1
|
Kazemi Asl S, Rahimzadegan M, Kazemi Asl A. Pharmacogenomics-based systematic review of coronary artery disease based on personalized medicine procedure. Heliyon 2024; 10:e28983. [PMID: 38601677 PMCID: PMC11004819 DOI: 10.1016/j.heliyon.2024.e28983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Background Coronary artery disease (CAD) is the most common reason for mortality and disability-adjusted life years (DALYs) lost globally. This study aimed to suggest a new gene list for the treatment of CAD by a systematic review of bioinformatics analyses of pharmacogenomics impacts of potential genes and variants. Methods PubMed search was filtered by the title including Coronary Artery Disease during 2020-2023. To find the genes with pharmacogenetic impact on the CAD, additional filtrations were considered according to the variant annotations. Protein-Protein Interactions (PPIs), Gene-miRNA Interactions (GMIs), Protein-Drug Interactions (PDIs), and variant annotation assessments (VAAs) performed by STRING-MODEL (ver. 12), Cytoscape (ver. 3.10), miRTargetLink.2., NetworkAnalyst (ver 0.3.0), and PharmGKB. Results Results revealed 5618 publications, 1290 papers were qualified, and finally, 650 papers were included. 4608 protein-coding genes were extracted, among them, 1432 unique genes were distinguished and 530 evidence-based repeated genes remained. 71 genes showed a pharmacogenetics-related variant annotation in at least (entirely 6331 annotations). Variant annotation assessment (VAA) showed 532 potential variants for the final report, and finally, the concluding PGs list represented 175 variants. Based on the function and MAF, 57 nonsynonymous variants of 29 Pharmacogenomics-related genes were associated with CAD. Conclusion Conclusively, evaluating circulating miR33a in individuals' plasma with CAD, and genotyping of rs2230806, rs2230808, rs2487032, rs12003906, rs2472507, rs2515629, and rs4149297 (ABCA1 variants) lead to precisely prescribing of well-known drugs. Also, the findings of this review can be used in both whole-genome sequencing (WGS) and whole-exome sequencing (WES) analysis in the prognosis and diagnosis of CAD.
Collapse
Affiliation(s)
- Siamak Kazemi Asl
- Deputy of Education, Ministry of Health and Medical Education, Tehran, Iran
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemi Asl
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Jaiswal V, Ang SP, Deb N, Hanif M, Batra N, Kanagala SG, Vojjala N, Rajak K, Roy P, Sharath M, Waleed MS, Wajid Z, Mattumpuram J. Association between Statin Use and Chemotherapy-Induced Cardiotoxicity: A Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:580. [PMID: 38674227 PMCID: PMC11052115 DOI: 10.3390/medicina60040580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 04/28/2024]
Abstract
Background: Chemotherapy-induced cardiac dysfunction (CIC) is a significant and concerning complication observed among cancer patients. Despite the demonstrated cardioprotective benefits of statins in various cardiovascular diseases, their effectiveness in mitigating CIC remains uncertain. Objective: This meta-analysis aims to comprehensively evaluate the potential cardioprotective role of statins in patients with CIC. Methods: A systematic literature search was conducted using PubMed, Embase, and Scopus databases to identify relevant articles published from inception until 10th May 2023. The outcomes were assessed using pooled odds ratio (OR) for categorical data and mean difference (MD) for continuous data, with corresponding 95% confidence intervals (95% CIs). Results: This meta-analysis comprised nine studies involving a total of 5532 patients, with 1904 in the statin group and 3628 in the non-statin group. The pooled analysis of primary outcome shows that patients who did not receive statin suffer a greater decline in the LVEF after chemotherapy compared to those who receive statin (MD, 3.55 (95% CI: 1.04-6.05), p = 0.01). Likewise, we observed a significantly higher final mean LVEF among chemotherapy patients with statin compared to the non-statin group of patients (MD, 2.08 (95% CI: 0.86-3.30), p > 0.001). Additionally, there was a lower risk of incident heart failure in the statin group compared to the non-statin group of patients (OR, 0.41 (95% CI: 0.27-0.62), p < 0.001). Lastly, the change in the mean difference for LVEDV was not statistically significant between the statin and non-statin groups (MD, 1.55 (95% CI: -5.22-8.33), p = 0.65). Conclusion: Among patients of CIC, statin use has shown cardioprotective benefits by improving left ventricular function and reducing the risk of heart failure.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Cardiovascular Research, Larkin Community Hospital, South Miami, FL 33143, USA
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA
| | - Novonil Deb
- North Bengal Medical College and Hospital, Darjeeling 734012, West Bengal, India
| | - Muhammad Hanif
- Department of Internal Medicine, SUNY Upstate Medical University, 750 E Adams St., Syracuse, NY 13210, USA
| | - Nitya Batra
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI 48073, USA
| | - Sai Gautham Kanagala
- Department of Internal Medicine, Metropolitan Hospital Center, New York, NY 10029, USA
| | - Nikhil Vojjala
- Internal Medicine Department, Trinity Health Oakland/Wayne State University, Detroit, MI 48341, USA
| | - Kripa Rajak
- Department of Internal Medicine, UPMC Harrisburgh, 111 S Front St., Harrisburg, PA 17101, USA
| | - Poulami Roy
- North Bengal Medical College and Hospital, Darjeeling 734012, West Bengal, India
| | - Medha Sharath
- Bangalore Medical College and Research Institute, Kalasipalya, Bengaluru 560002, Karnataka, India
| | - Madeeha Subhan Waleed
- Department of Internal Medicine, Lower Bucks Hospital, Bristo, 501 Bath Rd., Bristol, PA 19007, USA
| | - Zarghoona Wajid
- Department of Internal Medicine, Wayne State University School of Medicine, 540 E. Canfield Ave., Detroit, MI 48201, USA
| | - Jishanth Mattumpuram
- Division of Cardiology, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
3
|
Tiwari A, Haj N, Elgrably B, Berihu M, Laskov V, Barash S, Zigron S, Sason H, Shamay Y, Karni-Ashkenazi S, Holdengreber M, Saar G, Vandoorne K. Cross-Modal Imaging Reveals Nanoparticle Uptake Dynamics in Hematopoietic Bone Marrow during Inflammation. ACS NANO 2024; 18:7098-7113. [PMID: 38343099 PMCID: PMC10919094 DOI: 10.1021/acsnano.3c11201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Nanoparticles have been employed to elucidate the innate immune cell biology and trace cells accumulating at inflammation sites. Inflammation prompts innate immune cells, the initial responders, to undergo rapid turnover and replenishment within the hematopoietic bone marrow. Yet, we currently lack a precise understanding of how inflammation affects cellular nanoparticle uptake at the level of progenitors of innate immune cells in the hematopoietic marrow. To bridge this gap, we aimed to develop imaging tools to explore the uptake dynamics of fluorescently labeled cross-linked iron oxide nanoparticles in the bone marrow niche under varying degrees of inflammation. The inflammatory models included mice that received intramuscular lipopolysaccharide injections to induce moderate inflammation and streptozotocin-induced diabetic mice with additional intramuscular lipopolysaccharide injections to intensify inflammation. In vivo magnetic resonance imaging (MRI) and fluorescence imaging revealed an elevated level of nanoparticle uptake at the bone marrow as the levels of inflammation increased. The heightened uptake of nanoparticles within the inflamed marrow was attributed to enhanced permeability and retention with increased nanoparticle intake by hematopoietic progenitor cells. Moreover, intravital microscopy showed increased colocalization of nanoparticles within slowly patrolling monocytes in these inflamed hematopoietic marrow niches. Our discoveries unveil a previously unknown role of the inflamed hematopoietic marrow in enhanced storage and rapid deployment of nanoparticles, which can specifically target innate immune cells at their production site during inflammation. These insights underscore the critical function of the hematopoietic bone marrow in distributing iron nanoparticles to innate immune cells during inflammation. Our findings offer diagnostic and prognostic value, identifying the hematopoietic bone marrow as an imaging biomarker for early detection in inflammation imaging, advancing personalized clinical care.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Narmeen Haj
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Betsalel Elgrably
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Maria Berihu
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Viktor Laskov
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
- Third
Faculty of Medicine, Charles University, Prague 100 00, Czech Republic
| | - Sivan Barash
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Shachar Zigron
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Hagit Sason
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Yosi Shamay
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Shiri Karni-Ashkenazi
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| | - Maya Holdengreber
- Biomedical
Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Galit Saar
- Biomedical
Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Katrien Vandoorne
- Faculty
of Biomedical Engineering, Technion-Israel
Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
4
|
Calcagno C, David JA, Motaal AG, Coolen BF, Beldman T, Corbin A, Kak A, Ramachandran S, Pruzan A, Sridhar A, Soler R, Faries CM, Fayad ZA, Mulder WJM, Strijkers GJ. Self-gated, dynamic contrast-enhanced magnetic resonance imaging with compressed-sensing reconstruction for evaluating endothelial permeability in the aortic root of atherosclerotic mice. NMR IN BIOMEDICINE 2023; 36:e4823. [PMID: 36031706 PMCID: PMC10078106 DOI: 10.1002/nbm.4823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/01/2022] [Accepted: 08/21/2022] [Indexed: 05/16/2023]
Abstract
High-risk atherosclerotic plaques are characterized by active inflammation and abundant leaky microvessels. We present a self-gated, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) acquisition with compressed sensing reconstruction and apply it to assess longitudinal changes in endothelial permeability in the aortic root of Apoe-/- atherosclerotic mice during natural disease progression. Twenty-four, 8-week-old, female Apoe-/- mice were divided into four groups (n = 6 each) and imaged with self-gated DCE-MRI at 4, 8, 12, and 16 weeks after high-fat diet initiation, and then euthanized for CD68 immunohistochemistry for macrophages. Eight additional mice were kept on a high-fat diet and imaged longitudinally at the same time points. Aortic-root pseudo-concentration curves were analyzed using a validated piecewise linear model. Contrast agent wash-in and washout slopes (b1 and b2 ) were measured as surrogates of aortic root endothelial permeability and compared with macrophage density by immunohistochemistry. b2 , indicating contrast agent washout, was significantly higher in mice kept on an high-fat diet for longer periods of time (p = 0.03). Group comparison revealed significant differences between mice on a high-fat diet for 4 versus 16 weeks (p = 0.03). Macrophage density also significantly increased with diet duration (p = 0.009). Spearman correlation between b2 from DCE-MRI and macrophage density indicated a weak relationship between the two parameters (r = 0.28, p = 0.20). Validated piecewise linear modeling of the DCE-MRI data showed that the aortic root contrast agent washout rate is significantly different during disease progression. Further development of this technique from a single-slice to a 3D acquisition may enable better investigation of the relationship between in vivo imaging of endothelial permeability and atherosclerotic plaques' genetic, molecular, and cellular makeup in this important model of disease.
Collapse
Affiliation(s)
- Claudia Calcagno
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - John A David
- Amsterdam University Medical Centers, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Abdallah G Motaal
- Siemens Healthineers, Cardiovascular Care Group, Advanced Therapies Business, Erlangen, Germany
| | - Bram F Coolen
- Amsterdam University Medical Centers, Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Thijs Beldman
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexandra Corbin
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Arnav Kak
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarayu Ramachandran
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alison Pruzan
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Arthi Sridhar
- Department of Hematology/Oncology, UTHealth McGovern Medical School, Houston, TX, USA
| | - Raphael Soler
- CNRS, CRMBM, Marseille, France
- Department of Vascular and Endovascular Surgery, Hôpital Universitaire de la Timone, APHM, Marseille, France
| | - Christopher M Faries
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Zahi A Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Willem J M Mulder
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gustav J Strijkers
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Amsterdam University Medical Centers, Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Heiston EM, Hundley WG. Statins for Cardiac and Vascular Protection During and After Cancer Therapy. Curr Oncol Rep 2022; 24:555-561. [PMID: 35199294 PMCID: PMC9922479 DOI: 10.1007/s11912-022-01212-4] [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] [Accepted: 11/02/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Although cancer treatments have increased overall survival rates, the cardiovascular consequences of cancer therapy place patients at an increased risk of adverse outcomes. This manuscript presents data accumulated to date regarding cardiovascular outcomes relating to the administration of 3-hydroxy-3-methylglutarylcoenzyme-A reductase inhibitor (or statin) therapy in individuals receiving potentially cardiotoxic cancer treatments. RECENT FINDINGS Retrospective observational studies in humans and randomized controlled trials in animals suggest that statins may reduce cancer-specific and all-cause mortality. Further, statins may attenuate cancer therapy-induced declines in left ventricular ejection fraction (LVEF) and increases in blood pressure. Observational studies suggest a potential attenuation in LVEF decline in patients with cancer and primary or secondary indications to receive a statin for prevention of cardiovascular events. Large randomized clinical trials are warranted to understand the efficacy and potential impacts of statin class, dosage, and duration on cardiovascular outcomes in patients treated for cancer.
Collapse
Affiliation(s)
- Emily M Heiston
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, PO Box 980335, Richmond, VA, 23298, USA
| | - W Gregory Hundley
- Division of Cardiology, VCU Pauley Heart Center, Virginia Commonwealth University, PO Box 980335, Richmond, VA, 23298, USA.
| |
Collapse
|
6
|
Yan Y, Gao R, Zhang S, Gao Z, Chen A, Wang J, Zhang S, Dai W, Li F, Li X, Yang G, Liu L, Chen X. Hemoglobin A1c and Angiographic Severity with Coronary Artery Disease: A Cross-Sectional Study. Int J Gen Med 2022; 15:1485-1495. [PMID: 35210822 PMCID: PMC8857977 DOI: 10.2147/ijgm.s346525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/20/2022] [Indexed: 12/04/2022] Open
Abstract
Background Many studies have shown that glycated hemoglobin (HbA1c) is associated with coronary artery disease (CAD). HbA1c was independently related to angiographic severity in Chinese patients with CAD after adjusting for other covariates. Some traditional cardiovascular drugs may have an impact on this relationship. Methods This retrospective study enrolled a total of 572 CAD patients who underwent their coronary angiography and had their HbA1c levels measured at the Chinese Hospital. The complexity of the coronary artery lesions was evaluated using the Syntax score, and the subjects were divided into 4 inter quartiles according to HbA1c levels. Covariates included history of traditional cardiovascular drugs. Results The average age of selected participants was 61.00 ± 9.15 years old, and about 54.72% of them were male. Result of fully adjusted linear regression showed that HbA1c was positively associated with Syntax score after adjusting confounders (β = 1.09, 95% CI: 0.27, 1.91, P = 0.0096). By interaction and stratified analyses, the interactions were observed based on our specification including with the medication history of statins and angiotensin receptor blockers (ARBs) (P values for interaction <0.05). Conclusion In this study, we found a positive correlation between the HbA1c levels and the SYNTAX score among CAD individuals, and oral statins and ARBs medication could affect the correlation. Thus, HbA1c measurement could be used for the evaluation of the severity and complexity of coronary lesions among CAD patients.
Collapse
Affiliation(s)
- Yugang Yan
- School of Medical Engineering, Jining Medical University, Jining, Shandong, People’s Republic of China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| | - Ronghua Gao
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Shaohui Zhang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Zhencai Gao
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Anyong Chen
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Jianjun Wang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Shufang Zhang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Wen Dai
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Fen Li
- Electrocardiogram Room, Jining First People’s Hospital, Jining, Shandong, People’s Republic of China
| | - Xiangting Li
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Guoliang Yang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Lixin Liu
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Xueying Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
- Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, Shandong, People’s Republic of China
- Correspondence: Xueying Chen, Department of Cardiology, Affiliated Hospital of Jining Medical University, No. 89 Guhuai Road, Jining, 272029, Shandong, People’s Republic of China, Tel +86- 537- 2903508, Fax +86 537-2213030, Email
| |
Collapse
|
7
|
Tiwari A, Elgrably B, Saar G, Vandoorne K. Multi-Scale Imaging of Vascular Pathologies in Cardiovascular Disease. Front Med (Lausanne) 2022; 8:754369. [PMID: 35071257 PMCID: PMC8766766 DOI: 10.3389/fmed.2021.754369] [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: 08/06/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cardiovascular disease entails systemic changes in the vasculature. The endothelial cells lining the blood vessels are crucial in the pathogenesis of cardiovascular disease. Healthy endothelial cells direct the blood flow to tissues as vasodilators and act as the systemic interface between the blood and tissues, supplying nutrients for vital organs, and regulating the smooth traffic of leukocytes into tissues. In cardiovascular diseases, when inflammation is sensed, endothelial cells adjust to the local or systemic inflammatory state. As the inflamed vasculature adjusts, changes in the endothelial cells lead to endothelial dysfunction, altered blood flow and permeability, expression of adhesion molecules, vessel wall inflammation, thrombosis, angiogenic processes, and extracellular matrix production at the endothelial cell level. Preclinical multi-scale imaging of these endothelial changes using optical, acoustic, nuclear, MRI, and multimodal techniques has progressed, due to technical advances and enhanced biological understanding on the interaction between immune and endothelial cells. While this review highlights biological processes that are related to changes in the cardiac vasculature during cardiovascular diseases, it also summarizes state-of-the-art vascular imaging techniques. The advantages and disadvantages of the different imaging techniques are highlighted, as well as their principles, methodologies, and preclinical and clinical applications with potential future directions. These multi-scale approaches of vascular imaging carry great potential to further expand our understanding of basic vascular biology, to enable early diagnosis of vascular changes and to provide sensitive diagnostic imaging techniques in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Ashish Tiwari
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Betsalel Elgrably
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Galit Saar
- Biomedical Core Facility, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Katrien Vandoorne
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
8
|
Gencer B, Mach F. PCSK9 inhibition could be effective for acute myocardial infarction. Curr Med Chem 2021; 29:1016-1026. [PMID: 34348606 DOI: 10.2174/0929867328666210804091003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022]
Abstract
In this review, we will explore the role of PCSK9 and inhibition of PCSK9 in patients after acute myocardial infarction (MI). Despite the implementation of evidence-based therapies to improve outcomes, mortality at one-year remains at 12-15% and the need to further reduce complications related to MI persists. Mechanistic and epidemiologic studies suggest that the naturally occurring PCSK9 protein increases coronary plaque vulnerability through several pathways, including pro-inflammatory LDL-C oxidation and direct modification of plaque composition. PCSK9 inhibitors are a class of drugs with proven efficacy in patients with recent MI. The latest guidelines recommend the use of PCSK9 in patients with recent MI early in the process of care to reduce LDL-C values and associated morbidity. The use of PCSK9 inhibition could be beneficial for mortality reduction after an acute MI and should be tested in an appropriately powered randomized controlled trial.
Collapse
Affiliation(s)
- Baris Gencer
- Cardiology Division, Geneva University Hospitals. Switzerland
| | - François Mach
- Cardiology Division, Geneva University Hospitals. Switzerland
| |
Collapse
|
9
|
Techorueangwiwat C, Kanitsoraphan C, Hansrivijit P. Therapeutic implications of statins in heart failure with reduced ejection fraction and heart failure with preserved ejection fraction: a review of current literature. F1000Res 2021; 10:16. [PMID: 36873456 PMCID: PMC9982192 DOI: 10.12688/f1000research.28254.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2021] [Indexed: 11/20/2022] Open
Abstract
Statins are one of the standard treatments to prevent cardiovascular events such as coronary artery disease and heart failure (HF). However, data on the use of statins to improve clinical outcomes in patients with established HF remains controversial. We summarized available clinical studies which investigated the effects of statins on clinical outcomes in patients with HF with reduced ejection fraction (HFrEF) and HF with preserved ejection fraction (HFpEF). Statins possess many pleiotropic effects in addition to lipid-lowering properties that positively affect the pathophysiology of HF. In HFrEF, data from two large randomized placebo-controlled trials did not show benefits of statins on mortality of patients with HFrEF. However, more recent prospective cohort studies and meta-analyses have shown decreased risk of mortality as well as cardiovascular hospitalization with statins treatment. In HFpEF, most prospective and retrospective cohort studies as well as meta analyses have consistently reported positive effects of statins, including reducing mortality and improving other clinical outcomes. Current evidence also suggests better outcomes with lipophilic statins in patients with HF. In summary, statins might be effective in improving survival and other clinical outcomes in patients with HF, especially for patients with HFpEF. Lipophilic statins might also be more beneficial for HF patients. Based on current evidence, statins did not cause harm and should be continued in HF patients who are already taking the medication. Further randomized controlled trials are needed to clarify the benefits of statins in HF patients.
Collapse
|
10
|
Abstract
ABSTRACT Macrophage, as an integral component of the immune system and the first responder to local damage, is on the front line of defense against infection. Over the past century, the prevailing view of macrophage origin states that all macrophage populations resided in tissues are terminally differentiated and replenished by monocytes from bone-marrow progenitors. Nonetheless, this theory has been reformed by ground-breaking discoveries from the past decades. It is now believed that tissue-resident macrophages (TRMs) are originated from the embryonic precursors and seeded in tissue prenatally. They can replenish via self-renewal throughout the lifespan. Indeed, recent studies have demonstrated that tissue-resident macrophages should not be classified by the over-simplified macrophage polarization (M1/M2) dogma during inflammation. Moreover, multiple lines of evidence have indicated that tissue-resident macrophages play critical roles in maintaining tissue homeostasis and facilitating tissue repair through controlling infection and resolving inflammation. In this review, we summarize the properties of resident macrophages in the lung, spleen, and heart, and further highlight the impact of TRM populations on inflammation control and tissue repair. We also discuss the potential role of local proliferation in maintaining a physiologically stable TRM pool in response to acute inflammation.
Collapse
Affiliation(s)
- Xingjiang Mu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | | |
Collapse
|
11
|
Gunawardena T, Merinopoulos I, Wickramarachchi U, Vassiliou V, Eccleshall S. Endothelial Dysfunction and Coronary Vasoreactivity - A Review of the History, Physiology, Diagnostic Techniques, and Clinical Relevance. Curr Cardiol Rev 2021; 17:85-100. [PMID: 32552654 PMCID: PMC8142375 DOI: 10.2174/1573403x16666200618161942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/23/2020] [Accepted: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
The fervency for advancement and evolution in percutaneous coronary intervention has revolutionised the treatment of coronary artery disease. Historically, the focus of the interventional cardiologist was directed at the restoration of luminal patency of the major epicardial coronary arteries, yet whilst this approach is evolving with much greater utilisation of physiological assessment, it often neglects consideration of the role of the coronary microcirculation, which has been shown to clearly influence prognosis. In this review, we explore the narrative of the coronary circulation as more than just a simple conduit for blood but an organ with functional significance. We review organisation and physiology of the coronary circulation, as well as the current methods and techniques used to examine it. We discuss the studies exploring coronary artery endothelial function, appreciating that coronary artery disease occurs on a spectrum of disorder and that percutaneous coronary intervention has a latent effect on the coronary circulation with long-term consequences. It is concluded that greater recognition of the coronary artery endothelium and mechanisms of the coronary circulation should further guide revascularisation strategies.
Collapse
Affiliation(s)
- Tharusha Gunawardena
- Address correspondence to this author at the Department of Cardiology, Norfolk and Norwich University Hospital, Colney Lane NR4 7UY, Norwich, England; E-mail:
| | | | | | | | | |
Collapse
|
12
|
Sectm1a Facilitates Protection against Inflammation-Induced Organ Damage through Promoting TRM Self-Renewal. Mol Ther 2020; 29:1294-1311. [PMID: 33279722 DOI: 10.1016/j.ymthe.2020.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/11/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022] Open
Abstract
Tissue-resident macrophages (TRMs) are sentinel cells for maintaining tissue homeostasis and organ function. In this study, we discovered that lipopolysaccharide (LPS) administration dramatically reduced TRM populations and suppressed their self-renewal capacities in multiple organs. Using loss- and gain-of-function approaches, we define Sectm1a as a novel regulator of TRM self-renewal. Specifically, at the earlier stage of endotoxemia, Sectm1a deficiency exaggerated acute inflammation-induced reduction of TRM numbers in multiple organs by suppressing their proliferation, which was associated with more infiltrations of inflammatory monocytes/neutrophils and more serious organ damage. By contrast, administration of recombinant Sectm1a enhanced TRM populations and improved animal survival upon endotoxin challenge. Mechanistically, we identified that Sectm1a-induced upregulation in the self-renewal capacity of TRM is dependent on GITR-activated T helper cell expansion and cytokine production. Meanwhile, we found that TRMs may play an important role in protecting local vascular integrity during endotoxemia. Our study demonstrates that Sectm1a contributes to stabling TRM populations through maintaining their self-renewal capacities, which benefits the host immune response to acute inflammation. Therefore, Sectm1a may serve as a new therapeutic agent for the treatment of inflammatory diseases.
Collapse
|
13
|
Chen Z, Wang X, Hou X, Ding F, Yi K, Zhang P, You T. Knockdown of Long Non-Coding RNA AFAP1-AS1 Promoted Viability and Suppressed Death of Cardiomyocytes in Response to I/R In Vitro and In Vivo. J Cardiovasc Transl Res 2020; 13:996-1007. [PMID: 32406007 DOI: 10.1007/s12265-020-10016-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 04/24/2020] [Indexed: 12/13/2022]
Abstract
Long non-coding RNA (lncRNA) plays a pivotal role in the development of myocardial infarction (MI). The aim of this study was to investigate the effects of lncRNA actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) on cell cycle, proliferation, and apoptosis. RT-qPCR was used to detect the expression levels of AFAP1-AS1, miR-512-3p, and reticulon 3 (RTN3) in rat model of I/R. The simulated MI environment was constructed. MTT assay and flow cytometry were used to detect changes in cardiomyocyte viability and cell cycle/apoptosis after MI by AFAP1-AS1 silencing or RTN3 silencing. The targeting relationship of miR-512-3p and AFAP1-AS1 and RTN3 in cardiomyocytes was verified by dual luciferase reporter assay. The expression levels of AFAP1-AS1 and RTN3 were significantly upregulated in a rat model of LAD ligation (or MI) ligation, while the expression level of miR-512-3p was significantly reduced. Overexpressed AFAP1-AS1 and RTN3 promoted cardiomyocyte apoptosis and inhibited cardiomyocyte proliferation. MiR-512-3p was a direct target of AFAP1-AS1, and RTN3 was a direct target of miR-512-3p. AFAP1-AS1 promoted the progression of MI by targeting miR-512-3p. AFAP1-AS1 promoted the progression of MI by modulating the miR-512-3p/RTN3 axis. AFAP1-AS1 may be a potential therapy target for MI. Graphical Abstract The role of AFAP1-AS1 in regulating MI injury in vivo. (A) Effect of AFAP1-AS1 in MI injury in vivo. (B) The mRNA level of RTN3 in MI injury in vivo. (C) The protein level of RTN3 in MI injury in vivo. (D) Effect of miR-512-3p in MI model group. (E) TUNEL assay. *P < 0.05, **P < 0.01 vs the sham group; #P < 0.05, ##P < 0.01 vs the MI group.
Collapse
Affiliation(s)
- Zhigong Chen
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Department of Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Xinkuan Wang
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Xiaodong Hou
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Fan Ding
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Kang Yi
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Peng Zhang
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China
| | - Tao You
- Department of Cardiovascular Surgery, Gansu Provincial Hospital, No. 204, Dong gang West Road, Chengguan District, Lanzhou City, Gansu province, 730000, People's Republic of China.
- Congenital Heart Disease Diagnosis and Treatment Gansu Province International Science and Technology Cooperation Base, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
14
|
Nishimiya K, Matsumoto Y, Shimokawa H. Recent Advances in Vascular Imaging. Arterioscler Thromb Vasc Biol 2020; 40:e313-e321. [PMID: 33054393 DOI: 10.1161/atvbaha.120.313609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recent advances in vascular imaging have enabled us to uncover the underlying mechanisms of vascular diseases both ex vivo and in vivo. In the past decade, efforts have been made to establish various methodologies for evaluation of atherosclerotic plaque progression and vascular inflammatory changes in addition to biomarkers and clinical manifestations. Several recent publications in Arteriosclerosis, Thrombosis, and Vascular Biology highlighted the essential roles of in vivo and ex vivo vascular imaging, including magnetic resonance image, computed tomography, positron emission tomography/scintigraphy, ultrasonography, intravascular ultrasound, and most recently, optical coherence tomography, all of which can be used in bench and clinical studies at relative ease. With new methods proposed in several landmark studies, these clinically available imaging modalities will be used in the near future. Moreover, future development of intravascular imaging modalities, such as optical coherence tomography-intravascular ultrasound, optical coherence tomography-near-infrared autofluorescence, polarized-sensitive optical coherence tomography, and micro-optical coherence tomography, are anticipated for better management of patients with cardiovascular disease. In this review article, we will overview recent advances in vascular imaging and ongoing works for future developments.
Collapse
Affiliation(s)
- Kensuke Nishimiya
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuharu Matsumoto
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
15
|
Agrawal H, Choy HHK, Liu J, Auyoung M, Albert MA. Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2020; 40:e185-e192. [PMID: 32579480 DOI: 10.1161/atvbaha.120.313608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Harsh Agrawal
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| | - Ho-Hin K Choy
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Jason Liu
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Matthew Auyoung
- Division of Cardiology, Department of Medicine, California Pacific Medical Center, San Francisco (H.-h.K.C., J.L., M.A.)
| | - Michelle A Albert
- From the Center for the Study of Adversity and Cardiovascular Disease (NURTURE Center), Division of Cardiology, Department of Medicine, University of California San Francisco (H.A., M.A.A.)
| |
Collapse
|
16
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
17
|
van den Boomen M, Kause HB, van Assen HC, Dankers PYW, Bouten CVC, Vandoorne K. Triple-marker cardiac MRI detects sequential tissue changes of healing myocardium after a hydrogel-based therapy. Sci Rep 2019; 9:19366. [PMID: 31852978 PMCID: PMC6920418 DOI: 10.1038/s41598-019-55864-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/03/2019] [Indexed: 12/31/2022] Open
Abstract
Regenerative therapies based on injectable biomaterials, hold an unparalleled potential for treating myocardial ischemia. Yet, noninvasive evaluation of their efficacy has been lagging behind. Here, we report the development and longitudinal application of multiparametric cardiac magnetic resonance imaging (MRI) to evaluate a hydrogel-based cardiac regenerative therapy. A pH-switchable hydrogel was loaded with slow releasing insulin growth factor 1 and vascular endothelial growth factor, followed by intramyocardial injection in a mouse model of ischemia reperfusion injury. Longitudinal cardiac MRI assessed three hallmarks of cardiac regeneration: angiogenesis, resolution of fibrosis and (re)muscularization after infarction. The multiparametric approach contained dynamic contrast enhanced MRI that measured improved vessel features by assessing fractional blood volume and permeability*surface area product, T1-mapping that displayed reduced fibrosis, and tagging MRI that showed improved regional myocardial strain in hydrogel treated infarcts. Finally, standard volumetric MRI demonstrated improved left ventricular functioning in hydrogel treated mice followed over time. Histology confirmed MR-based vessel features and fibrotic measurements. Our novel triple-marker strategy enabled detection of ameliorated regeneration in hydrogel treated hearts highlighting the translational potential of these longitudinal MRI approaches.
Collapse
Affiliation(s)
- Maaike van den Boomen
- Department of Biomedical Engineering, Cell-Matrix Interaction for Cardiovascular Tissue Regeneration, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Hanne B Kause
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Hans C van Assen
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Patricia Y W Dankers
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Cell-Matrix Interaction for Cardiovascular Tissue Regeneration, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Katrien Vandoorne
- Department of Biomedical Engineering, Cell-Matrix Interaction for Cardiovascular Tissue Regeneration, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
18
|
Vandoorne K, Rohde D, Kim HY, Courties G, Wojtkiewicz G, Honold L, Hoyer FF, Frodermann V, Nayar R, Herisson F, Jung Y, Désogère PA, Vinegoni C, Caravan P, Weissleder R, Sosnovik DE, Lin CP, Swirski FK, Nahrendorf M. Imaging the Vascular Bone Marrow Niche During Inflammatory Stress. Circ Res 2019; 123:415-427. [PMID: 29980569 DOI: 10.1161/circresaha.118.313302] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Inflammatory stress induced by exposure to bacterial lipopolysaccharide causes hematopoietic stem cell expansion in the bone marrow niche, generating a cellular immune response. As an integral component of the hematopoietic stem cell niche, the bone marrow vasculature regulates the production and release of blood leukocytes, which protect the host against infection but also fuel inflammatory diseases. OBJECTIVE We aimed to develop imaging tools to explore vascular changes in the bone marrow niche during acute inflammation. METHODS AND RESULTS Using the TLR (Toll-like receptor) ligand lipopolysaccharide as a prototypical danger signal, we applied multiparametric, multimodality and multiscale imaging to characterize how the bone marrow vasculature adapts when hematopoiesis boosts leukocyte supply. In response to lipopolysaccharide, ex vivo flow cytometry and histology showed vascular changes to the bone marrow niche. Specifically, proliferating endothelial cells gave rise to new vasculature in the bone marrow during hypoxic conditions. We studied these vascular changes with complementary intravital microscopy and positron emission tomography/magnetic resonance imaging. Fluorescence and positron emission tomography integrin αVβ3 imaging signal increased during lipopolysaccharide-induced vascular remodeling. Vascular leakiness, quantified by albumin-based in vivo microscopy and magnetic resonance imaging, rose when neutrophils departed and hematopoietic stem and progenitor cells proliferated more vigorously. CONCLUSIONS Introducing a tool set to image bone marrow either with cellular resolution or noninvasively within the entire skeleton, this work sheds light on angiogenic responses that accompany emergency hematopoiesis. Understanding and monitoring bone marrow vasculature may provide a key to unlock therapeutic targets regulating systemic inflammation.
Collapse
Affiliation(s)
- Katrien Vandoorne
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - David Rohde
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Hye-Yeong Kim
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | | | - Gregory Wojtkiewicz
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Lisa Honold
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Friedrich Felix Hoyer
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Vanessa Frodermann
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Ribhu Nayar
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Fanny Herisson
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Yookyung Jung
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Wellman Center for Photomedicine (Y.J., C.P.L.)
| | - Pauline A Désogère
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.)
| | - Claudio Vinegoni
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Peter Caravan
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.)
| | - Ralph Weissleder
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Massachusetts General Hospital and Harvard Medical School, Charlestown; and Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.)
| | - David E Sosnovik
- Massachusetts General Hospital and Harvard Medical School, Boston; Department of Radiology, Martinos Center for Biomedical Imaging (P.A.D., P.C., D.E.S.).,Cardiovascular Research Center (D.E.S., M.N.)
| | - Charles P Lin
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Wellman Center for Photomedicine (Y.J., C.P.L.)
| | - Filip K Swirski
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Department of Imaging, Center for Systems Biology (K.V., D.R., H.-Y.K., G.G., G.W., L.H., F.F.H., V.F., R.N., F.H., Y.J., C.V., R.W., C.P.L., F.K.S., M.N.).,Cardiovascular Research Center (D.E.S., M.N.)
| |
Collapse
|
19
|
Liang J, Huang W, Jiang L, Paul C, Li X, Wang Y. Concise Review: Reduction of Adverse Cardiac Scarring Facilitates Pluripotent Stem Cell-Based Therapy for Myocardial Infarction. Stem Cells 2019; 37:844-854. [PMID: 30913336 PMCID: PMC6599570 DOI: 10.1002/stem.3009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine.
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xiangnan Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
20
|
Leoni G, Soehnlein O. (Re) Solving Repair After Myocardial Infarction. Front Pharmacol 2018; 9:1342. [PMID: 30534069 PMCID: PMC6275178 DOI: 10.3389/fphar.2018.01342] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases, including myocardial infarction and its complications such as heart failure, are the leading cause of death worldwide. To date, basic and translational research becomes necessary to unravel the mechanisms of cardiac repair post-myocardial infarction. The local inflammatory tissue response after acute myocardial infarction determines the subsequent healing process. The diversity of leukocytes such as neutrophils, macrophages and lymphocytes contribute to the clearance of dead cells while activating reparative pathways necessary for myocardial healing. Cardiomyocyte death triggers wall thinning, ventricular dilatation, and fibrosis that can cause left ventricular dysfunction and heart failure. The ultimate goal of cardiac repair is to regenerate functionally viable myocardium after myocardial infarction to prevent cardiac death. Current therapies for heart failure after myocardial infarction are limited and non-curative. At the moment in clinic, conventional surgical interventions such as coronary artery bypass graft or percutaneous coronary interventions are only able to partially restore heart function, with a minor improvement in the left ventricular ejection fraction. The goal of this review is to provide an overview of endogenous myocardial repair mechanisms possibly transferable to future treatment strategies. Among the innovative factors identified as essential in cardiac healing, we highlight specialized pro-resolving mediators as the emerging factors that provide the key molecular signals for the activation of the reparative cells in the myocardium.
Collapse
Affiliation(s)
- Giovanna Leoni
- Institute for Cardiovascular Prevention (IPEK), University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Stockholm, Sweden.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Liu YJ, Wang XZ, Wang Y, He RX, Yang L, Jing QM, Liu HW. Correlation between Sex and Prognosis of Acute Aortic Dissection in the Chinese Population. Chin Med J (Engl) 2018; 131:1430-1435. [PMID: 29893359 PMCID: PMC6006807 DOI: 10.4103/0366-6999.233943] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The prevalence, presentation, management, and prognosis of coronary heart disease differ according to sex. Greater understanding on the differences between men and women with acute aortic dissection (AAD) is needed. We aimed to investigate whether sex disparities are found in patients with AAD, and to study sex differences in complications, mortality in-hospital, and long-term. METHODS We included 884 patients enrolled in our institute between June 2002 and May 2016. Considering psychosocial factors, treatments, and the outcomes in men versus those in women with AAD, we explored the association of sex with psychosocial characteristics and mortality risk. For categorical variables, significant differences between groups were assessed with the Chi-square test or Fisher's exact test, and continuous parameters were assessed with Student's t-test. Univariate and stratified survival statistics were computed using Kaplan-Meier analysis. RESULTS A total of 884 patients (76.1% male, mean age 51.4 ± 11.8 years) were included in this study. There were fewer current smokers in female compared with male (17.5% vs. 67.2%, χ2 = 160.06, P < 0.05). The percentage of men who reported regular alcohol consumption was significantly higher than that in women (40.6% vs. 3.8%, χ2 = 100.18, P < 0.05). About 6.2% (55 of 884) of patients with AAD died before vascular or endovascular surgery was performed, 34.4% (304 of 884) of patients underwent surgical procedures, and 52.7% (466 of 884) and 12.8% (113 of 884) of patients received endovascular treatment and medication. Postoperative mortality similar (6.0% vs. 5.6%, respectively, χ2 = 0.03, P = 0.91) between men and women. Follow-up was completed in 653 of 829 patients (78.8%). Adjustment for age, history of coronary disease, hypertension, smoking and drinking, Type A and use of beta-blocker, angiotensin II receptor blockers, angiotensin converting enzyme (ACE) inhibitor, calcium-channel blockers and statins by multivariate logistic regression analysis suggested that age (odds ratios [OR s], 1.04; 95% confidence interval [CI], 1.01-1.07; P < 0.05), using of calcium-channel blockers (OR, 0.37; 95% CI, 0.18-0.74; P < 0.05), at discharge were independent predictors of late mortality, ACE inhibitors (OR, 1.91; 95% CI, 1.03-3.54; P = 0.04) was independent risk factor of late mortality. CONCLUSIONS In Chinese with AAD, sex is not independently associated with long-term clinical outcomes. Age, the intake of calcium-channel blockers at discharge might help to improve long-term outcomes.
Collapse
Affiliation(s)
- Yan-Jie Liu
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Xiao-Zeng Wang
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Ya Wang
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Rui-Xia He
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Lin Yang
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Quan-Min Jing
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Hai-Wei Liu
- Department of Cardiology, Institute of Cardiovascular Research, General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| |
Collapse
|