1
|
Wu Y, Xu Y, Xu L. Pharmacological therapy targeting the immune response in atherosclerosis. Int Immunopharmacol 2024; 141:112974. [PMID: 39168023 DOI: 10.1016/j.intimp.2024.112974] [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: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by the formation of atherosclerotic plaques that consist of numerous cells including smooth muscle cells, endothelial cells, immune cells, and foam cells. The most abundant innate and adaptive immune cells, including neutrophils, monocytes, macrophages, B cells, and T cells, play a pivotal role in the inflammatory response, lipoprotein metabolism, and foam cell formation to accelerate atherosclerotic plaque formation. In this review, we have discussed the underlying mechanisms of activated immune cells in promoting AS and reviewed published clinical trials for the treatment of AS by suppressing immune cell activation. We have also presented some crucial shortcomings of current clinical trials. Lastly, we have discussed the therapeutic potential of novel compounds, including herbal medicine and dietary food, in alleviating AS in animals. Despite these limitations, further clinical trials and experimental studies will enhance our understanding of the mechanisms modulated by immune cells and promote widespread drug use to treat AS by suppressing immune system-induced inflammation.
Collapse
Affiliation(s)
- Yirong Wu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China.
| | - Linhao Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Translational Medicine Research Center, Hangzhou First People's Hospital, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
2
|
Ozawa K, Packwood W, Muller MA, Qi Y, Xie A, Varlamov O, McCarty OJ, Chung D, López JA, Lindner JR. Removal of endothelial surface-associated von villebrand factor suppresses accelerate datherosclerosis after myocardial infarction. J Transl Med 2024; 22:412. [PMID: 38693516 PMCID: PMC11062912 DOI: 10.1186/s12967-024-05231-6] [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: 02/01/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Thromboinflammation involving platelet adhesion to endothelial surface-associated von Willebrand factor (VWF) has been implicated in the accelerated progression of non-culprit plaques after MI. The aim of this study was to use arterial endothelial molecular imaging to mechanistically evaluate endothelial-associated VWF as a therapeutic target for reducing remote plaque activation after myocardial infarction (MI). METHODS Hyperlipidemic mice deficient for the low-density lipoprotein receptor and Apobec-1 underwent closed-chest MI and were treated chronically with either: (i) recombinant ADAMTS13 which is responsible for proteolytic removal of VWF from the endothelial surface, (ii) N-acetylcysteine (NAC) which removes VWF by disulfide bond reduction, (iii) function-blocking anti-factor XI (FXI) antibody, or (iv) no therapy. Non-ischemic controls were also studied. At day 3 and 21, ultrasound molecular imaging was performed with probes targeted to endothelial-associated VWF A1-domain, platelet GPIbα, P-selectin and vascular cell adhesion molecule-1 (VCAM-1) at lesion-prone sites of the aorta. Histology was performed at day 21. RESULTS Aortic signal for P-selectin, VCAM-1, VWF, and platelet-GPIbα were all increased several-fold (p < 0.01) in post-MI mice versus sham-treated animals at day 3 and 21. Treatment with NAC and ADAMTS13 significantly attenuated the post-MI increase for all four molecular targets by > 50% (p < 0.05 vs. non-treated at day 3 and 21). On aortic root histology, mice undergoing MI versus controls had 2-4 fold greater plaque size and macrophage content (p < 0.05), approximately 20-fold greater platelet adhesion (p < 0.05), and increased staining for markers of platelet transforming growth factor-β1 signaling. Accelerated plaque growth and inflammatory activation was almost entirely prevented by ADAMTS13 and NAC. Inhibition of FXI had no significant effect on molecular imaging signal or plaque morphology. CONCLUSIONS Plaque inflammatory activation in remote arteries after MI is strongly influenced by VWF-mediated platelet adhesion to the endothelium. These findings support investigation into new secondary preventive therapies for reducing non-culprit artery events after MI.
Collapse
Affiliation(s)
- Koya Ozawa
- Sydney Medical School Nepean, Faculty of Medicine and Health, Department of Cardiology, The University of Sydney, Nepean Hospital, Sydney, NSW, Australia
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Matthew A Muller
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Aris Xie
- Cardiovascular Division and Robert M. Berne Cardiovascular Research Center, University of Virginia, Box 801394, 415 Lane Rd, Charlottesville, VA, 22908, USA
| | - Oleg Varlamov
- Oregon National Primate Research Center, Portland, OR, USA
| | - Owen J McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, USA
| | - Dominic Chung
- BloodWorks Research Institute, University of Washington, Seattle, WA, USA
| | - José A López
- BloodWorks Research Institute, University of Washington, Seattle, WA, USA
| | - Jonathan R Lindner
- Cardiovascular Division and Robert M. Berne Cardiovascular Research Center, University of Virginia, Box 801394, 415 Lane Rd, Charlottesville, VA, 22908, USA.
| |
Collapse
|
3
|
Amadio P, Sandrini L, Zarà M, Barbieri SS, Ieraci A. NADPH-oxidases as potential pharmacological targets for thrombosis and depression comorbidity. Redox Biol 2024; 70:103060. [PMID: 38310682 PMCID: PMC10848036 DOI: 10.1016/j.redox.2024.103060] [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: 12/08/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/06/2024] Open
Abstract
There is a complex interrelationship between the nervous system and the cardiovascular system. Comorbidities of cardiovascular diseases (CVD) with mental disorders, and vice versa, are prevalent. Adults with mental disorders such as anxiety and depression have a higher risk of developing CVD, and people with CVD have an increased risk of being diagnosed with mental disorders. Oxidative stress is one of the many pathways associated with the pathophysiology of brain and cardiovascular disease. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is one of the major generators of reactive oxygen species (ROS) in mammalian cells, as it is the enzyme that specifically produces superoxide. This review summarizes recent findings on the consequences of NOX activation in thrombosis and depression. It also discusses the therapeutic effects and pharmacological strategies of NOX inhibitors in CVD and brain disorders. A better comprehension of these processes could facilitate the development of new therapeutic approaches for the prevention and treatment of the comorbidity of thrombosis and depression.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060, Novedrate (CO), Italy; Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
| |
Collapse
|
4
|
Hu X, Zhao P, Zhang J, Zhu Y, Zhou W, Hong K, Sun R, Wang Y, Lu Y, Liu Y. Ultrasound-assisted biomimetic nanobubbles for targeted treatment of atherosclerosis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 51:102682. [PMID: 37105342 DOI: 10.1016/j.nano.2023.102682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/13/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
Cardiovascular disease caused by atherosclerosis remains the main reason of death in the worldwide scale. Although oxidative stress plays a key role in the initiation and progression of atherosclerosis, current antioxidant drugs have limited efficacy. To resolve this problem, we constructed Nox2 siRNA-loaded nanobubbles (PNBs-siNox2) coated with platelet membranes to utilize their antioxidant stress activity and targeting effect for atherosclerosis treatment. After platelet membranes modification, the capacity of PNB to target collagen, foam cells, or human umbilical vein endothelial cells (HUVECs) was significantly increased. Moreover, our study demonstrated that under ultrasonic irradiation, biomimetic nanobubbles were more effective at targeting atherosclerotic plaques and delivering genes into cells. In the present study, we provided a biomimetic gene loading strategy based on nanoplatform for noninvasive, precise and efficient therapy of atherosclerosis, which further improved the efficiency of gene transfection and effectively slowed the progression of atherosclerotic plaques when combined with ultrasound.
Collapse
Affiliation(s)
- Xin Hu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Pengxuan Zhao
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Jun Zhang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Ying Zhu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Wei Zhou
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Kai Hong
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Ruiying Sun
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China
| | - Yuxue Wang
- Department of Ultrasound, The Affifiliated Hospital of Yunnan University, Kunming 650021, China
| | - Yongping Lu
- Department of Ultrasound, The Affifiliated Hospital of Yunnan University, Kunming 650021, China.
| | - Yani Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan 430030, China.
| |
Collapse
|
5
|
Li Z, Bi R, Sun S, Chen S, Chen J, Hu B, Jin H. The Role of Oxidative Stress in Acute Ischemic Stroke-Related Thrombosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8418820. [PMID: 36439687 PMCID: PMC9683973 DOI: 10.1155/2022/8418820] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/13/2022] [Accepted: 11/02/2022] [Indexed: 09/22/2023]
Abstract
Acute ischemic stroke is a serious life-threatening disease that affects almost 600 million people each year throughout the world with a mortality of more than 10%, while two-thirds of survivors remain disabled. However, the available treatments for ischemic stroke are still limited to thrombolysis and/or mechanical thrombectomy, and there is an urgent need for developing new therapeutic target. Recently, intravascular oxidative stress, derived from endothelial cells, platelets, and leukocytes, has been found to be tightly associated with stroke-related thrombosis. It not only promotes primary thrombus formation by damaging endothelial cells and platelets but also affects thrombus maturation and stability by modifying fibrin components. Thus, oxidative stress is expected to be a novel target for the prevention and treatment of ischemic stroke. In this review, we first discuss the mechanisms by which oxidative stress promotes stroke-related thrombosis, then summarize the oxidative stress biomarkers of stroke-related thrombosis, and finally put forward an antithrombotic therapy targeting oxidative stress in ischemic stroke.
Collapse
Affiliation(s)
- Zhifang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuai Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shengcai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiefang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Platelet Redox Imbalance in Hypercholesterolemia: A Big Problem for a Small Cell. Int J Mol Sci 2022; 23:ijms231911446. [PMID: 36232746 PMCID: PMC9570056 DOI: 10.3390/ijms231911446] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance between reactive oxygen species (ROS) synthesis and their scavenging by anti-oxidant defences is the common soil of many disorders, including hypercholesterolemia. Platelets, the smallest blood cells, are deeply involved in the pathophysiology of occlusive arterial thrombi associated with myocardial infarction and stroke. A great deal of evidence shows that both increased intraplatelet ROS synthesis and impaired ROS neutralization are implicated in the thrombotic process. Hypercholesterolemia is recognized as cause of atherosclerosis, cerebro- and cardiovascular disease, and, closely related to this, is the widespread acceptance that it strongly contributes to platelet hyperreactivity via direct oxidized LDL (oxLDL)-platelet membrane interaction via scavenger receptors such as CD36 and signaling pathways including Src family kinases (SFK), mitogen-activated protein kinases (MAPK), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In turn, activated platelets contribute to oxLDL generation, which ends up propagating platelet activation and thrombus formation through a mechanism mediated by oxidative stress. When evaluating the effect of lipid-lowering therapies on thrombogenesis, a large body of evidence shows that the effects of statins and proprotein convertase subtilisin/kexin type 9 inhibitors are not limited to the reduction of LDL-C but also to the down-regulation of platelet reactivity mainly by mechanisms sensitive to intracellular redox balance. In this review, we will focus on the role of oxidative stress-related mechanisms as a cause of platelet hyperreactivity and the pathophysiological link of the pleiotropism of lipid-lowering agents to the beneficial effects on platelet function.
Collapse
|
7
|
Tyagi T, Jain K, Gu SX, Qiu M, Gu VW, Melchinger H, Rinder H, Martin KA, Gardiner EE, Lee AI, Ho Tang W, Hwa J. A guide to molecular and functional investigations of platelets to bridge basic and clinical sciences. NATURE CARDIOVASCULAR RESEARCH 2022; 1:223-237. [PMID: 37502132 PMCID: PMC10373053 DOI: 10.1038/s44161-022-00021-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/17/2022] [Indexed: 07/29/2023]
Abstract
Platelets have been shown to be associated with pathophysiological process beyond thrombosis, demonstrating critical additional roles in homeostatic processes, such as immune regulation, and vascular remodeling. Platelets themselves can have multiple functional states and can communicate and regulate other cells including immune cells and vascular smooth muscle cells, to serve such diverse functions. Although traditional platelet functional assays are informative and reliable, they are limited in their ability to unravel platelet phenotypic heterogeneity and interactions. Developments in methods such as electron microscopy, flow cytometry, mass spectrometry, and 'omics' studies, have led to new insights. In this Review, we focus on advances in platelet biology and function, with an emphasis on current and promising methodologies. We also discuss technical and biological challenges in platelet investigations. Using coronavirus disease 2019 (COVID-19) as an example, we further describe the translational relevance of these approaches and the possible 'bench-to-bedside' utility in patient diagnosis and care.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Miaoyun Qiu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Hannah Melchinger
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth E Gardiner
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
8
|
Lee BK, Jee HJ, Jung YS. Aβ 1-40-Induced Platelet Adhesion Is Ameliorated by Rosmarinic Acid through Inhibition of NADPH Oxidase/PKC-δ/Integrin α IIbβ 3 Signaling. Antioxidants (Basel) 2021; 10:antiox10111671. [PMID: 34829541 PMCID: PMC8615194 DOI: 10.3390/antiox10111671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/31/2023] Open
Abstract
In platelets, oxidative stress reportedly increases platelet adhesion to vessels, thus promoting the vascular pathology of various neurodegenerative diseases, including Alzheimer’s disease (AD). Recently, it has been shown that β-amyloid (Aβ) can increase oxidative stress in platelets; however, the underlying mechanism remains elusive. In the present study, we aimed to elucidate the signaling pathway of platelet adhesion induced by Aβ1–40, the major form of circulating Aβ, through Western blotting, immunofluorescence confocal microscopy, and fluorescence-activated cell sorting analysis. Additionally, we examined whether rosmarinic acid (RA), a natural polyphenol antioxidant, can modulate these processes. Our results show that Aβ1–40-induced platelet adhesion is mediated through NADPH oxidase/ROS/PKC-δ/integrin αIIbβ3 signaling, and these signaling pathways are significantly inhibited by RA. Collectively, these results suggest that RA may have beneficial effects on platelet-associated vascular pathology in AD.
Collapse
Affiliation(s)
- Bo Kyung Lee
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); (H.J.J.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - Hye Jin Jee
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); (H.J.J.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- KIURI Research Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); (H.J.J.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
9
|
Zhang X, Veliky CV, Birru RL, Barinas-Mitchell E, Magnani JW, Sekikawa A. Potential Protective Effects of Equol (Soy Isoflavone Metabolite) on Coronary Heart Diseases-From Molecular Mechanisms to Studies in Humans. Nutrients 2021; 13:3739. [PMID: 34835997 PMCID: PMC8622975 DOI: 10.3390/nu13113739] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/31/2022] Open
Abstract
Equol, a soy isoflavone-derived metabolite of the gut microbiome, may be the key cardioprotective component of soy isoflavones. Systematic reviews have reported that soy isoflavones have no to very small effects on traditional cardiovascular disease risk factors. However, the potential mechanistic mode of action of equol on non-traditional cardiovascular risk factors has not been systematically reviewed. We searched the PubMed through to July 2021 by using terms for equol and each of the following markers: inflammation, oxidation, endothelial function, vasodilation, atherosclerosis, arterial stiffness, and coronary heart disease. Of the 231 records identified, 69 articles met the inclusion criteria and were summarized. Our review suggests that equol is more lipophilic, bioavailable, and generally more potent compared to soy isoflavones. Cell culture, animal, and human studies show that equol possesses antioxidative, anti-inflammatory, and vasodilatory properties and improves arterial stiffness and atherosclerosis. Many of these actions are mediated through the estrogen receptor β. Overall, equol may have a greater cardioprotective benefit than soy isoflavones. Clinical studies of equol are warranted because equol is available as a dietary supplement.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.Z.); (C.V.V.); (R.L.B.); (E.B.-M.)
| | - Cole V. Veliky
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.Z.); (C.V.V.); (R.L.B.); (E.B.-M.)
| | - Rahel L. Birru
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.Z.); (C.V.V.); (R.L.B.); (E.B.-M.)
| | - Emma Barinas-Mitchell
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.Z.); (C.V.V.); (R.L.B.); (E.B.-M.)
| | - Jared W. Magnani
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Akira Sekikawa
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; (X.Z.); (C.V.V.); (R.L.B.); (E.B.-M.)
| |
Collapse
|
10
|
Brown E, Ozawa K, Moccetti F, Vinson A, Hodovan J, Nguyen TA, Bader L, López JA, Kievit P, Shaw GD, Chung DW, Osborn W, Fu X, Chen J, Lindner JR. Arterial Platelet Adhesion in Atherosclerosis-Prone Arteries of Obese, Insulin-Resistant Nonhuman Primates. J Am Heart Assoc 2021; 10:e019413. [PMID: 33880941 PMCID: PMC8200741 DOI: 10.1161/jaha.120.019413] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Platelet–endothelial interactions are thought to contribute to early atherogenesis. These interactions are potentiated by oxidative stress. We used in vivo molecular imaging to test the hypothesis that platelet–endothelial interactions occur at early stages of plaque development in obese, insulin‐resistant nonhuman primates, and are suppressed by NADPH‐oxidase‐2 inhibition. Methods and Results Six adult rhesus macaques fed a Western‐style diet for a median of 4.0 years were studied at baseline and after 8 weeks of therapy with the NADPH‐oxidase‐2‐inhibitor apocynin (50 mg/kg per day). Six lean control animals were also studied. Measurements included intravenous glucose tolerance test, body composition by dual‐energy X‐ray absorptiometry, carotid intimal medial thickness, carotid artery contrast ultrasound molecular imaging for platelet GPIbα (glycoprotein‐ Ibα) and vascular cell adhesion molecule‐1, and blood oxidative markers on mass spectrometry. Compared with lean controls, animals on a Western‐style diet were obese (median body mass: 16.0 versus 8.7 kg, P=0.003; median truncal fat: 49% versus 20%, P=0.002), were insulin resistant (4‐fold higher insulin–glucose area under the curve on intravenous glucose tolerance test, P=0.002), had 40% larger carotid intimal medial thickness (P=0.004), and exhibited oxidative signatures on proteomics. In obese but not lean animals, signal enhancement on molecular imaging was significantly elevated for GPIbα and vascular cell adhesion molecule‐1. The signal correlated modestly with intimal medial thickness but not with the degree of insulin resistance. Apocynin significantly (P<0.01) reduced median signal for GPIbα by >80% and vascular cell adhesion molecule‐1 signal by 75%, but did not affect intimal medial thickness, body mass, or intravenous glucose tolerance test results. Conclusion In nonhuman primates, diet‐induced obesity and insulin resistance leads to platelet–endothelial adhesion at early atherosclerotic lesion sites, which is associated with the expression of pro‐inflammatory adhesion molecules. These responses appear to be mediated, in part, through oxidative pathways.
Collapse
Affiliation(s)
- Eran Brown
- Knight Cardiovascular Institute Portland OR
| | - Koya Ozawa
- Knight Cardiovascular Institute Portland OR
| | | | - Amanda Vinson
- Oregon National Primate Research CenterOregon Health & Science University Portland OR
| | | | | | - Lindsay Bader
- Oregon National Primate Research CenterOregon Health & Science University Portland OR
| | | | - Paul Kievit
- Oregon National Primate Research CenterOregon Health & Science University Portland OR
| | | | | | | | - Xiaoyun Fu
- Bloodworks Research Institute Seattle WA
| | | | - Jonathan R Lindner
- Knight Cardiovascular Institute Portland OR.,Oregon National Primate Research CenterOregon Health & Science University Portland OR
| |
Collapse
|
11
|
Ozawa K, Muller MA, Varlamov O, Tavori H, Packwood W, Mueller PA, Xie A, Ruggeri Z, Chung D, López JA, Lindner JR. Proteolysis of Von Willebrand Factor Influences Inflammatory Endothelial Activation and Vascular Compliance in Atherosclerosis. ACTA ACUST UNITED AC 2020; 5:1017-1028. [PMID: 33145464 PMCID: PMC7591934 DOI: 10.1016/j.jacbts.2020.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
Abstract
In murine models of atherosclerosis, excess endothelial-associated vWF results not only in platelet adhesion, but also endothelial expression of leukocyte adhesion molecules, indicating a role of platelets in endothelial activation. The events triggered by excess endothelial-associated vWF lead to accelerated plaque growth and abnormal arterial mechanical properties. The cellular and molecular events described herein can be assessed noninvasively through molecular imaging.
This study used in vivo molecular imaging to characterize endotheliall activation attributable to von Willebrand factor (vWF)-mediated platelet adhesion in atherosclerosis. In atherosclerotic mice lacking the low-density lipoprotein receptor on Western diet, the additional genetic deletion of the ADAMTS13, which cleaves endothelial-associated vWF, produced greater aortic molecular imaging signal for not only vWF and platelets, but also for endothelial adhesion molecules VCAM1 and P-selectin, larger plaque size, and lower aortic distensibility. Sustained ADAMTS13 therapy reduced signal for all 4 molecular targets and plaque size. We conclude that excess endothelial-associated vWF contributes to not only platelet adhesion, but also to up-regulation of endothelial cell adhesion molecules.
Collapse
Key Words
- AD13−/−, deficient for ADAMTS13
- Apo-E−/−, deficient for apolipoprotein-E
- BP, blood pressure
- GPIbα, glycoprotein-Ibα
- LDL, low-density lipoprotein
- LDL-R, low-density lipoprotein receptor
- LDL-R−/−, deficient for low-density lipoprotein receptor
- MB, microbubble
- NFκB, nuclear factor κ-light-chain-enhancer of activated B cells
- WSD, Western-style diet
- atherosclerosis
- molecular imaging
- platelets
- vWF, von Willebrand factor
- von Willebrand factor
Collapse
Affiliation(s)
- Koya Ozawa
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Matthew A. Muller
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Oleg Varlamov
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon
| | - Hagai Tavori
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Paul A. Mueller
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Zaverio Ruggeri
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, California
| | | | | | - Jonathan R. Lindner
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon
- Address for correspondence: Dr. Jonathan R. Lindner, Cardiovascular Division, UHN-62, Oregon Health and Science University, 3181 Southwest Sam Jackson Park Road, Portland, Oregon 97239.
| |
Collapse
|
12
|
Molecular Ultrasound Imaging. NANOMATERIALS 2020; 10:nano10101935. [PMID: 32998422 PMCID: PMC7601169 DOI: 10.3390/nano10101935] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
In the last decade, molecular ultrasound imaging has been rapidly progressing. It has proven promising to diagnose angiogenesis, inflammation, and thrombosis, and many intravascular targets, such as VEGFR2, integrins, and selectins, have been successfully visualized in vivo. Furthermore, pre-clinical studies demonstrated that molecular ultrasound increased sensitivity and specificity in disease detection, classification, and therapy response monitoring compared to current clinically applied ultrasound technologies. Several techniques were developed to detect target-bound microbubbles comprising sensitive particle acoustic quantification (SPAQ), destruction-replenishment analysis, and dwelling time assessment. Moreover, some groups tried to assess microbubble binding by a change in their echogenicity after target binding. These techniques can be complemented by radiation force ultrasound improving target binding by pushing microbubbles to vessel walls. Two targeted microbubble formulations are already in clinical trials for tumor detection and liver lesion characterization, and further clinical scale targeted microbubbles are prepared for clinical translation. The recent enormous progress in the field of molecular ultrasound imaging is summarized in this review article by introducing the most relevant detection technologies, concepts for targeted nano- and micro-bubbles, as well as their applications to characterize various diseases. Finally, progress in clinical translation is highlighted, and roadblocks are discussed that currently slow the clinical translation.
Collapse
|
13
|
Jani V, Shivaram P, Porter TR, Kutty S. Ultrasound Theranostics in Adult and Pediatric Cardiovascular Research. Cardiovasc Drugs Ther 2020; 35:185-190. [PMID: 32495071 DOI: 10.1007/s10557-020-07016-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Theranostics, the practice of systematically integrating diagnostics with treatment, has evolved as a field of medicine. In the context of ultrasound based theranostics, both traditional microbubbles and inorganic nanoparticles have emerged as technologies of clinical interest. Ultrasound induced microbubble cavitation has demonstrated efficacy in a variety of applications, including thrombolysis, tumor ablation, targeted microvascular flow enhancement, and targeted drug and gene delivery. This commentary summarizes the mechanisms and applications of ultrasound-based theranostics in cardiovascular medicine, including its impact in pediatric cardiology. It also provides an overview of ongoing clinical trials for theranostics in cardiovascular medicine.
Collapse
Affiliation(s)
- Vivek Jani
- The Helen B. Taussig Heart Center, The Johns Hopkins Hospital, The Johns Hopkins University School of Medicine, M2315, 1800 Orleans St, Baltimore, MD, 21287, USA
| | - Pushpa Shivaram
- Division of Cardiology, Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Thomas R Porter
- Division of Cardiology, University of Nebraska Medical Center College of Medicine, Omaha, NE, USA
| | - Shelby Kutty
- The Helen B. Taussig Heart Center, The Johns Hopkins Hospital, The Johns Hopkins University School of Medicine, M2315, 1800 Orleans St, Baltimore, MD, 21287, USA.
| |
Collapse
|
14
|
Kosareva A, Abou-Elkacem L, Chowdhury S, Lindner JR, Kaufmann BA. Seeing the Invisible-Ultrasound Molecular Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:479-497. [PMID: 31899040 DOI: 10.1016/j.ultrasmedbio.2019.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
Ultrasound molecular imaging has been developed in the past two decades with the goal of non-invasively imaging disease phenotypes on a cellular level not depicted on anatomic imaging. Such techniques already play a role in pre-clinical research for the assessment of disease mechanisms and drug effects, and are thought to in the future contribute to earlier diagnosis of disease, assessment of therapeutic effects and patient-tailored therapy in the clinical field. In this review, we first describe the chemical composition and structure as well as the in vivo behavior of the ultrasound contrast agents that have been developed for molecular imaging. We then discuss the strategies that are used for targeting of contrast agents to specific cellular targets and protocols used for imaging. Next we describe pre-clinical data on imaging of thrombosis, atherosclerosis and microvascular inflammation and in oncology, including the pathophysiological principles underlying the selection of targets in each area. Where applicable, we also discuss efforts that are currently underway for translation of this technique into the clinical arena.
Collapse
Affiliation(s)
- Alexandra Kosareva
- Cardiovascular Molecular Imaging, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lotfi Abou-Elkacem
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California, USA
| | - Sayan Chowdhury
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford, California, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Portland, Oregon, USA; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Beat A Kaufmann
- Cardiovascular Molecular Imaging, Department of Biomedicine, University of Basel, Basel, Switzerland; Department of Cardiology, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
15
|
Ultrasound molecular imaging: insights into cardiovascular pathology. J Echocardiogr 2020; 18:86-93. [PMID: 32056137 PMCID: PMC7244457 DOI: 10.1007/s12574-020-00463-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/16/2020] [Accepted: 01/27/2020] [Indexed: 01/06/2023]
Abstract
Similar to what has already occurred in cancer medicine, the management of cardiovascular conditions will likely be improved by non-invasive molecular imaging technologies that can provide earlier or more accurate diagnosis. These techniques are already having a positive impact in pre-clinical research by providing insight into pathophysiology or efficacy of new therapies. Contrast enhanced ultrasound (CEU) molecular imaging is a technique that relies on the ultrasound detection of targeted microbubble contrast agents to examine molecular or cellular events that occur at the blood pool-endothelial interface. CEU molecular imaging techniques have been developed that are able to provide unique information on atherosclerosis, ischemia reperfusion injury, angiogenesis, vascular inflammation, and thrombus formation. Accordingly, CEU has the potential to be used in a wide variety of circumstances to detect disease early or at the bedside, and to guide appropriate therapy based on vascular phenotype. This review will describe the physical basis for CEU molecular imaging, and the specific disease processes for the pre-clinical translational research experience.
Collapse
|
16
|
Moccetti F, Brown E, Xie A, Packwood W, Qi Y, Ruggeri Z, Shentu W, Chen J, López JA, Lindner JR. Myocardial Infarction Produces Sustained Proinflammatory Endothelial Activation in Remote Arteries. J Am Coll Cardiol 2019; 72:1015-1026. [PMID: 30139430 DOI: 10.1016/j.jacc.2018.06.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/07/2018] [Accepted: 06/10/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND In the months after acute myocardial infarction (MI), risk for acute atherothrombotic events in nonculprit arteries increases several fold. OBJECTIVES This study investigated whether sustained proinflammatory and prothrombotic endothelial alterations occur in remote vessels after MI. METHODS Wild-type mice, atherosclerotic mice with double knockout (DKO) of the low-density lipoprotein receptor and Apobec-1, and DKO mice treated with the Nox-inhibitor apocynin were studied at baseline and at 3 and 21 days after closed-chest MI. Ultrasound molecular imaging of P-selectin, vascular cell adhesion molecule (VCAM)-1, von Willebrand factor (VWF) A1-domain, and platelet GPIbα was performed. Intravital microscopy was used to characterize post-MI leukocyte and platelet recruitment in the remote microcirculation after MI. RESULTS Aortic molecular imaging for P-selectin, VCAM-1, VWF-A1, and platelets was increased several-fold (p < 0.01) 3 days post-MI for both wild-type and DKO mice. At 21 days, these changes resolved in wild-type mice but persisted in DKO mice. Signal for platelet adhesion was abolished 1 h after administration of ADAMTS13, which regulates VWF multimerization. In DKO and wild-type mice, apocynin significantly attenuated the post-MI increase for molecular targets, and platelet depletion significantly reduced P-selectin and VCAM-1 signal. On intravital microscopy, MI resulted in remote vessel leukocyte adhesion and platelet string or net complexes. On histology, high-risk inflammatory features in aortic plaque increased in DKO mice 21 days post-MI, which were completely prevented by apocynin. CONCLUSIONS Acute MI stimulates a spectrum of changes in remote vessels, including up-regulation of endothelial inflammatory adhesion molecules and platelet-endothelial adhesion from endothelial-associated VWF multimers. These remote arterial alterations persist longer in the presence of hyperlipidemia, are associated with accelerated plaque growth and inflammation, and are attenuated by Nox inhibition.
Collapse
Affiliation(s)
- Federico Moccetti
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Eran Brown
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Zaverio Ruggeri
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, California
| | - Weihui Shentu
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | | | | | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
17
|
Abstract
Noninvasive imaging has played an increasing role in the process of cardiovascular drug development. This review focuses specifically on the use of molecular imaging, which has been increasingly applied to improve and accelerate certain preclinical steps in drug development, including the identification of appropriate therapeutic targets, evaluation of on-target and off-target effects of candidate therapies, assessment of dose response, and the evaluation of drug or biological biodistribution and pharmacodynamics. Unlike the case in cancer medicine, in cardiovascular medicine, molecular imaging has not been used as a primary surrogate clinical end point for drug approval. However, molecular imaging has been applied in early clinical trials, particularly in phase 0 studies, to demonstrate proof-of-concept or to explain variation in treatment effect. Many of these applications where molecular imaging has been used in drug development have involved the retasking of technologies that were originally intended as clinical diagnostics. With greater experience and recognition of the rich information provided by in vivo molecular imaging, it is anticipated that it will increasingly be used to address the enormous time and costs associated with bringing a new drug to clinical launch.
Collapse
Affiliation(s)
- Jonathan R Lindner
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland.
| | - Jeanne Link
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland
| |
Collapse
|
18
|
Oceans as a Source of Immunotherapy. Mar Drugs 2019; 17:md17050282. [PMID: 31083446 PMCID: PMC6562586 DOI: 10.3390/md17050282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023] Open
Abstract
Marine flora is taxonomically diverse, biologically active, and chemically unique. It is an excellent resource, which offers great opportunities for the discovery of new biopharmaceuticals such as immunomodulators and drugs targeting cancerous, inflammatory, microbial, and fungal diseases. The ability of some marine molecules to mediate specific inhibitory activities has been demonstrated in a range of cellular processes, including apoptosis, angiogenesis, and cell migration and adhesion. Immunomodulators have been shown to have significant therapeutic effects on immune-mediated diseases, but the search for safe and effective immunotherapies for other diseases such as sinusitis, atopic dermatitis, rheumatoid arthritis, asthma and allergies is ongoing. This review focuses on the marine-originated bioactive molecules with immunomodulatory potential, with a particular focus on the molecular mechanisms of specific agents with respect to their targets. It also addresses the commercial utilization of these compounds for possible drug improvement using metabolic engineering and genomics.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Non-invasive molecular imaging is currently used as a research technique to better understand disease pathophysiology. There are also many potential clinical applications where molecular imaging may provide unique information that allows either earlier or more definitive diagnosis, or can guide precision medicine-based decisions on therapy. Contrast-enhanced ultrasound (CEU) with targeted microbubble contrast agents is one such technique that has been developed that has the unique properties of providing rapid information and revealing information only on events that occur within the vascular space. RECENT FINDINGS CEU molecular probes have been developed for a wide variety of disease states including atherosclerosis, vascular inflammation, thrombosis, tumor neovascularization, and ischemic injury. While the technique has not yet been adapted to clinical use, it has been used to reveal pathological processes, to identify new therapeutic targets, and to test the efficacy of novel treatments. This review will explore the physical basis for CEU molecular imaging, its strengths and limitations compared to other molecular imaging modalities, and the pre-clinical translational research experience.
Collapse
Affiliation(s)
- Eran Brown
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cardiovascular Institute, UHN-62, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, UHN-62, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA. .,Oregon National Primate Research Center (J.R.L.), Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
20
|
Ozawa K, Packwood W, Varlamov O, Qi Y, Xie A, Wu MD, Ruggeri Z, López J, Lindner JR. Molecular Imaging of VWF (von Willebrand Factor) and Platelet Adhesion in Postischemic Impaired Microvascular Reflow. Circ Cardiovasc Imaging 2018; 11:e007913. [PMID: 30571316 PMCID: PMC6309798 DOI: 10.1161/circimaging.118.007913] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/13/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Complete mechanistic understanding of impaired microvascular reflow after myocardial infarction will likely lead to new therapies for reducing infarct size. Myocardial contrast echocardiography perfusion imaging and molecular imaging were used to evaluate the contribution of microvascular endothelial-associated VWF (von Willebrand factor) and platelet adhesion to microvascular no-reflow. METHODS AND RESULTS Myocardial infarction was produced by transient LAD ligation in WT (wild type) mice, WT mice treated with the VWF proteolytic enzyme ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13), and ADAMTS13-deficient (ADAMTS13-/-) mice. Myocardial contrast echocardiography perfusion imaging and molecular imaging of VWF and platelet GP (glycoprotein) Ibα were performed 30 minutes after ischemia-reperfusion. Infarct size was measured at 3 days. Mortality during ischemia-reperfusion incrementally increased in WT+ADAMTS13, WT, and ADAMTS13-/- mice (14%, 43%, and 63%, respectively; P<0.05). For WT mice, molecular imaging signal for platelets and VWF in the postischemic risk area was 4- to 5-fold higher ( P<0.05) compared with both the remote nonischemic regions or to sham-treated mice. Signal enhancement in the risk area was completely abolished by ADAMTS13 treatment for both platelets (12.8±3.3 versus -1.0±4.4 IU; P<0.05) and VWF (13.9±4.0 versus -1.0±3.0 IU; P<0.05). ADAMTS13-/- compared with WT mice had 2- to 3-fold higher risk area signal for platelets (33.1±8.5 IU) and VWF (30.9±1.9 IU). Microvascular reflow in the risk area incrementally decreased for WT+ADAMTS13, WT, and ADAMTS13-/- mice ( P<0.05), whereas infarct size incrementally increased ( P<0.05). CONCLUSIONS Mechanistic information on microvascular no-reflow is possible by combining perfusion and molecular imaging. In reperfused myocardial infarction, excess endothelial-associated VWF and secondary platelet adhesion in the risk area microcirculation contribute to impaired reflow and are modifiable.
Collapse
Affiliation(s)
- Koya Ozawa
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Oleg Varlamov
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
| | - Melinda D. Wu
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
- Doernbecher’s Children’s Hospital, Oregon Health & Science University, Portland, OR
| | - Zaverio Ruggeri
- Department of Molecular & Experimental Medicine, Scripps Research Institute, La Jolla, CA
| | | | - Jonathan R. Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| |
Collapse
|
21
|
Atkinson T, Packwood W, Xie A, Liang S, Qi Y, Ruggeri Z, Lopez J, Davidson BP, Lindner JR. Assessment of Novel Antioxidant Therapy in Atherosclerosis by Contrast Ultrasound Molecular Imaging. J Am Soc Echocardiogr 2018; 31:1252-1259.e1. [PMID: 30213420 PMCID: PMC6218294 DOI: 10.1016/j.echo.2018.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ultrasound molecular imaging was used to evaluate the therapeutic effects of antioxidant therapy with EUK-207, which has superoxide dismutase and catalase activities, on suppressing high-risk atherosclerotic features. METHODS Mice with age-dependent atherosclerosis produced by deletion of the low-density lipoprotein receptor and Apobec-1 were studied at 20 and 40 weeks of age. EUK-207 or vehicle was administered for the preceding 8 weeks. Therapy for 28 weeks was also studied for 40-week-old mice. Ultrasound molecular imaging of the thoracic aorta was performed with contrast agents targeted to endothelial P-selectin, von Willebrand factor A1-domain, and platelet glycoprotein Ibα or control agent. Aortic plaque area and macrophage content were assessed by histology. RESULTS In 20-week-old double-knockout mice, EUK-207 compared with sham therapy produced only nonsignificant trends for reduction in molecular imaging signal for endothelial P-selectin, von Willebrand factor A1-domain, and platelet adhesion. At 40 weeks, EUK-207 given for 8 or 28 weeks significantly (P < .05) reduced signal for all three endothelial-associated events essentially to background levels, with the exception of glycoprotein Ibα signal after 8 weeks (P = .06). On aortic histology, EUK-207 therapy for 8 weeks did not affect plaque area or macrophage content at either age. However, EUK-207 for 28 weeks almost completely suppressed plaque development (350 ± 258 vs 4 ± 6 × 103 μm2, P = .014) and macrophage content (136 ± 103 vs 3 ± 2 × 103 μm2, P = .002) compared with control mice at 40 weeks. CONCLUSIONS Molecular imaging can be used to assess vascular responses to antioxidants and has demonstrated that certain antioxidants reduce vascular endothelial activation and platelet adhesion, but reductions in plaque size and macrophage content occurs only with long-duration therapy that is started early.
Collapse
Affiliation(s)
- Tamara Atkinson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Portland VA Medical Center, Portland, Oregon
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Sherry Liang
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Zaverio Ruggeri
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, California
| | | | - Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Portland VA Medical Center, Portland, Oregon
| | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
22
|
NADPH oxidase 2 (NOX2): A key target of oxidative stress-mediated platelet activation and thrombosis. Trends Cardiovasc Med 2018; 28:429-434. [DOI: 10.1016/j.tcm.2018.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/24/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
|
23
|
Moccetti F, Weinkauf CC, Davidson BP, Belcik JT, Marinelli ER, Unger E, Lindner JR. Ultrasound Molecular Imaging of Atherosclerosis Using Small-Peptide Targeting Ligands Against Endothelial Markers of Inflammation and Oxidative Stress. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:1155-1163. [PMID: 29548756 DOI: 10.1016/j.ultrasmedbio.2018.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study was to evaluate a panel of endothelium-targeted microbubble (MB) ultrasound contrast agents bearing small peptide ligands as a human-ready approach for molecular imaging of markers of high-risk atherosclerotic plaque. Small peptide ligands with established affinity for human P-selectin, VCAM-1, LOX-1 and von Willebrand factor (VWF) were conjugated to the surface of lipid-stabilized MBs. Contrast-enhanced ultrasound (CEUS) molecular imaging of the thoracic aorta was performed in wild-type and gene-targeted mice with advanced atherosclerosis (DKO). Histology was performed on carotid endarterectomy samples from patients undergoing surgery for unstable atherosclerosis to assess target expression in humans. In DKO mice, CEUS signal for all four targeted MBs was significantly higher than that for control MBs, and was three to sevenfold higher than in wild-type mice, with the highest signal achieved for VCAM-1 and VWF. All molecular targets were present on the patient plaque surface but expression was greatest for VCAM-1 and VWF. We conclude that ultrasound contrast agents bearing small peptide ligands feasible for human use can be targeted against endothelial cell adhesion molecules for inflammatory cells and platelets for imaging advanced atherosclerotic disease.
Collapse
Affiliation(s)
- Federico Moccetti
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Craig C Weinkauf
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Brian P Davidson
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Portland Veterans Affairs Medical Center, Portland, Oregon, USA
| | - J Todd Belcik
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
24
|
Porter TR, Mulvagh SL, Abdelmoneim SS, Becher H, Belcik JT, Bierig M, Choy J, Gaibazzi N, Gillam LD, Janardhanan R, Kutty S, Leong-Poi H, Lindner JR, Main ML, Mathias W, Park MM, Senior R, Villanueva F. Clinical Applications of Ultrasonic Enhancing Agents in Echocardiography: 2018 American Society of Echocardiography Guidelines Update. J Am Soc Echocardiogr 2018; 31:241-274. [DOI: 10.1016/j.echo.2017.11.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
25
|
Yan F, Sun Y, Mao Y, Wu M, Deng Z, Li S, Liu X, Xue L, Zheng H. Ultrasound Molecular Imaging of Atherosclerosis for Early Diagnosis and Therapeutic Evaluation through Leucocyte-like Multiple Targeted Microbubbles. Am J Cancer Res 2018; 8:1879-1891. [PMID: 29556362 PMCID: PMC5858506 DOI: 10.7150/thno.22070] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/02/2018] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases resulting from atherosclerosis have become a serious threat to human health. It is well-known that an ongoing inflammatory response is involved during atherosclerosis progression that ultimately results in the accumulation of lipids and formation of plaques. Monitoring the pathological changes during the inflammatory response will be of great significance for early diagnosis and therapeutic evaluation of atherosclerosis. Targeted contrast-enhanced ultrasonography has been shown to be a promising noninvasive imaging technique for evaluating the degree of atherosclerosis and may potentially be translated to clinical imaging in the future. However, inadequate cell adhesion of targeted microbubbles (MBs) in large arterial vessels still remains a great challenge. Methods: By mimicking the leucocytes that are recruited to the vessel wall during the initiation of atherosclerosis through selectin-dependent arrest and cell adhesion molecule-mediated firm cell adhesion, we developed VCAM-1/ICAM-1/P-selectin-targeted MBVIS by integrating VCAM-1 and ICAM-1 antibodies and synthetic polymeric sialyl Lewis X (sLex) onto the MB surface. Results: The resulting MBVIS had a high affinity to inflammatory bEnd.3 cells in both static and dynamic flow conditions. Significantly enhanced ultrasound imaging signals were achieved by MBVIS in detecting the atherosclerosis progress when compared with the single- or dual-targeted MBs. Taking advantage of the artificial MBVIS, less ultrasound imaging signals were found in the atorvastatin-treated, but not placebo-treated, ApoE-deficient mice with atherosclerosis, revealing a potential therapeutic efficacy of atorvastatin for early stage atherosclerosis. This was further confirmed by histologic staining examination. Conclusions: Our study provides a promising ultrasound molecular imaging probe for early-stage diagnosis and therapeutic evaluation of atherosclerosis.
Collapse
|
26
|
Weinkauf CC, Concha-Moore K, Lindner JR, Marinelli ER, Hadinger KP, Bhattacharjee S, Berman SS, Goshima K, Leon LR, Matsunaga TO, Unger E. Endothelial vascular cell adhesion molecule 1 is a marker for high-risk carotid plaques and target for ultrasound molecular imaging. J Vasc Surg 2018; 68:105S-113S. [PMID: 29452833 DOI: 10.1016/j.jvs.2017.10.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Molecular imaging of carotid plaque vulnerability to atheroembolic events is likely to lead to improvements in selection of patients for carotid endarterectomy (CEA). The aims of this study were to assess the relative value of endothelial inflammatory markers for this application and to develop molecular ultrasound contrast agents for their imaging. METHODS Human CEA specimens were obtained prospectively from asymptomatic (30) and symptomatic (30) patients. Plaques were assessed by semiquantitative immunohistochemistry for vascular cell adhesion molecule 1 (VCAM-1), lectin-like oxidized low-density lipoprotein receptor 1, P-selectin, and von Willebrand factor. Established small peptide ligands to each of these targets were then synthesized and covalently conjugated to the surface of lipid-shelled microbubble ultrasound contrast agents, which were then evaluated in a flow chamber for binding kinetics to activated human aortic endothelial cells under variable shear conditions. RESULTS Expression of VCAM-1 on the endothelium of CEA specimens from symptomatic patients was 2.4-fold greater than that from asymptomatic patients (P < .01). Expression was not significantly different between groups for P-selectin (P = .43), von Willebrand factor (P = .59), or lectin-like oxidized low-density lipoprotein receptor 1 (P = .99). Although most plaques from asymptomatic patients displayed low VCAM-1 expression, approximately one in five expressed high VCAM-1 similar to plaques from symptomatic patients. In vitro flow chamber experiments demonstrated that VCAM-1-targeted microbubbles bind cells that express VCAM-1, even under high-shear conditions that approximate those found in human carotid arteries, whereas binding efficiency was lower for the other agents. CONCLUSIONS VCAM-1 displays significantly higher expression on high-risk (symptomatic) vs low-risk (asymptomatic) carotid plaques. Ultrasound contrast agents bearing ligands for VCAM-1 can sustain high-shear attachment and may be useful for identifying patients in whom more aggressive treatment is warranted.
Collapse
Affiliation(s)
- Craig C Weinkauf
- Division of Vascular and Endovascular Surgery, University of Arizona, Tucson, Ariz
| | | | - Jonathan R Lindner
- Division of Cardiovascular Medicine, Oregon Health Sciences University, Portland, Ore
| | | | - Kyle P Hadinger
- Department of Biomedical Engineering, University of Arizona, Tucson, Ariz
| | | | | | - Kay Goshima
- Division of Vascular and Endovascular Surgery, University of Arizona, Tucson, Ariz
| | - Luis R Leon
- Division of Vascular and Endovascular Surgery, University of Arizona, Tucson, Ariz
| | - Terry O Matsunaga
- Department of Biomedical Engineering, University of Arizona, Tucson, Ariz; Department of Medical Imaging, University of Arizona, Tucson, Ariz
| | - Evan Unger
- NuvOx Pharmaceuticals, Tucson, Ariz; Department of Medical Imaging, University of Arizona, Tucson, Ariz.
| |
Collapse
|
27
|
Chen HJ, Roy TL, Wright GA. Perfusion measures for symptom severity and differential outcome of revascularization in limb ischemia: Preliminary results with arterial spin labeling reactive hyperemia. J Magn Reson Imaging 2017; 47:1578-1588. [PMID: 29193492 DOI: 10.1002/jmri.25910] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/13/2017] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Previously, a theoretical model based on microvascular physiology was established to facilitate the interpretation of calf perfusion dynamics recorded by arterial spin labeling (ASL). PURPOSE To investigate the clinical relevance of novel perfusion indices by comparing them to the symptoms, response to revascularization, and short-term functional outcome in patients with peripheral arterial disease (PAD). STUDY TYPE Prospective cohort study. POPULATION Nineteen patients with PAD. FIELD STRENGTH/SEQUENCE Pulsed ASL at 3T. ASSESSMENT The mid-calf reactive hyperemia induced by 2 minutes of arterial occlusion was recorded in PAD patients. The perfusion responses were characterized by the peak, time-to-peak, and physiological model-derived indices including the baseline perfusion fr , arterial resistance Ra , and compliance Ca , and sensitivity gATP and response time τATP of downstream microvasculature to metabolic stress. These indices were compared to the disease severity and outcome within 6 months after revascularization assessed by self-reported symptoms and the ankle-brachial index. Disease severity was categorized as asymptomatic, claudication, or critical limb ischemia. The outcome was categorized as symptom resolved or limited improvement. STATISTICAL TESTS Severity and outcome groups were compared using Mann-Whitney and Kruskal-Wallis tests with Holm-Sidak adjustments. RESULTS The peak perfusion decreased and model arterial resistance increased progressively with increasing severity of limb ischemia (P = 0.0402 and 0.0413, respectively). Eleven patients had a successful endovascular procedure, including six patients who had symptoms resolved, four patients who had remaining leg pain, and one patient lost to follow-up. The subjects with limited improvement had significantly lower preintervention microvascular sensitivity gATP than those with symptoms resolved (8.72 ± 1.46 vs. 4.93 ± 0.91, P = 0.0466). DATA CONCLUSION ASL reactive hyperemia reflects multiple aspects of the pathophysiology. Measures of macrovascular arterial disease are related to the manifested symptom severity, whereas preintervention gATP associated with microvascular dysfunction is related to prognosis following revascularization. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 4 J. Magn. Reson. Imaging 2018;47:1578-1588.
Collapse
Affiliation(s)
- Hou-Jen Chen
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Schulich Heart Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Trisha L Roy
- Schulich Heart Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Division of Vascular Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Graham A Wright
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Schulich Heart Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Maier A, Plaza-Heck P, Meixner F, Guenther F, Kaufmann BA, Kramer M, Heidt T, Zirlik A, Hilgendorf I, Reinöhl J, Stachon P, Bronsert P, Birkemeyer R, Neudorfer I, Peter K, Bode C, von zur Mühlen C. A molecular intravascular ultrasound contrast agent allows detection of activated platelets on the surface of symptomatic human plaques. Atherosclerosis 2017; 267:68-77. [DOI: 10.1016/j.atherosclerosis.2017.10.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022]
|
29
|
Jiang ZZ, Liu XT, Ma CY, He C, Li XY, Hou CL, Cheng ZS, Xia GY. Detection of Atherosclerotic Plaques in the Rabbit Aorta Using Ultrasound Microbubbles Conjugated to Interleukin-18 Antibodies. Med Sci Monit 2017; 23:5446-5454. [PMID: 29142190 PMCID: PMC5701460 DOI: 10.12659/msm.907572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The purpose of the study was to investigate the ability of microbubbles (MBs) targeting interleukin-18 (IL-18) to detect plaques in a rabbit atherosclerotic plaque model. MATERIAL AND METHODS A rabbit atherosclerotic plaque model was established. The locations of the atherosclerotic plaques were verified by two-dimensional scanning and color Doppler flow imaging. An IL-18 antibody was conjugated to naked MBs (MBc) using the biotin-streptavidin conjugation method, resulting in the formation of MBIL-18. MBc and MBIL-18 were then used for contrast-enhanced ultrasound (CEUS) studies. The locations of CD34 and IL-18 within the plaques were determined by immunohistochemistry, and IL-18 expression levels in the plaques were determined by Western blot analysis. The relationships between IL-18 expression and the contrast intensity of the 2 MBs were analyzed. RESULTS MBc and MBIL-18 were both uniformly dispersed. Fluorescence microscopy and flow cytometry revealed that IL-18 was successfully conjugated to MBs. CEUS images showed that the intensity of the MBIL-18 signal was substantially enhanced and prolonged compared with that of the MBc signal. Immunohistochemistry showed that CD34 expression was significantly increased in the plaques and that IL-18 was mainly located in the inner parts and base of the atherosclerotic plaques. Western blot analysis revealed that IL-18 expression was higher in the plaque regions. Correlation analysis showed that IL-18 expression was correlated with the contrast intensity of MBIL-18 (r=0.903, P<0.05) but not with MBc (r=0.540, P>0.05). CONCLUSIONS MBs targeting IL-18 may be a novel, noninvasive method of diagnosing atherosclerotic plaques.
Collapse
Affiliation(s)
- Zhen-Zhen Jiang
- Department of Ultrasound, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Xia-Tian Liu
- Department of Ultrasound, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Cai-Ye Ma
- Department of Ultrasound, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital, Shaoxing, Zhejiang, China (mainland)
| | - Xing-Yun Li
- Department of Ultrasound, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Chuan-Lin Hou
- Department of Pathology, Shaoxing People`s Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| | - Zu-Sheng Cheng
- Department of Radiology, Shaoxing Seventh Hospital, Shaoxing, Zhejiang, China (mainland)
| | - Guo-Yuan Xia
- Department of Ultrasound, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China (mainland)
| |
Collapse
|
30
|
Violi F, Loffredo L, Carnevale R, Pignatelli P, Pastori D. Atherothrombosis and Oxidative Stress: Mechanisms and Management in Elderly. Antioxid Redox Signal 2017; 27:1083-1124. [PMID: 28816059 DOI: 10.1089/ars.2016.6963] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The incidence of cardiovascular events (CVEs) increases with age, representing the main cause of death in an elderly population. Aging is associated with overproduction of reactive oxygen species (ROS), which may affect clotting and platelet activation, and impair endothelial function, thus predisposing elderly patients to thrombotic complications. Recent Advances: There is increasing evidence to suggest that aging is associated with an imbalance between oxidative stress and antioxidant status. Thus, upregulation of ROS-producing enzymes such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and myeloperoxidase, along with downregulation of antioxidant enzymes, such as superoxide dismutase and glutathione peroxidase, occurs during aging. This imbalance may predispose to thrombosis by enhancing platelet and clotting activation and eliciting endothelial dysfunction. Recently, gut-derived products, such as trimethylamine N-oxide (TMAO) and lipopolysaccharide, are emerging as novel atherosclerotic risk factors, and gut microbiota composition has been shown to change by aging, and may concur with the increased cardiovascular risk in the elderly. CRITICAL ISSUES Antioxidant treatment is ineffective in patients at risk or with cardiovascular disease. Further, anti-thrombotic treatment seems to work less in the elderly population. FUTURE DIRECTIONS Interventional trials with antioxidants targeting enzymes implicated in aging-related atherothrombosis are warranted to explore whether modulation of redox status is effective in lowering CVEs in the elderly. Antioxid. Redox Signal. 27, 1083-1124.
Collapse
Affiliation(s)
- Francesco Violi
- 1 I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Roma, Italy
| | - Lorenzo Loffredo
- 1 I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Roma, Italy
| | - Roberto Carnevale
- 1 I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Roma, Italy .,2 Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome , Latina, Italy
| | - Pasquale Pignatelli
- 1 I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Roma, Italy
| | - Daniele Pastori
- 1 I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome , Roma, Italy
| |
Collapse
|
31
|
Sun R, Tian J, Zhang J, Wang L, Guo J, Liu Y. Monitoring inflammation injuries in the progression of atherosclerosis with contrast enhanced ultrasound molecular imaging. PLoS One 2017; 12:e0186155. [PMID: 28982198 PMCID: PMC5628944 DOI: 10.1371/journal.pone.0186155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/26/2017] [Indexed: 01/22/2023] Open
Abstract
Purpose The upregulation of vascular cell adhesion molecule-1(VCAM-1) on vascular endothelium plays a great role in the progression of atherosclerosis (AS). In this study, ultrasound molecular imaging was performed to monitor the inflammation injuries in the onset and progression of atherosclerosis with microbubbles targeted to VCAM-1. Methods Mice deficient for the apolipoprotein E (ApoE-/-mice) with high-cholesterol diet were studied as an age-dependent model of atherosclerosis. At 8, 16, 24, and 32 weeks of age, contrast enhanced ultrasound (CEU) molecular imaging of proximal ascending aorta was performed with microbubbles targeted to VCAM-1. Plaque size, monocytes infiltration and the expression of VCAM-1 in the proximal ascending aorta were assessed by histology and western blot analysis, separately. Results In ApoE-/- mice, molecular imaging for VCAM-1 detected selective signal enhancement (P<0.01 versus non-targeted microbubbles) at all ages of ApoE-/- mice. Moreover, signals from targeted microbubbles increased from 8wks to 32wks age (P<0.05 for trend) in ApoE-/- mice, indicating the upregulation of VCAM-1 with the progression of atherosclerosis. Consistent with CEU imaging results, both western blot analysis and immunohistochemistry revealed the expression of VCAM-1 and monocytes infiltration were age-dependent in ApoE-/- mice. Conclusions CEU molecular imaging can be used to noninvasively detect the VCAM-1 expression on the endothelium in the progression of atherosclerosis. By investigating specific molecular biomarkers, it could help to monitor the inflammation and the progression of AS, which may in some extent contribute to the prediction of vulnerable plaque.
Collapse
Affiliation(s)
- Ruiying Sun
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Tian
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Zhang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liping Wang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Guo
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yani Liu
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
32
|
Ajith Y, Dimri U, Dixit SK, Singh SK, Gopalakrishnan A, Madhesh E, Rajesh JB, Sangeetha SG. Immunomodulatory basis of antioxidant therapy and its future prospects: an appraisal. Inflammopharmacology 2017; 25:10.1007/s10787-017-0393-5. [PMID: 28864996 DOI: 10.1007/s10787-017-0393-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 08/23/2017] [Indexed: 02/06/2023]
Abstract
Antioxidants are agents which can modulate oxidant-antioxidant profile of body system by neutralizing pro-oxidant molecules. The current scientific knowledge on mechanisms of antioxidant activity of biomolecules was critically reviewed with a special emphasis on immunomodulation. The immuno-oxidative wreckage of animals in various disease conditions and the role of biomodulators in curbing the oxidative stress through immune pathways were analyzed. The critical role of immunomodulatory mechanisms in controlling oxidative damage was identified. Selection of antioxidant therapy considering the immunopharmacology of the drug as well as immunological basis of disease may reduce treatment failure and adverse health effects. Hence, it is suggested that future studies on antioxidants may focus on the immuno-oxidative pathobiology to better understand its clinical effects and effective disease management.
Collapse
Affiliation(s)
- Y Ajith
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India.
| | - U Dimri
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| | - S K Dixit
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| | - Shanker K Singh
- Department of Veterinary Medicine, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, UP, 281 001, India
| | - A Gopalakrishnan
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| | - E Madhesh
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| | - J B Rajesh
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| | - S G Sangeetha
- Division of Medicine, Indian Veterinary Research Institute, Izatnagar, UP, 243122, India
| |
Collapse
|
33
|
Apocynin suppressed the nuclear factor-κB pathway and attenuated lung injury in a rat hemorrhagic shock model. J Trauma Acute Care Surg 2017; 82:566-574. [DOI: 10.1097/ta.0000000000001337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Liang G, Vo D, Nguyen PK. Fundamentals of Cardiovascular Molecular Imaging: a Review of Concepts and Strategies. CURRENT CARDIOVASCULAR IMAGING REPORTS 2017. [DOI: 10.1007/s12410-017-9403-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
35
|
Platelets and von Willebrand factor in atherogenesis. Blood 2017; 129:1415-1419. [PMID: 28174163 DOI: 10.1182/blood-2016-07-692673] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/13/2017] [Indexed: 12/17/2022] Open
Abstract
The role of platelet adhesion, activation, and aggregation in acute atherothrombotic events such as myocardial infarction and stroke is well established. There is increasing evidence that platelet-endothelial interactions also contribute to early atherosclerotic plaque initiation and growth. Through these interactions, platelet-derived factors can contribute to the proinflammatory and mitogenic status of resident mural cells. Among the many putative mechanisms for platelet-endothelial interactions, increased endothelial-associated von Willebrand factor, particularly in a multimerized form, which interacts with platelet glycoproteins and integrins, is a major factor and represents a therapeutic target in early atherogenesis.
Collapse
|
36
|
Violi F, Carnevale R, Loffredo L, Pignatelli P, Gallin JI. NADPH Oxidase-2 and Atherothrombosis: Insight From Chronic Granulomatous Disease. Arterioscler Thromb Vasc Biol 2016; 37:218-225. [PMID: 27932349 DOI: 10.1161/atvbaha.116.308351] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023]
Abstract
The phagocytic cell enzyme NADPH oxidase-2 (Nox2) is critical for killing micro-organisms via production of reactive oxygen species and thus is a key element of the innate immune system. Nox2 is also detectable in endothelial cells and platelets where it has vasoconstrictive and aggregating properties, respectively. Patients with X-linked chronic granulomatous disease with hereditary Nox2 deficiency not only have impaired bacterial killing but, in association with loss of Nox2 function, also have enhanced carotid artery dilation, impaired platelet-related thrombosis, and reduced carotid atherosclerotic burden. Experimental studies corroborated these reports in chronic granulomatous disease by demonstrating (1) Nox2 is upregulated in atherosclerotic plaque, and this upregulation significantly correlates with oxidative stress and (2) pharmacological inhibition of Nox2 is associated with a delayed atherosclerotic progression in animal models. Furthermore, the role of Nox2 in platelet-associated thrombosis was substantiated by experiments showing impaired platelet activation in animals treated with a Nox2 inhibitor or impaired platelet aggregation along with reduced platelet-related thrombosis in the mouse knockout model of Nox2. Interestingly, in chronic granulomatous disease patients and in the mouse knockout model of Nox2, no defects of primary hemostasis were detected. This review analyses experimental and clinical data suggesting Nox2 is a potential target for counteracting the atherothrombotic process.
Collapse
Affiliation(s)
- Francesco Violi
- From the Division of I Clinica Medica, Policlinico Umberto I, Sapienza University, Rome, Italy (F.V., L.L., P.P.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C.); and Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.I.G.).
| | - Roberto Carnevale
- From the Division of I Clinica Medica, Policlinico Umberto I, Sapienza University, Rome, Italy (F.V., L.L., P.P.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C.); and Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.I.G.)
| | - Lorenzo Loffredo
- From the Division of I Clinica Medica, Policlinico Umberto I, Sapienza University, Rome, Italy (F.V., L.L., P.P.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C.); and Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.I.G.)
| | - Pasquale Pignatelli
- From the Division of I Clinica Medica, Policlinico Umberto I, Sapienza University, Rome, Italy (F.V., L.L., P.P.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C.); and Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.I.G.)
| | - John I Gallin
- From the Division of I Clinica Medica, Policlinico Umberto I, Sapienza University, Rome, Italy (F.V., L.L., P.P.); Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (R.C.); and Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD (J.I.G.)
| |
Collapse
|
37
|
Zhao W, Wu C, Li S, Chen X. Adiponectin protects palmitic acid induced endothelial inflammation and insulin resistance via regulating ROS/IKKβ pathways. Cytokine 2016; 88:167-176. [DOI: 10.1016/j.cyto.2016.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 12/24/2022]
|
38
|
Guo Y, Liu XC, Wang YJ, Li Q, Yang Q, Weng XG, Chen Y, Cai WY, Kan XX, Chen X, Huang HF, Zhu XX, Li YJ. Effects of Shenlian extract on experimental atherosclerosis in ApoE-deficient mice based on ultrasound biomicroscopy. Altern Ther Health Med 2016; 16:469. [PMID: 27846838 PMCID: PMC5111256 DOI: 10.1186/s12906-016-1449-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 11/07/2016] [Indexed: 11/10/2022]
Abstract
Background This study directly and dynamically investigated the effects of SL extract (i.e., a combination of Radix Salviae miltiorrhizae and Andrographis paniculata extract) on plaque progression in vivo by high resolution ultrasound biomicroscopy (UBM). Methods An atherosclerosis model was established by placing a perivascular collar on the right common carotid artery in apolipoprotein E-deficient (ApoE-/-) mice. Thickness, plaque area and local blood flow were observed by UBM, pathological changes were observed by histochemical staining, and lipid levels were measured by respective commercially available kits. Results Compared with the model group, the SL extract groups showed reduced wall thickness of the aortic arch (GC: P = 0.001, P = 0.002, and P < 0.001; LC: P < 0.001, P < 0.001, and P < 0.001; BC: P = 0.027, P = 0.017, and P = 0.003; respectively), which presented with retarded plaque progression of the cartoid artery with concordantly increased blood flow (P = 0.002 and P < 0.001) as visualized in vivo by UBM. Histological analysis confirmed the reduction of carotid atherosclerosis. Conclusions The SL extract inhibited the formation of atherosclerotic plaques in an ApoE-/- mice model by UBM analysis, and did so by effects that ameliorated local blood flow and improved blood lipid levels.
Collapse
|
39
|
Chatterjee M, Gawaz M. Clinical significance of receptor shedding-platelet GPVI as an emerging diagnostic and therapeutic tool. Platelets 2016; 28:362-371. [PMID: 27753514 DOI: 10.1080/09537104.2016.1227062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Platelet membrane bedecked with a wide array of receptors offers a platform to regulate platelet responsiveness, thrombotic propensity, inflammatory disposition, and immune reactivity under diverse pathophysiological conditions. Ectopic proteolytic cleavage of such receptors irreversibly inactivates receptor-mediated intracellular signaling governing cellular functions, further releases soluble fragments into circulation which might modulate functions of target cells. Glycoprotein VI-(GPVI) is a membrane glycoprotein expressed in platelets and megakaryocytes. Platelet GPVI surface expression is enhanced following acute ischemic events like myocardial infarction and cerebral stroke, serves as an imminent diagnostic tool independent of markers of tissue necrosis, and is associated with poor prognosis. Platelets undergo GPVI shedding and thereby contribute to soluble plasma levels of sGPVI, with distinct diagnostic and prognostic attributes. This review summarizes the functional significance and mechanistic basis whereby GPVI surface availability is up- or downregulated on platelets and the impact of GPVI in diagnostic, prognostic, and therapeutic strategies in diseases where platelets play a regulatory role. Further, we also highlight how novel non-invasive platelet-based diagnostic and therapeutic strategies have evolved utilizing GPVI for lesion-directed antithrombotic therapy or to counteract atherosclerotic disposition to ameliorate care of patients particularly in the context of cardio-cerebro-vascular medicine.
Collapse
Affiliation(s)
- Madhumita Chatterjee
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| | - Meinrad Gawaz
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| |
Collapse
|
40
|
Molecular Imaging of Vulnerable Atherosclerotic Plaques in Animal Models. Int J Mol Sci 2016; 17:ijms17091511. [PMID: 27618031 PMCID: PMC5037788 DOI: 10.3390/ijms17091511] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis is characterized by intimal plaques of the arterial vessels that develop slowly and, in some cases, may undergo spontaneous rupture with subsequent heart attack or stroke. Currently, noninvasive diagnostic tools are inadequate to screen atherosclerotic lesions at high risk of acute complications. Therefore, the attention of the scientific community has been focused on the use of molecular imaging for identifying vulnerable plaques. Genetically engineered murine models such as ApoE−/− and ApoE−/−Fbn1C1039G+/− mice have been shown to be useful for testing new probes targeting biomarkers of relevant molecular processes for the characterization of vulnerable plaques, such as vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, intercellular adhesion molecule (ICAM)-1, P-selectin, and integrins, and for the potential development of translational tools to identify high-risk patients who could benefit from early therapeutic interventions. This review summarizes the main animal models of vulnerable plaques, with an emphasis on genetically altered mice, and the state-of-the-art preclinical molecular imaging strategies.
Collapse
|
41
|
Daeichin V, Kooiman K, Skachkov I, Bosch JG, Theelen TL, Steiger K, Needles A, Janssen BJ, Daemen MJAP, van der Steen AFW, de Jong N, Sluimer JC. Quantification of Endothelial αvβ3 Expression with High-Frequency Ultrasound and Targeted Microbubbles: In Vitro and In Vivo Studies. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2283-2293. [PMID: 27302657 DOI: 10.1016/j.ultrasmedbio.2016.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 06/06/2023]
Abstract
Angiogenesis is a critical feature of plaque development in atherosclerosis and might play a key role in both the initiation and later rupture of plaques. The precursory molecular or cellular pro-angiogenic events that initiate plaque growth and that ultimately contribute to plaque instability, however, cannot be detected directly with any current diagnostic modality. This study was designed to investigate the feasibility of ultrasound molecular imaging of endothelial αvβ3 expression in vitro and in vivo using αvβ3-targeted ultrasound contrast agents (UCAs). In the in vitro study, αvβ3 expression was confirmed by immunofluorescence in a murine endothelial cell line and detected using the targeted UCA and ultrasound imaging at 18-MHz transmit frequency. In the in vivo study, expression of endothelial αvβ3 integrin in murine carotid artery vessels and microvessels of the salivary gland was quantified using targeted UCA and high-frequency ultrasound in seven animals. Our results indicated that endothelial αvβ3 expression was significantly higher in the carotid arterial wall containing atherosclerotic lesions than in arterial segments without any lesions. We also found that the salivary gland can be used as an internal positive control for successful binding of targeted UCA to αvβ3 integrin. In conclusion, αvβ3-targeted UCA allows non-invasive assessment of the expression levels of αvβ3 on the vascular endothelium and may provide potential insights into early atherosclerotic plaque detection and treatment monitoring.
Collapse
Affiliation(s)
- Verya Daeichin
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands.
| | - Klazina Kooiman
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Ilya Skachkov
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Johan G Bosch
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands
| | - Thomas L Theelen
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Ben J Janssen
- Department of Pharmacology, MUMC, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Antonius F W van der Steen
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands; Shenzhen Institute of Advanced Technologies, Shenzhen, China
| | - Nico de Jong
- Biomedical Engineering, Thorax Center, Erasmus MC, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands; Netherlands Heart Institute, Utrecht, The Netherlands
| | - Judith C Sluimer
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
42
|
Maulucci G, Bačić G, Bridal L, Schmidt HH, Tavitian B, Viel T, Utsumi H, Yalçın AS, De Spirito M. Imaging Reactive Oxygen Species-Induced Modifications in Living Systems. Antioxid Redox Signal 2016; 24:939-58. [PMID: 27139586 PMCID: PMC4900226 DOI: 10.1089/ars.2015.6415] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Reactive Oxygen Species (ROS) may regulate signaling, ion channels, transcription factors, and biosynthetic processes. ROS-related diseases can be due to either a shortage or an excess of ROS. RECENT ADVANCES Since the biological activity of ROS depends on not only concentration but also spatiotemporal distribution, real-time imaging of ROS, possibly in vivo, has become a need for scientists, with potential for clinical translation. New imaging techniques as well as new contrast agents in clinically established modalities were developed in the previous decade. CRITICAL ISSUES An ideal imaging technique should determine ROS changes with high spatio-temporal resolution, detect physiologically relevant variations in ROS concentration, and provide specificity toward different redox couples. Furthermore, for in vivo applications, bioavailability of sensors, tissue penetration, and a high signal-to-noise ratio are additional requirements to be satisfied. FUTURE DIRECTIONS None of the presented techniques fulfill all requirements for clinical translation. The obvious way forward is to incorporate anatomical and functional imaging into a common hybrid-imaging platform. Antioxid. Redox Signal. 24, 939-958.
Collapse
Affiliation(s)
- Giuseppe Maulucci
- 1 Institute of Physics, Catholic University of Sacred Heart , Roma, Italy
| | - Goran Bačić
- 2 Faculty of Physical Chemistry, University of Belgrade , Belgrade, Serbia
| | - Lori Bridal
- 3 Laboratoire d'Imagerie Biomédicale, Sorbonne Universités and UPMC Univ Paris 06 and CNRS and INSERM , Paris, France
| | - Harald Hhw Schmidt
- 4 Department of Pharmacology and Personalised Medicine, CARIM, Faculty of Health, Medicine & Life Science, Maastricht University , Maastricht, the Netherlands
| | - Bertrand Tavitian
- 5 Laboratoire de Recherche en Imagerie, Université Paris Descartes, Hôpital Européen Georges Pompidou , Service de Radiologie, Paris, France
| | - Thomas Viel
- 5 Laboratoire de Recherche en Imagerie, Université Paris Descartes, Hôpital Européen Georges Pompidou , Service de Radiologie, Paris, France
| | - Hideo Utsumi
- 6 Innovation Center for Medical Redox Navigation, Kyushu University , Fukuoka, Japan
| | - A Süha Yalçın
- 7 Department of Biochemistry, School of Medicine, Marmara University , İstanbul, Turkey
| | - Marco De Spirito
- 1 Institute of Physics, Catholic University of Sacred Heart , Roma, Italy
| |
Collapse
|
43
|
Liu D, Chen Y, Sun P, Bai W, Gao A. STAT3 methylation in white blood cells as a novel sensitive biomarker for the toxic effect of low-dose benzene exposure. Toxicol Res (Camb) 2016; 5:800-807. [PMID: 30090390 PMCID: PMC6061912 DOI: 10.1039/c5tx00445d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/20/2016] [Indexed: 12/13/2022] Open
Abstract
Alterations in DNA methylation patterns play an essential role in disease process and are associated with cancer risk. To explore the toxic effect and early sensitive biomarker of the health effects of low-dose benzene exposure (LDBE), and investigate the correlation between DNA methylation and the toxic effect of LDBE, a cross-sectional study was conducted in a sample of 571 workers; 312 workers who were exposed to a 1.82 ± 1.16 mg m-3 air benzene concentration were assigned to the LDBE group, while 259 non-known benzene exposure (NBE) workers were assigned to the control group, with an air benzene concentration of 0.06 ± 0.01 mg m-3. Routine blood indexes, alanine transaminase (ALT), oxidative stress parameters and signal transducer and activator of transcription 3 (STAT3) methylation were detected. Compared with the NBE population, the STAT3 methylation level (P = 0.001), Platelets (PLTs) (P = 0.002) and 8-isoprostane-PGFs (8-iso-PGF2a) (P = 0.001) manifested a significant reduction, while ALT (P = 0.002) and 8-hydroxy-2 deoxyguanosine (8-OHdG) (P = 0.002) showed a significant rise in the LDBE population. In addition, a significant correlation was observed between STAT3 methylation and oxidative stress, namely 8-OhdG and 8-iso-PGF2a. Furthermore, a multivariate analysis showed that the STAT3 methylation (structure loadings = 0.909) was the most strongly correlated with the other set of variables, especially with white blood cells (WBCs) (structure loadings = 0.675). Taken together, STAT3 methylation may be the underlying mechanism involved in the early toxic effect of LDBE, therefore, STAT3 methylation can be a novel sensitive biomarker for the toxic effect of low-dose benzene exposure.
Collapse
Affiliation(s)
- Di Liu
- Department of Occupational Health and Environmental Health , School of Public Health , Capital Medical University , Beijing 100069 , China . ; ; Tel: +86-10-83911509
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Yujiao Chen
- Department of Occupational Health and Environmental Health , School of Public Health , Capital Medical University , Beijing 100069 , China . ; ; Tel: +86-10-83911509
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Pengling Sun
- Department of Occupational Health and Environmental Health , School of Public Health , Capital Medical University , Beijing 100069 , China . ; ; Tel: +86-10-83911509
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Wenlin Bai
- Department of Occupational Health and Environmental Health , School of Public Health , Capital Medical University , Beijing 100069 , China . ; ; Tel: +86-10-83911509
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Ai Gao
- Department of Occupational Health and Environmental Health , School of Public Health , Capital Medical University , Beijing 100069 , China . ; ; Tel: +86-10-83911509
- Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| |
Collapse
|
44
|
Abstract
Molecular imaging offers great potential for noninvasive visualization and quantitation of the cellular and molecular components involved in atherosclerotic plaque stability. In this chapter, we review emerging molecular imaging modalities and approaches for quantitative, noninvasive detection of early biological processes in atherogenesis, including vascular endothelial permeability, endothelial adhesion molecule up-regulation, and macrophage accumulation, with special emphasis on mouse models. We also highlight a number of targeted imaging nanomaterials for assessment of advanced atherosclerotic plaques, including extracellular matrix degradation, proteolytic enzyme activity, and activated platelets using mouse models of atherosclerosis. The potential for clinical translation of molecular imaging nanomaterials for assessment of atherosclerotic plaque biology, together with multimodal approaches is also discussed.
Collapse
|
45
|
Wang X, Gkanatsas Y, Palasubramaniam J, Hohmann JD, Chen YC, Lim B, Hagemeyer CE, Peter K. Thrombus-Targeted Theranostic Microbubbles: A New Technology towards Concurrent Rapid Ultrasound Diagnosis and Bleeding-free Fibrinolytic Treatment of Thrombosis. Am J Cancer Res 2016; 6:726-38. [PMID: 27022419 PMCID: PMC4805666 DOI: 10.7150/thno.14514] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/08/2016] [Indexed: 12/29/2022] Open
Abstract
Rationale: Myocardial infarction and stroke are leading causes of morbidity/mortality. The typical underlying pathology is the formation of thrombi/emboli and subsequent vessel occlusion. Systemically administered fibrinolytic drugs are the most effective pharmacological therapy. However, bleeding complications are relatively common and this risk as such limits their broader use. Furthermore, a rapid non-invasive imaging technology is not available. Thereby, many thrombotic events are missed or only diagnosed when ischemic damage has already occurred. Objective: Design and preclinical testing of a novel 'theranostic' technology for the rapid non-invasive diagnosis and effective, bleeding-free treatment of thrombosis. Methods and Results: A newly created, innovative theranostic microbubble combines a recombinant fibrinolytic drug, an echo-enhancing microbubble and a recombinant thrombus-targeting device in form of an activated-platelet-specific single-chain antibody. After initial in vitro proof of functionality, we tested this theranostic microbubble both in ultrasound imaging and thrombolytic therapy using a mouse model of ferric-chloride-induced thrombosis in the carotid artery. We demonstrate the reliable highly sensitive detection of in vivo thrombi and the ability to monitor their size changes in real time. Furthermore, these theranostic microbubbles proofed to be as effective in thrombolysis as commercial urokinase but without the prolongation of bleeding time as seen with urokinase. Conclusions: We describe a novel theranostic technology enabling simultaneous diagnosis and treatment of thrombosis, as well as monitoring of success or failure of thrombolysis. This technology holds promise for major progress in rapid diagnosis and bleeding-free thrombolysis thereby potentially preventing the often devastating consequences of thrombotic disease in many patients.
Collapse
|
46
|
Abstract
Atherosclerotic lesions initiate in regions characterized by low shear stress and reduced activity of endothelial atheroprotective molecules such as nitric oxide, which is the key molecule managing vascular homeostasis. The generation of reactive oxygen species from the vascular endothelium is strongly related to various enzymes, such as xanthine oxidase, endothelial nitric oxide synthase and nicotinamide-adenine dinucleotide phosphate oxidase. Several pharmaceutical agents, including angiotensin converting enzyme inhibitors, angiotensin receptors blockers and statins, along with a variety of other agents, have demonstrated additional antioxidant properties beyond their principal role. Reports regarding the antioxidant role of vitamins present controversial results, especially those based on large scale studies. In addition, there is growing interest on the role of dietary flavonoids and their potential to improve endothelial function by modifying the oxidative stress status. However, the vascular-protective role of flavonoids and especially their antioxidant properties are still under investigation. Indeed, further research is required to establish the impact of the proposed new therapeutic strategies in atherosclerosis.
Collapse
|
47
|
Shim CY, Liu YN, Atkinson T, Xie A, Foster T, Davidson BP, Treible M, Qi Y, López JA, Munday A, Ruggeri Z, Lindner JR. Molecular Imaging of Platelet-Endothelial Interactions and Endothelial von Willebrand Factor in Early and Mid-Stage Atherosclerosis. Circ Cardiovasc Imaging 2015; 8:e002765. [PMID: 26156014 DOI: 10.1161/circimaging.114.002765] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Nonthrombotic platelet-endothelial interactions may contribute to atherosclerotic plaque development, although in vivo studies examining mechanism without platelet preactivation are lacking. Using in vivo molecular imaging at various stages of atherosclerosis, we quantified platelet-endothelial interactions and evaluated the contribution of major adhesion pathways. METHODS AND RESULTS Mice deficient for the low-density lipoprotein receptor and Apobec-1 were studied as an age-dependent model of atherosclerosis at 10, 20, 30, and 40 weeks of age, which provided progressive increase in stage from early fatty streak (10 weeks) to large complex plaques without rupture (40 weeks). Platelet-targeted contrast ultrasound molecular imaging of the thoracic aorta performed with microbubbles targeted to GPIbα demonstrated selective signal enhancement as early as 10 weeks of age. This signal increased progressively with age (almost 8-fold increase from 10 to 40 weeks, analysis of variance P<0.001). Specificity for platelet targeting was confirmed by the reduction in platelet-targeted signal commensurate with the decrease in platelet count after immunodepletion with anti-GPIb or anti-CD41 antibody. Inhibition of P-selectin in 20 and 40 weeks atherosclerotic mice resulted in a small (15% to 30%) reduction in platelet signal. Molecular imaging with microbubbles targeted to the A1 domain of von Willebrand factor demonstrated selective signal enhancement at all time points, which did not significantly increase with age. Treatment of 20 and 40 week mice with recombinant ADAMTS13 eliminated platelet and von Willebrand factor molecular imaging signal. CONCLUSIONS Platelet-endothelial interactions occur in early atherosclerosis. These interactions are in part caused by endothelial von Willebrand factor large multimers, which can be reversed with exogenous ADAMTS13.
Collapse
Affiliation(s)
- Chi Young Shim
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Ya Ni Liu
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Tamara Atkinson
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Aris Xie
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Ted Foster
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Brian P Davidson
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Mackenzie Treible
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Yue Qi
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - José A López
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Adam Munday
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Zaverio Ruggeri
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.)
| | - Jonathan R Lindner
- From the Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR (C.Y.S., Y.N.L., T.A., A.X., T.F., B.P.D., M.T., Y.Q., J.R.L.); Puget Sound Blood Center Research Institute, Seattle, WA (J.A.L., A.M.); and Department of Molecular and Experimental Medicine, Roon Research Center for Arteriosclerosis and Thrombosis, The Scripps Research Institute, La Jolla, CA (Z.R.).
| |
Collapse
|
48
|
Violi F, Pignatelli P. Clinical Application of NOX Activity and Other Oxidative Biomarkers in Cardiovascular Disease: A Critical Review. Antioxid Redox Signal 2015; 23:514-32. [PMID: 24382131 DOI: 10.1089/ars.2013.5790] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SIGNIFICANCE The oxidative stress theory of atherosclerosis is based on the assumption that the production of reactive oxidant species (ROS) by blood, as well as resident cells of the artery wall, elicits the formation of oxidized low-density lipoproteins (ox-LDL), which, in turn, promotes a series of inflammatory responses, ultimately leading to atherosclerotic plaque. This theory prompted the development of new laboratory methodologies that aimed at assessing the relationship between oxidative stress and clinical progression of human atherosclerosis. CRITICAL ISSUES Markers assessing the oxidation of phospholipid and protein components of LDL were among the first to be developed. Clinical trials included cross-sectional as well as retrospective and prospective studies that, however, provided equivocal results. Thus, clear evidence that oxidative biomarkers add more to the risk stratification by common atherosclerotic risk factors is still lacking. RECENT ADVANCES More recently, the analysis of oxidative stress focused on enzymatic pathways generating ROS, such as NADPH oxidase and myeloperoxidase (MPO). Experimental and clinical studies suggest that both enzymes may be implicated in promoting atherosclerotic disease. Novel laboratory methodologies have been, therefore, developed to study NADPH oxidase and MPO in patients with stable atherosclerosis as well as in patients with acute coronary and cerebro-vascular syndromes. FUTURE DIRECTIONS This review will analyze the strengths and weaknesses of the current methodology to study these enzymes in human atherosclerosis with particular regard to their clinical application in several settings of cardiovascular disease. Clinical methodology and results of previous studies with regard to markers of LDL oxidation have also been reviewed as a useful background for the future development of clinical trials.
Collapse
Affiliation(s)
- Francesco Violi
- I Clinica Medica , Department of Internal Medicine and Medical Specialties, Rome, Italy
| | - Pasquale Pignatelli
- I Clinica Medica , Department of Internal Medicine and Medical Specialties, Rome, Italy
| |
Collapse
|
49
|
Quesada IM, Lucero A, Amaya C, Meijles DN, Cifuentes ME, Pagano PJ, Castro C. Selective inactivation of NADPH oxidase 2 causes regression of vascularization and the size and stability of atherosclerotic plaques. Atherosclerosis 2015; 242:469-75. [PMID: 26298737 DOI: 10.1016/j.atherosclerosis.2015.08.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/23/2015] [Accepted: 08/06/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND A variety of NADPH oxidase (Nox) isoforms including Noxs 1, 2, 4 and 5 catalyze the formation of reactive oxygen species (ROS) in the vascular wall. The Nox2 isoform complex has arguably received the greatest attention in the progression of atherogenesis in animal models. Thus, in the current study we postulated that specific Nox2 oxidase inhibition could reverse or attenuate atherosclerosis in mice fed a high-fat diet. METHODS We evaluated the effect of isoform-selective Nox2 assembly inhibitor on the progression and vascularization of atheromatous plaques. Apolipoprotein E-deficient mice (ApoE-/-) were fed a high fat diet for two months and treated over 15 days with Nox2ds-tat or control sequence (scrambled); 10 mg/kg/day, i.p. Mice were sacrificed and superoxide production in arterial tissue was detected by cytochrome C reduction assay and dihydroethidium staining. Plaque development was evaluated and the angiogenic markers VEGF, HIF1-α and visfatin were quantified by real time qRT-PCR. MMP-9 protein release and gelatinolytic activity was determined as a marker for vascularization. RESULTS Nox2ds-tat inhibited Nox-derived superoxide determined by cytochrome C in carotid arteries (2.3 ± 0.1 vs 1.7 ± 0.1 O2(•-) nmol/min*mg protein; P < 0.01) and caused a significant regression in atherosclerotic plaques in aorta (66 ± 6 μm(2) vs 37 ± 1 μm(2); scrmb vs. Nox2ds-tat; P < 0.001). Increased VEGF, HIF-1α, MMP-9 and visfatin expression in arterial tissue in response to high-fat diet were significantly attenuated by Nox2ds-tat which in turn impaired both MMP-9 protein expression and activity. CONCLUSION Given these results, it is quite evident that selective Nox inhibitors can reverse vascular pathology arising with atherosclerosis.
Collapse
Affiliation(s)
- I M Quesada
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - A Lucero
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina
| | - C Amaya
- Cellular and Molecular Biology Lab, Institute of Histology and Embryology (IHEM) CONICET, Mendoza, Argentina
| | - D N Meijles
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - M E Cifuentes
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - P J Pagano
- Department of Pharmacology & Chemical Biology and Vascular Medicine Institute, 200 Lothrop Street, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - C Castro
- Vascular Biology Lab, Institute of Medicine and Experimental Biology of Cuyo (IMBECU) CONICET, School of Medical Sciences, National University of Cuyo, Mendoza, Argentina.
| |
Collapse
|
50
|
Tofik R, Swärd P, Ekelund U, Struglics A, Torffvit O, Rippe B, Bakoush O. Plasma pro-inflammatory cytokines, IgM-uria and cardiovascular events in patients with chest pain: A comparative study. Scandinavian Journal of Clinical and Laboratory Investigation 2015; 75:638-45. [PMID: 26174976 DOI: 10.3109/00365513.2015.1057218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Risk stratification of patients presenting with acute chest pain is crucial for immediate and long-term management. Traditional predictors are suboptimal; therefore inflammatory biomarkers are studied for clinical assessment of patients at risk. Recently, we reported the association of IgM-uria with worse cardiovascular outcome in patients with acute chest pain. In this study, in the same cohort of patients with chest pain, we compared the value of IgM-uria to pro-inflammatory cytokines in predicting the occurrence of subsequent cardiovascular events. METHODS A total of 178 consecutive patients presenting with acute chest pain to the emergency department at the University Hospital of Lund, were recruited. Twenty-seven of 57 patients with acute coronary syndrome (ACS), and 18 of 118 patients with non-specific chest pain at baseline developed a subsequent major cardiovascular event during the 18 months follow-up. Urinary proteins (IgM-uria and Microalbuminuria) and plasma inflammatory markers (IL-6, Il-8, IL-10, IFN-γ and TNF-α) were measured at time of admission. RESULTS Using the receiver operating characteristic curves, the area under the curve for predicting cardiovascular events was 0.71 (95%CI 0.61-0.81) for IgM-uria, 0.61 (95%CI 0.51-0.71) for IL-6, 0.63 (95%CI 0.53-0.72) for IL-8, 0.65 (95%CI 0.56-0.74) for IL-10, and 0.64 (95% CI 0.54-0.74) for TNF-α. In multivariate Cox-regression analysis adjusted for age, microalbuminuria, IgM-uria, IL-10, TNF-α, troponin T, hsCRP and ACS at baseline; IgM-uria was the only biomarker that remained an independent predictor of outcome (HR = 4.2, 95%CI 2.2-7.8, p < 0.001). CONCLUSION In patients with chest pain with or without acute coronary syndrome, IgM-uria could better predict the occurrence of cardiovascular events than plasma pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Rafid Tofik
- a Department of Emergency Medicine , Skåne University Hospital , Lund , Sweden.,b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Per Swärd
- c Department of Orthopaedics , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Ulf Ekelund
- a Department of Emergency Medicine , Skåne University Hospital , Lund , Sweden
| | - André Struglics
- c Department of Orthopaedics , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Ole Torffvit
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Bengt Rippe
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden
| | - Omran Bakoush
- b Department of Nephrology , Clinical Sciences Lund, Lund University , Lund , Sweden.,d Department of Internal Medicine , College of Medicine, UAE University , UAE
| |
Collapse
|