51
|
Hong LZ, Xue Q, Shao H. Inflammatory Markers Related to Innate and Adaptive Immunity in Atherosclerosis: Implications for Disease Prediction and Prospective Therapeutics. J Inflamm Res 2021; 14:379-392. [PMID: 33628042 PMCID: PMC7897977 DOI: 10.2147/jir.s294809] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/21/2021] [Indexed: 12/20/2022] Open
Abstract
Several lines of evidence have linked a dysregulated inflammatory setting to the pathogenesis of atherosclerosis, which is a form of chronic vascular inflammation. Various inflammatory biomarkers have been associated with inflammation and are recognized as potential tools to monitor the progression of atherosclerosis. A well-studied inflammatory marker in the context of cardiovascular diseases is C-reactive protein (CRP) or, more accurately, highly sensitive-CRP (hs-CRP), which has been established as an inflammatory biomarker for atherosclerotic events. In addition, a growing body of investigations has attempted to disclose the potential of inflammatory cytokines, enzymes, and genetic polymorphisms related to innate and adaptive immunity as biomarkers for predicting the development of atherosclerosis. In this review article, we clarify both traditional and novel inflammatory biomarkers related to components of the innate and adaptive immune system that may mirror the progression or phases of atherosclerotic inflammation/lesions. Furthermore, the contribution of the inflammatory biomarkers in developing potential therapeutics against atherosclerotic treatment will be discussed.
Collapse
Affiliation(s)
- Ling-Zhi Hong
- Emergency Department, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, 311700, Zhejiang Province, People’s Republic of China
| | - Qi Xue
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| |
Collapse
|
52
|
Tian D, Qin Q, Li M, Li X, Xu Q, Lv Q. Homocysteine Impairs Endothelial Cell Barrier Function and Angiogenic Potential via the Progranulin/EphA2 Pathway. Front Pharmacol 2021; 11:614760. [PMID: 33510642 PMCID: PMC7836014 DOI: 10.3389/fphar.2020.614760] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/04/2020] [Indexed: 12/26/2022] Open
Abstract
Hyperhomocysteinemia is a well-recognized independent risk factor for cardiovascular disease. To date, the mechanism of pathological plasma homocysteine (Hcy) level elevation remains to be elucidated. We aimed to investigate the levels of progranulin (PGRN), Eph-receptor tyrosine kinase-type A2 (EphA2), vascular cell adhesion molecule-1 (VCAM-1), and Hcy in patients with arteriosclerosis and investigate their functions in Hcy-injured human umbilical vein endothelial cells (HUVECs). EphA2 knockdown was induced in HUVECs by shRNA lentivirus infection with EphA2-RNAi, and bulk RNA-seq assay was performed. Then we investigated the mechanism underlying the effect of recombinant human PGRN (rhPGRN) combined with shRNA interference of EphA2 on cell proliferation, migration, and angiogenesis in Hcy-injured HUVECs. Results showed that serum EphA2, VCAM-1, and Hcy levels in acute coronary syndrome patients were significantly higher than those in chronic coronary syndrome patients (p = 0.000; p = 0.000; p = 0.033, respectively). In vitro, we demonstrated that knockdown of EphA2 significantly impaired cell adhesion and inhibited HUVECs migration and angiogenesis (p < 0.001), which was associated with reduction in VCAM1 and VE-cadherin (p < 0.05). Hcy modulated the expression of PGRN and EphA2 in a time-and dose-dependent manner. However, rhPGRN ameliorated the Hcy-induced reduction in cell viability and migration (p < 0.05). Mechanistically, we found that PGRN/EphA2 and its downstream AKT/NF-κB signaling might be the primary signal transduction pathways underlying Hcy-induced injury. The present study illustrated that PGRN plays a previously unrecognized role in Hcy-induced endothelial injury, which is achieved through its interaction with EphA2 signaling, implying a promising therapeutic target for cardiovascular disease.
Collapse
Affiliation(s)
- Dan Tian
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Qin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Disease, Shanghai, China
| | - Mingfei Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Disease, Shanghai, China
| | - Xiaoyu Li
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Xu
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianzhou Lv
- Department of Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
53
|
Noguchi H, Yamada S, Hirano KI, Yamaguchi S, Suzuki A, Guo X, Zaima N, Li M, Kobayashi K, Ikeda Y, Nakayama T, Sasaguri Y. Outside-in signaling by femoral cuff injury induces a distinct vascular lesion in adipose triglyceride lipase knockout mice. Histol Histopathol 2021; 36:91-100. [PMID: 33231284 DOI: 10.14670/hh-18-285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Genetic deficiency of adipose triglyceride lipase (ATGL), a rate-limiting enzyme for intracellular triglyceride (TG) hydrolysis, causes TG-deposit cardiomyovasculopathy (TGCV), a recently identified rare cardiovascular disorder (ORPHA code: 565612) in humans. One of the major characteristics of TGCV is a novel type of diffuse and concentric coronary atherosclerosis with ATGL-deficient smooth muscle cells (SMCs). Patients with TGCV have intractable coronary artery disease. Therefore, it is crucial to investigate the mechanisms underlying vascular lesions in ATGL deficiency using animal models. Cuff injury is an experimental procedure to induce vascular remodeling with neointimal formation with SMCs after placing a cuff around the adventitial side of the artery without direct influence on endothelium. We report the effect of cuff injury on femoral arteries of ATGL-knockout (ATGL⁻/⁻) mice. Cuff-induced concentric neointimal formation with migrating SMCs was exacerbated in ATGL⁻/⁻ mice, mimicking atherosclerotic lesions in patients with TGCV. In the media, cell death of SMCs and loss of elastic fibers increased. Perivascular infiltrating cells expressing tumor necrosis factor-α (TNF-α) were more prominent in ATGL⁻/⁻ mice than in wild-type (WT) mice. In Boyden chamber experiments, a greater number of ATGL⁻/⁻ SMCs migrated in response to TNF-α compared to WT SMCs. These data, for the first time, demonstrated that outside-in signaling by cuff-induced neointimal formation where paracrine stimuli from adventitial infiltrating cells may lead to neointimal formation and mediolysis in ATGL-deficient conditions. Cuff injury might be a valuable model for understanding the mechanisms underlying the development of atherosclerotic lesions in patients with TGCV.
Collapse
Grants
- 16ek0109092h0002 Japan Agency for Medical Research and Development (AMED)
- 24790394 Ministry of Education, Culture, Sports, Science and Technology, Tokyo, Japan
- 16K08750 Ministry of Education, Culture, Sports, Science and Technology, Tokyo, Japan
Collapse
Affiliation(s)
- Hirotsugu Noguchi
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kagoshima, Japan
- Department of Pathology, Field of Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Sohsuke Yamada
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kagoshima, Japan
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Kanazawa, Japan
| | - Ken-Ichi Hirano
- Laboratory of Cardiovascular Disease, Novel, Non-invasive, and Nutritional Therapeutics (CNT) and Triglyceride Research Center (TGRC), Department of Triglyceride Science, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Satoshi Yamaguchi
- Laboratory of Cardiovascular Disease, Novel, Non-invasive, and Nutritional Therapeutics (CNT) and Triglyceride Research Center (TGRC), Department of Triglyceride Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Akira Suzuki
- Laboratory of Cardiovascular Disease, Novel, Non-invasive, and Nutritional Therapeutics (CNT) and Triglyceride Research Center (TGRC), Department of Triglyceride Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Xin Guo
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, Kanazawa, Japan
| | - Nobuhiro Zaima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Kindai, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Kindai, Japan
| | - Ming Li
- Laboratory of Cardiovascular Disease, Novel, Non-invasive, and Nutritional Therapeutics (CNT) and Triglyceride Research Center (TGRC), Department of Triglyceride Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kunihisa Kobayashi
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University Chikushi Hospital, Fukuoka, Japan
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kagoshima, Japan
| | - Yasuyuki Sasaguri
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kagoshima, Japan
| |
Collapse
|
54
|
Alberti S, Zhang Q, D'Agostino I, Bruno A, Tacconelli S, Contursi A, Guarnieri S, Dovizio M, Falcone L, Ballerini P, Münch G, Yu Y, Patrignani P. The antiplatelet agent revacept prevents the increase of systemic thromboxane A 2 biosynthesis and neointima hyperplasia. Sci Rep 2020; 10:21420. [PMID: 33293599 PMCID: PMC7722842 DOI: 10.1038/s41598-020-77934-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/13/2020] [Indexed: 11/21/2022] Open
Abstract
Neointima hyperplasia is a crucial component of restenosis after coronary angioplasty. We have hypothesized that enhanced generation of platelet-derived thromboxane (TX)A2 in response to vascular damage plays a critical role in neointimal hyperplasia and that antiplatelet agents may mitigate it. In cocultures of human platelets and coronary artery smooth muscle cells (CASMC), we found that platelets induced morphologic changes and enhanced the migration of CASMC. The exposure of platelets to Aspirin [an inhibitor of cyclooxygenase (COX)-1] reduced the generation of TXA2 and prevented the morphological and functional changes induced by platelets in CASMC. Platelet-derived TXA2 induced COX-2 and enhanced prostaglandin (PG)E2 biosynthesis in CASMC, a known mechanism promoting neointimal hyperplasia. COX-2 induction was prevented by different antiplatelet agents, i.e., Aspirin, the TP antagonist SQ29,548, or Revacept (a dimeric soluble GPVI-Fc fusion protein). The administration of the novel antiplatelet agent Revacept to C57BL/6 mice, beginning three days before femoral artery denudation, and continuing up to seven days after injury, prevented the increase of the systemic biosynthesis di TXA2 and reduced femoral artery intima-to-media area and the levels of markers of cell proliferation and macrophage infiltration. Revacept might serve as a therapeutic agent for percutaneous coronary angioplasty and stent implantation.
Collapse
Affiliation(s)
- Sara Alberti
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Qianqian Zhang
- International Peace Maternity and Child Health Hospital of China Welfare Institution, Shanghai, China
| | - Ilaria D'Agostino
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Annalisa Bruno
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Stefania Tacconelli
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Annalisa Contursi
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Simone Guarnieri
- CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Melania Dovizio
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy.,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Lorenza Falcone
- CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy
| | - Patrizia Ballerini
- CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy.,Department of Innovative Technologies in Medicine and Dentistry, "G. D'Annunzio" University, Chieti, Italy
| | | | - Ying Yu
- Shanghai Institute for Biological Sciences, Chinese Academy of Science, Shanghai, China.,Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Science, "G. D'Annunzio" University, Chieti, Italy. .,CAST (Center for Advanced Studies and Technology) (Ex CeSI-MeT), "G. D'Annunzio" University, Via dei Vestini 31, 66100, Chieti, Italy.
| |
Collapse
|
55
|
Kuang H, Wang Y, Shi Y, Yao W, He X, Liu X, Mo X, Lu S, Zhang P. Construction and performance evaluation of Hep/silk-PLCL composite nanofiber small-caliber artificial blood vessel graft. Biomaterials 2020; 259:120288. [DOI: 10.1016/j.biomaterials.2020.120288] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/06/2020] [Accepted: 08/01/2020] [Indexed: 11/29/2022]
|
56
|
Deveci B, Gazi E. Relation Between Globulin, Fibrinogen, and Albumin With the Presence and Severity of Coronary Artery Disease. Angiology 2020; 72:174-180. [PMID: 32996325 DOI: 10.1177/0003319720959985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The albumin to globulin ratio (AGR) is used as a prognostic marker in acute ischemic cardiovascular events. We investigated whether serum AGR, fibrinogen, and fibrinogen to albumin ratio (FAR) are related to the presence and severity of coronary artery disease (CAD). Patients who underwent coronary angiography procedures were analyzed retrospectively. The severity of CAD was assessed by the Gensini score. The study population (3031 patients; 1071 females and 1960 males) was divided into 3 tertiles based on AGR values. Gensini score, lipid levels, diabetes mellitus (DM), hypertension (HT), age, and fibrinogen level were higher in the low AGR group. Pearson correlation analysis showed that AGR (r = -0.068, P < .001) was negatively and fibrinogen (r = 0.187, P < .001) was positively correlated with the Gensini score. Male gender, HT, smoking, DM, age, high triglyceride (TG) level, low-density lipoprotein cholesterol (LDL-C) >160 mg/dL, estimated glomerular filtration rate (eGFR) <60 mL/min, and fibrinogen level >3.5 g/L were independent predictors of CAD. Male gender, age, eGFR, DM, LDL-C, TG, and FAR had an independent positive relation to the Gensini score. In conclusion, similar to traditional risk factors, plasma fibrinogen and albumin levels showed a close relation with the presence and severity of CAD.
Collapse
Affiliation(s)
- Bülent Deveci
- Department of Cardiology, 448249Sağlık Bilimleri University, Ankara, Turkey
| | - Emine Gazi
- Department of Cardiology, 52950Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
57
|
Extracellular vesicle signalling in atherosclerosis. Cell Signal 2020; 75:109751. [PMID: 32860954 PMCID: PMC7534042 DOI: 10.1016/j.cellsig.2020.109751] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a major cardiovascular disease and in 2016, the World Health Organisation (WHO) estimated 17.5 million global deaths, corresponding to 31% of all global deaths, were driven by inflammation and deposition of lipids into the arterial wall. This leads to the development of plaques which narrow the vessel lumen, particularly in the coronary and carotid arteries. Atherosclerotic plaques can become unstable and rupture, leading to myocardial infarction or stroke. Extracellular vesicles (EVs) are a heterogeneous population of vesicles secreted from cells with a wide range of biological functions. EVs participate in cell-cell communication and signalling via transport of cargo including enzymes, DNA, RNA and microRNA in both physiological and patholophysiological settings. EVs are present in atherosclerotic plaques and have been implicated in cellular signalling processes in atherosclerosis development, including immune responses, inflammation, cell proliferation and migration, cell death and vascular remodeling during progression of the disease. In this review, we summarise the current knowledge regarding EV signalling in atherosclerosis progression and the potential of utilising EV signatures as biomarkers of disease.
Collapse
|
58
|
Wang W, Ma F, Zhang H. MicroRNA-374 is a potential diagnostic biomarker for atherosclerosis and regulates the proliferation and migration of vascular smooth muscle cells. Cardiovasc Diagn Ther 2020; 10:687-694. [PMID: 32968625 DOI: 10.21037/cdt-20-444] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background The occurrence and development of atherosclerosis (AS) are closely related to the abnormality of vascular smooth muscle cells (VSMCs), and multiple microRNAs (miRNAs) have been reported to participate in the pathogenesis of AS. This study explored the expression and clinical value of miR-374 in the serum of AS patients, and analyzed its effect on the proliferation and migration of VSMCs. Methods The expression levels of miR-374 in the serum of 102 asymptomatic patients with AS and 89 healthy patients were detected by fluorescence quantitative PCR. The diagnostic value of miR-374 was evaluated through the receiver operating characteristic (ROC) curve. What's more, CCK-8 and Transwell assays were used to analyze the effects of miR-374 on the proliferation and migration of VSMCs. Results The expression level of miR-374 in the serum of AS patients was significantly higher than that of the control group. At the same time, the expression of miR-374 in AS patients was positively correlated with carotid intima-media thickness (CIMT). The area under the ROC curve is 0.824. Furthermore, overexpression of miR-374 significantly promoted the proliferation and migration of VSMCs, whereas reducing miR-374 inhibited the proliferation and migration of VSMCs. Conclusions The high expression of miR-374 may be a potential diagnostic marker for AS, and overexpression of miR-374 may play a role in AS by promoting the proliferation and migration of VSMCs.
Collapse
Affiliation(s)
- Weihong Wang
- Department of Healthcare, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fenghua Ma
- Department of Healthcare, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongyan Zhang
- Department of Thoracic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
59
|
Minelli S, Minelli P, Montinari MR. Reflections on Atherosclerosis: Lesson from the Past and Future Research Directions. J Multidiscip Healthc 2020; 13:621-633. [PMID: 32801729 PMCID: PMC7398886 DOI: 10.2147/jmdh.s254016] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022] Open
Abstract
The clinical manifestations of atherosclerosis are nowadays the main cause of death in industrialized countries, but atherosclerotic disease was found in humans who lived thousands of years ago, before the spread of current risk factors. Atherosclerotic lesions were identified on a 5300-year-old mummy, as well as in Egyptian mummies and other ancient civilizations. For many decades of the twentieth century, atherosclerosis was considered a degenerative disease, mainly determined by a passive lipid storage, while the most recent theory of atherogenesis is based on endothelial dysfunction. The importance of inflammation and immunity in atherosclerosis’s pathophysiology was realized around the turn of the millennium, when in 1999 the famous pathologist Russell Ross published in the New England Journal of Medicine an article entitled “Atherosclerosis – an inflammatory disease”. In the following decades, inflammation has been a topic of intense basic research in atherosclerosis, albeit its importance has ancient scientific roots. In fact, in 1856 Rudolph Virchow was the first proponent of this hypothesis, but evidence of the key role of inflammation in atherogenesis occurred only in 2017. It seemed interesting to retrace the key steps of atherosclerosis in a historical context: from the teachings of the physicians of the Roman Empire to the response-to-injury hypothesis, up to the key role of inflammation and immunity at various stages of disease. Finally, we briefly discussed current knowledge and future trajectories of atherosclerosis research and its therapeutic implications.
Collapse
Affiliation(s)
- Sergio Minelli
- Department of Cardiology, Local Health Unit Lecce, Lecce, Italy
| | - Pierluca Minelli
- Faculty of Medicine and Surgery "A. Gemelli", Catholic University of the Sacred Heart, Rome, Italy
| | - Maria Rosa Montinari
- Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| |
Collapse
|
60
|
Subbotin VM. Pattern of organ remodeling in chronic non-communicable diseases is due to endogenous regulations and falls under the category of Kauffman's self-organization: A case of arterial neointimal pathology. Med Hypotheses 2020; 143:110106. [PMID: 32759005 DOI: 10.1016/j.mehy.2020.110106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/07/2020] [Accepted: 07/11/2020] [Indexed: 01/10/2023]
Abstract
Clinical diagnosis is based on analysis of pathologic findings that may result in perceived patterns. The same is true for diagnostic pathology: Pattern analysis is a foundation of the histopathology-based diagnostic system and, in conjunction with clinical and laboratory findings, forms a basis for the classification of diseases. Any histopathology diagnosis is based on the explicit assumption that the same diseased condition should result in formation of the same (or highly similar) morphologic patterns in different individuals; it is a standard approach in microscopic pathology, including that of non-communicable chronic diseases with organ remodeling. During fifty years of examining diseased tissues under microscopy, I keep asking the same question: Why is a similarity of patterns expected for chronic organ remodeling? For infection diseases, xenobiotic toxicity and deficiencies forming an identical pathologic pattern in different individuals is understandable and logical: The same infection, xenobiotic, or deficiency strikes the same target, which results in identical pathology. The same is true for Mendelian diseases: The same mutations lead to the same altered gene expressions and the same pathologic pattern. But why does this regularity hold true for chronic diseases with organ remodeling? Presumable causes (or risk factors) for a particular chronic disease differ in magnitude and duration between individuals, which should result in various series of transformations. Yet, mysteriously enough, pathological remodeling in a particular chronic disease always falls into a main dominating pattern, perpetuating and progressing in a similar fashion in different patients. Furthermore, some chronic diseases of different etiologies and dissimilar causes/risk factors manifest as identical or highly similar patterns of pathologic remodeling. HYPOTHESIS: I hypothesize that regulations governing a particular organ's chronic remodeling were selected in evolution as the safest response to various insults and physiologic stress conditions. This hypothesis implies that regulations directing diseased chronic remodeling always preexist but normally are controlled; this control can be disrupted by a diverse range of non-specific signals, liberating the pathway for identical pathologic remodeling. This hypothesis was tested in an analysis of arterial neointimal formation, the identical pathology occurring in different diseases and pathological conditions: graft vascular disease in organ transplantation, in-stent restenosis, peripheral arterial diseases, idiopathic intimal hyperplasia, Kawasaki disease, coronary atherosclerosis and as reaction to drugs. The hypothesis suggests that arterial intimal cells are poised between only two alternative pathways: the pathway with controlled intimal cell proliferation or the pathway where such control is disrupted, ultimately leading to the progressive neointimal pathology. By this property the arterial neointimal formation constitutes a special case of Kauffman's self-organization. This new hypothesis gives a parsimonious explanation for identical pathological patterns of arterial remodeling (neointimal formation), which occurs in diseases of different etiologies and due to dissimilar causes/risk factors, or without any etiology and causes/risk factors at all. This new hypothesis also suggests that regulation facilitating intimal cell proliferation cannot be overwritten or annulled because this feature is vital for arterial differentiation, cell renewal, and integrity. This hypothesis suggests that studying numerous, and likely interchangeable, non-specific signals that disrupt regulation controlling intimal cell proliferation is unproductive; instead, a study of the controlling regulation(s) itself should be a priority of our research.
Collapse
Affiliation(s)
- Vladimir M Subbotin
- University of Pittsburgh, Pittsburgh, PA 15260, USA; University of Wisconsin, Madison, WI 53705, USA; Arrowhead Parmaceuticals, Madison, WI 53719, USA.
| |
Collapse
|
61
|
Rosales C, Gillard BK, Gotto AM, Pownall HJ. The Alcohol-High-Density Lipoprotein Athero-Protective Axis. Biomolecules 2020; 10:E987. [PMID: 32630283 PMCID: PMC7408510 DOI: 10.3390/biom10070987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/22/2023] Open
Abstract
Ingestion of alcohol is associated with numerous changes in human energy metabolism, especially that of plasma lipids and lipoproteins. Regular moderate alcohol consumption is associated with reduced atherosclerotic cardiovascular disease (ASCVD), an effect that has been attributed to the concurrent elevations of plasma high-density lipoprotein-cholesterol (HDL-C) concentrations. More recent evidence has accrued against the hypothesis that raising plasma HDL concentrations prevents ASCVD so that other metabolic processes associated with alcohol consumption have been considered. This review explored the roles of other metabolites induced by alcohol consumption-triglyceride-rich lipoproteins, non-esterified free fatty acids, and acetate, the terminal alcohol metabolite in athero-protection: Current evidence suggests that acetate has a key role in athero-protection but additional studies are needed.
Collapse
Affiliation(s)
| | | | | | - Henry J. Pownall
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; (C.R.); (B.K.G.); (A.M.G.J.)
| |
Collapse
|
62
|
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
63
|
Corti A, Gaucher C, Pompella A. Editorial: Targeting Monocytes/Macrophages to Treat Atherosclerotic Inflammation. Front Pharmacol 2020; 11:86. [PMID: 32116730 PMCID: PMC7033601 DOI: 10.3389/fphar.2020.00086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 01/27/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Alessandro Corti
- Dipartimento di Ricerca Traslazionale NTMC, Scuola Medica dell'Università di Pisa, Pisa, Italy
| | | | - Alfonso Pompella
- Dipartimento di Ricerca Traslazionale NTMC, Scuola Medica dell'Università di Pisa, Pisa, Italy
| |
Collapse
|
64
|
Buja LM. Innovators in atherosclerosis research: A historical review. Int J Cardiol 2020; 307:8-14. [PMID: 32070483 DOI: 10.1016/j.ijcard.2020.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/23/2020] [Accepted: 02/06/2020] [Indexed: 12/20/2022]
Abstract
This review presents a retrospective analysis of the significance of the contributions of pathologists and kindred investigators in the latter half of the twentieth century to the advancement of understanding of atherosclerosis, a major disease and affliction of humankind. These outstanding investigators contributed importantly to the development of a large body of evidence encompassing population-based autopsy studies, experimental animal studies and cell biological investigations that, coupled with insights from epidemiological studies, serve as the underpinning for the current dominant response to injury theory of atherogenesis. Their collective contributions have been highly meritorious and will remain seminally important into the future.
Collapse
Affiliation(s)
- L Maximilian Buja
- McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Chief, Cardiovascular Pathology, Texas Heart Institute, Houston, TX, United States of America..
| |
Collapse
|
65
|
Wang D, Zhang Y, Shen C. Research update on the association between SFRP5, an anti-inflammatory adipokine, with obesity, type 2 diabetes mellitus and coronary heart disease. J Cell Mol Med 2020; 24:2730-2735. [PMID: 32004418 PMCID: PMC7077606 DOI: 10.1111/jcmm.15023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Secreted frizzled-related protein 5 (SFRP5), an anti-inflammatory adipokine secreted by adipocytes, has been demonstrated to exert its anti-inflammatory effect via antagonizing the non-canonical wingless-type family member 5A (WNT5A) signalling pathways. The WNT5A protein, as a potent pro-inflammatory signalling molecule, is strongly involved in a variety of inflammatory disorders such as obesity, type 2 diabetes mellitus (T2DM) and atherosclerosis. In this review, we systematically outlined the current understanding on the roles of SFRP5 in the pathogenesis of three inflammatory diseases including obesity, T2DM and coronary heart disease (CHD). Our review might stimulate future research using SFRP5 as a promising novel therapeutic target for the treatment of obesity, T2DM and CHD.
Collapse
Affiliation(s)
- Di Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
66
|
Basatemur GL, Jørgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat Rev Cardiol 2019; 16:727-744. [PMID: 31243391 DOI: 10.1038/s41569-019-0227-9] [Citation(s) in RCA: 697] [Impact Index Per Article: 116.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type present at all stages of an atherosclerotic plaque. According to the 'response to injury' and 'vulnerable plaque' hypotheses, contractile VSMCs recruited from the media undergo phenotypic conversion to proliferative synthetic cells that generate extracellular matrix to form the fibrous cap and hence stabilize plaques. However, lineage-tracing studies have highlighted flaws in the interpretation of former studies, revealing that these studies had underestimated both the content and functions of VSMCs in plaques and have thus challenged our view on the role of VSMCs in atherosclerosis. VSMCs are more plastic than previously recognized and can adopt alternative phenotypes, including phenotypes resembling foam cells, macrophages, mesenchymal stem cells and osteochondrogenic cells, which could contribute both positively and negatively to disease progression. In this Review, we present the evidence for VSMC plasticity and summarize the roles of VSMCs and VSMC-derived cells in atherosclerotic plaque development and progression. Correct attribution and spatiotemporal resolution of clinically beneficial and detrimental processes will underpin the success of any therapeutic intervention aimed at VSMCs and their derivatives.
Collapse
Affiliation(s)
- Gemma L Basatemur
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Helle F Jørgensen
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Murray C H Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Bennett
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, UK.
- INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
67
|
Abstract
OBJECTIVE This study aimed to investigate the effect of nifedipine on radial artery (RA) relaxation before puncture for coronary intervention. METHODS In all, 120 patients were randomly assigned to nifedipine or control group. The diameter, resistance index (RI), and peak systolic velocity (PSV) of the RA were observed at 5, 15, and 30 min after nifedipine administration by a Doppler ultrasound examination. RESULTS The greatest effect on RA diameter, PSV and resistance index (RI) was observed 5 minutes after sublingual nifedipine administration. The RA parameter were associated with the baseline diameter. CONCLUSIONS Sublingual nifedipine administration before RA puncture has an obvious dilatation effect on small diameter RAs.
Collapse
|
68
|
Yang S, Li X, Yang F, Zhao R, Pan X, Liang J, Tian L, Li X, Liu L, Xing Y, Wu M. Gut Microbiota-Dependent Marker TMAO in Promoting Cardiovascular Disease: Inflammation Mechanism, Clinical Prognostic, and Potential as a Therapeutic Target. Front Pharmacol 2019; 10:1360. [PMID: 31803054 PMCID: PMC6877687 DOI: 10.3389/fphar.2019.01360] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, especially in developed countries, and atherosclerosis (AS) is the common pathological basis of many cardiovascular diseases (CVDs) such as coronary heart disease (CHD). The role of the gut microbiota in AS has begun to be appreciated in recent years. Trimethylamine N-oxide (TMAO), an important gut microbe-dependent metabolite, is generated from dietary choline, betaine, and L-carnitine. Multiple studies have suggested a correlation between plasma TMAO levels and the risk of AS. However, the mechanism underlying this relationship is still unclear. In this review, we discuss the TMAO-involved mechanisms of atherosclerotic CVD from the perspective of inflammation, inflammation-related immunity, cholesterol metabolism, and atherothrombosis. We also summarize available clinical studies on the role of TMAO in predicting prognostic outcomes, including major adverse cardiovascular events (MACE), in patients presenting with AS. Finally, since TMAO may be a novel therapeutic target for AS, several therapeutic strategies including drugs, dietary, etc. to lower TMAO levels that are currently being explored are also discussed.
Collapse
Affiliation(s)
- Shengjie Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinye Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Fan Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran Zhao
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Xiandu Pan
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Jiaqi Liang
- Department of Cardiovascular, Beijing Longfu Hospital, Beijing, China
| | - Li Tian
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoya Li
- Beijing University of Chinese Medicine, Beijing, China.,Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
69
|
Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int J Mol Sci 2019; 20:E5694. [PMID: 31739395 PMCID: PMC6888164 DOI: 10.3390/ijms20225694] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.
Collapse
Affiliation(s)
- Armand Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Koen Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Abraham Kroon
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
70
|
Liu B, Wang Y, Zhang Y, Yan B. Mechanisms of Protective Effects of SGLT2 Inhibitors in Cardiovascular Disease and Renal Dysfunction. Curr Top Med Chem 2019; 19:1818-1849. [PMID: 31456521 DOI: 10.2174/1568026619666190828161409] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus is one of the most common forms of the disease worldwide. Hyperglycemia and insulin resistance play key roles in type 2 diabetes mellitus. Renal glucose reabsorption is an essential feature in glycaemic control. Kidneys filter 160 g of glucose daily in healthy subjects under euglycaemic conditions. The expanding epidemic of diabetes leads to a prevalence of diabetes-related cardiovascular disorders, in particular, heart failure and renal dysfunction. Cellular glucose uptake is a fundamental process for homeostasis, growth, and metabolism. In humans, three families of glucose transporters have been identified, including the glucose facilitators GLUTs, the sodium-glucose cotransporter SGLTs, and the recently identified SWEETs. Structures of the major isoforms of all three families were studied. Sodium-glucose cotransporter (SGLT2) provides most of the capacity for renal glucose reabsorption in the early proximal tubule. A number of cardiovascular outcome trials in patients with type 2 diabetes have been studied with SGLT2 inhibitors reducing cardiovascular morbidity and mortality. The current review article summarises these aspects and discusses possible mechanisms with SGLT2 inhibitors in protecting heart failure and renal dysfunction in diabetic patients. Through glucosuria, SGLT2 inhibitors reduce body weight and body fat, and shift substrate utilisation from carbohydrates to lipids and, possibly, ketone bodies. These pleiotropic effects of SGLT2 inhibitors are likely to have contributed to the results of the EMPA-REG OUTCOME trial in which the SGLT2 inhibitor, empagliflozin, slowed down the progression of chronic kidney disease and reduced major adverse cardiovascular events in high-risk individuals with type 2 diabetes. This review discusses the role of SGLT2 in the physiology and pathophysiology of renal glucose reabsorption and outlines the unexpected logic of inhibiting SGLT2 in the diabetic kidney.
Collapse
Affiliation(s)
- Ban Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuliang Wang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Biao Yan
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
71
|
Chen L, Qu H, Guo M, Zhang Y, Cui Y, Yang Q, Bai R, Shi D. ANRIL and atherosclerosis. J Clin Pharm Ther 2019; 45:240-248. [PMID: 31703157 DOI: 10.1111/jcpt.13060] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/26/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE The 3.8-kb-long antisense non-coding RNA at the INK4 locus (ANRIL) is transcribed from the short arm of human chromosome 9 on P21 and is associated with malfunction of the vascular endothelium, vascular smooth muscle cell (VSMC) proliferation/migration/senescence/apoptosis, mononuclear cell adhesion and proliferation, glycolipid metabolism disorder and DNA damage. Hence, ANRIL plays an important role in atherogenesis. Moreover, genome-wide association studies (GWAS) have identified ANRIL as a biomarker that is closely related to coronary heart disease (CHD). The objective of this review was to discuss the pathological mechanism of ANRIL in atherosclerotic development and its significance as a predictor of cardiovascular disease. METHODS Review of the PubMed, EMBASE and Cochrane databases for articles demonstrating the roles of ANRIL in the development of atherosclerotic diseases. RESULTS AND DISCUSSION The abnormal expression of ANRIL is linked to vascular endothelium injury; the proliferation, migration, senescence and apoptosis of VSMCs; mononuclear cell adhesion and proliferation; glycolipid metabolism disorder; DNA damage; and competing endogenous RNAs. Moreover, ANRIL accelerates the progression of CHD by regulating its single nucleotide polymorphisms (SNPs). WHAT IS NEW AND CONCLUSION Considering that ANRIL accelerates atherosclerosis (AS) development and is a risk factor for CHD, it is reasonable for us to explore an efficacious ANRIL-based therapy for AS in CHD.
Collapse
Affiliation(s)
- Li Chen
- Peking University Traditional Chinese Medicine Clinical Medical School (Xi yuan), Beijing, China
| | - Hua Qu
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ming Guo
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Zhang
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanyuan Cui
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaoning Yang
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruina Bai
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Dazhuo Shi
- Cardiovascular Diseases Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
72
|
Libby P, Hansson GK. From Focal Lipid Storage to Systemic Inflammation: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 74:1594-1607. [PMID: 31537270 PMCID: PMC6910128 DOI: 10.1016/j.jacc.2019.07.061] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/12/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022]
Abstract
Concepts of atherogenesis have evolved considerably with time. Early animal experiments showed that a cholesterol-rich diet could induce fatty lesion formation in arteries. The elucidation of lipoprotein metabolism ultimately led to demonstrating the clinical benefits of lipid lowering. The view of atheromata as bland accumulations of smooth muscle cells that elaborated an extracellular matrix that could entrap lipids then expanded to embrace inflammation as providing pathways that could link risk factors to atherogenesis. The characterization of leukocyte adhesion molecules and their control by proinflammatory cytokines, the ability of chemokines to recruit leukocytes, and the identification of inflammatory cell subtypes in lesions spurred the unraveling of innate and adaptive immune pathways that contribute to atherosclerosis and its thrombotic complications. Such pathophysiologic insights have led to the identification of biomarkers that can define categories of risk and direct therapies and to the development of new treatments.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Göran K Hansson
- Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
73
|
Abstract
Inflammation is an important driver of atherosclerosis, the underlying pathology of cardiovascular diseases. Therefore, therapeutic targeting of inflammatory pathways is suggested to improve cardiovascular outcomes in patients with cardiovascular diseases. This concept was recently proven by CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study), which demonstrated the therapeutic potential of the monoclonal IL (interleukin)-1β-neutralizing antibody canakinumab. IL-1β and other IL-1 family cytokines are important vascular and systemic inflammatory mediators, which contribute to atherogenesis. The NLRP3 (NOD [nucleotide oligomerization domain]-, LRR [leucine-rich repeat]-, and PYD [pyrin domain]-containing protein 3) inflammasome, an innate immune signaling complex, is the key mediator of IL-1 family cytokine production in atherosclerosis. NLRP3 is activated by various endogenous danger signals abundantly present in atherosclerotic lesions, such as oxidized low-density lipoprotein and cholesterol crystals. Consequently, NLRP3 inflammasome activation contributes to the vascular inflammatory response driving atherosclerosis development and progression. Here, we review the mechanisms of NLRP3 inflammasome activation and proinflammatory IL-1 family cytokine production in the context of atherosclerosis and discuss treatment possibilities in light of the positive outcomes of the CANTOS trial.
Collapse
Affiliation(s)
- Alena Grebe
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.)
| | - Florian Hoss
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.)
| | - Eicke Latz
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.) .,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.).,German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.).,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (E.L.)
| |
Collapse
|
74
|
Pashirzad M, Shafiee M, Avan A, Ryzhikov M, Fiuji H, Bahreyni A, Khazaei M, Soleimanpour S, Hassanian SM. Therapeutic potency of crocin in the treatment of inflammatory diseases: Current status and perspective. J Cell Physiol 2019; 234:14601-14611. [PMID: 30673132 DOI: 10.1002/jcp.28177] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Crocin is the major component of saffron, which is used in phytomedicine for the treatment of several diseases including diabetes, fatty liver, depression, menstruation disorders, and, of special interest in this review, inflammatory diseases. Promising selective anti-inflammatory properties of this pharmacological active component have been observed in several studies. Saffron has been shown to exert anti-inflammatory properties against several inflammatory diseases and can be used as a novel therapeutic agent for the treatment of inflammatory diseases either alone or in combination with other standard anti-inflammatory agents. This review summarizes the protective role of saffron and its pharmacologically active constituents in the pathogenesis of inflammatory diseases including digestive diseases, dermatitis, asthma, atherosclerosis, and neurodegenerative diseases for a better understanding and hence a better management of these diseases.
Collapse
Affiliation(s)
- Mehran Pashirzad
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Shafiee
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, Missouri
| | - Hamid Fiuji
- Department of Biochemistry, Payame-Noor University, Mashhad, Iran
| | - Amirhossein Bahreyni
- Department of Clinical Biochemistry and Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
75
|
Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat Med 2019; 25:1280-1289. [PMID: 31359001 PMCID: PMC7274198 DOI: 10.1038/s41591-019-0512-5] [Citation(s) in RCA: 525] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 06/05/2019] [Indexed: 12/12/2022]
Abstract
In response to various stimuli, vascular smooth muscle cells (SMCs) can
de-differentiate, proliferate and migrate in a process known as phenotypic
modulation. However, the phenotype of modulated SMCs in vivo during
atherosclerosis and the influence of this process on coronary artery disease
(CAD) risk have not been clearly established. Using single cell RNA sequencing,
we comprehensively characterized the transcriptomic phenotype of modulated SMCs
in vivo in atherosclerotic lesions of both mouse and human arteries and found
that these cells transform into unique fibroblast-like cells, termed
“fibromyocytes”, rather than into a classical macrophage
phenotype. SMC-specific knockout of TCF21, a causal CAD gene,
markedly inhibited SMC phenotypic modulation in mice, leading to the presence of
fewer fibromyocytes within lesions as well as within the protective fibrous cap
of the lesions. Moreover, TCF21 expression was strongly
associated with SMC phenotypic modulation in diseased human coronary arteries,
and higher levels of TCF21 expression were associated with
decreased CAD risk human CAD-relevant tissues. These results establish a
protective role for both TCF21 and SMC phenotypic modulation in
this disease.
Collapse
|
76
|
Manshadi MKD, Saadat M, Mohammadi M, Shamsi M, Dejam M, Kamali R, Sanati-Nezhad A. Delivery of magnetic micro/nanoparticles and magnetic-based drug/cargo into arterial flow for targeted therapy. Drug Deliv 2019; 25:1963-1973. [PMID: 30799655 PMCID: PMC6292362 DOI: 10.1080/10717544.2018.1497106] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Magnetic drug targeting (MDT) and magnetic-based drug/cargo delivery are emerging treatment methods which attracting the attention of many researchers for curing different cancers and artery diseases such as atherosclerosis. Herein, computational studies are accomplished by utilizing magnetic approaches for cancer and artery atherosclerosis drug delivery, including nanomagnetic drug delivery and magnetic-based drug/cargo delivery. For the first time, the four-layer structural model of the artery tissue and its porosity parameters are modeled in this study which enables the interaction of particles with the tissue walls in blood flow. The effects of parameters, including magnetic field strength (MFS), magnet size, particle size, the initial position of particles, and the relative magnetic permeability of particles, on the efficacy of MDT through the artery walls are characterized. The magnetic particle penetration into artery layers and fibrous cap (the covering layer over the inflamed part of the artery) is further simulated. The MDT in healthy and diseased arteries demonstrates that some of the particles stuck in these tissues due to the collision of particles or blood flow deviation in the vicinity of the inflamed part of the artery. Therefore the geometry of artery and porosity of its layers should be considered to show the real interaction of particles with the artery walls. Also, the results show that increasing the particles/drug/cargo size and MFS leads to more particles/drug/cargo retention within the tissue. The present work provides insights into the decisive factors in arterial MDT with an obvious impact on locoregional cancer treatment, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Mohammad K D Manshadi
- a Department of Mechanical and Manufacturing Engineering , University of Calgary , Calgary , Alberta , Canada
| | - Mahsa Saadat
- b Department of Chemical Engineering, College of Engineering , Shahid Bahonar University of Kerman , Kerman , Iran
| | - Mehdi Mohammadi
- a Department of Mechanical and Manufacturing Engineering , University of Calgary , Calgary , Alberta , Canada.,c Department of Biological Science , University of Calgary , Calgary , Alberta , Canada.,d Center for Bioengineering Research and Education , University of Calgary , Calgary , Alberta , Canada
| | - Milad Shamsi
- c Department of Biological Science , University of Calgary , Calgary , Alberta , Canada.,d Center for Bioengineering Research and Education , University of Calgary , Calgary , Alberta , Canada
| | - Morteza Dejam
- e Department of Petroleum Engineering College of Engineering and Applied Science , University of Wyoming , Laramie , WY , USA
| | - Reza Kamali
- f Department of Mechanical Engineering , Shiraz University , Shiraz , Iran
| | - Amir Sanati-Nezhad
- c Department of Biological Science , University of Calgary , Calgary , Alberta , Canada.,d Center for Bioengineering Research and Education , University of Calgary , Calgary , Alberta , Canada
| |
Collapse
|
77
|
Steinberg I, Huland DM, Vermesh O, Frostig HE, Tummers WS, Gambhir SS. Photoacoustic clinical imaging. PHOTOACOUSTICS 2019; 14:77-98. [PMID: 31293884 PMCID: PMC6595011 DOI: 10.1016/j.pacs.2019.05.001] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/09/2019] [Accepted: 05/30/2019] [Indexed: 05/18/2023]
Abstract
Photoacoustic is an emerging biomedical imaging modality, which allows imaging optical absorbers in the tissue by acoustic detectors (light in - sound out). Such a technique has an immense potential for clinical translation since it allows high resolution, sufficient imaging depth, with diverse endogenous and exogenous contrast, and is free from ionizing radiation. In recent years, tremendous developments in both the instrumentation and imaging agents have been achieved. These opened avenues for clinical imaging of various sites allowed applications such as brain functional imaging, breast cancer screening, diagnosis of psoriasis and skin lesions, biopsy and surgery guidance, the guidance of tumor therapies at the reproductive and urological systems, as well as imaging tumor metastases at the sentinel lymph nodes. Here we survey the various clinical and pre-clinical literature and discuss the potential applications and hurdles that still need to be overcome.
Collapse
Affiliation(s)
- Idan Steinberg
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Department of Bioengineering, At Stanford University, School of Medicine, Stanford, CA, United States
| | - David M. Huland
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, At Stanford University, School of Medicine, Stanford, CA, United States
| | - Ophir Vermesh
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, At Stanford University, School of Medicine, Stanford, CA, United States
| | - Hadas E. Frostig
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, At Stanford University, School of Medicine, Stanford, CA, United States
| | - Willemieke S. Tummers
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, At Stanford University, School of Medicine, Stanford, CA, United States
| | - Sanjiv S. Gambhir
- Department of Radiology, At Stanford University, School of Medicine, Stanford, CA, United States
- Department of Bioengineering, At Stanford University, School of Medicine, Stanford, CA, United States
- Department of Materials Science & Engineering, At Stanford University, School of Medicine, Stanford, CA, United States
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection, At Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
78
|
Pownall HJ, Gotto AM. Cholesterol: Can't Live With It, Can't Live Without It. Methodist Debakey Cardiovasc J 2019; 15:9-15. [PMID: 31049144 DOI: 10.14797/mdcj-15-1-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Given its role in many biochemical processes essential to life, cholesterol remains a topic of intense research. Of all the plasma lipids, cholesterol is distinctive because it is a precursor to steroidogenic molecules, some of which regulate metabolism, and its blood concentration in the form of low- and high-density lipoprotein cholesterol (HDL-C) are positive and negative risk factors for atherosclerotic cardiovascular disease (ASCVD). New research, however, has challenged the widely held belief that high HDL-C levels are atheroprotective and is showing that both low and high plasma HDL-C levels confer an increased risk of ASCVD. Furthermore, it is disputing the widely cited mechanism involved in reverse cholesterol transport. This review explores the evolution of cholesterol research starting with the Gofman and Framingham studies, the development of traditional and emerging lipid-lowering therapies, and the role of reverse cholesterol transport in HDL cardioprotection.
Collapse
Affiliation(s)
- Henry J Pownall
- HOUSTON METHODIST RESEARCH INSTITUTE, HOUSTON, TEXAS; WEILL CORNELL MEDICINE, NEW YORK, NEW YORK
| | - Antonio M Gotto
- HOUSTON METHODIST RESEARCH INSTITUTE, HOUSTON, TEXAS; WEILL CORNELL MEDICINE, NEW YORK, NEW YORK
| |
Collapse
|
79
|
Yousefi M, Mamipour M, Sokullu SE, Ghaderi S, Amini H, Rahbarghazi R. Toll-like receptors in the functional orientation of cardiac progenitor cells. J Cell Physiol 2019; 234:19451-19463. [PMID: 31025370 DOI: 10.1002/jcp.28738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
Cardiac progenitor cells (CPCs) have the potential to differentiate into several cell lineages with the ability to restore in cardiac tissue. Multipotency and self-renewal activity are the crucial characteristics of CPCs. Also, CPCs have promising therapeutic roles in cardiac diseases such as valvular disease, thrombosis, atherosclerosis, congestive heart failure, and cardiac remodeling. Toll-like receptors (TLRs), as the main part of the innate immunity, have a key role in the development and differentiation of immune cells. Some reports are found regarding the effect of TLRs in the maturation of stem cells. This article tried to find the potential role of TLRs in the dynamics of CPCs. By showing possible crosstalk between the TLR signaling pathways and CPCs dynamics, we could achieve a better conception related to TLRs in the regeneration of cardiac tissue.
Collapse
Affiliation(s)
- Mohammadreza Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Mina Mamipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Sadiye E Sokullu
- Engineering Sciences, Bioengineering Department, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| | - Shahrooz Ghaderi
- Department of System Physiology, Ruhr University, Bochum, Germany
| | - Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
80
|
Higashi Y, Gautam S, Delafontaine P, Sukhanov S. IGF-1 and cardiovascular disease. Growth Horm IGF Res 2019; 45:6-16. [PMID: 30735831 PMCID: PMC6504961 DOI: 10.1016/j.ghir.2019.01.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/17/2018] [Accepted: 01/30/2019] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is an inflammatory arterial pathogenic condition, which leads to ischemic cardiovascular diseases, such as coronary artery disease and myocardial infarction, stroke, and peripheral arterial disease. Atherosclerosis is a multifactorial disorder and its pathophysiology is highly complex. Changes in expression of multiple genes coupled with environmental and lifestyle factors initiate cascades of adverse events involving multiple types of cells (e.g. vascular endothelial cells, smooth muscle cells, and macrophages). IGF-1 is a pleiotropic factor, which is found in the circulation (endocrine IGF-1) and is also produced locally in arteries (endothelial cells and smooth muscle cells). IGF-1 exerts a variety of effects on these cell types in the context of the pathogenesis of atherosclerosis. In fact, there is an increasing body of evidence suggesting that IGF-1 has beneficial effects on the biology of atherosclerosis. This review will discuss recent findings relating to clinical investigations on the relation between IGF-1 and cardiovascular disease and basic research using animal models of atherosclerosis that have elucidated some of the mechanisms underlying atheroprotective effects of IGF-1.
Collapse
Affiliation(s)
- Yusuke Higashi
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States; Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States.
| | - Sandeep Gautam
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Patrick Delafontaine
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Sergiy Sukhanov
- Department of Medicine, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
81
|
Schuchardt M, Prüfer N, Tu Y, Herrmann J, Hu XP, Chebli S, Dahlke K, Zidek W, van der Giet M, Tölle M. Dysfunctional high-density lipoprotein activates toll-like receptors via serum amyloid A in vascular smooth muscle cells. Sci Rep 2019; 9:3421. [PMID: 30833653 PMCID: PMC6399289 DOI: 10.1038/s41598-019-39846-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 02/01/2019] [Indexed: 01/12/2023] Open
Abstract
Serum amyloid A (SAA) is an uremic toxin and acute phase protein. It accumulates under inflammatory conditions associated with high cardiovascular morbidity and mortality in patients with sepsis or end-stage renal disease (ESRD). SAA is an apolipoprotein of the high-density lipoprotein (HDL). SAA accumulation turns HDL from an anti-inflammatory to a pro-inflammatory particle. SAA activates monocyte chemoattractant protein-1 (MCP-1) in vascular smooth muscle cells. However, the SAA receptor-mediated signaling pathway in vascular cells is poorly understood. Therefore, the SAA-mediated signaling pathway for MCP-1 production was investigated in this study. The SAA-induced MCP-1 production is dependent on the activation of TLR2 and TLR4 as determined by studies with specific receptor antagonists and agonists or siRNA approach. Experiments were confirmed in tissues from TLR2 knockout, TLR4 deficient and TLR2 knock-out/TLR4 deficient mice. The intracellular signaling pathway is IκBα and subsequently NFκB dependent. The MCP-1 production induced by SAA-enriched HDL and HDL isolated from septic patients with high SAA content is also TLR2 and TLR4 dependent. Taken together, the TLR2 and TLR4 receptors are functional SAA receptors mediating MCP-1 release. Furthermore, the TLR2 and TLR4 are receptors for dysfunctional HDL. These results give a further inside in SAA as uremic toxin involved in uremia-related pro-inflammatory response in the vascular wall.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Nicole Prüfer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Yuexing Tu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany.,Zhejiang Provincial People´s Hospital, Intensive Care Unit, Hangzhou, China
| | - Jaqueline Herrmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Xiu-Ping Hu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Sarah Chebli
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Katja Dahlke
- Deutsches Institut für Ernaehrungsforschung, Department of Gastrointestinal Microbiology, Arthur-Scheunert-Allee 114-116, 14558, Nuthethal, Germany
| | - Walter Zidek
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Markus van der Giet
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany.
| | - Markus Tölle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology, Hindenburgdamm 30, 12203, Berlin, Germany
| |
Collapse
|
82
|
Bibli SI, Hu J, Sigala F, Wittig I, Heidler J, Zukunft S, Tsilimigras DI, Randriamboavonjy V, Wittig J, Kojonazarov B, Schürmann C, Siragusa M, Siuda D, Luck B, Abdel Malik R, Filis KA, Zografos G, Chen C, Wang DW, Pfeilschifter J, Brandes RP, Szabo C, Papapetropoulos A, Fleming I. Cystathionine γ Lyase Sulfhydrates the RNA Binding Protein Human Antigen R to Preserve Endothelial Cell Function and Delay Atherogenesis. Circulation 2019; 139:101-114. [DOI: 10.1161/circulationaha.118.034757] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Fragiska Sigala
- First Propedeutic Department of Surgery, Vascular Surgery Division, National and Kapodistrian University of Athens Medical School, Greece (F.S., D.I.T., K.A.F., G.Z.)
| | - Ilka Wittig
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Juliana Heidler
- Functional Proteomics, SFB 815 Core Unit (J.W., J. Heidler), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Sven Zukunft
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Diamantis I. Tsilimigras
- First Propedeutic Department of Surgery, Vascular Surgery Division, National and Kapodistrian University of Athens Medical School, Greece (F.S., D.I.T., K.A.F., G.Z.)
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Janina Wittig
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- Functional Proteomics, SFB 815 Core Unit (J.W., J. Heidler), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Baktybek Kojonazarov
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Justus Liebig University, Germany (B.K.)
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology (C. Schürmann, R.P.B.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Daniel Siuda
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Bert Luck
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Randa Abdel Malik
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| | - Konstantinos A. Filis
- First Propedeutic Department of Surgery, Vascular Surgery Division, National and Kapodistrian University of Athens Medical School, Greece (F.S., D.I.T., K.A.F., G.Z.)
| | - George Zografos
- First Propedeutic Department of Surgery, Vascular Surgery Division, National and Kapodistrian University of Athens Medical School, Greece (F.S., D.I.T., K.A.F., G.Z.)
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.C., D.W.W.)
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.C., D.W.W.)
| | - Josef Pfeilschifter
- Pharmacentre Frankfurt/Zentrum für Arzneimittelforschung, Entwicklung und –Sicherheit (J.P.), Goethe University, Frankfurt am Main, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology (C. Schürmann, R.P.B.), Goethe University, Frankfurt am Main, Germany
| | - Csaba Szabo
- Laboratory of Pharmacology, Department of Medicine, University of Fribourg, Switzerland (C. Szabo)
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Greece (A.P.)
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece (A.P.)
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine (S.-I.B., J. Hu, S.Z., V.R., J.W., M.S., D.S., B.L., R.A.M., I.F.), Goethe University, Frankfurt am Main, Germany
- German Center of Cardiovascular Research, Partner site RheinMain, Frankfurt am Main, Germany (S.-I.B., J. Hu, I.W., J. Heidler, S.Z., V.R., J.W., C. Schürmann, M.S., D.S., B.L., R.A.M., R.P.B., I.F.)
| |
Collapse
|
83
|
Mittal R, Jhaveri VM, Kay SIS, Greer A, Sutherland KJ, McMurry HS, Lin N, Mittal J, Malhotra AK, Patel AP. Recent Advances in Understanding the Pathogenesis of Cardiovascular Diseases and Development of Treatment Modalities. Cardiovasc Hematol Disord Drug Targets 2019; 19:19-32. [PMID: 29737266 DOI: 10.2174/1871529x18666180508111353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/15/2017] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
Cardiovascular Diseases (CVDs) are a leading cause of morbidity and mortality worldwide. The underlying pathology for cardiovascular disease is largely atherosclerotic in nature and the steps include fatty streak formation, plaque progression and plaque rupture. While there is optimal drug therapy available for patients with CVD, there are also underlying drug delivery obstacles that must be addressed. Challenges in drug delivery warrant further studies for the development of novel and more efficacious medical therapies. An extensive understanding of the molecular mechanisms of disease in combination with current challenges in drug delivery serves as a platform for the development of novel drug therapeutic targets for CVD. The objective of this article is to review the pathogenesis of atherosclerosis, first-line medical treatment for CVD, and key obstacles in an efficient drug delivery.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Vasanti M Jhaveri
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Sae-In Samantha Kay
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida FL, United States
| | - Aubrey Greer
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Kyle J Sutherland
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Hannah S McMurry
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Nicole Lin
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Arul K Malhotra
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, Florida FL, United States
| | - Amit P Patel
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida FL, United States
| |
Collapse
|
84
|
Alderfer L, Wei A, Hanjaya-Putra D. Lymphatic Tissue Engineering and Regeneration. J Biol Eng 2018; 12:32. [PMID: 30564284 PMCID: PMC6296077 DOI: 10.1186/s13036-018-0122-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
The lymphatic system is a major circulatory system within the body, responsible for the transport of interstitial fluid, waste products, immune cells, and proteins. Compared to other physiological systems, the molecular mechanisms and underlying disease pathology largely remain to be understood which has hindered advancements in therapeutic options for lymphatic disorders. Dysfunction of the lymphatic system is associated with a wide range of disease phenotypes and has also been speculated as a route to rescue healthy phenotypes in areas including cardiovascular disease, metabolic syndrome, and neurological conditions. This review will discuss lymphatic system functions and structure, cell sources for regenerating lymphatic vessels, current approaches for engineering lymphatic vessels, and specific therapeutic areas that would benefit from advances in lymphatic tissue engineering and regeneration.
Collapse
Affiliation(s)
- Laura Alderfer
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Alicia Wei
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Donny Hanjaya-Putra
- Department of Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556 USA
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46656 USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556 USA
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN 46556 USA
- Advanced Diagnostics and Therapeutics, University of Notre Dame, Notre Dame, IN 46556 USA
- Center for Nanoscience and Technology (NDnano), University of Notre Dame, Notre Dame, IN 46556 USA
| |
Collapse
|
85
|
Fulop T, Witkowski JM, Olivieri F, Larbi A. The integration of inflammaging in age-related diseases. Semin Immunol 2018; 40:17-35. [PMID: 30287177 DOI: 10.1016/j.smim.2018.09.003] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
|
86
|
Yang Q, Xu J, Ma Q, Liu Z, Sudhahar V, Cao Y, Wang L, Zeng X, Zhou Y, Zhang M, Xu Y, Wang Y, Weintraub NL, Zhang C, Fukai T, Wu C, Huang L, Han Z, Wang T, Fulton DJ, Hong M, Huo Y. PRKAA1/AMPKα1-driven glycolysis in endothelial cells exposed to disturbed flow protects against atherosclerosis. Nat Commun 2018; 9:4667. [PMID: 30405100 PMCID: PMC6220207 DOI: 10.1038/s41467-018-07132-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022] Open
Abstract
Increased aerobic glycolysis in endothelial cells of atheroprone areas of blood vessels has been hypothesized to drive increased inflammation and lesion burden but direct links remain to be established. Here we show that endothelial cells exposed to disturbed flow in vivo and in vitro exhibit increased levels of protein kinase AMP-activated (PRKA)/AMP-activated protein kinases (AMPKs). Selective deletion of endothelial Prkaa1, coding for protein kinase AMP-activated catalytic subunit alpha1, reduces glycolysis, compromises endothelial cell proliferation, and accelerates the formation of atherosclerotic lesions in hyperlipidemic mice. Rescue of the impaired glycolysis in Prkaa1-deficient endothelial cells through Slc2a1 overexpression enhances endothelial cell viability and integrity of the endothelial cell barrier, and reverses susceptibility to atherosclerosis. In human endothelial cells, PRKAA1 is upregulated by disturbed flow, and silencing PRKAA1 reduces glycolysis and endothelial viability. Collectively, these results suggest that increased glycolysis in the endothelium of atheroprone arteries is a protective mechanism.
Collapse
Affiliation(s)
- Qiuhua Yang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jiean Xu
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Qian Ma
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Zhiping Liu
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Varadarajan Sudhahar
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yapeng Cao
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Lina Wang
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Xianqiu Zeng
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yaqi Zhou
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Min Zhang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yiming Xu
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, China
| | - Yong Wang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, 610075, Chengdu, China
| | - Neal L Weintraub
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Chunxiang Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Tohru Fukai
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77840, USA
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - Tao Wang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, 518036, Shenzhen, China
| | - David J Fulton
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Mei Hong
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 518055, Shenzhen, China.
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
87
|
Siegel G, Berkholz J, Klüßendorf D, Knosalla C, Zakrzewicz A, Ermilov E, Malmsten M, Lindman B. Atherogenesis and plaque rupture, surface/interface-related phenomena. Colloids Surf A Physicochem Eng Asp 2018. [DOI: 10.1016/j.colsurfa.2018.05.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
88
|
Libby P, Loscalzo J, Ridker PM, Farkouh ME, Hsue PY, Fuster V, Hasan AA, Amar S. Inflammation, Immunity, and Infection in Atherothrombosis: JACC Review Topic of the Week. J Am Coll Cardiol 2018; 72:2071-2081. [PMID: 30336831 PMCID: PMC6196735 DOI: 10.1016/j.jacc.2018.08.1043] [Citation(s) in RCA: 406] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022]
Abstract
Observations on human and experimental atherosclerosis, biomarker studies, and now a large-scale clinical trial support the operation of immune and inflammatory pathways in this disease. The factors that incite innate and adaptive immune responses implicated in atherogenesis and in lesion complication include traditional risk factors such as protein and lipid components of native and modified low-density lipoprotein, angiotensin II, smoking, visceral adipose tissue, and dysmetabolism. Infectious processes and products of the endogenous microbiome might also modulate atherosclerosis and its complications either directly, or indirectly by eliciting local and systemic responses that potentiate disease expression. Trials with antibiotics have not reduced recurrent cardiovascular events, nor have vaccination strategies yet achieved clinical translation. However, anti-inflammatory interventions such as anticytokine therapy and colchicine have begun to show efficacy in this regard. Thus, inflammatory and immune mechanisms can link traditional and emerging risk factors to atherosclerosis, and offer novel avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Joseph Loscalzo
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paul M Ridker
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael E Farkouh
- Peter Munk Cardiac Centre and the Heart and Stroke Richard Lewar Centre, University of Toronto, Toronto, Ontario, Canada
| | - Priscilla Y Hsue
- University of California, San Francisco General Hospital, San Francisco, California
| | | | - Ahmed A Hasan
- The National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Salomon Amar
- Departments of Pharmacology, Immunology and Microbiology, New York Medical College, Valhalla, New York
| |
Collapse
|
89
|
Tsifaki M, Kelaini S, Caines R, Yang C, Margariti A. Regenerating the Cardiovascular System Through Cell Reprogramming; Current Approaches and a Look Into the Future. Front Cardiovasc Med 2018; 5:109. [PMID: 30177971 PMCID: PMC6109758 DOI: 10.3389/fcvm.2018.00109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular disease (CVD), despite the advances of the medical field, remains one of the leading causes of mortality worldwide. Discovering novel treatments based on cell therapy or drugs is critical, and induced pluripotent stem cells (iPS Cells) technology has made it possible to design extensive disease-specific in vitro models. Elucidating the differentiation process challenged our previous knowledge of cell plasticity and capabilities and allows the concept of cell reprogramming technology to be established, which has inspired the creation of both in vitro and in vivo techniques. Patient-specific cell lines provide the opportunity of studying their pathophysiology in vitro, which can lead to novel drug development. At the same time, in vivo models have been designed where in situ transdifferentiation of cell populations into cardiomyocytes or endothelial cells (ECs) give hope toward effective cell therapies. Unfortunately, the efficiency as well as the concerns about the safety of all these methods make it exceedingly difficult to pass to the clinical trial phase. It is our opinion that creating an ex vivo model out of patient-specific cells will be one of the most important goals in the future to help surpass all these hindrances. Thus, in this review we aim to present the current state of research in reprogramming toward the cardiovascular system's regeneration, and showcase how the development and study of a multicellular 3D ex vivo model will improve our fighting chances.
Collapse
Affiliation(s)
- Marianna Tsifaki
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Sophia Kelaini
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rachel Caines
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Chunbo Yang
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andriana Margariti
- The Wellcome-Wolfson Building, Centre for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
90
|
Wang T, Ouyang H, Zhou H, Xia L, Wang X, Wang T. Pro‑atherogenic activation of A7r5 cells induced by the oxLDL/β2GPI/anti‑β2GPI complex. Int J Mol Med 2018; 42:1955-1966. [PMID: 30085340 PMCID: PMC6108850 DOI: 10.3892/ijmm.2018.3805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
A previous study has revealed that oxidized low‑density lipoprotein (oxLDL)/β2‑glycoprotein I (β2GPI)/anti‑β2‑glycoprotein I (anti‑β2GPI), an immune complex, is able to activate the Toll‑like receptor 4 (TLR4)/nuclear factor κβ (NF‑κβ) inflammatory signaling pathway in macrophages, and consequently enhance foam cell formation and the secretion of prothrombin activators. However, the effects of the oxLDL/β2GPI/anti‑β2GPI complex on vascular smooth muscle cells have yet to be investigated. The present study investigated whether the oxLDL/β2GPI/anti‑β2GPI complex was able to reinforce the pro‑atherogenic activities of a rat thoracic aorta smooth muscle cell line (A7r5) and examined the underlying molecular mechanisms. The results revealed that the oxLDL/β2GPI/anti‑β2GPI complex treatment significantly (P<0.05 vs. the media, oxLDL, oxLDL/β2GPI and β2GPI/anti‑β2GPI groups) enhanced the pro‑atherogenic activation of A7r5 cells, including intracellular lipid loading, Acyl‑coenzyme A cholesterol acyltransferase mRNA expression, migration, matrix metalloproteinase‑9 and monocyte chemoattractant protein 1 secretion, all via TLR4. In addition, the expression of TLR4 and the phosphorylation of NF‑κβ p65, p38 and ERK1/2 were also upregulated in oxLDL/β2GPI/anti‑β2GPI complex‑treated A7r5 cells. Pre‑treatment with TAK‑242, a TLR4 inhibitor, was able to partly attenuate the oxLDL/β2GPI/anti‑β2GPI complex‑induced phosphorylation of NF‑κβ p65; however, it had no effect on the phosphorylation of extracellular regulated kinase 1/2 (ERK1/2) and p38. Meanwhile, the NF‑κβ p65 inhibitor ammonium pyrrolidinedithiocarbamate and the ERK1/2 inhibitor U0126, but not the p38 inhibitor SB203580, were able to block oxLDL/β2GPI/anti‑β2GPI complex‑induced foam cell formation and migration in A7r5 cells. Hence, it was demonstrated that the oxLDL/β2GPI/anti‑β2GPI complex is able to enhance the lipid uptake, migration and active molecule secretion of A7r5 cells via TLR4, and finally deteriorate atherosclerosis plaques. Additionally, it was demonstrated that oxLDL/β2GPI/anti‑β2GPI complex‑induced foam cell formation and migration may be partly mediated by the TLR4/NF‑κβ signaling pathway and that ERK1/2 may also participate in the process.
Collapse
Affiliation(s)
- Ting Wang
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hang Ouyang
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hong Zhou
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Longfei Xia
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaoyan Wang
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ting Wang
- Jiangsu Key Laboratory of Medicine Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
91
|
Abou Ziki MD, Mani A. The interplay of canonical and noncanonical Wnt signaling in metabolic syndrome. Nutr Res 2018; 70:18-25. [PMID: 30049588 DOI: 10.1016/j.nutres.2018.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 12/22/2022]
Abstract
Metabolic syndrome is a cluster of inherited metabolic traits, which centers around obesity and insulin resistance and is a major contributor to the growing prevalence of cardiovascular disease. The factors that underlie the association of metabolic traits in this syndrome are poorly understood due to disease heterogeneity and complexity. Genetic studies of kindreds with severe manifestation of metabolic syndrome have led to the identification of casual rare mutations in the LDL receptor-related protein 6, which serves as a co-receptor with frizzled protein receptors for Wnt signaling ligands. Extensive investigations have since unraveled the significance of the Wnt pathways in regulating body mass, glucose metabolism, de novo lipogenesis, low-density lipoprotein clearance, vascular smooth muscle plasticity, liver fat, and liver inflammation. The impaired canonical Wnt signaling observed in the R611C mutation carriers and the ensuing activation of noncanonical Wnt signaling constitute the underlying mechanism for these cardiometabolic abnormalities. Transcription factor 7-like 2 is a key transcription factor activated through LDL receptor-related protein 6 canonical Wnt and reciprocally inhibited by the noncanonical pathway. TC7L2 increases insulin receptor expression, decreases low-density lipoprotein and triglyceride synthesis, and inhibits vascular smooth muscle proliferation. Canonical Wnt also inhibits noncanonical protein kinase C, Ras homolog gene family member A, and Rho associated coiled-coil containing protein kinase 2 activation, thus inhibiting steatohepatitis and transforming growth factor β-mediated extracellular matrix deposition and hepatic fibrosis. Therefore, dysregulation of the highly conserved Wnt signaling pathway underlies the pleiotropy of metabolic traits of the metabolic syndrome and the subsequent end-organ complications.
Collapse
Affiliation(s)
- Maen D Abou Ziki
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510; Deparetment of Genetics, Yale University School of Medicine, New Haven, CT, 06510.
| |
Collapse
|
92
|
Miteva K, Madonna R, De Caterina R, Van Linthout S. Innate and adaptive immunity in atherosclerosis. Vascul Pharmacol 2018; 107:S1537-1891(17)30464-0. [PMID: 29684642 DOI: 10.1016/j.vph.2018.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/03/2018] [Accepted: 04/15/2018] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disorder of the large and medium-size arteries characterized by the subendothelial accumulation of cholesterol, immune cells, and extracellular matrix. At the early onset of atherogenesis, endothelial dysfunction takes place. Atherogenesis is further triggered by the accumulation of cholesterol-carrying low-density lipoproteins, which acquire properties of damage-associated molecular patterns and thereby trigger an inflammatory response. Following activation of the innate immune response, mainly governed by monocytes and macrophages, the adaptive immune response is started which further promotes atherosclerotic plaque formation. In this review, an overview is given describing the role of damage-associated molecular patterns, NLRP3 inflammasome activation, and innate and adaptive immune cells in the atherogenesis process.
Collapse
Affiliation(s)
- Kapka Miteva
- Department of Biomedical Sciences, Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Rozzano, Milano, Italy
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Raffaele De Caterina
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy
| | - Sophie Van Linthout
- Berlin-Brandenburg Center for Regenerative Therapies, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany; Department of Cardiology, Charité, University Medicine Berlin, Campus Virchow Klinikum, Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
93
|
Gillard BK, Rosales C, Xu B, Gotto AM, Pownall HJ. Rethinking reverse cholesterol transport and dysfunctional high-density lipoproteins. J Clin Lipidol 2018; 12:849-856. [PMID: 29731282 DOI: 10.1016/j.jacl.2018.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/26/2018] [Accepted: 04/03/2018] [Indexed: 12/31/2022]
Abstract
Human plasma high-density lipoprotein cholesterol concentrations are a negative risk factor for atherosclerosis-linked cardiovascular disease. Pharmacological attempts to reduce atherosclerotic cardiovascular disease by increasing plasma high-density lipoprotein cholesterol have been disappointing so that recent research has shifted from HDL quantity to HDL quality, that is, functional vs dysfunctional HDL. HDL has varying degrees of dysfunction reflected in impaired reverse cholesterol transport (RCT). In the context of atheroprotection, RCT occurs by 2 mechanisms: one is the well-known trans-hepatic pathway comprising macrophage free cholesterol (FC) efflux, which produces early forms of FC-rich nascent HDL (nHDL). Lecithin:cholesterol acyltransferase converts HDL-FC to HDL-cholesteryl ester while converting nHDL from a disc to a mature spherical HDL, which transfers its cholesteryl ester to the hepatic HDL receptor, scavenger receptor B1 for uptake, conversion to bile salts, or transfer to the intestine for excretion. Although widely cited, current evidence suggests that this is a minor pathway and that most HDL-FC and nHDL-FC rapidly transfer directly to the liver independent of lecithin:cholesterol acyltransferase activity. A small fraction of plasma HDL-FC enters the trans-intestinal efflux pathway comprising direct FC transfer to the intestine. SR-B1-/- mice, which have impaired trans-hepatic FC transport, are characterized by high plasma levels of a dysfunctional FC-rich HDL that increases plasma FC bioavailability in a way that produces whole-body hypercholesterolemia and multiple pathologies. The design of future therapeutic strategies to improve RCT will have to be formulated in the context of these dual RCT mechanisms and the role of FC bioavailability.
Collapse
Affiliation(s)
- Baiba K Gillard
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| | - Corina Rosales
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| | - Bingqing Xu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Antonio M Gotto
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA
| | - Henry J Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA; Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
94
|
Goikuria H, Freijo MDM, Vega Manrique R, Sastre M, Elizagaray E, Lorenzo A, Vandenbroeck K, Alloza I. Characterization of Carotid Smooth Muscle Cells during Phenotypic Transition. Cells 2018; 7:cells7030023. [PMID: 29562638 PMCID: PMC5870355 DOI: 10.3390/cells7030023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 02/07/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are central players in carotid atherosclerosis plaque development. Although the precise mechanisms involved in plaque destabilization are not completely understood, it is known that VSMC proliferation and migration participate in plaque stabilization. In this study, we analyzed expression patterns of genes involved in carotid atherosclerosis development (e.g., transcription factors of regulation of SMC genes) of VSMCs located inside or outside the plaque lesion that may give clues about changes in phenotypic plasticity during atherosclerosis. VSMCs were isolated from 39 carotid plaques extracted from symptomatic and asymptomatic patients by endarterectomy. Specific biomarker expression, related with VSMC phenotype, was analyzed by qPCR, western immunoblot, and confocal microscopy. MYH11, CNN1, SRF, MKL2, and CALD1 were significantly underexpressed in VSMCs from plaques compared with VSMCs from a macroscopically intact (MIT) region, while SPP1, KLF4, MAPLC3B, CD68, and LGALS3 were found significantly upregulated in plaque VSMCs versus MIT VSMCs. The gene expression pattern of arterial VSMCs from a healthy donor treated with 7-ketocholesterol showed high similarity with the expression pattern of carotid plaque VSMCs. Our results indicate that VSMCs isolated from plaque show a typical SMC dedifferentiated phenotype with macrophage-like features compared with VSMCs isolated from a MIT region of the carotid artery. Additionally, MYH11, KLF5, and SPP1 expression patterns were found to be associated with symptomatology of human carotid atherosclerosis.
Collapse
Affiliation(s)
- Haize Goikuria
- Neurogenomiks Neuroscience Department, Faculty of Medicine and Nursing, Basque Country University, 48940 Leioa, Spain.
- ACHUCARRO Basque Center for Neuroscience, Basque Country University, 48940 Leioa, Spain.
| | | | | | - María Sastre
- Neurogenomiks Neuroscience Department, Faculty of Medicine and Nursing, Basque Country University, 48940 Leioa, Spain.
- ACHUCARRO Basque Center for Neuroscience, Basque Country University, 48940 Leioa, Spain.
| | | | - Ana Lorenzo
- Neurology Unit, Basurto University Hospital (BUH), 48013 Bilbao, Spain.
| | - Koen Vandenbroeck
- Neurogenomiks Neuroscience Department, Faculty of Medicine and Nursing, Basque Country University, 48940 Leioa, Spain.
- ACHUCARRO Basque Center for Neuroscience, Basque Country University, 48940 Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| | - Iraide Alloza
- Neurogenomiks Neuroscience Department, Faculty of Medicine and Nursing, Basque Country University, 48940 Leioa, Spain.
- ACHUCARRO Basque Center for Neuroscience, Basque Country University, 48940 Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
95
|
Sargolzaei J, Chamani E, Kazemi T, Fallah S, Soori H. The role of adiponectin and adipolin as anti-inflammatory adipokines in the formation of macrophage foam cells and their association with cardiovascular diseases. Clin Biochem 2018; 54:1-10. [PMID: 29452073 DOI: 10.1016/j.clinbiochem.2018.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023]
Abstract
Obesity is one of the major public health concerns that is closely associated with obesity-related disorders such as type 2 diabetes mellitus (T2DM), hypertension, and atherosclerosis. Atherosclerosis is a chronic disease characterized by excess cholesterol deposition in the arterial intima and the formation of foam cells. Adipocytokines or adipokines are secreted by the adipose tissue as endocrine glands; adiponectin and adipolin are among these adipokines that are associated with obese and insulin-resistant phenotypes. Adipolin and adiponectin are cytokines that exert substantial impact on obesity, progression of atherosclerosis, insulin resistance, and glucose metabolism. In this paper, we review the formation of macrophage foam cells, which are associated with atherosclerosis, and the macrophage mechanism, which includes uptake, esterification, and release. We also summarize current information on adipose tissue-derived hormone and energy homeostasis in obesity. Finally, the role of adipokines, e.g., adipoline and adiponectin, in regulating metabolic, cardiovascular diseases is discussed.
Collapse
Affiliation(s)
- Javad Sargolzaei
- Department of Biochemistry, Institute Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Elham Chamani
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Tooba Kazemi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Soudabeh Fallah
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hosna Soori
- Department of Biochemistry, Institute Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
96
|
Laminar Flow Attenuates Macrophage Migration Inhibitory Factor Expression in Endothelial Cells. Sci Rep 2018; 8:2360. [PMID: 29403061 PMCID: PMC5799263 DOI: 10.1038/s41598-018-20885-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/25/2018] [Indexed: 12/20/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a non-canonical cytokine that is involved in multiple inflammatory diseases, including atherosclerosis. High MIF expression found in leukocytes which facilitates the initiation and progression of atherosclerosis. However, little is known about biomechanical forces in the induction of MIF in endothelial cells (ECs). Here, we show that laminar shear stress (LS) inhibits the expression of MIF in ECs. By profiling the whole transcriptome of human coronary artery ECs under different shear stress, we found that athero-protective LS attenuates the expression of MIF whereas pro-atherosclerotic oscillatory shear stress (OS) significantly increased the expression of MIF. En face staining of rabbit aorta revealed high MIF immunoreactivity in lesser curvature as well as arterial bifurcation areas where OS is predominant. Mechanistically, we found that Krüpple like factor 2 (KLF2) is required for inhibition of MIF expression in ECs in the context of shear stress. Knockdown of KLF2 abolishes LS-dependent MIF inhibition while overexpression of KLF2 significantly attenuated MIF expression. Overall, the present work showed that MIF is a shear stress-sensitive cytokine and is transcriptionally regulated by KLF2, suggesting that LS exerts its athero-protective effect in part by directly inhibiting pro-inflammatory MIF expression.
Collapse
|
97
|
Missimer A, Fernandez ML, DiMarco DM, Norris GH, Blesso CN, Murillo AG, Vergara-Jimenez M, Lemos BS, Medina-Vera I, Malysheva OV, Caudill MA. Compared to an Oatmeal Breakfast, Two Eggs/Day Increased Plasma Carotenoids and Choline without Increasing Trimethyl AmineN-Oxide Concentrations. J Am Coll Nutr 2018; 37:140-148. [DOI: 10.1080/07315724.2017.1365026] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Amanda Missimer
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Diana M. DiMarco
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Gregory H. Norris
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Christopher N. Blesso
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Ana Gabriela Murillo
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | | | - Bruno S. Lemos
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Isabel Medina-Vera
- Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Olga V. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | - Marie A. Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| |
Collapse
|
98
|
Docherty CK, Carswell A, Friel E, Mercer JR. Impaired mitochondrial respiration in human carotid plaque atherosclerosis: A potential role for Pink1 in vascular smooth muscle cell energetics. Atherosclerosis 2018; 268:1-11. [PMID: 29156421 PMCID: PMC6565844 DOI: 10.1016/j.atherosclerosis.2017.11.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/30/2017] [Accepted: 11/10/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS DNA damage and mitochondrial dysfunction are thought to play an essential role in ageing and the energetic decline of vascular smooth muscle cells (VSMCs) essential for maintaining plaque integrity. We aimed to better understand VSMCs and identify potentially useful compensatory pathways that could extend their lifespan. Moreover, we wanted to assess if defects in mitochondrial respiration exist in human atherosclerotic plaques and to identify the appropriate markers that may reflect a switch in VSMC energy metabolism. METHODS Human plaque tissue and cells were assessed for composition and evidence of DNA damage, repair capacity and mitochondrial dysfunction. Fresh plaque tissue was evaluated using high resolution oxygen respirometry to assess oxidative metabolism. Recruitment and processing of the mitochondrial regulator of autophagy Pink1 kinase was investigated in combination with transcriptional and protein markers associated with a potential switch to a more glycolytic metabolism. RESULTS Human VSMC have increased nuclear (nDNA) and mitochondrial (mtDNA) damage and reduced repair capacity. A subset of VSMCs within plaque cap had decreased oxidative phosphorylation and expression of Pink1 kinase. Plaque cells demonstrated increased glycolytic activity in response to loss of mitochondrial function. A potential compensatory glycolytic program may act as energetic switch via AMP kinase (AMPK) and hexokinase 2 (Hex2). CONCLUSIONS We have identified a subset of plaque VSMCs required for plaque stability that have increased mitochondrial dysfunction and decreased oxidative phosphorylation. Pink1 kinase may initiate a cellular response to promote a compensatory glycolytic program associated with upregulation of AMPK and Hex2.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Carotid Arteries/enzymology
- Carotid Arteries/pathology
- Carotid Artery Diseases/enzymology
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/pathology
- Cells, Cultured
- DNA Damage
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Energy Metabolism
- Glycolysis
- Hexokinase/metabolism
- Humans
- Mitochondria, Muscle/enzymology
- Mitochondria, Muscle/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Oxidative Phosphorylation
- Oxidative Stress
- Plaque, Atherosclerotic
- Protein Kinases/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Craig K Docherty
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University Avenue, University of Glasgow, Glasgow, G12 8TA, Scotland, United Kingdom
| | - Andy Carswell
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University Avenue, University of Glasgow, Glasgow, G12 8TA, Scotland, United Kingdom
| | - Elaine Friel
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University Avenue, University of Glasgow, Glasgow, G12 8TA, Scotland, United Kingdom
| | - John R Mercer
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University Avenue, University of Glasgow, Glasgow, G12 8TA, Scotland, United Kingdom.
| |
Collapse
|
99
|
Lv JX, Kong Q, Ma X. Current advances in circulating inflammatory biomarkers in atherosclerosis and related cardio-cerebrovascular diseases. Chronic Dis Transl Med 2017; 3:207-212. [PMID: 29354803 PMCID: PMC5747494 DOI: 10.1016/j.cdtm.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis (AS) is a systemic chronic disease affecting both the coronary and cerebral arteries. Inflammation plays a key role in the initiation and progression of AS, and numerous inflammatory factors have been proposed as potential biomarkers. This article reviews recent research in studies on major circulating inflammatory biomarkers to identify surrogates that may reflect processes associated with AS development and the risk of AS-related vascular events, such as Von Willebrand factor, lectin-like oxidized low-density-lipoprotein receptor-1, soluble urokinase plasminogen activator receptor, regulated upon activation, normal T-cell expressed and secreted, and microparticles, which may provide new perspectives for clinical AS evaluation and risk stratification.
Collapse
Affiliation(s)
- Jun-Xuan Lv
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Qi Kong
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Xin Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| |
Collapse
|
100
|
Desyatova A, Poulson W, Deegan P, Lomneth C, Seas A, Maleckis K, MacTaggart J, Kamenskiy A. Limb flexion-induced twist and associated intramural stresses in the human femoropopliteal artery. J R Soc Interface 2017; 14:rsif.2017.0025. [PMID: 28330991 DOI: 10.1098/rsif.2017.0025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 02/24/2017] [Indexed: 01/18/2023] Open
Abstract
High failure rates of femoropopliteal artery (FPA) interventions are often attributed to severe mechanical deformations that occur with limb movement. Torsion of the FPA likely plays a significant role, but is poorly characterized and the associated intramural stresses are currently unknown. FPA torsion in the walking, sitting and gardening postures was characterized in n = 28 in situ FPAs using intra-arterial markers. Principal mechanical stresses and strains were quantified in the superficial femoral artery (SFA), adductor hiatus segment (AH) and the popliteal artery (PA) using analytical modelling. The FPA experienced significant torsion during limb flexion that was most severe in the gardening posture. The associated mechanical stresses were non-uniformly distributed along the length of the artery, increasing distally and achieving maximum values in the PA. Maximum twist in the SFA ranged 10-13° cm-1, at the AH 8-16° cm-1, and in the PA 14-26° cm-1 in the walking, sitting and gardening postures. Maximum principal stresses were 30-35 kPa in the SFA, 27-37 kPa at the AH and 39-43 kPa in the PA. Understanding torsional deformations and intramural stresses in the FPA can assist with device selection for peripheral arterial disease interventions and may help guide the development of devices with improved characteristics.
Collapse
Affiliation(s)
- Anastasia Desyatova
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - William Poulson
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul Deegan
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carol Lomneth
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Andreas Seas
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland, Baltimore, MD, USA
| | - Kaspars Maleckis
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jason MacTaggart
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Alexey Kamenskiy
- Department of Surgery, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|