologia, Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain;
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
| | - Noemí Lamonja-Vicente
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
- Departament de Psicologia Clínica i Psicobiologia, Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| | - Maria Mataró
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
- Departament de Psicologia Clínica i Psicobiologia, Institut de Neurociències, Universitat de Barcelona, 08035 Barcelona, Spain
| | - Pere Torán-Montserrat
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
| | - M. Teresa Alzamora
- Unitat de Suport a la Recerca Metropolitana Nord, Fundació Institut Universitari per a la recerca a l’Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), 08303 Mataró, Spain; (G.P.); (R.F.); (A.C.-G.); (A.H.); (N.L.-V.); (P.T.-M.); (M.T.A.)
- Centre d’Atenció Primària Riu Nord-Riu Sud Santa Coloma de Gramenet, Direcció d’Atenció Primària Barcelonés Nord i Maresme, Institut Català de la Salut, 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
7
|
Benítez-Camacho J, Ballesteros A, Beltrán-Camacho L, Rojas-Torres M, Rosal-Vela A, Jimenez-Palomares M, Sanchez-Gomar I, Durán-Ruiz MC. Endothelial progenitor cells as biomarkers of diabetes-related cardiovascular complications. Stem Cell Res Ther 2023; 14:324. [PMID: 37950274 PMCID: PMC10636846 DOI: 10.1186/s13287-023-03537-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
Diabetes mellitus (DM) constitutes a chronic metabolic disease characterized by elevated levels of blood glucose which can also lead to the so-called diabetic vascular complications (DVCs), responsible for most of the morbidity, hospitalizations and death registered in these patients. Currently, different approaches to prevent or reduce DM and its DVCs have focused on reducing blood sugar levels, cholesterol management or even changes in lifestyle habits. However, even the strictest glycaemic control strategies are not always sufficient to prevent the development of DVCs, which reflects the need to identify reliable biomarkers capable of predicting further vascular complications in diabetic patients. Endothelial progenitor cells (EPCs), widely known for their potential applications in cell therapy due to their regenerative properties, may be used as differential markers in DVCs, considering that the number and functionality of these cells are affected under the pathological environments related to DM. Besides, drugs commonly used with DM patients may influence the level or behaviour of EPCs as a pleiotropic effect that could finally be decisive in the prognosis of the disease. In the current review, we have analysed the relationship between diabetes and DVCs, focusing on the potential use of EPCs as biomarkers of diabetes progression towards the development of major vascular complications. Moreover, the effects of different drugs on the number and function of EPCs have been also addressed.
Collapse
Affiliation(s)
- Josefa Benítez-Camacho
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
| | - Antonio Ballesteros
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain
| | - Lucía Beltrán-Camacho
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Córdoba, Spain
- Cell Biology, Physiology and Immunology Department, Córdoba University, Córdoba, Spain
| | - Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
| | - Margarita Jimenez-Palomares
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
| | - Ismael Sanchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Science Faculty, Cádiz University, Torre Sur. Avda. República Saharaui S/N, Polígono Río San Pedro, Puerto Real, 11519, Cádiz, Spain.
- Biomedical Research and Innovation Institute of Cadiz (INIBICA), Cádiz, Spain.
| |
Collapse
|
8
|
Di Stolfo G, Pacilli MA, Seripa D, De Luca G, Urbano M, Coli C, Gravina C, Greco A, Potenza DR, Salvatori MP, Schernthaner GH, Poredos P, Catalano M, Mastroianno S. Involvement of APOE in Incidence of Revascularization in Patients Affected by Peripheral Arterial Disease: A Prospective Study from Southern Italy. J Clin Med 2023; 12:5178. [PMID: 37629219 PMCID: PMC10455789 DOI: 10.3390/jcm12165178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION Atherosclerosis is a complex multifactorial disease and apolipoprotein E (APOE) polymorphism has been associated with cardiovascular events. The APOE gene, located on chromosome 19q13.2, has an important role in lipid metabolism, in particular on circulating cholesterol levels, implying further pleiotropic effects; from its polymorphism are derived three alleles (ε2, ε3 and ε4), which induce different phenotypes, while its impact on carotid and femoral atherosclerosis is still controversial. OBJECTIVES The aim of the study is to investigate the relationship between APOE genotypes and peripheral revascularization in a cohort of patients affected by advanced peripheral arterial disease (PAD) at a prolonged follow-up. MATERIALS AND METHODS Some 332 patients (259 males and 73 females; mean age 70.86 ± 7.95 years) with severe PAD were enrolled in a longitudinal study, with a 90.75 ± 32.25 month follow-up, assessing major adverse cardiovascular events (MACE). RESULTS As compared with ε3/ε3, in ε4 patients we observed a significant higher incidence of carotid (13.2% vs. 5.6%; HR = 2.485, 95% CI 1.062-5.814; p = 0.036) and lower limb (11.8% vs. 4.3%; HR = 2.765, 95% CI 1.091-7.008; p = 0.032) revascularizations and, accordingly, a higher incidence of total peripheral revascularizations (13.5% vs. 9.5%; HR = 2.705, 95% CI 1.420-5.151; p = 0.002). HR remained statistically significant even when adjusted for classic cardiovascular risk factors. CONCLUSIONS In our observational study, we confirm that the ε4 allele is associated with higher total peripheral revascularization in patients with advanced atherosclerotic vascular disease at prolonged follow-up.
Collapse
Affiliation(s)
- Giuseppe Di Stolfo
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Michele Antonio Pacilli
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Davide Seripa
- Complex Structure of Geriatrics, Medical Sciences Department, Fondazione IRCCS Casa Sollievodella Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Giovanni De Luca
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Maria Urbano
- Complex Structure of Geriatrics, Medical Sciences Department, Fondazione IRCCS Casa Sollievodella Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Carlo Coli
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Carolina Gravina
- Complex Structure of Geriatrics, Medical Sciences Department, Fondazione IRCCS Casa Sollievodella Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Antonio Greco
- Complex Structure of Geriatrics, Medical Sciences Department, Fondazione IRCCS Casa Sollievodella Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Domenico Rosario Potenza
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Mauro Pellegrino Salvatori
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Gerit-Holger Schernthaner
- Division of Angiology, Department of Internal Medicine 2, Medical University of Vienna, 1090 Vienna, Austria
| | - Pavel Poredos
- Department of Vascular Diseases, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
| | - Mariella Catalano
- Research Center on Vascular Diseases and Angiology Unit, University of Milan, L. Sacco Hospital, 20157 Milan, Italy
| | - Sandra Mastroianno
- Cardiovascular Department, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| |
Collapse
|
9
|
Cullina S, Wojcik GL, Shemirani R, Klarin D, Gorman BR, Sorokin EP, Gignoux CR, Belbin GM, Pyarajan S, Asgari S, Tsao PS, Damrauer SM, Abul-Husn NS, Kenny EE. Admixture mapping of peripheral artery disease in a Dominican population reveals a putative risk locus on 2q35. Front Genet 2023; 14:1181167. [PMID: 37600667 PMCID: PMC10432698 DOI: 10.3389/fgene.2023.1181167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Peripheral artery disease (PAD) is a form of atherosclerotic cardiovascular disease, affecting ∼8 million Americans, and is known to have racial and ethnic disparities. PAD has been reported to have a significantly higher prevalence in African Americans (AAs) compared to non-Hispanic European Americans (EAs). Hispanic/Latinos (HLs) have been reported to have lower or similar rates of PAD compared to EAs, despite having a paradoxically high burden of PAD risk factors; however, recent work suggests prevalence may differ between sub-groups. Here, we examined a large cohort of diverse adults in the BioMe biobank in New York City. We observed the prevalence of PAD at 1.7% in EAs vs. 8.5% and 9.4% in AAs and HLs, respectively, and among HL sub-groups, the prevalence was found at 11.4% and 11.5% in Puerto Rican and Dominican populations, respectively. Follow-up analysis that adjusted for common risk factors demonstrated that Dominicans had the highest increased risk for PAD relative to EAs [OR = 3.15 (95% CI 2.33-4.25), p < 6.44 × 10-14]. To investigate whether genetic factors may explain this increased risk, we performed admixture mapping by testing the association between local ancestry and PAD in Dominican BioMe participants (N = 1,813) separately from European, African, and Native American (NAT) continental ancestry tracts. The top association with PAD was an NAT ancestry tract at chromosome 2q35 [OR = 1.96 (SE = 0.16), p < 2.75 × 10-05) with 22.6% vs. 12.9% PAD prevalence in heterozygous NAT tract carriers versus non-carriers, respectively. Fine-mapping at this locus implicated tag SNP rs78529201 located within a long intergenic non-coding RNA (lincRNA) LINC00607, a gene expression regulator of key genes related to thrombosis and extracellular remodeling of endothelial cells, suggesting a putative link of the 2q35 locus to PAD etiology. Efforts to reproduce the signal in other Hispanic cohorts were unsuccessful. In summary, we showed how leveraging health system data helped understand nuances of PAD risk across HL sub-groups and admixture mapping approaches elucidated a putative risk locus in a Dominican population.
Collapse
Affiliation(s)
- Sinead Cullina
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Genevieve L. Wojcik
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Ruhollah Shemirani
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Derek Klarin
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Vascular Surgery, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Bryan R. Gorman
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, United States
- Booz Allen Hamilton, McLean, VA, United States
| | - Elena P. Sorokin
- Department of Genetics, Stanford University, Stanford, CA, United States
| | - Christopher R. Gignoux
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Colorado Center for Personalized Medicine, Aurora, CO, United States
| | - Gillian M. Belbin
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, United States
- Department of Medicine, Brigham Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Samira Asgari
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Philip S. Tsao
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
| | - Scott M. Damrauer
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Noura S. Abul-Husn
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Division of Genomic Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
10
|
Cullina S, Wojcik GL, Shemirani R, Klarin D, Gorman BR, Sorokin EP, Gignoux CR, Belbin GM, Pyarajan S, Asgari S, Tsao PS, Damrauer SM, Abul-Husn NS, Kenny EE. Admixture Mapping of Peripheral Artery Disease in a Dominican Population Reveals a Novel Risk Locus on 2q35. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.27.23287788. [PMID: 37034679 PMCID: PMC10081406 DOI: 10.1101/2023.03.27.23287788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Peripheral artery disease (PAD) is a form of atherosclerotic cardiovascular disease, affecting ∼8 million Americans, and is known to have racial and ethnic disparities. PAD has been reported to have significantly higher prevalence in African Americans (AAs) compared to non-Hispanic European Americans (EAs). Hispanic/Latinos (HLs) have been reported to have lower or similar rates of PAD compared to EAs, despite having a paradoxically high burden of PAD risk factors, however recent work suggests prevalence may differ between sub-groups. Here we examined a large cohort of diverse adults in the Bio Me biobank in New York City (NYC). We observed the prevalence of PAD at 1.7% in EAs vs 8.5% and 9.4% in AAs and HLs, respectively; and among HL sub-groups, at 11.4% and 11.5% in Puerto Rican and Dominican populations, respectively. Follow-up analysis that adjusted for common risk factors demonstrated that Dominicans had the highest increased risk for PAD relative to EAs (OR=3.15 (95% CI 2.33-4.25), P <6.44×10 -14 ). To investigate whether genetic factors may explain this increased risk, we performed admixture mapping by testing the association between local ancestry (LA) and PAD in Dominican Bio Me participants (N=1,940) separately for European (EUR), African (AFR) and Native American (NAT) continental ancestry tracts. We identified a NAT ancestry tract at chromosome 2q35 that was significantly associated with PAD (OR=2.05 (95% CI 1.51-2.78), P <4.06×10 -6 ) with 22.5% vs 12.5% PAD prevalence in heterozygous NAT tract carriers versus non-carriers, respectively. Fine-mapping at this locus implicated tag SNP rs78529201 located within a long intergenic non-coding RNA (lincRNA) LINC00607 , a gene expression regulator of key genes related to thrombosis and extracellular remodeling of endothelial cells, suggesting a putative link of the 2q35 locus to PAD etiology. In summary, we showed how leveraging health systems data helped understand nuances of PAD risk across HL sub-groups and admixture mapping approaches elucidated a novel risk locus in a Dominican population.
Collapse
|
11
|
Zhabin S, Lazarenko V, Azarova I, Klyosova E, Bykanova M, Chernousova S, Bashkatov D, Gneeva E, Polonikova A, Churnosov M, Solodilova M, Polonikov A. The Joint Link of the rs1051730 and rs1902341 Polymorphisms and Cigarette Smoking to Peripheral Artery Disease and Atherosclerotic Lesions of Different Arterial Beds. Life (Basel) 2023; 13:496. [PMID: 36836853 PMCID: PMC9961460 DOI: 10.3390/life13020496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Genome-wide association studies (GWAS) have discovered numerous single nucleotide polymorphisms (SNP) contributing to peripheral artery disease (PAD), but their joint effects with risk factors like cigarette smoking (CS) on disease susceptibility have not been systematically investigated. The present study looked into whether CS mediates the effects of GWAS loci on the development of PAD and atherosclerotic lesions in different arterial beds. DNA samples from 1263 unrelated individuals of Slavic origin including 620 PAD patients and 643 healthy subjects were genotyped by the MassArray-4 system for rs1051730, rs10134584, rs1902341, rs10129758 which are known as PAD-associated GWAS loci. The rs1051730 polymorphism was strongly associated with an increased risk of PAD (p = 5.1 × 10-6), whereas rs1902341 did not show an association with disease risk. The rs1051730 polymorphism was associated with increased plasma levels of LDL cholesterol (p = 0.001), and conferred a greater risk of PAD in cigarette smokers than in nonsmokers (p < 0.01). Interestingly, the rs1902341T allele was associated with an increased risk of PAD in smokers and a decreased disease risk in nonsmokers. SNPs and CS were both linked to unilateral and/or bilateral atherosclerotic lesions of peripheral vessels, as well as the abdominal aorta, coronary, and cerebral arteries. The studied polymorphisms exert pleiotropic and cigarette smoking-mediated effects on atherosclerotic lesions of different arterial beds.
Collapse
Affiliation(s)
- Sergey Zhabin
- Department of Surgical Diseases №1, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Victor Lazarenko
- Department of Surgical Diseases of Institute of Continuing Education, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russia
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russia
| | - Marina Bykanova
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russia
| | - Svetlana Chernousova
- Department of Surgical Diseases №1, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Daniil Bashkatov
- Department of Surgical Diseases №1, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Ekaterina Gneeva
- Department of Surgical Diseases №1, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Anna Polonikova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State University, 85 Pobedy Street, Belgorod 308015, Russia
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russia
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russia
| |
Collapse
|
12
|
The Genetic Architecture of the Etiology of Lower Extremity Peripheral Artery Disease: Current Knowledge and Future Challenges in the Era of Genomic Medicine. Int J Mol Sci 2022; 23:ijms231810481. [PMID: 36142394 PMCID: PMC9499674 DOI: 10.3390/ijms231810481] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
Lower extremity artery disease (LEAD), caused by atherosclerotic obstruction of the arteries of the lower limb extremities, has exhibited an increase in mortality and morbidity worldwide. The phenotypic variability of LEAD is correlated with its complex, multifactorial etiology. In addition to traditional risk factors, it has been shown that the interaction between genetic factors (epistasis) or between genes and the environment potentially have an independent role in the development and progression of LEAD. In recent years, progress has been made in identifying genetic variants associated with LEAD, by Genome-Wide Association Studies (GWAS), Whole Exome Sequencing (WES) studies, and epigenetic profiling. The aim of this review is to present the current knowledge about the genetic factors involved in the etiopathogenic mechanisms of LEAD, as well as possible directions for future research. We analyzed data from the literature, starting with candidate gene-based association studies, and then continuing with extensive association studies, such as GWAS and WES. The results of these studies showed that the genetic architecture of LEAD is extremely heterogeneous. In the future, the identification of new genetic factors will allow for the development of targeted molecular therapies, and the use of polygenic risk scores (PRS) to identify individuals at an increased risk of LEAD will allow for early prophylactic measures and personalized therapy to improve their prognosis.
Collapse
|
13
|
Ghanzouri I, Amal S, Ho V, Safarnejad L, Cabot J, Brown-Johnson CG, Leeper N, Asch S, Shah NH, Ross EG. Performance and usability testing of an automated tool for detection of peripheral artery disease using electronic health records. Sci Rep 2022; 12:13364. [PMID: 35922657 PMCID: PMC9349186 DOI: 10.1038/s41598-022-17180-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/21/2022] [Indexed: 11/18/2022] Open
Abstract
Peripheral artery disease (PAD) is a common cardiovascular disorder that is frequently underdiagnosed, which can lead to poorer outcomes due to lower rates of medical optimization. We aimed to develop an automated tool to identify undiagnosed PAD and evaluate physician acceptance of a dashboard representation of risk assessment. Data were derived from electronic health records (EHR). We developed and compared traditional risk score models to novel machine learning models. For usability testing, primary and specialty care physicians were recruited and interviewed until thematic saturation. Data from 3168 patients with PAD and 16,863 controls were utilized. Results showed a deep learning model that utilized time engineered features outperformed random forest and traditional logistic regression models (average AUCs 0.96, 0.91 and 0.81, respectively), P < 0.0001. Of interviewed physicians, 75% were receptive to an EHR-based automated PAD model. Feedback emphasized workflow optimization, including integrating risk assessments directly into the EHR, using dashboard designs that minimize clicks, and providing risk assessments for clinically complex patients. In conclusion, we demonstrate that EHR-based machine learning models can accurately detect risk of PAD and that physicians are receptive to automated risk detection for PAD. Future research aims to prospectively validate model performance and impact on patient outcomes.
Collapse
Affiliation(s)
- I Ghanzouri
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - S Amal
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - V Ho
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - L Safarnejad
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - J Cabot
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - C G Brown-Johnson
- Department of Medicine, Primary Care and Population Health, Stanford, CA, USA
| | - N Leeper
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - S Asch
- Department of Medicine, Primary Care and Population Health, Stanford, CA, USA
| | - N H Shah
- Department of Medicine, Center for Biomedical Informatics Research, Stanford University School of Medicine, 780 Welch Road, CJ350, Stanford, CA, 94305, USA
| | - E G Ross
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Medicine, Center for Biomedical Informatics Research, Stanford University School of Medicine, 780 Welch Road, CJ350, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Yang Q, Li C, Chen Q. SS31 Ameliorates Oxidative Stress via the Restoration of Autophagic Flux to Protect Aged Mice From Hind Limb Ischemia. Front Cardiovasc Med 2022; 9:789331. [PMID: 35497980 PMCID: PMC9046554 DOI: 10.3389/fcvm.2022.789331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/28/2022] [Indexed: 11/25/2022] Open
Abstract
Background Oxidative stress and impaired autophagic flux play important roles in the development of peripheral artery disease (PAD). SS31 is considered an important antioxidant peptide and autophagy regulator. We aimed to investigate the role of SS31 in PAD myopathy and its possible mechanism both in vivo and in vitro. Methods A hind limb ischemia (HLI) model was established with old C57BL/6 (14-month-old) mice. Mice in the SS31 group were intraperitoneally injected with SS31 (3 mg/kg) for 4 weeks. We examined skeletal muscle function and histomorphology, autophagy-related protein levels and reactive oxygen species (ROS) content. For the in vitro experiments, after C2C12 myotubes were treated with CoCl2, SS31, and chloroquine (CQ) or rapamycin (RAPA), we measured ROS content, autophagy-related protein levels and antioxidant enzyme expression. Results SS31 treatment effectively enhanced the recovery of skeletal muscle function, alleviated skeletal muscle injury and suppressed mitochondrial ROS production in ischemic limbs. SS31 reduced apoptosis and oxidative stress, and SS31 restored impaired autophagic flux by inhibiting the AKT-mTOR pathway. In vitro studies showed that SS31 restored autophagic flux and improved oxidative stress in C2C12 cells. Moreover, phosphorylated AKT (p-AKT) and phosphorylated mTOR (p-mTOR) levels were reduced. Conclusion These experiments indicated that SS31 can inhibit oxidative stress by restoring autophagic flux to reverse hypoxia-induced injury in vivo and in vitro.
Collapse
|
15
|
Liu YT, Lin CC, Wang L, Nfor ON, Hsu SY, Lung CC, Tantoh DM, Chang HR, Liaw YP. Peripheral Vascular Disease Susceptibility Based on Diabetes Mellitus and rs17367504 Polymorphism of the MTHFR Gene. Diabetes Metab Syndr Obes 2021; 14:2381-2388. [PMID: 34079316 PMCID: PMC8165095 DOI: 10.2147/dmso.s309242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/28/2021] [Indexed: 11/29/2022] Open
Abstract
PURPOSE Peripheral vascular disease (PVD) is a life-threatening condition affecting the lower extremities. Common risk factors include type 2 diabetes (T2D), hypertension, dyslipidemia, smoking, and older age. There is a little-documented research on the genetic basis of the disease in Taiwan. We examined the impact of T2D and the blood pressure-associated rs17367504 variant of the Methylenetetrahydrofolate reductase (MTHFR) gene on PVD risk. MATERIALS AND METHODS In this population-based association study, we linked data from 8992 participants in Taiwan Biobank (TWB) to their medical records in the National Health Insurance Research Database (NHIRD). Participants were 30 to 70 years old at recruitment and included those assessed between 2008 and 2015. We tested for association of PVD with rs17367504 and T2D using multiple logistic regression models. The rs17367504 variant was assessed using the Axiom-Taiwan Biobank Array Plate (TWB chip: Affymetrix, Inc., Santa Clara, CA, USA). RESULTS Among cases with T2D (n = 1294), 158 (12.21%) were identified with PVD. T2D was associated with PVD (odds ratio [OR], 1.52; 95% confidence interval [CI], 1.21-1.91; p<0.001) whereas rs17367504 variant was not (OR, 0.96; CI, 0.76-1.21; p = 0.728 in AG/GG compared to AA homozygotes). However, T2D and rs17367504 had an interactive effect on PVD (p for interaction = 0.0076). Results from our stratified analyses displayed OR of 1.75 (CI, 1.35-2.26; p<0.001) in AA individuals with DM and 0.94 (CI, 0.56-1.58; p = 0.811) in AG+GG individuals with T2D. Using the AA genotype and no T2D as the reference group, the respective OR of PVD was 1.77 (CI, 1.38-2.28; p<0.001) in AA individuals with T2D; 1.18 (CI, 0.91-1.55; p = 0.215) in AG+GG individuals with no T2D, and 1.03 (CI, 0.66-1.60; p = 0.892) in AG+GG individuals with T2D . CONCLUSION We found that type 2 diabetes was associated with increased risk of peripheral vascular disease, particularly in AA genotype carriers of the rs17367504 variant in Taiwan.
Collapse
Affiliation(s)
- Yin-Tso Liu
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan
- Department of Cardiovascular Surgery, Asia University Hospital, Taichung, 40201, Taiwan
| | - Chuan-Chao Lin
- Department of Physical Medicine and Rehabilitation, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Lee Wang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Shu-Yi Hsu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Chia-Chi Lung
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
| | - Disline Manli Tantoh
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Horng-Rong Chang
- Institute of Medicine, Chung Shan Medical University, Taichung, 40201, Taiwan
- Division of Nephrology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| |
Collapse
|
16
|
Liu F, Du J, Nie M, Fu J, Sun J. 5,10-methylenetetrahydrofolate reductase C677T gene polymorphism and peripheral arterial disease: A meta-analysis. Vascular 2020; 29:913-919. [PMID: 33357155 DOI: 10.1177/1708538120982698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Peripheral arterial disease is one common vascular disease most caused by atherosclerosis. As with stroke and coronary heart disease, peripheral arterial disease is one clinical type of atherosclerotic cardiovascular disease with many unmeasured environmental and genetic components. MTHFR C677T polymorphism is associated with the increased risk of ischemic stroke and coronary heart disease. MTHFR C677T polymorphism is associated with decreasing enzyme activity and increasing homocysteine levels. Meta-analysis of studies had demonstrated an association between elevated plasma homocysteine levels and peripheral arterial disease. Elevated plasma homocysteine level is closely related to MTHFR C677T polymorphism. Recent studies had clarified the relationship of MTHFR C677T polymorphism and peripheral arterial disease. So we performed a meta-analysis to investigate the association between MTHFR C677T polymorphism and peripheral arterial disease. MATERIALS AND METHODS We searched the database PubMed, Embase, and Cochrane Library for all English-language articles related to peripheral arterial disease and MTHFR C677T through 30 June 2020. Analysis results were shown by forest plot. Publication bias was estimated using funnel plot. RESULTS A total of 15 studies comprising 1929 patients with peripheral arterial disease and 2952 healthy controls were included in the meta-analysis. Significant associations between MTHFR C677T genetic polymorphism and peripheral arterial disease were found (OR = 1.31, 95% CI: 1.09-1.58, P <0.01). But there was no significant association (poor OR = 1.11, 95% CI: 0.98-1.26, P =0.11) between the T allele carrier and peripheral arterial disease. CONCLUSION Our meta-analysis suggested that MTHFR C677T genetic polymorphism TT genotype may be associated with increased peripheral arterial disease risk. But further studies with large sample sizes are needed to confirm our findings.
Collapse
Affiliation(s)
- Fanyun Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Menglin Nie
- Department of Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Fu
- Department of Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianming Sun
- Department of Vascular Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Safarova MS, Fan X, Austin EE, van Zuydam N, Hopewell J, Schaid DJ, Kullo IJ. Targeted Sequencing Study to Uncover Shared Genetic Susceptibility Between Peripheral Artery Disease and Coronary Heart Disease-Brief Report. Arterioscler Thromb Vasc Biol 2020; 39:1227-1233. [PMID: 31070467 DOI: 10.1161/atvbaha.118.312128] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective- It is unclear to what extent genetic susceptibility variants are shared between peripheral artery disease (PAD) and coronary heart disease (CHD), both manifestations of atherosclerotic vascular disease. We investigated whether common and low-frequency/rare variants in loci associated with CHD are also associated with PAD. Approach and Results- Targeted sequencing of 41 genomic regions associated with CHD in genome-wide association studies was performed in 1749 PAD cases (65±11 years, 61% men) and 1855 controls (60±11 years, 56% men) of European ancestry. PAD cases had a resting/postexercise ankle-brachial index ≤0.9, or history of lower extremity revascularization; controls had no history of PAD. We tested the association of common (defined as minor allele frequency ≥5%) variants with PAD assuming an additive genetic model with adjustment for age and sex. To identify low-frequency/rare variants (minor allele frequency <5%) associated with PAD, we conducted gene-level analyses using sequence kernel association test and permutation test. After Bonferroni correction, we found common variants in SH2B3, ABO, and ZEB2 to be associated with PAD ( P<4.5×10-5). At the gene level, the strongest associations were for LPL and SH2B3. Conclusions- Targeted sequencing of 41 genomic regions associated with CHD revealed several common variants/genes to be associated with PAD, highlighting the basis of shared genetic susceptibility between CHD and PAD.
Collapse
Affiliation(s)
- Maya S Safarova
- From the Department of Cardiovascular Medicine (M.S.S., X.F., E.E.A., I.J.K.), Mayo Clinic, Rochester, MN
| | - Xiao Fan
- From the Department of Cardiovascular Medicine (M.S.S., X.F., E.E.A., I.J.K.), Mayo Clinic, Rochester, MN
| | - Erin E Austin
- From the Department of Cardiovascular Medicine (M.S.S., X.F., E.E.A., I.J.K.), Mayo Clinic, Rochester, MN
| | - Natalie van Zuydam
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom (N.v.Z.)
| | - Jemma Hopewell
- Nuffield Department of Population Health, Oxford, United Kingdom (J.H.)
| | - Daniel J Schaid
- Department of Health Sciences Research (D.J.S.), Mayo Clinic, Rochester, MN
| | - Iftikhar J Kullo
- From the Department of Cardiovascular Medicine (M.S.S., X.F., E.E.A., I.J.K.), Mayo Clinic, Rochester, MN.,Gonda Vascular Center (I.J.K.), Mayo Clinic, Rochester, MN
| |
Collapse
|
18
|
Bogucka-Kocka A, Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Bogucki J, Komsta Ł, Kołodziej P, Zubilewicz T, Feldo M, Kocki J. Dysregulation of MicroRNA Regulatory Network in Lower Extremities Arterial Disease. Front Genet 2019; 10:1200. [PMID: 31827490 PMCID: PMC6892359 DOI: 10.3389/fgene.2019.01200] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/29/2019] [Indexed: 01/12/2023] Open
Abstract
Atherosclerosis and its comorbidities are the major contributors to the global burden of death worldwide. Lower extremities arterial disease (LEAD) is a common manifestation of atherosclerotic disease of arteries of lower extremities. MicroRNAs belong to epigenetic factors that regulate gene expression and have not yet been extensively studied in LEAD. We aimed to indicate the most promising microRNA and gene expression signatures of LEAD, to identify interactions between microRNA and genes and to describe potential effect of modulated gene expression. High-throughput sequencing was employed to examine microRNAome and transcriptome of peripheral blood mononuclear cells of patients with LEAD, in relation to controls. Statistical significance of microRNAs and genes analysis results was evaluated using DESeq2 and uninformative variable elimination by partial least squares methods. Altered expression of 26 microRNAs (hsa-let-7f-1-3p, hsa-miR-34a-5p, -122-5p, -3591-3p, -34a-3p, -1261, -21-5p, -15a-5p, -548d-5p, -34b-5p, -424-3p, -548aa, -548t-3p, -4423-3p, -196a-5p, -330-3p, -766-3p, -30e-3p, -125b-5p, -1301-3p, -3184-5p, -423-3p, -339-3p, -138-5p, -99a-3p, and -6087) and 14 genes (AK5, CD248, CDS2, FAM129A, FBLN2, GGT1, NOG, NRCAM, PDE7A, RP11-545E17.3, SLC12A2, SLC16A10, SLC4A10, and ZSCAN18) were the most significantly differentially expressed in LEAD group compared to controls. Discriminative value of revealed microRNAs and genes were confirmed by receiver operating characteristic analysis. Dysregulations of 26 microRNAs and 14 genes were used to propose novel biomarkers of LEAD. Regulatory interactions between biomarker microRNAs and genes were studied in silico using R multiMiR package. Functional analysis of genes modulated by proposed biomarker microRNAs was performed using DAVID 6.8 tools and revealed terms closely related to atherosclerosis and, interestingly, the processes involving nervous system. The study provides new insight into microRNA-dependent regulatory mechanisms involved in pathology of LEAD. Proposed microRNA and gene biomarkers of LEAD may provide new diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Daniel P Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Karol P Ruszel
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| | - Andrzej Stępniewski
- Ecotech Complex, Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, University of Marie Curie-Sklodowska, Lublin, Poland
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Jacek Bogucki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| | - Łukasz Komsta
- Chair and Department of Medicinal Chemistry, Medical University of Lublin, Lublin, Poland
| | - Przemysław Kołodziej
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Tomasz Zubilewicz
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Marcin Feldo
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
19
|
Alvelo JL, Papademetris X, Mena-Hurtado C, Jeon S, Sumpio BE, Sinusas AJ, Stacy MR. Radiotracer Imaging Allows for Noninvasive Detection and Quantification of Abnormalities in Angiosome Foot Perfusion in Diabetic Patients With Critical Limb Ischemia and Nonhealing Wounds. Circ Cardiovasc Imaging 2019; 11:e006932. [PMID: 29748311 PMCID: PMC5951395 DOI: 10.1161/circimaging.117.006932] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
Abstract
Background: Single photon emission computed tomography (SPECT)/computed tomography (CT) imaging allows for assessment of skeletal muscle microvascular perfusion but has not been quantitatively assessed in angiosomes, or 3-dimensional vascular territories, of the foot. This study assessed and compared resting angiosome foot perfusion between healthy subjects and diabetic patients with critical limb ischemia (CLI). Additionally, the relationship between SPECT/CT imaging and the ankle–brachial index—a standard tool for evaluating peripheral artery disease—was assessed. Methods and Results: Healthy subjects (n=9) and diabetic patients with CLI and nonhealing ulcers (n=42) underwent SPECT/CT perfusion imaging of the feet. CT images were segmented into angiosomes for quantification of relative radiotracer uptake, expressed as standardized uptake values. Standardized uptake values were assessed in ulcerated angiosomes of patients with CLI and compared with whole-foot standardized uptake values in healthy subjects. Serial SPECT/CT imaging was performed to assess uptake kinetics of technetium-99m-tetrofosmin. The relationship between angiosome perfusion and ankle–brachial index was assessed via correlational analysis. Resting perfusion was significantly lower in CLI versus healthy subjects (P=0.0007). Intraclass correlation coefficients of 0.95 (healthy) and 0.93 (CLI) demonstrated excellent agreement between serial perfusion measurements. Correlational analysis, including healthy and CLI subjects, demonstrated a significant relationship between ankle–brachial index and SPECT/CT (P=0.01); however, this relationship was not significant for diabetic CLI patients only (P=0.2). Conclusions: SPECT/CT imaging assesses regional foot perfusion and detects abnormalities in microvascular perfusion that may be undetectable by conventional ankle–brachial index in patients with diabetes mellitus. SPECT/CT may provide a novel approach for evaluating responses to targeted therapies.
Collapse
Affiliation(s)
- Jessica L Alvelo
- Department of Internal Medicine (J.L.A., C.M.-H., B.E.S., A.J.S., M.R.S.)
| | - Xenophon Papademetris
- Department of Radiology and Biomedical Imaging (X.P., B.E.S., A.J.S.).,Department of Biomedical Engineering (X.P.)
| | | | | | - Bauer E Sumpio
- Department of Internal Medicine (J.L.A., C.M.-H., B.E.S., A.J.S., M.R.S.).,Department of Radiology and Biomedical Imaging (X.P., B.E.S., A.J.S.).,and Department of Surgery (B.E.S.) Yale University School of Medicine, New Haven, CT
| | - Albert J Sinusas
- Department of Internal Medicine (J.L.A., C.M.-H., B.E.S., A.J.S., M.R.S.).,Department of Radiology and Biomedical Imaging (X.P., B.E.S., A.J.S.)
| | - Mitchel R Stacy
- Department of Internal Medicine (J.L.A., C.M.-H., B.E.S., A.J.S., M.R.S.)
| |
Collapse
|
20
|
Searching for the Genetic Determinants of Peripheral Arterial Disease: A Review of the Literature and Future Directions. Cardiol Rev 2019; 27:145-152. [PMID: 30946061 DOI: 10.1097/crd.0000000000000231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Peripheral arterial disease (PAD) is a significant but under-recognized disease that is poorly understood despite population-scale genetic studies. To address this morbid disease, clinicians need additional tools to identify, prevent, and treat patients at risk for PAD. Genetic studies of coronary artery disease have yielded promising results for clinical application, which have thus far been lacking in PAD. In this article, we review recent findings, discuss limitations, and propose future directions of genomic study and clinical application. However, despite many studies, we still lack definitive genetic markers for PAD. This can be attributed to the heterogeneity of PAD's pathogenesis and clinical manifestations, as well as inconsistencies in study methodologies, limitations of current genetic assessment techniques, incompletely comprehended molecular pathophysiology, and confounding generalized atherosclerotic risk factors. The goals of this review are to evaluate the limitations of our current genetic knowledge of PAD and to propose approaches to expedite the identification of valuable markers of PAD.
Collapse
|
21
|
Okba AM, Abd El Raouf Raafat M, Nazmy Farres M, Abd El Nour Melek N, Amin MM, Gendy NN. Expanded peripheral CD4 +CD28 null T cells and its association with atherosclerotic changes in patients with end stage renal disease on hemodialysis. Hum Immunol 2019; 80:748-754. [PMID: 30853362 DOI: 10.1016/j.humimm.2019.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
End-stage renal disease (ESRD) patients, including those on hemodialysis, possess a high risk for cardiovascular diseases, as the first leading cause of death among them. Traditional risk factors do not utterly elucidate this. Throughout the last two decades, CD4+CD28null T cells; an unusual subset of T lymphocytes, was detected high with excess cardiovascular (CV) mortality. We aimed to investigate the circulating CD4+CD28null T cells frequency in ESRD patients on hemodialysis and to evaluate their relationship with atherosclerotic changes. High-resolution carotid ultrasonography was done to assess the common carotid artery intima media thickness in a number of ESRD patients, accordingly patients were selected and subdivided into two groups; 30 with atherosclerosis (mean [SD] age, 51.6 [6.3] years) and 30 without (mean [SD] age, 48.9 [5.5] years). Another 30 healthy individuals (mean [SD] age, 48.5 [6.8] years) were enrolled. Analysis of CD4+CD28null T-cells frequency by flow-cytometry was performed in all studied subjects. CD4+CD28null T cell percentage was significantly higher in ESRD patients, (mean [SD], 7.3 [2.7] %) compared to healthy individuals (mean [SD], 3.0 [0.8] %), (p < 0.001). Additionally, the expansion of these unusual T lymphocytes was significantly higher in ESRD patients with atherosclerotic changes (mean [SD], 9.47 [0.75] %) compared to those without atherosclerosis (mean [SD], 5.22 [2.14] %), (p < 0.001). In conclusion circulating CD4+CD28null T lymphocyte population showed expansion in ESRD patients, and of interest in correlation to preclinical atherosclerotic changes.
Collapse
Affiliation(s)
- Ashraf Mahmoud Okba
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | | | - Mohamed Nazmy Farres
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | - Nermine Abd El Nour Melek
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt
| | - Mariam Maged Amin
- Department of Internal Medicine, Clinical Immunology and Allergy, Faculty of Medicine, Ain Shams University, Egypt.
| | - Nelly Nader Gendy
- Department of Internal Medicine and Nephrology, Theodor Bilharz Research Institute, Egypt
| |
Collapse
|
22
|
Hicks CW, Yang C, Ndumele CE, Folsom AR, Heiss G, Black JH, Selvin E, Matsushita K. Associations of Obesity With Incident Hospitalization Related to Peripheral Artery Disease and Critical Limb Ischemia in the ARIC Study. J Am Heart Assoc 2018; 7:e008644. [PMID: 30369315 PMCID: PMC6201405 DOI: 10.1161/jaha.118.008644] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
Background We conducted an analysis of data from the ARIC (Atherosclerosis Risk in Communities) study to assess the independent association of obesity with peripheral artery disease ( PAD ) and critical limb ischemia ( CLI ). Methods and Results All black and white ARIC participants without prevalent PAD at baseline (1987-1989) were included. We used Cox proportional hazards models adjusting for potential confounders and then potential mediators to quantify the association between body mass index ( BMI ) and incident hospitalizations related to PAD without CLI and with CLI through 2013. Our analysis included 13 988 men and women followed for a median of 24 years. Incident PAD without CLI and PAD with CLI occurred in 373 and 201 participants, respectively. After adjusting for potential confounders, higher BMI at baseline was associated with increased risk of PAD without CLI when BMI was modeled continuously (hazard ratio per 1- SD increment in BMI: 1.23; 95% confidence interval, 1.11-1.37) and with PAD with CLI regardless of whether BMI was modeled categorically ( P<0.05) or continuously (hazard ratio per 1- SD increment in BMI: 1.51; 95% confidence interval, 1.34-1.69). The associations of BMI with PAD without CLI and with CLI were attenuated after further accounting for potential mediators but remained significant for PAD with CLI when BMI was linearly modeled (hazard ratio per 1- SD increment in BMI: 1.19; 95% confidence interval, 1.04-1.36). The positive association between BMI and PAD with CLI was stronger than the association between BMI and PAD without CLI for all models ( P<0.001). Conclusions In the general population, BMI is positively associated with incident hospitalized PAD after adjusting for potential confounders, particularly its most severe form of CLI . Maintaining an optimal weight, in addition to controlling other cardiovascular risk factors, may play a role in reducing risk of PAD with CLI .
Collapse
Affiliation(s)
- Caitlin W. Hicks
- Division of Vascular Surgery and Endovascular TherapyJohns Hopkins University School of MedicineBaltimoreMD
| | - Chao Yang
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Chiadi E. Ndumele
- Division of CardiologyJohns Hopkins University School of MedicineBaltimoreMD
| | - Aaron R. Folsom
- Division of Epidemiology & Community HealthUniversity of MinnesotaMinneapolisMN
| | - Gerardo Heiss
- Department of EpidemiologyGillings School of Global Public HealthUniversity of North CarolinaChapel HillNC
| | - James H. Black
- Division of Vascular Surgery and Endovascular TherapyJohns Hopkins University School of MedicineBaltimoreMD
| | - Elizabeth Selvin
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| | - Kunihiro Matsushita
- Department of EpidemiologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMD
| |
Collapse
|
23
|
McGillivray P, Clarke D, Meyerson W, Zhang J, Lee D, Gu M, Kumar S, Zhou H, Gerstein M. Network Analysis as a Grand Unifier in Biomedical Data Science. Annu Rev Biomed Data Sci 2018. [DOI: 10.1146/annurev-biodatasci-080917-013444] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Biomedical data scientists study many types of networks, ranging from those formed by neurons to those created by molecular interactions. People often criticize these networks as uninterpretable diagrams termed hairballs; however, here we show that molecular biological networks can be interpreted in several straightforward ways. First, we can break down a network into smaller components, focusing on individual pathways and modules. Second, we can compute global statistics describing the network as a whole. Third, we can compare networks. These comparisons can be within the same context (e.g., between two gene regulatory networks) or cross-disciplinary (e.g., between regulatory networks and governmental hierarchies). The latter comparisons can transfer a formalism, such as that for Markov chains, from one context to another or relate our intuitions in a familiar setting (e.g., social networks) to the relatively unfamiliar molecular context. Finally, key aspects of molecular networks are dynamics and evolution, i.e., how they evolve over time and how genetic variants affect them. By studying the relationships between variants in networks, we can begin to interpret many common diseases, such as cancer and heart disease.
Collapse
Affiliation(s)
- Patrick McGillivray
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - Declan Clarke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - William Meyerson
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut 06520, USA
| | - Jing Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut 06520, USA
| | - Donghoon Lee
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut 06520, USA
| | - Mengting Gu
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut 06520, USA
- Department of Computer Science, Yale University, New Haven, Connecticut 06520, USA
| | - Sushant Kumar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - Holly Zhou
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
| | - Mark Gerstein
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut 06520, USA
- Department of Computer Science, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
24
|
A Network-Biology Informed Computational Drug Repositioning Strategy to Target Disease Risk Trajectories and Comorbidities of Peripheral Artery Disease. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2018; 2017:108-117. [PMID: 29888052 PMCID: PMC5961807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Currently, drug discovery approaches focus on the design of therapies that alleviate an index symptom by reengineering the underlying biological mechanism in agonistic or antagonistic fashion. For example, medicines are routinely developed to target an essential gene that drives the disease mechanism. Therapeutic overloading where patients get multiple medications to reduce the primary and secondary side effect burden is standard practice. This single-symptom based approach may not be scalable, as we understand that diseases are more connected than random and molecular interactions drive disease comorbidities. In this work, we present a proof-of-concept drug discovery strategy by combining network biology, disease comorbidity estimates, and computational drug repositioning, by targeting the risk factors and comorbidities of peripheral artery disease, a vascular disease associated with high morbidity and mortality. Individualized risk estimation and recommending disease sequelae based therapies may help to lower the mortality and morbidity of peripheral artery disease.
Collapse
|
25
|
Kozarova M, Malachovska Z, Zidzik J, Javorsky M, Demkova K, Habalova V, Tkac I. Risk allele of gene variant rs6584389 is associated with increased intima-media thickness in patients with type 2 diabetes. VASA 2018; 47:279-284. [PMID: 29671694 DOI: 10.1024/0301-1526/a000705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Genome-wide association studies identified several gene variants associated with peripheral arterial disease (PAD). Among them, rs6584389 A>C was significantly associated with PAD defined by decreased ankle-brachial index (ABI). The aim of this study was to investigate whether the rs6584389 variant is also associated with the earlier stages of atherosclerosis assessed by intima-media thickness (IMT) or pulse-wave velocity (PWV) in clinically asymptomatic subjects with type 2 diabetes (T2DM), a group of patients with a high cardiovascular risk. PATIENTS AND METHODS In total, 111 patients with T2DM (56 females, 55 males) with a mean age 63.0 ± 9.1 years were consecutively included in the study. IMT was measured by ultrasound using 7 MHz linear transducer. PWV was measured using a piezoelectric method. Genotyping for rs6584389 was performed by PCR-HRMA method. RESULTS The carriers of the risk C-allele of rs6584389 variant had significantly higher mean left-side IMT (AA: 0.67 ± 0.12, AC 0.77 ± 0.21, CC 0.78 ± 0.22 mm; p = 0.04). In multiple linear regression analysis, rs6586389 genotype was significantly associated with all measured IMT parameters. The presence of each risk C-allele predicted an increase in left-side IMT by 0.056 mm (p = 0.017), right-side IMT by 0.053 mm (p = 0.039), average IMT by 0.054 mm (p = 0.023), and maximal IMT by 0.058 mm (p = 0.021). Age and HbA1c levels were also significantly associated with increased IMT in all multivariate models. CONCLUSIONS Gene variant rs6584389 A>C near to PAX2 gene was associated with increased carotid IMT in patients with type 2 diabetes independently of the other main risk factors for atherosclerosis.
Collapse
Affiliation(s)
- Miriam Kozarova
- 1 Safarik University in Kosice, Faculty of Medicine, Department of Internal Medicine 4, Košice, Slovakia.,a These authors contributed equally to this paper
| | - Zuzana Malachovska
- 1 Safarik University in Kosice, Faculty of Medicine, Department of Internal Medicine 4, Košice, Slovakia.,a These authors contributed equally to this paper
| | - Jozef Zidzik
- 2 Safarik University in Kosice, Faculty of Medicine, Department of Medical Biology, Košice, Slovakia
| | - Martin Javorsky
- 1 Safarik University in Kosice, Faculty of Medicine, Department of Internal Medicine 4, Košice, Slovakia
| | - Katarina Demkova
- 1 Safarik University in Kosice, Faculty of Medicine, Department of Internal Medicine 4, Košice, Slovakia
| | - Viera Habalova
- 2 Safarik University in Kosice, Faculty of Medicine, Department of Medical Biology, Košice, Slovakia
| | - Ivan Tkac
- 1 Safarik University in Kosice, Faculty of Medicine, Department of Internal Medicine 4, Košice, Slovakia
| |
Collapse
|
26
|
Posadas-Sánchez R, Angeles-Martínez J, Pérez-Hernández N, Rodríguez-Pérez JM, López-Bautista F, Flores-Dominguez C, Fragoso JM, Posadas-Romero C, Vargas-Alarcón G. The IL-10-1082 (rs1800896) G allele is associated with a decreased risk of developing premature coronary artery disease and some IL-10 polymorphisms were associated with clinical and metabolic parameters. The GEA study. Cytokine 2018. [PMID: 29525679 DOI: 10.1016/j.cyto.2018.02.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Interleukin 10 (IL-10) is an anti-inflammatory cytokine with a protective role in the formation and the development of the atherosclerotic plaque. The aim of the present study was to establish if IL-10 gene polymorphisms are associated with the development of premature coronary artery disease (pCAD) and cardiovascular risk factors in Mexican individuals. Three IL-10 gene polymorphisms [-592C/A (rs1800872), -819C/T (rs1800871), and -1082 A/G (rs1800896)] and IL-10 plasma levels were analyzed in 2266 individuals (1160 pCAD patients and 1106 healthy controls). Under recessive and co-dominant2 models, the -1082 A/G (rs1800896) G allele was associated with decreased risk of developing pCAD (OR = 0.572, Prec = 0.022 and OR = 0.567, Pcod2 = 0.023). In pCAD patients, the polymorphisms were associated with hyperinsulinemia, small and dense LDLs, hypertension, and diabetes mellitus. In the control group, the polymorphisms were associated with hypertension, hyperuricemia, and small and dense LDLs. pCAD patients have significantly higher IL-10 plasma levels than healthy controls [0.91 (0.55-1.67) pg/mL vs 0.45 (0.24-0.98) pg/mL, respectively, P < 0.0001]. Nevertheless, these levels were not associated with the genotypes analyzed in the present study. The results suggest that the IL-10-1082 A/G (rs1800896) G allele is associated with a decreased risk of developing pCAD. In patients and controls, the polymorphisms analyzed were associated with some cardiovascular risk factors. Although, in pCAD patients the IL-10 plasma levels were higher, they were not associated with the genotypes of the polymorphisms examined.
Collapse
Affiliation(s)
| | - Javier Angeles-Martínez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Nonanzit Pérez-Hernández
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | | | - Fabiola López-Bautista
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Carmina Flores-Dominguez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Carlos Posadas-Romero
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|
27
|
Shu L, Arneson D, Yang X. Bioinformatics Principles for Deciphering Cardiovascular Diseases. ENCYCLOPEDIA OF CARDIOVASCULAR RESEARCH AND MEDICINE 2018:273-292. [DOI: 10.1016/b978-0-12-809657-4.99576-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
28
|
Okeke E, Dokun AO. Role of genetics in peripheral arterial disease outcomes; significance of limb-salvage quantitative locus-1 genes. Exp Biol Med (Maywood) 2017; 243:190-197. [PMID: 29199462 DOI: 10.1177/1535370217743460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Peripheral artery disease is a major health care problem with significant morbidity and mortality. Humans with peripheral artery disease exhibit two major and differential clinical manifestations - intermittent claudication and critical limb ischemia. Individuals with intermittent claudication or critical limb ischemia have overlapping risk factors and objective measures of blood flow. Hence, we hypothesized that variation in genetic make-up may be an important determinant in the severity of peripheral artery disease. Previous studies have identified polymorphism in genes, contributing to extent of atherosclerosis but much less is known about polymorphisms associated with genes that can influence peripheral artery disease severity. This review outlines some of the progress made up-to-date to unravel the molecular mechanisms underlining differential peripheral artery disease severity. By exploring the recovery phenotype of different mouse strains following experimental peripheral artery disease, our group identified the limb salvage-associated quantitative trait locus 1 on mouse chromosome 7 as the first genetic modifier of perfusion recovery and tissue necrosis phenotypes. Furthermore, a number of genes within LSq-1, such as ADAM12, IL-21Rα, and BAG3 were identified as genetic modifiers of peripheral artery disease severity that function through preservation of endothelial and skeletal muscle cells during ischemia. Taken together, these studies suggest manipulation of limb salvage-associated quantitative trait locus 1 genes show great promise as therapeutic targets in the management of peripheral artery disease. Impact statement Peripheral artery disease (PAD) is a major health care problem with significant morbidity and mortality. Individuals with similar atherosclerosis burden do display different severity of disease. This review outlines some of the progress made up-to-date in unraveling the molecular mechanisms underlining differential PAD severity with a focus on the role of the Limb Salvage-associated Quantitative trait locus 1 (LSq-1), a key locus in adaptation to ischemia in PAD.
Collapse
Affiliation(s)
- Emmanuel Okeke
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, 12325 University of Tennessee Health Sciences Center , Memphis, TN 38163, USA
| | - Ayotunde O Dokun
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, 12325 University of Tennessee Health Sciences Center , Memphis, TN 38163, USA
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW In this paper, we review the progress made thus far in research related to the genetics of peripheral arterial disease (PAD) by detailing efforts to date in heritability, linkage analyses, and candidate gene studies. We further summarize more contemporary genome-wide association studies (GWAS) and epigenetic studies of PAD. Finally, we review current challenges and future avenues of advanced research in PAD genetics including whole genome sequencing studies. RECENT FINDINGS Studies have estimated the heritability of PAD to be moderate, though the contribution to this heritability that is independent of traditional cardiovascular risk factors remains unclear. Recent efforts have identified SNPs associated with PAD in GWAS analyses, but these have yet to be replicated in independent studies. Much remains to be discovered in the field of PAD genetics. An improved understanding of the genetic foundation for PAD will allow for earlier diagnosis of disease and a more complete pathophysiological understanding of the mechanisms of the disease leading to novel therapeutic interventions. Future avenues for success will likely arise from very large-scale GWAS, whole genome sequencing, and epigenetic studies involving very well-characterized cohorts.
Collapse
Affiliation(s)
- Nathan Belkin
- Division of Vascular and Endovascular Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA, 19104, USA
| | - Scott M Damrauer
- Division of Vascular and Endovascular Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 4 Maloney, Philadelphia, PA, 19104, USA. .,Department of Surgery, Corporal Michael Crescenz VA Medical Center, 3900 Woodland Ave., Philadelphia, PA, 19104, USA.
| |
Collapse
|
30
|
Posadas-Sánchez R, Pérez-Hernández N, Rodríguez-Pérez JM, Coral-Vázquez RM, Roque-Ramírez B, Llorente L, Lima G, Flores-Dominguez C, Villarreal-Molina T, Posadas-Romero C, Vargas-Alarcón G. Interleukin-27 polymorphisms are associated with premature coronary artery disease and metabolic parameters in the Mexican population: the genetics of atherosclerotic disease (GEA) Mexican study. Oncotarget 2017; 8:64459-64470. [PMID: 28969085 PMCID: PMC5610017 DOI: 10.18632/oncotarget.16223] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/03/2017] [Indexed: 11/25/2022] Open
Abstract
Several studies suggest an important role of Interleukin-27 in the development of atherosclerosis. The aim of this study was to establish whether the IL-27p28 gene polymorphisms are associated with premature coronary artery disease and/or other cardiovascular risk factors. Four IL-27p28 gene polymorphisms were selected and genotyped in 1162 premature coronary artery disease cases and 1107 controls. rs26528 T and rs40837 A alleles were significantly associated with a lower risk of premature coronary artery disease under different inheritance models (Pdominant = 0.046; Pover-dominant = 0.002; Pco-dominant1 = 0.007 for rs26528T; Pover-dominant = 0.008 and Pco-dominant1 = 0.031 for rs40837). The rs40837 A allele was also associated with a lower risk of insulin resistance, in cases (Pover-dominant = 0.037) and controls (Padditive = 0.008; Pdominant = 0.047; Precessive = 0.014; Pco-dominant2 = 0.006), while the rs26528 T allele was associated with a lower risk of insulin resistance only in the control group (Precessive = 0.016; Pco-dominant2 = 0.021). Interleukin-27 plasma levels were measured in 450 controls and 450 cases, and were significantly higher in cases compared to controls (P = 0.004). However, Interleukin-27 plasma levels were not associated with IL-27p28 polymorphisms. Luciferase assays showed that co-transfection of the rs40837 A allele and miR-379-5p significantly decreased luciferase gene expression. Our study shows for the first time, that IL-27p28 gene polymorphisms are associated with premature coronary artery disease and with some metabolic parameters. The rs40837 A allele in presence of miR-379-5p significantly decreased luciferase gene expression.
Collapse
Affiliation(s)
- Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico D.F., México
| | - Nonanzit Pérez-Hernández
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico D.F., México
| | | | - Ramón M. Coral-Vázquez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico D.F., México
| | | | - Luis Llorente
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico D.F., México
| | - Guadalupe Lima
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico D.F., México
| | | | - Teresa Villarreal-Molina
- Laboratorio de Genómica Cardiovascular, Instituto Nacional de Medicina Genómica, Mexico D.F., México
| | - Carlos Posadas-Romero
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico D.F., México
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico D.F., México
| |
Collapse
|
31
|
McClung JM, McCord TJ, Ryan TE, Schmidt CA, Green TD, Southerland KW, Reinardy JL, Mueller SB, Venkatraman TN, Lascola CD, Keum S, Marchuk DA, Spangenburg EE, Dokun A, Annex BH, Kontos CD. BAG3 (Bcl-2-Associated Athanogene-3) Coding Variant in Mice Determines Susceptibility to Ischemic Limb Muscle Myopathy by Directing Autophagy. Circulation 2017; 136:281-296. [PMID: 28442482 PMCID: PMC5537727 DOI: 10.1161/circulationaha.116.024873] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Critical limb ischemia is a manifestation of peripheral artery disease that carries significant mortality and morbidity risk in humans, although its genetic determinants remain largely unknown. We previously discovered 2 overlapping quantitative trait loci in mice, Lsq-1 and Civq-1, that affected limb muscle survival and stroke volume after femoral artery or middle cerebral artery ligation, respectively. Here, we report that a Bag3 variant (Ile81Met) segregates with tissue protection from hind-limb ischemia. METHODS We treated mice with either adeno-associated viruses encoding a control (green fluorescent protein) or 2 BAG3 (Bcl-2-associated athanogene-3) variants, namely Met81 or Ile81, and subjected the mice to hind-limb ischemia. RESULTS We found that the BAG3 Ile81Met variant in the C57BL/6 (BL6) mouse background segregates with protection from tissue necrosis in a shorter congenic fragment of Lsq-1 (C.B6-Lsq1-3). BALB/c mice treated with adeno-associated virus encoding the BL6 BAG3 variant (Ile81; n=25) displayed reduced limb-tissue necrosis and increased limb tissue perfusion compared with Met81- (n=25) or green fluorescent protein- (n=29) expressing animals. BAG3Ile81, but not BAG3Met81, improved ischemic muscle myopathy and muscle precursor cell differentiation and improved muscle regeneration in a separate, toxin-induced model of injury. Systemic injection of adeno-associated virus-BAG3Ile81 (n=9), but not BAG3Met81 (n=10) or green fluorescent protein (n=5), improved ischemic limb blood flow and limb muscle histology and restored muscle function (force production). Compared with BAG3Met81, BAG3Ile81 displayed improved binding to the small heat shock protein (HspB8) in ischemic skeletal muscle cells and enhanced ischemic muscle autophagic flux. CONCLUSIONS Taken together, our data demonstrate that genetic variation in BAG3 plays an important role in the prevention of ischemic tissue necrosis. These results highlight a pathway that preserves tissue survival and muscle function in the setting of ischemia.
Collapse
Affiliation(s)
- Joseph M McClung
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville.
| | - Timothy J McCord
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Terence E Ryan
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Cameron A Schmidt
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Tom D Green
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Kevin W Southerland
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Jessica L Reinardy
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sarah B Mueller
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Talaignair N Venkatraman
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Lascola
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Sehoon Keum
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Douglas A Marchuk
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Espen E Spangenburg
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Ayotunde Dokun
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Brian H Annex
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| | - Christopher D Kontos
- From Department of Physiology and Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC (J.M.M., T.E.R., C.A.S., T.D.G., E.E.S); Department of Medicine, Division of Cardiology (T.J.M., J.L.R., S.B.M., C.D.K.), Department of Surgery, Division of General Surgery (K.W.S.), Department of Pharmacology and Cancer Biology (J.L.R., S.B.M., C.D.K.), Department of Radiology (T.N.V., C.D.L.), and Department of Molecular Genetics and Microbiology (S.K., D.A.M.), Duke University Medical Center, Durham, NC; and Department of Medicine, Division of Endocrinology (A.D., B.H.A.), Division of Cardiovascular Medicine (B.H.A.), and Robert M. Berne Cardiovascular Research Center (B.H.A.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
32
|
Schwindt AG, Bennett JG, Crowder WH, Dohad S, Janzer SF, George JC, Tedder B, Davis TP, Cawich IM, Gammon RS, Muck PE, Pigott JP, Dishmon DA, Lopez LA, Golzar JA, Chamberlin JR, Moulton MJ, Zakir RM, Kaki AK, Fishbein GJ, McDaniel HB, Hezi-Yamit A, Simpson JB, Desai A. Lower Extremity Revascularization Using Optical Coherence Tomography-Guided Directional Atherectomy: Final Results of the EValuatIon of the PantheriS OptIcal COherence Tomography ImagiNg Atherectomy System for Use in the Peripheral Vasculature (VISION) Study. J Endovasc Ther 2017; 24:355-366. [PMID: 28393673 DOI: 10.1177/1526602817701720] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE To evaluate the safety and efficacy of a novel optical coherence tomography (OCT)-guided atherectomy catheter in treating patients with symptomatic femoropopliteal disease. METHODS The VISION trial ( ClinicalTrials.gov identifier NCT01937351) was a single-arm, multicenter, global investigational device exemption study enrolling 158 subjects (mean age 67.2±10.5 years; 87 men) across 20 participating sites. In this cohort, 198 lesions were treated with an average length of 53±40 mm using the Pantheris catheter alone or Pantheris + adjunctive therapy. The primary safety endpoint was the composite of major adverse events (MAEs) through 6 months (objective performance goal 43.2%). Technical success (primary efficacy outcome) was defined as the percent of target lesions with a residual diameter stenosis ≤50% after treatment with the Pantheris device alone (objective performance goal 87.0%). Procedural success was defined as reduction in stenosis to ≤30% after Pantheris ± adjunctive therapy. Tissue specimens retrieved from each treated lesion were histologically analyzed to evaluate the accuracy and precision of OCT image guidance. RESULTS The primary efficacy outcome was achieved in 192 (97.0%) of the 198 lesions treated with the Pantheris catheter. Across all lesions, mean diameter stenosis was reduced from 78.7%±15.1% at baseline to 30.3%±11.8% after Pantheris alone (p<0.001) and to 22.4%±9.9% after Pantheris ± adjunctive therapy (p<0.001). Of the 198 target lesions, 104 (52.5%) were treated with the Pantheris alone, 84 (42.4%) were treated with Pantheris + adjunctive angioplasty, and 10 (5.1%) with Pantheris + angioplasty + stenting. The composite MAE outcome through 6 months occurred in 25 (16.6%) of 151 subjects. There were no clinically significant perforations, 1 (0.5%) catheter-related dissection, 4 (2%) embolic events, and a 6.4% clinically driven target lesion revascularization rate at 6 months. The 40-lesion chronic total occlusion (CTO) subset (mean lesion length 82±38 mm) achieved a similar significant reduction in stenosis to 35.5%±13.6% after Pantheris alone (p<0.001). Histological analysis of atherectomy specimens confirmed <1% adventitia in 82.1% of the samples, highlighting the precision of OCT guidance. Characterization of the OCT-guided lesions revealed evidence of an underestimation of disease burden when using fluoroscopy. CONCLUSION OCT-guided atherectomy for femoropopliteal disease is safe and effective. Additionally, the precision afforded by OCT guidance leads to greater removal of plaque during atherectomy while sparing the adventitia.
Collapse
Affiliation(s)
- Arne G Schwindt
- 1 Department of Vascular Surgery, St Franziskus-Hospital Münster, Germany
| | - J Gray Bennett
- 2 St Dominic-Jackson Memorial Hospital, Jackson, MI, USA
| | | | - Suhail Dohad
- 3 Cedars Sinai Medical Center, West Hollywood, CA, USA
| | | | - Jon C George
- 5 Deborah Heart and Lung Center, Brown Mills, NJ, USA
| | - Barry Tedder
- 6 St Bernards Medical Center, Jonesboro, AR, USA
| | | | | | | | | | - John P Pigott
- 11 Jobst Vascular Institute Promedica Toledo Hospital, Toledo, OH, USA
| | | | - Lou A Lopez
- 13 St Joseph's Hospital, Fort Wayne, IN, USA
| | - Jaafer A Golzar
- 14 Advocate Christ Hospital and Medical Center, Oak Lawn, IL, USA
| | | | | | - Ramzan M Zakir
- 17 St Peters University Hospital, New Brunswick, NJ, USA
| | - Amir K Kaki
- 18 DMC Cardiovascular Institute Harper-Hutzel Hospital, Detroit, MI, USA
| | | | | | | | - John B Simpson
- 21 Avinger Inc, Redwood City, CA, USA.,22 Sequoia Hospital, Redwood City, CA, USA
| | - Arjun Desai
- 21 Avinger Inc, Redwood City, CA, USA.,23 Stanford University Medical Center, Stanford, CA, USA
| |
Collapse
|
33
|
ANNEX BRIANH, BELLER GEORGEA. TOWARDS THE DEVELOPMENT OF NOVEL THERAPEUTICS FOR PERIPHERAL ARTERY DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2016; 127:224-234. [PMID: 28066055 PMCID: PMC5216482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the Western world. Peripheral artery disease (PAD) has been less studied then coronary artery disease but is nearly as common. PAD impairs blood flow to the leg(s) and causes functional impairment, leg pain, and amputation. The last drug approved for PAD was in 1999. Blood flow to leg proceeds through one major artery and in PAD total occlusions in the course of that vessel are common. Thus, the extent of new blood vessel growth determines a patients' clinical course. Promoting the growth of new blood vessels (therapeutic angiogenesis) was a major goal of therapy. Results from studies using cytokine growth factors have shown disappointing results. Using clinical and preclinical studies, our laboratory has identified several novel therapeutic approaches. One, a modulator of innate immunity, will be reviewed as an approach that has the potential to create new therapies for PAD.
Collapse
Affiliation(s)
- BRIAN H. ANNEX
- Correspondence and reprint requests: Brian H. Annex, MD,
Department of Medicine, University of Virginia, 1215 Lee Street, PO Box 800158, Charlottesville, VA 22908434-982-0853434-982-1998
| | | |
Collapse
|
34
|
Ryan TE, Schmidt CA, Green TD, Brown DA, Neufer PD, McClung JM. Mitochondrial Regulation of the Muscle Microenvironment in Critical Limb Ischemia. Front Physiol 2015; 6:336. [PMID: 26635622 PMCID: PMC4649016 DOI: 10.3389/fphys.2015.00336] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/02/2015] [Indexed: 01/11/2023] Open
Abstract
Critical limb ischemia (CLI) is the most severe clinical presentation of peripheral arterial disease and manifests as chronic limb pain at rest and/or tissue necrosis. Current clinical interventions are largely ineffective and therapeutic angiogenesis based trials have shown little efficacy, highlighting the dire need for new ideas and novel therapeutic approaches. Despite a decade of research related to skeletal muscle as a determinant of morbidity and mortality outcomes in CLI, very little progress has been made toward an effective therapy aimed directly at the muscle myopathies of this disease. Within the muscle cell, mitochondria are well positioned to modulate the ischemic cellular response, as they are the principal sites of cellular energy production and the major regulators of cellular redox charge and cell death. In this mini review, we update the crucial importance of skeletal muscle to CLI pathology and examine the evolving influence of muscle and endothelial cell mitochondria in the complex ischemic microenvironment. Finally, we discuss the novelty of muscle mitochondria as a therapeutic target for ischemic pathology in the context of the complex co-morbidities often associated with CLI.
Collapse
Affiliation(s)
- Terence E Ryan
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Cameron A Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Tom D Green
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - David A Brown
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina University Greenville, NC, USA ; East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University Greenville, NC, USA
| |
Collapse
|
35
|
Genome-Wide Association Study of Peripheral Arterial Disease in a Japanese Population. PLoS One 2015; 10:e0139262. [PMID: 26488411 PMCID: PMC4619060 DOI: 10.1371/journal.pone.0139262] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Characteristics of peripheral arterial disease (PAD) are the occlusion or stenosis of multiple vessel sites caused mainly by atherosclerosis and chronic lower limb ischemia. To identify PAD susceptible loci, we conducted a genome-wide association study (GWAS) with 785 cases and 3,383 controls in a Japanese population using 431,666 single nucleotide polymorphisms (SNP). After staged analyses including a total of 3,164 cases and 20,134 controls, we identified 3 novel PAD susceptibility loci at IPO5/RAP2A, EDNRA and HDAC9 with genome wide significance (combined P = 6.8 x 10−14, 5.3 x 10−9 and 8.8 x 10−8, respectively). Fine-mapping at the IPO5/RAP2A locus revealed that rs9584669 conferred risk of PAD. Luciferase assay showed that the risk allele at this locus reduced expression levels of IPO5. To our knowledge, these are the first genetic risk factors for PAD.
Collapse
|
36
|
Kullo IJ, Leeper NJ. The genetic basis of peripheral arterial disease: current knowledge, challenges, and future directions. Circ Res 2015; 116:1551-60. [PMID: 25908728 DOI: 10.1161/circresaha.116.303518] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Several risk factors for atherosclerotic peripheral arterial disease (PAD), such as dyslipidemia, diabetes mellitus, and hypertension, are heritable. However, predisposition to PAD may be influenced by genetic variants acting independently of these risk factors. Identification of such genetic variants will provide insights into underlying pathophysiologic mechanisms and facilitate the development of novel diagnostic and therapeutic approaches. In contrast to coronary heart disease, relatively few genetic variants that influence susceptibility to PAD have been discovered. This may be, in part, because of greater clinical and genetic heterogeneity in PAD. In this review, we (1) provide an update on the current state of knowledge about the genetic basis of PAD, including results of family studies and candidate gene, linkage as well as genome-wide association studies; (2) highlight the challenges in investigating the genetic basis of PAD and possible strategies to overcome these challenges; and (3) discuss the potential of genome sequencing, RNA sequencing, differential gene expression, epigenetic profiling, and systems biology in increasing our understanding of the molecular genetics of PAD.
Collapse
Affiliation(s)
- Iftikhar J Kullo
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (I.J.K.); and Department of Vascular Surgery, Stanford, Stanford, CA (N.J.L.).
| | - Nicholas J Leeper
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (I.J.K.); and Department of Vascular Surgery, Stanford, Stanford, CA (N.J.L.)
| |
Collapse
|
37
|
Wang T, Cunningham A, Dokun AO, Hazarika S, Houston K, Chen L, Lye RJ, Spolski R, Leonard WJ, Annex BH. Loss of interleukin-21 receptor activation in hypoxic endothelial cells impairs perfusion recovery after hindlimb ischemia. Arterioscler Thromb Vasc Biol 2015; 35:1218-25. [PMID: 25838422 PMCID: PMC4865891 DOI: 10.1161/atvbaha.115.305476] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/10/2015] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Surgical hindlimb ischemia (HLI) in mice has become a valuable preclinical model to study peripheral arterial disease. We previously identified that the different phenotypic outcomes after HLI across inbred mouse strains is related to a region on the short arm of mouse chromosome 7. The gene coding the interleukin-21 receptor (IL-21R) lies at the peak of association in this region. APPROACH AND RESULTS With quantitative real-time polymerase chain reaction, we found that a mouse strain with a greater ability to upregulate IL-21R after HLI had better perfusion recovery than a strain with no upregulation after HLI. Immunofluorescent staining of ischemic hindlimb tissue showed IL-21R expression on endothelial cells (ECs) from C57BL/6 mice. An EC-enriched fraction isolated from ischemic hindlimb muscle showed higher Il-21R levels than an EC-enriched fraction from nonischemic limbs. In vitro, human umbilical vein ECs showed elevated IL-21R expression after hypoxia and serum starvation. Under these conditions, IL-21 treatment increased cell viability, decreased cell apoptosis, and augmented tube formation. In vivo, either knockout Il21r or blocking IL-21 signaling by treating with IL-21R-Fc (fusion protein that blocks IL-21 binding to its receptor) in C57BL/6 mice resulted in less perfusion recovery after HLI. Both in vitro and in vivo modulation of the IL-21/IL-21R axis under hypoxic conditions resulted in increased signal transducer and activator of transcription 3 phosphorylation and a subsequent increase in the B-cell lymphoma leukemia-2/BCL-2-associated X protein ratio. CONCLUSION Our data indicate that IL-21R upregulation and ligand activation in hypoxic ECs may help perfusion recovery by limiting/preventing apoptosis and favoring cell survival and angiogenesis through the signal transducer and activator of transcription 3 pathway.
Collapse
Affiliation(s)
- Tao Wang
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Alexis Cunningham
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Ayotunde O Dokun
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Surovi Hazarika
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Kevin Houston
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Lingdan Chen
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - R John Lye
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Rosanne Spolski
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Warren J Leonard
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.)
| | - Brian H Annex
- From the Robert M. Berne Cardiovascular Research Center (T.W., A.C., A.O.D., S.H., K.H., L.C., R.J.L., B.H.A.) and Division of Cardiovascular Medicine, Department of Medicine (S.H., B.H.A.), University of Virginia, Charlottesville; and Laboratory of Molecular Immunology and the Immunology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD (R.S., W.J.L.).
| |
Collapse
|
38
|
Chen YW, Wang YY, Zhao D, Yu CG, Xin Z, Cao X, Shi J, Yang GR, Yuan MX, Yang JK. High prevalence of lower extremity peripheral artery disease in type 2 diabetes patients with proliferative diabetic retinopathy. PLoS One 2015; 10:e0122022. [PMID: 25822410 PMCID: PMC4379174 DOI: 10.1371/journal.pone.0122022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/06/2015] [Indexed: 11/30/2022] Open
Abstract
Little is known about the relationship between lower extremity peripheral arterial disease (PAD) and proliferative diabetic retinopathy (PDR) in type 2 diabetes (T2D). Here, we explored the relationship between sight-threatening PDR and PAD. We screened for diabetic retinopathy (DR) and PAD in hospitalized patients with T2D. Patients with a diabetic duration of more than 10 years, HbA1c ≥7.5%, eGFR ≥60mL/min/1.73m2 and with PDR or with no diabetic retinopathy (NDR) were eligible for this cross-sectional study. Severities of DR were graded by digital retinal photographs according to the Early Treatment Diabetic Retinopathy Study (ETDRS) scale. We assessed PAD by measuring Ankle Brachial Index (ABI), Toe Brachial Index (TBI) and Doppler ultrasound. Statistical analyses were performed using SPSS 17.0 software. Of the 1544 patients, 169 patients with extreme eye (57 PDR and 112 NDR) phenotypes met the inclusion criteria. Patients with PDR had a significantly higher proportion of low ABI (≤0.99) and high ABI (≥1.3) than patients with NDR (28.1% and 15.8% vs. 14.3% and 6.2% respectively, P<0.05). PDR patients also had lower TBI than NDR patients (0.56±0.09 vs. 0.61±0.08, P<0.01). The proportion of patients with abnormal duplex ultrasound was higher in PDR than in NDR (21.1% vs. 9.8%, P<0.001). This showed that PDR associated with PAD could be defined in multiple ways: abnormal ABI (≤0.9) (OR = 3.61, 95% CI: 1.15–11.26), abnormal TBI (OR = 2.84, 95% CI: 1.19–6.64), abnormal duplex (OR = 3.28, 95% CI: 1.00–10.71), and critical limb ischemia (OR = 5.52, 95% CI: 2.14–14.26). Moreover, PDR was a stronger independent correlation factor for PAD than a diabetic duration of 10 years. In conclusion, PAD is more common in PDR than in NDR. It implies that PDR and PAD are mostly concomitant in T2D. We should focus on screening PAD in patients with PDR in clinical practice.
Collapse
Affiliation(s)
- Yi-Wen Chen
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Geriatrics, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ying-Ying Wang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Dong Zhao
- Department of Endocrinology, Beijing Tongzhou District Luhe Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Cai-Guo Yu
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Endocrinology, Beijing Tongzhou District Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhong Xin
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Xi Cao
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jing Shi
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Guang-Ran Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Ming-Xia Yuan
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Diabetes Research and Care, Beijing, China
- * E-mail:
| |
Collapse
|
39
|
Armingohar Z, Jørgensen JJ, Kristoffersen AK, Schenck K, Dembic Z. Polymorphisms in the interleukin-10 gene and chronic periodontitis in patients with atherosclerotic and aortic aneurysmal vascular diseases. J Oral Microbiol 2015; 7:26051. [PMID: 25700628 PMCID: PMC4336353 DOI: 10.3402/jom.v7.26051] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/22/2014] [Accepted: 01/13/2015] [Indexed: 12/18/2022] Open
Abstract
Background Chronic periodontitis (CP), atherosclerotic and aortic aneurysmal vascular diseases (VD) are chronic inflammatory conditions with multifactorial etiologies, including involvement of predisposing genetic factors. In a previous study, polymorphisms in the gene for the anti-inflammatory interleukin-1 receptor antagonist were associated with CP in patients with VD. Objective This study investigates whether polymorphisms in the gene for the anti-inflammatory interleukin-10 (IL10) could be related to CP in the same manner. Methods Seventy-two patients with VD of whom 35 had CP were genotyped for single nucleotide polymorphisms (SNPs) in the IL10 −592 (rs1800872), −819 (rs1800871), and −1,082 (rs1800896) gene by Taqman rtPCR method and by DNA sequencing. Results The C alleles and C/C genotypes of IL10 −592 and IL10 −819 frequencies were significantly higher, while the frequencies of the IL10 −592 (C/A) and IL10 −819 (C/T) heterozygote genotypes were significantly lower in the VD group with CP compared to those without CP. The IL10 haplotype ATA frequency (−1,082, −819, −592) showed a trend to a significant difference between the two groups indicating protection against CP. Conclusions Taken together, our findings suggest an independent association of genetic polymorphisms in the IL-10 gene locus with CP in patients with VD. Development of CP and the implications on vascular disease emphasize the importance of early detection and adequate treatment of periodontitis among these patients.
Collapse
Affiliation(s)
- Zahra Armingohar
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway;
| | | | - Anne K Kristoffersen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Karl Schenck
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Zlatko Dembic
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
40
|
Di Minno G, Spadarella G, Cafaro G, Petitto M, Lupoli R, Di Minno A, de Gaetano G, Tremoli E. Systematic reviews and meta-analyses for more profitable strategies in peripheral artery disease. Ann Med 2014; 46:475-89. [PMID: 25045928 PMCID: PMC4245179 DOI: 10.3109/07853890.2014.932618] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the peripheral arteries, a thrombus superimposed on atherosclerosis contributes to the progression of peripheral artery disease (PAD), producing intermittent claudication (IC), ischemic necrosis, and, potentially, loss of the limb. PAD with IC is often undiagnosed and, in turn, undertreated. The low percentage of diagnosis (∼30%) in this setting of PAD is of particular concern because of the potential worsening of PAD (amputation) and the high risk of adverse vascular outcomes (vascular death, coronary artery disease, stroke). A Medline literature search of the highest-quality systematic reviews and meta-analyses of randomized controlled trials documents that, due to risk of bias, imprecision, and indirectness, the overall quality of the evidence concerning diagnostic tools and antithrombotic interventions in PAD is generally low. Areas of research emerge from the information collected. Appropriate treatments for PAD patients will only derive from ad-hoc studies. Innovative imaging techniques are needed to identify PAD subjects at the highest vascular risk. Whether IC unresponsive to physical exercise and smoking cessation identifies those with a heritable predisposition to more severe vascular events deserves to be addressed. Devising ways to improve prevention of vascular events in patients with PAD implies a co-ordinated approach in vascular medicine.
Collapse
Affiliation(s)
- Giovanni Di Minno
- Department of Clinical Mediine and Surgery, Università degli Studi di Napoli , Naples , Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kullo IJ, Shameer K, Jouni H, Lesnick TG, Pathak J, Chute CG, de Andrade M. The ATXN2-SH2B3 locus is associated with peripheral arterial disease: an electronic medical record-based genome-wide association study. Front Genet 2014; 5:166. [PMID: 25009551 PMCID: PMC4070196 DOI: 10.3389/fgene.2014.00166] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/19/2014] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES In contrast to coronary heart disease (CHD), genetic variants that influence susceptibility to peripheral arterial disease (PAD) remain largely unknown. BACKGROUND We performed a two-stage genomic association study leveraging an electronic medical record (EMR) linked-biorepository to identify genetic variants that mediate susceptibility to PAD. METHODS PAD was defined as a resting/post-exercise ankle-brachial index (ABI) ≤0.9 or ≥1.4 and/or history of lower extremity revascularization. Controls were patients without history of PAD. In Stage I we performed a genome-wide association analysis adjusting for age and sex, of 537, 872 SNPs in 1641 PAD cases (66 ± 11 years, 64% men) and 1604 control subjects (61 ± 7 year, 60% men) of European ancestry. In Stage II we genotyped the top 48 SNPs that were associated with PAD in Stage I, in a replication cohort of 740 PAD cases (70 ± 11 year, 63% men) and 1051 controls (70 ± 12 year, 61% men). RESULTS The SNP rs653178 in the ATXN2-SH2B3 locus was significantly associated with PAD in the discovery cohort (OR = 1.23; P = 5.59 × 10(-5)), in the replication cohort (OR = 1.22; 8.9 × 10(-4)) and in the combined cohort (OR = 1.22; P = 6.46 × 10(-7)). In the combined cohort this SNP remained associated with PAD after additional adjustment for cardiovascular risk factors including smoking (OR = 1.22; P = 2.15 × 10(-6)) and after excluding patients with ABI > 1.4 (OR = 1.24; P = 3.98 × 10(-7)). The SNP is in near-complete linkage disequilibrium (LD) (r (2) = 0.99) with a missense SNP (rs3184504) in SH2B3, a gene encoding an adapter protein that plays a key role in immune and inflammatory response pathways and vascular homeostasis. The SNP has pleiotropic effects and has been previously associated with multiple phenotypes including myocardial infarction. CONCLUSIONS Our findings suggest that the ATXN2-SH2B3 locus influences susceptibility to PAD.
Collapse
Affiliation(s)
- Iftikhar J Kullo
- Division of Cardiovascular Diseases, Mayo Clinic Rochester, MN, USA
| | - Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic Rochester, MN, USA
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic Rochester, MN, USA
| | - Timothy G Lesnick
- Biomedical Statistics and Informatics, Health-Related Sciences, Mayo Clinic Rochester, MN, USA
| | - Jyotishman Pathak
- Biomedical Statistics and Informatics, Health-Related Sciences, Mayo Clinic Rochester, MN, USA
| | - Christopher G Chute
- Biomedical Statistics and Informatics, Health-Related Sciences, Mayo Clinic Rochester, MN, USA
| | - Mariza de Andrade
- Biomedical Statistics and Informatics, Health-Related Sciences, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
42
|
Chang P, Nead KT, Olin JW, Cooke JP, Leeper NJ. Clinical and socioeconomic factors associated with unrecognized peripheral artery disease. Vasc Med 2014; 19:289-296. [PMID: 24872403 DOI: 10.1177/1358863x14535475] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peripheral artery disease (PAD) is a highly prevalent condition that frequently goes undetected and untreated. Socioeconomic factors associated with unrecognized PAD are not known. The ankle-brachial index (ABI) was calculated in 1656 study participants undergoing non-emergent coronary angiography with PAD defined as an ABI <0.9. Subjects were followed for mortality and cardiovascular outcomes. Compared to those without PAD, those with unrecognized PAD at enrollment were older, had higher rates of cardiovascular comorbidities, and had higher major adverse cardiovascular events (MACE) (p<0.03 for all). Among those enrolling without a reported history of PAD, there was a higher prevalence of PAD with decreasing income (p=0.004), education level (p<0.001), social isolation (p=0.027) and depression (p=0.034); 50% of these individuals reported symptoms suggestive of claudication. In conclusion, the prevalence of unrecognized PAD is high amongst a cohort of high-risk individuals referred for coronary angiography. A profile of lower socioeconomic status is associated with unrecognized PAD. These subjects will report symptoms suggestive of claudication and impaired walking ability when directly queried.
Collapse
Affiliation(s)
- Peter Chang
- Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin T Nead
- Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey W Olin
- Zena and Michael A Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - John P Cooke
- Methodist Hospital Research Institute, Houston, TX, USA
| | - Nicholas J Leeper
- Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA, USA Stanford Cardiovascular Institute, Stanford, CA, USA
| |
Collapse
|
43
|
Abstract
Vascular endothelium has important regulatory functions in the cardiovascular system and a pivotal role in the maintenance of vascular health and metabolic homeostasis. It has long been recognized that endothelial dysfunction participates in the pathogenesis of atherosclerosis from early, preclinical lesions to advanced, thrombotic complications. In addition, endothelial dysfunction has been recently implicated in the development of insulin resistance and type 2 diabetes mellitus (T2DM). Considering that states of insulin resistance (eg, metabolic syndrome, impaired fasting glucose, impaired glucose tolerance, and T2DM) represent the most prevalent metabolic disorders and risk factors for atherosclerosis, it is of considerable scientific and clinical interest that both metabolic and vascular disorders have endothelial dysfunction as a common background. Importantly, endothelial dysfunction has been associated with adverse outcomes in patients with established cardiovascular disease, and a growing body of evidence indicates that endothelial dysfunction also imparts adverse prognosis in states of insulin resistance. In this review, we discuss the association of insulin resistance and T2DM with endothelial dysfunction and vascular disease, with a focus on the underlying mechanisms and prognostic implications of the endothelial dysfunction in metabolic and vascular disorders. We also address current therapeutic strategies for the improvement of endothelial dysfunction.
Collapse
|
44
|
Armingohar Z, Jørgensen JJ, Kristoffersen AK, Schenck K, Dembic Z. Polymorphisms in the Interleukin-1 Gene Locus and Chronic Periodontitis in Patients with Atherosclerotic and Aortic Aneurysmal Vascular Diseases. Scand J Immunol 2014; 79:338-45. [DOI: 10.1111/sji.12166] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/14/2014] [Indexed: 02/07/2023]
Affiliation(s)
- Z. Armingohar
- Department of Oral Biology; Faculty of Dentistry; University of Oslo; Oslo Norway
| | - J. J. Jørgensen
- Department of Vascular Surgery; Oslo University Hospital; Aker and University of Oslo; Oslo Norway
| | - A. K. Kristoffersen
- Department of Oral Biology; Faculty of Dentistry; University of Oslo; Oslo Norway
| | - K. Schenck
- Department of Oral Biology; Faculty of Dentistry; University of Oslo; Oslo Norway
| | - Z. Dembic
- Department of Oral Biology; Faculty of Dentistry; University of Oslo; Oslo Norway
| |
Collapse
|
45
|
Lathe R, Sapronova A, Kotelevtsev Y. Atherosclerosis and Alzheimer--diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr 2014; 14:36. [PMID: 24656052 PMCID: PMC3994432 DOI: 10.1186/1471-2318-14-36] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/26/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Aging is accompanied by increasing vulnerability to pathologies such as atherosclerosis (ATH) and Alzheimer disease (AD). Are these different pathologies, or different presentations with a similar underlying pathoetiology? DISCUSSION Both ATH and AD involve inflammation, macrophage infiltration, and occlusion of the vasculature. Allelic variants in common genes including APOE predispose to both diseases. In both there is strong evidence of disease association with viral and bacterial pathogens including herpes simplex and Chlamydophila. Furthermore, ablation of components of the immune system (or of bone marrow-derived macrophages alone) in animal models restricts disease development in both cases, arguing that both are accentuated by inflammatory/immune pathways. We discuss that amyloid β, a distinguishing feature of AD, also plays a key role in ATH. Several drugs, at least in mouse models, are effective in preventing the development of both ATH and AD. Given similar age-dependence, genetic underpinnings, involvement of the vasculature, association with infection, Aβ involvement, the central role of macrophages, and drug overlap, we conclude that the two conditions reflect different manifestations of a common pathoetiology. MECHANISM Infection and inflammation selectively induce the expression of cholesterol 25-hydroxylase (CH25H). Acutely, the production of 'immunosterol' 25-hydroxycholesterol (25OHC) defends against enveloped viruses. We present evidence that chronic macrophage CH25H upregulation leads to catalyzed esterification of sterols via 25OHC-driven allosteric activation of ACAT (acyl-CoA cholesterol acyltransferase/SOAT), intracellular accumulation of cholesteryl esters and lipid droplets, vascular occlusion, and overt disease. SUMMARY We postulate that AD and ATH are both caused by chronic immunologic challenge that induces CH25H expression and protection against particular infectious agents, but at the expense of longer-term pathology.
Collapse
Affiliation(s)
- Richard Lathe
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Pieta Research, PO Box 27069, Edinburgh EH10 5YW, UK
| | - Alexandra Sapronova
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Optical Research Group, Laboratory of Evolutionary Biophysics of Development, Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Yuri Kotelevtsev
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Biomedical Centre for Research Education and Innovation (CREI), Skolkovo Institute of Science and Technology, Skolkovo 143025, Russia
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Little France, Edinburgh EH16 4TJ, UK
| |
Collapse
|
46
|
Vargas-Alarcón G, Posadas-Romero C, Villarreal-Molina T, Alvarez-León E, Angeles-Martinez J, Posadas-Sanchez R, Monroy-Muñoz I, Luna-Fuentes S, González-Salazar C, Ramirez-Bello J, Cardoso-Saldaña G, Medina-Urrutia A, Kimura-Hayama E. IL-24 gene polymorphisms are associated with cardiometabolic parameters and cardiovascular risk factors but not with premature coronary artery disease: the genetics of atherosclerotic disease Mexican study. J Interferon Cytokine Res 2014; 34:659-66. [PMID: 24552169 DOI: 10.1089/jir.2013.0081] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Coronary artery disease (CAD) is a multifactorial and polygenic disorder that results from an excessive inflammatory response. We analyzed whether interleukin-24 (IL-24) gene polymorphisms are associated with premature CAD in a case-control association study. Four polymorphisms (rs1150253, rs1150256, rs1150258, and rs3762344) of the IL-24 gene were analyzed by 5' exonuclease TaqMan genotyping assays in a group of 952 patients with premature CAD, 284 individuals with subclinical atherosclerosis (SA), and 912 controls. The studied polymorphisms were not associated with the risk of premature CAD or SA (P>0.05). Under dominant models adjusted for age, sex, body mass index, and medication, the polymorphisms were associated with cardiometabolic parameters and cardiovascular risk factors. Three polymorphisms (rs1150253, rs1150256, and rs3762344) were associated with hypertension and increased levels of systolic blood pressure in controls. In SA, 2 polymorphisms (rs1150256 and rs3762344) were associated with type 2 diabetes mellitus, gamma-glutamyl transpeptidase (GGT), and alkaline phosphatase, whereas rs1150253 was associated with GGT and type 2 diabetes mellitus and rs1150258 with GGT and alkaline phosphatase. In premature CAD, the 4 polymorphisms were associated with total cholesterol >200 mg/dL, low-density lipoprotein cholesterol (LDL-C), and GGT, whereas rs1150256 was associated also with ApoA. On the other hand, rs1150258 was associated with ApoA, LDL-C >100 mg/dL, and apoB/apoA ratio, and rs3762344 with ApoA, apoB/apoA ratio, LDL-C >100 mg/dL, and total cholesterol. On the basis of single-nucleotide polymorphism functional prediction software, rs1150253 and rs1150258 polymorphisms seem to be functional. The 4 studied polymorphisms were in linkage disequilibrium and had a similar haplotype distribution in patients and controls. Our study demonstrates the association of IL-24 polymorphisms with metabolic and cardiovascular risk factors in individuals with premature CAD, SA, and controls.
Collapse
Affiliation(s)
- Gilberto Vargas-Alarcón
- 1 Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez , Mexico City, Mexico
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Priest JR, Nead KT, Wehner MR, Cooke JP, Leeper NJ. Self-reported history of childhood smoking is associated with an increased risk for peripheral arterial disease independent of lifetime smoking burden. PLoS One 2014; 9:e88972. [PMID: 24558458 PMCID: PMC3928325 DOI: 10.1371/journal.pone.0088972] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/06/2014] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic disorders are well known to be associated with obesity, lipid profile, smoking, hypertension and other medical comorbidities, and large cohort studies have explored the childhood correlates to these adult risk factors. However, there has been little investigation into the childhood risk factors for peripheral arterial disease (PAD). We endeavored to better understand the role of smoking in childhood in the risk for PAD in a well described cohort of 1,537 adults at high risk for cardiovascular disease. In a multivariate regression model, we observed an increased risk of PAD among those who reported a history of smoking during childhood (OR = 2.86; 95% CI, 1.99-4.11; P<0.001), which remained statistically significant after controlling for lifetime smoking burden (OR = 1.55; 95% CI, 1.00-2.41; P = 0.049). Our novel observation of disproportionate risk of PAD conferred by a history of childhood smoking may reflect an unrecognized biological mechanism such as a unique susceptibility to vascular injury or an unaccounted for covariate such as secondhand smoke exposure in childhood. This observation suggests further investigation is required into the pathophysiology of smoking in the developing vasculature and the need for detailed clinical data about patterns of childhood smoking and smoke exposure.
Collapse
Affiliation(s)
- James R. Priest
- Division of Pediatric Cardiology, Stanford University School of Medicine, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Kevin T. Nead
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mackenzie R. Wehner
- Stanford University School of Medicine, Stanford, California, United States of America
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Methodist DeBakey Heart and Vascular Center, Houston, Texas, United States of America
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
| | - Nicholas J. Leeper
- Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Abstract
Peripheral artery disease (PAD) usually refers to ischemia of the lower limb vessels. Currently, the estimated number of cases in the world is 202 million. PAD is the third leading cause of atherosclerotic cardiovascular morbidity. The measurement of the ankle-brachial index (ABI) is recommended as a first-line noninvasive test for screening and diagnosis of PAD. An ABI <0.90 is an independent predictor of cardiovascular events and this measurement is useful to identify patients at moderate to high risk of cardiovascular disease. However, there is insufficient evidence to assess the benefits and harms of screening for PAD with the ABI in asymptomatic adults. Lifestyle modifications, including smoking cessation, dietary changes and physical activity, are currently the most cost-effective interventions. Inverse associations with PAD have been reported for some subtypes of dietary fats, fiber, antioxidants (vitamins E and C), folate, vitamins B6, B12 and D, flavonoids, and fruits and vegetables. A possible inverse association between better adherence to the Mediterranean diet and the risk of symptomatic PAD has also been reported in a large randomized clinical trial. Therefore, a Mediterranean-style diet could be effective in the primary and secondary prevention of PAD, although further experimental studies are needed to better clarify this association. (Circ J 2014; 78: 553-559).
Collapse
Affiliation(s)
- Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra
| | | |
Collapse
|
49
|
Downing KP, Nead KT, Kojima Y, Assimes T, Maegdefessel L, Quertermous T, Cooke JP, Leeper NJ. The combination of 9p21.3 genotype and biomarker profile improves a peripheral artery disease risk prediction model. Vasc Med 2013; 19:3-8. [PMID: 24323119 DOI: 10.1177/1358863x13514791] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Peripheral artery disease (PAD) is a highly morbid condition affecting more than 8 million Americans. Frequently, PAD patients are unrecognized and therefore do not receive appropriate therapies. Therefore, new methods to identify PAD have been pursued, but have thus far had only modest success. Here we describe a new approach combining genomic and metabolic information to enhance the diagnosis of PAD. We measured the genotype of the chromosome 9p21 cardiovascular-risk polymorphism rs10757269 as well as the biomarkers C-reactive protein, cystatin C, β2-microglobulin, and plasma glucose in a study population of 393 patients undergoing coronary angiography. The rs10757269 allele was associated with PAD status (ankle-brachial index < 0.9) independent of biomarkers and traditional cardiovascular risk factors (odds ratio = 1.92; 95% confidence interval, 1.29-2.85). Importantly, compared to a previously validated risk factor-based PAD prediction model, the addition of biomarkers and rs10757269 significantly and incrementally improved PAD risk prediction as assessed by the net reclassification index (NRI = 33.5%; p = 0.001) and integrated discrimination improvement (IDI = 0.016; p = 0.017). In conclusion, a model including a panel of biomarkers, which includes both genomic information (which is reflective of heritable risk) and metabolic information (which integrates environmental exposures), predicts the presence or absence of PAD better than established risk models, suggesting clinical utility for the diagnosis of PAD.
Collapse
Affiliation(s)
- Kelly P Downing
- Division of Vascular Surgery, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Peripheral vascular disease (PVD) is an atherosclerotic disease affecting the lower extremities, resulting in skeletal muscle ischemia, intermittent claudication, and, in more severe stages of disease, limb amputation and death. The evaluation of therapy in this patient population can be challenging, as the standard clinical indices are insensitive to assessment of regional alterations in skeletal muscle physiology. Radiotracer imaging of the lower extremities with techniques such as PET and SPECT can provide a noninvasive quantitative technique for the evaluation of the pathophysiology associated with PVD and may complement clinical indices and other imaging approaches. This review discusses the progress in radiotracer-based evaluation of PVD and highlights recent advancements in molecular imaging with potential for clinical application.
Collapse
Affiliation(s)
- Mitchel R. Stacy
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Wunan Zhou
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Albert J. Sinusas
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|