401
|
Liang Y, Li L, Chen Y, Xiao J, Wei D. PD-1/PD-L1 immune checkpoints: Tumor vs atherosclerotic progression. Clin Chim Acta 2021; 519:70-75. [PMID: 33872608 DOI: 10.1016/j.cca.2021.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/13/2022]
Abstract
Immunotherapy has become one of the most attraction cancer therapy strategies. The PD-1/PD-L1 pathway plays key roles in immune responses and autoimmunity by regulating T cell activity. Overactivation of this pathway dampens T cell and immune function, which allows tumor cells immune escape. Antibody or inhibitors of PD-1/PD-L1 immune targets have been implicated in clinic anti-cancer therapy and gain great clinic outcoming for their high efficiency. However, recent studies showed that the PD-1/PD-L1 immunotherapy in some tumor patients was found to accelerate T cell-driven inflammatory and the progression of atherosclerotic lesions. This article reviews the research progression of PD-1/PD-L1 in tumors and atherosclerosis, and the possible mechanisms of anti-PD-1/PD-L1 immunotherapy increasing the risk of atherosclerotic lesions.
Collapse
Affiliation(s)
- Yamin Liang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Lu Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Yanmei Chen
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Jinyan Xiao
- YueYang Maternal-Child Medicine Health Hospital Hunan Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women & Children's Medical Center, Yueyang, Hunan 414000, China.
| | - Dangheng Wei
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
402
|
BMP4-mediated browning of perivascular adipose tissue governs an anti-inflammatory program and prevents atherosclerosis. Redox Biol 2021; 43:101979. [PMID: 33895484 PMCID: PMC8099561 DOI: 10.1016/j.redox.2021.101979] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 01/13/2023] Open
Abstract
Loss of perivascular adipose tissue (PVAT) impairs endothelial function and enhances atherosclerosis. However, the roles of PVAT thermoregulation in vascular inflammation and the development of atherosclerosis remains unclear. Bone morphogenetic protein 4 (BMP4) transforms white adipocyte to beige adipocyte, while promotes a brown-to-white shift in inter-scapular brown adipose tissue (BAT). Here, we found that knockdown of BMP4 in PVAT reduced expression of brown adipocyte-characteristic genes and increased endothelial inflammation in vitro co-culture system. Ablating BMP4 expression either in adipose tissues or specifically in BAT in ApoE−/− mice demonstrated a marked exacerbation of atherosclerotic plaque formation in vivo. We further demonstrated that proinflammatory factors (especially IL-1β) increased in the supernatant of BMP4 knockdown adipocytes. Overexpression of BMP4 in adipose tissues promotes browning of PVAT and protects against atherosclerosis in ApoE−/− mice. These findings uncover an organ crosstalk between PVAT and blood endothelial cells that is engaged in atherosclerosis. BMP4 expression positively correlates with browning but negatively with coronary artery disease. BMP4 KO leads to impaired PVAT metabolism and atherosclerosis. Impaired PVAT metabolism mainly induces atherosclerosis by activating inflammation rather than increasing blood lipids. Impaired PVAT metabolism drive local inflammation by inducing the secretion of adipocyte-derived proinflammatory factors.
Collapse
|
403
|
Iannuzzo G, Gentile M, Bresciani A, Mallardo V, Di Lorenzo A, Merone P, Cuomo G, Pacileo M, Sarullo FM, Venturini E, D’Andrea A, Vigorito C, Giallauria F. Inhibitors of Protein Convertase Subtilisin/Kexin 9 (PCSK9) and Acute Coronary Syndrome (ACS): The State-of-the-Art. J Clin Med 2021; 10:1510. [PMID: 33916362 PMCID: PMC8038577 DOI: 10.3390/jcm10071510] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 01/12/2023] Open
Abstract
Acute Coronary Syndrome (ACS) remains one of the most frequent causes of morbidity and mortality in the world. Although the age- and gender-adjusted incidence of ACS is decreasing, the mortality associated with this condition remains high, especially 1-year after the acute event. Several studies demonstrated that PCSK9 inhibitors therapy determine a significant reduction of major adverse cardiovascular events (MACE) in post-ACS patients, through a process of plaque modification, by intervening in lipid metabolism and platelet aggregation and finally determining an improvement in endothelial function. In the EVACS (Evolocumab in Acute Coronary Syndrome) study, evolocumab allows >90% of patients to achieve LDL-C < 55 mg/dL according to ESC/EAS guidelines compared to 11% of patients who only receive statins. In the EVOPACS (EVOlocumab for Early Reduction of low-density lipoprotein (LDL)-cholesterol Levels in Patients With Acute Coronary Syndromes) study, evolocumab determined LDL levels reduction of 40.7% (95% CI: 45.2 to 36.2; p < 0.001) and allowed 95.7% of patients to achieve LDL levels <55 mg/dL. In ODYSSEY Outcome trial, alirocumab reduced the overall risk of MACE by 15% (HR = 0.85; CI: 0.78-0.93; p = 0.0003), with a reduced risk of all-cause mortality (HR = 0.85; CI: 0.73-0.98: nominal p = 0026), and fewer deaths for coronary heart disease (CHD) compared to the control group (HR = 0.92; CI: 0.76-1.11; p = 0.38). The present review aimed at describing the beneficial effect of PCSK9 inhibitors therapy early after ACS in reducing LDL circulating levels (LDL-C) and the risk of major adverse cardiovascular events, which was very high in the first year and persists higher later after the acute event.
Collapse
Affiliation(s)
- Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.G.); (V.M.)
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.G.); (V.M.)
| | - Alessandro Bresciani
- Department of Medicine and Medical Specialties, A. Cardarelli Hospital, 80131 Naples, Italy;
| | - Vania Mallardo
- Department of Clinical Medicine and Surgery, “Federico II” University, 80131 Naples, Italy; (M.G.); (V.M.)
| | - Anna Di Lorenzo
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (A.D.L.); (P.M.); (G.C.); (C.V.); (F.G.)
| | - Pasquale Merone
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (A.D.L.); (P.M.); (G.C.); (C.V.); (F.G.)
| | - Gianluigi Cuomo
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (A.D.L.); (P.M.); (G.C.); (C.V.); (F.G.)
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, “Umberto I” Hospital, Viale San Francesco, 84014 Nocera Inferiore, Italy; (M.P.); (A.D.)
| | - Filippo M. Sarullo
- Cardiovascular Rehabilitation Unit, Buccheri La Ferla Fatebenefratelli Hospital, 90123 Palermo, Italy;
| | - Elio Venturini
- Cardiac Rehabilitation Unit, Azienda USL Toscana Nord-Ovest, Cecina Civil Hospital, 57023 Cecina, Italy;
| | - Antonello D’Andrea
- Unit of Cardiology and Intensive Care, “Umberto I” Hospital, Viale San Francesco, 84014 Nocera Inferiore, Italy; (M.P.); (A.D.)
| | - Carlo Vigorito
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (A.D.L.); (P.M.); (G.C.); (C.V.); (F.G.)
| | - Francesco Giallauria
- Department of Translational Medical Sciences, “Federico II” University of Naples, Via S. Pansini 5, 80131 Naples, Italy; (A.D.L.); (P.M.); (G.C.); (C.V.); (F.G.)
| |
Collapse
|
404
|
Does Familial Mediterranean Fever Provoke Atherosclerosis in Children? Evaluation of Arterial Stiffness and Serum Endocan Levels. Clin Rheumatol 2021; 40:4199-4206. [PMID: 33813620 DOI: 10.1007/s10067-021-05721-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES This study aimed to evaluate the risk for atherosclerosis by using echocardiographic arterial stiffness (AS) parameters and serum endocan levels, as a biomarker of endothelial dysfunction (ED) in children with FMF. METHODS Seventy-nine children with FMF (12-18 years) and 41 healthy children were included, and clinical features (age at the first attack, age at the time of diagnosis, diagnosis delay time, colchicine dose, biological agent usage, MEFV mutations, and symptoms of attacks) of patients were noted. Arterial stiffness parameters were calculated by using echocardiographic aortic measurements with blood pressure monitoring. Hemogram parameters, acute phase reactants, blood glucose and lipid levels of 12 hours of fasting, and serum endocan levels were evaluated for all participants. RESULTS There were no statistically significance regarding demographic features, acute phase reactants, and hemogram parameters. Blood glucose and lipid levels were similar, except for HDL (lower in FMF group, p=0.029). Serum endocan levels did not differ in two groups (p=0.906). Only stiffness of descending aorta was lower in FMF group (p=0.028), and the other AS parameters were similar between two groups (p>0.05 for each parameters). CONCLUSION Good disease control could be preventive for atherosclerosis in children with FMF. On the other hand, screening for cardiovascular diseases is essential, particularly for uncontrolled cases. Distribution of MEFV gene mutations KEY POINTS: • Exaggerated inflammation is the prominent feature of FMF attacks; moreover, it is shown that subclinical inflammation might also continue in attack-free periods. • Chronic inflammation contributes to atherosclerotic process in almost all stages by activating endothelial cells, producing reactive oxygen species, and accelerating foam cell and atherosclerotic plaque formations. • However, the results of this study showed that there was no difference in terms of atherosclerotic markers such as serum endocan levels and arterial stiffness parameters between pediatric FMF patients and healthy peers. • Good disease control in pediatric FMF patients may prevent early atherosclerotic changes during childhood, which then may lead a probable decreased risk of subsequent CVD in adulthood.
Collapse
|
405
|
Han X, Wang H, Li Y, Liu L, Gao S. A 2 miRNAs-based signature for the diagnosis of atherosclerosis. BMC Cardiovasc Disord 2021; 21:150. [PMID: 33761890 PMCID: PMC7988968 DOI: 10.1186/s12872-021-01960-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a leading cause of vascular disease worldwide. MicroRNAs (miRNAs) play an essential role in the development of AS. However, the miRNAs-based biomarkers for the diagnosis of AS are still limited. Here, we aimed to identify the miRNAs significantly related to AS and construct the predicting model based on these miRNAs for distinguishing the AS patients from healthy cases. METHODS The miRNA and mRNA expression microarray data of blood samples from patients with AS and healthy cases were obtained from the GSE59421 and GSE20129 of Gene Expression Omnibus (GEO) database, respectively. Weighted Gene Co-expression Network Analysis (WGCNA) was performed to evaluate the correlation of the miRNAs and mRNAs with AS and identify the miRNAs and mRNAs significantly associated with AS. The potentially critical miRNAs were further optimized by functional enrichment analysis. The logistic regression models were constructed based on these optimized miRNAs and validated by threefold cross-validation method. RESULTS WGCNA revealed 42 miRNAs and 532 genes significantly correlated with AS. Functional enrichment analysis identified 12 crucial miRNAs in patients with AS. Moreover, 6 miRNAs among the identified 12 miRNAs, were selected using a stepwise regression model, in which four miRNAs, including hsa-miR-654-5p, hsa-miR-409-3p, hsa-miR-485-5p and hsa-miR-654-3p, were further identified through multivariate regression analysis. The threefold cross-validation method showed that the AUC of logistic regression model based on the four miRNAs was 0.7308, 0.8258, and 0.7483, respectively, with an average AUC of 0.7683. CONCLUSION We identified a total of four miRNAs, including hsa-miR-654-5p and hsa-miR-409-3p, are identified as the potentially critical biomarkers for AS. The logistic regression model based on the identified 2 miRNAs could reliably distinguish the patients with AS from normal cases.
Collapse
Affiliation(s)
- Xiujiang Han
- Department of Geriatrics, Tianjin NanKai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin City, 300100, China
| | - Huimin Wang
- Department of Neurology, Tianjin NanKai Hospital, Tianjin City, 300100, China
| | - Yongjian Li
- First Department of Cardiovascular Medicine, Tianjin NanKai Hospital, Tianjin City, 300100, China
| | - Lina Liu
- Department of Geriatrics, Tianjin NanKai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin City, 300100, China
| | - Sheng Gao
- Nankai University, No. 94 Weijin Road, Nankai District, Tianjin City, 300071, China.
| |
Collapse
|
406
|
Zhang P, Liang T, Wang X, Wu T, Xie Z, Yu Y, Yu H. Serum-Derived Exosomes from Patients with Coronary Artery Disease Induce Endothelial Injury and Inflammation in Human Umbilical Vein Endothelial Cells. Int Heart J 2021; 62:396-406. [PMID: 33731537 DOI: 10.1536/ihj.20-641] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Endothelial injury and inflammation have been found to be essential in the pathogenesis of coronary artery disease (CAD). Circulating exosomes are of great value as novel biomarkers for CAD. However, the role of circulating exosomes in the pathogenesis of CAD remains unclear. Thus, in this study, we aimed to examine whether circulating exosomes from CAD are involved in the endothelial injury and inflammation. The serum-derived exosomes were isolated from CAD and controls using an ExoQuick reagent, and these were then quantified by measuring the protein levels using BCA methods. The uptake of exosomes by human umbilical vein endothelial cells (HUVECs) was observed by laser scanning microscope and analyzed via flow cytometry. Then, HUVECs were treated with vehicle, exosomes from CAD (CAD-exo), and controls (ctrl-exo) in the absence and presence of vascular endothelial growth factor (VEGF). Cell viability, migration, and angiogenesis were evaluated using CCK-8 assay, scratch assay, and tube formation assay. Inflammatory factors including IL-1β, IL-6, TNF-α, ICAM-1, and VCAM-1 levels were detected via qPCR. As per our findings, no significant differences were noted in uptake of ctrl-exo and CAD-exo by HUVECs. CAD-exo suppressed cell viability in a dose-dependent manner. Compared with ctrl-exo, CAD-exo-treated HUVECs significantly suppressed migration and angiogenesis. However, CAD-exo had a stronger inhibitory effect on VEGF-induced migration and angiogenesis compared with ctrl-exo. Moreover, IL-1β, TNF-α, and ICAM-1 were determined to be significantly upregulated in HUVECs treated with CAD-exo, but IL-6 and VCAM-1 expressions were not affected. Overall, our results suggest that CAD-exo are involved in endothelial injury and inflammation, which may, in turn, cause endothelial dysfunction and potentially promote the development of CAD.
Collapse
Affiliation(s)
- Ping Zhang
- Second School of Clinical Medicine, Southern Medical University.,Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science
| | - Tao Liang
- Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science.,Guangdong General Hospital, School of Medicine, South China University of Technology
| | - Xuan Wang
- Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science.,Guangdong General Hospital, School of Medicine, South China University of Technology
| | - Tianlong Wu
- Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science
| | - Zhixin Xie
- Second School of Clinical Medicine, Southern Medical University.,Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science
| | - Yanhong Yu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University
| | - Huimin Yu
- Second School of Clinical Medicine, Southern Medical University.,Department of Cardiovascular, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Science
| |
Collapse
|
407
|
Tang M, Hong L, Li H, Chen W, Tai L, Minshall R, Huang W, Chen J. Stiffness of aortic arch and carotid arteries increases in ApoE-knockout mice with high-fat diet: evidence from echocardiography. Am J Transl Res 2021; 13:1352-1364. [PMID: 33841661 PMCID: PMC8014403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/26/2020] [Indexed: 06/12/2023]
Abstract
Arterial stiffness is an effective predictor of atherosclerosis. Measurement of pulse-wave velocity (PWV) is a gold-standard approach to study arterial stiffness. This study aims to examine arterial stiffness and heart functions via echocardiography at an early stage of atherosclerosis. A model of atherosclerosis in ApoE-knockout (ApoE-/- ) mice fed on high-fat diet (HFD) was used, with normal chow diet (ND) as a control. Stiffness of aortic arch and carotid arteries and left ventricular (LV) systolic/diastolic functions were measured by echocardiography. The plasma cholesterol levels and atherosclerotic plaque areas in the aortas were measured. The PWV values of aortic arch and carotid arteries were compared at 2, 4, 6 and 8 weeks with different diets. Compared with ND mice, PWV values in aortic arch and carotid arteries were significantly increased in HFD mice after 8 weeks (Aortic arch: 516.65 ± 216.89 cm/s vs. 192.53 ± 71.71 cm/s; Carotid arteries: 514.26 ± 211.01 cm/s vs. 188.03 ± 75.14 cm/s, respectively; both P < 0.01) accompanied by the decrease in LV systolic/diastolic functions. These were well correlated with the increase in plasma cholesterol levels. Echo-based PWV measurement in the aortic arch was found more sensitive to predict atherosclerosis than in the carotid arteries in ApoE-/- mice. Measuring aortic arch PWV via echocardiography could represent a new diagnostic strategy for early detection of atherosclerosis.
Collapse
Affiliation(s)
- Ming Tang
- Department of Medicine, University of Illinois at ChicagoChicago, IL 60612, USA
- Center for Cardiovascular Research, University of Illinois at ChicagoChicago, IL 60612, USA
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Liang Hong
- Department of Medicine, University of Illinois at ChicagoChicago, IL 60612, USA
| | - Haibin Li
- Department of Medicine, University of Illinois at ChicagoChicago, IL 60612, USA
- Center for Cardiovascular Research, University of Illinois at ChicagoChicago, IL 60612, USA
- Department of Pathology and Institute of Precision Medicine, Jining Medical UniversityJining 272067, China
| | - Wanshi Chen
- Department of Cardiology, The Children’s Hospital of Chongqing Medical UniversityChongqing 400014, China
| | - Leon Tai
- Department of Anatomy and Cell Biology, University of Illinois at ChicagoChicago, IL 60612, USA
| | - Richard Minshall
- Department of Anesthesiology, University of Illinois at ChicagoChicago, IL 60612, USA
| | - Wei Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Jiwang Chen
- Department of Medicine, University of Illinois at ChicagoChicago, IL 60612, USA
- Center for Cardiovascular Research, University of Illinois at ChicagoChicago, IL 60612, USA
| |
Collapse
|
408
|
From Mitochondria to Atherosclerosis: The Inflammation Path. Biomedicines 2021; 9:biomedicines9030258. [PMID: 33807807 PMCID: PMC8000234 DOI: 10.3390/biomedicines9030258] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key process in metazoan organisms due to its relevance for innate defense against infections and tissue damage. However, inflammation is also implicated in pathological processes such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease of the arterial wall where unstable atherosclerotic plaque rupture causing platelet aggregation and thrombosis may compromise the arterial lumen, leading to acute or chronic ischemic syndromes. In this review, we will focus on the role of mitochondria in atherosclerosis while keeping inflammation as a link. Mitochondria are the main source of cellular energy. Under stress, mitochondria are also capable of controlling inflammation through the production of reactive oxygen species (ROS) and the release of mitochondrial components, such as mitochondrial DNA (mtDNA), into the cytoplasm or into the extracellular matrix, where they act as danger signals when recognized by innate immune receptors. Primary or secondary mitochondrial dysfunctions are associated with the initiation and progression of atherosclerosis by elevating the production of ROS, altering mitochondrial dynamics and energy supply, as well as promoting inflammation. Knowing and understanding the pathways behind mitochondrial-based inflammation in atheroma progression is essential to discovering alternative or complementary treatments.
Collapse
|
409
|
Xu S, Zhang J, Liu J, Ye J, Xu Y, Wang Z, Yu J, Ye D, Zhao M, Feng Y, Pan W, Wang M, Wan J. The role of interleukin-10 family members in cardiovascular diseases. Int Immunopharmacol 2021; 94:107475. [PMID: 33662690 DOI: 10.1016/j.intimp.2021.107475] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 12/15/2022]
Abstract
Interleukin (IL)-10 cytokine family members, including IL-10, IL-19, IL-20, IL-22, IL-24, IL-26 and the distantly related IL-28A, IL-28B, and IL-29, play critical roles in the regulation of inflammation. The occurrence and progression of cardiovascular diseases closely correlate with the regulation of inflammation, which may provide novel strategies for the treatment of cardiovascular diseases. In recent years, studies have focused on the association between the IL-10 cytokine family and the physiological and pathological progression of cardiovascular diseases. The aim of this review is to summarize relevant studies and clarify whether the IL-10 cytokine family contributes to the regulation of cardiovascular diseases.
Collapse
Affiliation(s)
- Shuwan Xu
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junping Yu
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yongqi Feng
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Jun Wan
- The First Clinical College of Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
410
|
Yang L, Chen L, Fang Y, Ma S. Downregulation of GSK-3β Expression via Ultrasound-Targeted Microbubble Destruction Enhances Atherosclerotic Plaque Stability in New Zealand Rabbits. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:710-722. [PMID: 33261913 DOI: 10.1016/j.ultrasmedbio.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/14/2020] [Accepted: 11/01/2020] [Indexed: 06/12/2023]
Abstract
Accumulating evidence suggests that atherosclerosis (AS) is the underlying cause of vascular diseases, including heart disease and stroke. Ultrasound-targeted microbubble destruction (UTMD) technology provides a tolerable, efficient and effective system for drug delivery and gene transfection, which has broad application prospects in the treatment of AS. In addition, glycogen synthase kinase (GSK)-3β has been implicated as a potentially valuable therapeutic agent for AS treatment; however, the specific molecular mechanisms remain unknown. Therefore, this study was conducted to explore the effect of downregulation of GSK-3β expression via UTMD on atherosclerotic plaque stability. We established a THP-1 macrophage-derived foam cell model in vitro and an atherosclerotic plaque model in the right common carotid artery of New Zealand rabbits. We determined levels of the relevant vulnerable plaque stability elements. The results indicate that GSK-3β was upregulated in the foam cells and in atherosclerotic rabbits. Downregulation of GSK-3β expression by UTMD suppressed vulnerable plaque factors and inflammation in vitro and in vivo, changed the cytoskeleton of the foam cells in vitro, increased Young's modulus and decreased the peak intensity of atherosclerotic plaque in vivo. Moreover, GSK-3β inhibition by UTMD did not influence the viability of the foam cells. Collectively, our results indicate that GSK-3β could be a potential target for anti-atherogenic interventions and, in particular, can improve the stability of AS plaques in combination with UTMD.
Collapse
Affiliation(s)
- Lifei Yang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Lingzi Chen
- Ningbo University School of Medicine, Ningbo, China
| | - Ye Fang
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Suya Ma
- Department of Ultrasound, Ningbo Urology and Nephrology Hospital, Ningbo, China.
| |
Collapse
|
411
|
Mata R, Yao Y, Cao W, Ding J, Zhou T, Zhai Z, Gao C. The Dynamic Inflammatory Tissue Microenvironment: Signality and Disease Therapy by Biomaterials. RESEARCH 2021; 2021:4189516. [PMID: 33623917 PMCID: PMC7879376 DOI: 10.34133/2021/4189516] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022]
Abstract
Tissue regeneration is an active multiplex process involving the dynamic inflammatory microenvironment. Under a normal physiological framework, inflammation is necessary for the systematic immunity including tissue repair and regeneration as well as returning to homeostasis. Inflammatory cellular response and metabolic mechanisms play key roles in the well-orchestrated tissue regeneration. If this response is dysregulated, it becomes chronic, which in turn causes progressive fibrosis, improper repair, and autoimmune disorders, ultimately leading to organ failure and death. Therefore, understanding of the complex inflammatory multiple player responses and their cellular metabolisms facilitates the latest insights and brings novel therapeutic methods for early diseases and modern health challenges. This review discusses the recent advances in molecular interactions of immune cells, controlled shift of pro- to anti-inflammation, reparative inflammatory metabolisms in tissue regeneration, controlling of an unfavorable microenvironment, dysregulated inflammatory diseases, and emerging therapeutic strategies including the use of biomaterials, which expand therapeutic views and briefly denote important gaps that are still prevailing.
Collapse
Affiliation(s)
- Rani Mata
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wangbei Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
412
|
HDAC3 protects against atherosclerosis through inhibition of inflammation via the microRNA-19b/PPARγ/NF-κB axis. Atherosclerosis 2021; 323:1-12. [PMID: 33756273 DOI: 10.1016/j.atherosclerosis.2021.02.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is one of the leading causes of cardiovascular diseases. Studies have revealed critical roles of microRNAs (miRNAs) in the progression of AS. This study was conducted to elucidate the role and mechanism by which miR-19b influences AS. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with oxidized-low-density lipoprotein (ox-LDL), and an AS mouse model was generated with the help of ApoE-/- mice using a high-fat diet regimen. The expression patterns of peroxisome proliferator-activated receptor γ (PPARγ), nuclear factor κB (NF-κB)/p65, miR-19b and histone deacetylase 3 (HDAC3) were then characterized by reverse transcription quantitative polymerase chain reaction and Western blot analysis. In addition, the relationship among PPARγ, NF-κB/p65, miR-19b and HDAC3 was evaluated by co-immunoprecipitation, chromatin immunoprecipitation and dual-luciferase reporter gene assays. Gain- and loss-of-function experiments were also performed to examine their functional significance on ox-LDL-induced inflammation in HUVECs. Enzyme-linked immunosorbent assay was applied to determine the expression patterns of inflammatory factors in AS mice. RESULTS PPARγ and HDAC3 were poorly expressed, while miR-19b and NF-κB/p65 were highly expressed in ox-LDL-induced HUVECs and arterial tissues of AS mice. PPARγ inhibited ox-LDL-induced inflammation in HUVECs by ubiquitination and degradation of NF-κB/p65. miR-19b, downregulated by HDAC3, targeted PPARγ and negatively-regulated its expression. Upregulated PPARγ or HDAC3 or downregulated miR-19b or NF-κB/p65 reduced TNF-α and IL-1β expression levels in ox-LDL-induced HUVECs and AS mice. CONCLUSIONS Collectively, the results show that HDAC3 upregulation prevents inflammation to inhibit AS by inactivating NF-κB/p65 via upregulation of miR-19b-mediated PPARγ, providing a basic therapeutic consideration for AS treatment.
Collapse
|
413
|
Liu MN, Luo G, Gao WJ, Yang SJ, Zhou H. miR-29 family: A potential therapeutic target for cardiovascular disease. Pharmacol Res 2021; 166:105510. [PMID: 33610720 DOI: 10.1016/j.phrs.2021.105510] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 01/11/2023]
Abstract
Cardiovascular disease (CVD), including heart failure, myocardial fibrosis and myocardial infarction, etc, remains one of the leading causes of mortality worldwide. Evidence shows that miRNA plays an important role in the pathogenesis of CVD. miR-29 family is one of miRNA, and over the past decades, many studies have demonstrated that miR-29 is involved in maintaining the integrity of arteries and in the regulation of atherosclerosis, especially in the process of myocardial fibrosis. Besides, heart failure, myocardial fibrosis and myocardial infarction are inseparable from the regulatory role of miR-29. Here, we comprehensively review recent studies regarding miR-29 and CVD, illustrate the possibility of miR-29 as a potential marker for prevention, treatment and prognostic observation.
Collapse
Affiliation(s)
- Meng-Nan Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Wan-Jiao Gao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Si-Jin Yang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China; National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, Sichuan, China.
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China.
| |
Collapse
|
414
|
Non- 18F-FDG/ 18F-NaF Radiotracers Proposed for the Diagnosis and Management of Diseases of the Heart and Vasculature. PET Clin 2021; 16:273-284. [PMID: 33589388 DOI: 10.1016/j.cpet.2020.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
18F-fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) are front-runners in PET. However, these tracers have limitations in the imaging of diseases in the heart. A multitude of other radiotracers have been identified as potentially useful PET agents in the identification of cardiovascular disease. This critical review examines recent studies with the use of non-18F-FDG/18F-NaF radiotracers in the identification and surveillance of cardiovascular diseases. We highlight the need for further investigation into alternative PET radiotracers to demonstrate their clinical value in the management of these pathologies.
Collapse
|
415
|
Liu N, Su D, Liu K, Liu B, Wang S, Zhang X. The effects of IL-17/IL-17R inhibitors on atherosclerosis in psoriasis and psoriatic arthritis: A protocol for systematic review and meta analysis. Medicine (Baltimore) 2021; 100:e24549. [PMID: 33578549 PMCID: PMC7886472 DOI: 10.1097/md.0000000000024549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Psoriasis (PSO) is a systemic inflammatory disorder that presents with erythematous scaling of the skin and is associated with autoimmune dysfunction. Atherosclerosis is one of the major comorbidities of PSO. PSO-associated inflammatory factor IL-17 could lead to vascular endothelial cell injury and atherosclerosis. While some research results show that IL-17 helps stabilize plaque formation. Efficacy and safety on PSO and psoriatic arthritis (PSA) of existing IL-17/IL-17R biologics (secukinumab, ixekizumab, brodalumab, and bimekizumab) have been clinically validated, but whether they can improve atherosclerotic outcomes in psoriatic patients remains controversial. METHODS Seven electronic search engines will be searched from inception to December 1, 2020, including PubMed, Embase, Scopus, PsycINFO, Global Health, Web of Science and the Cochrane Library. Clinical trial registries, potential grey literature, relevant conference abstracts, and reference lists of identified studies will also be searched. Literature selection, data extraction, and quality assessment will be done by 2 independent authors. Based on the heterogeneity test, the fixed effect or random effect model will be used for data synthesis. Changes in lung function will be evaluated as the primary outcome. Assessment of symptoms, quality of life, medication use, exacerbations and adverse events will be assessed as secondary outcomes. RevMan V. 5.3.5 (The Nordic Cochrane Centre, Copenhagen, Denmark) will be used for meta-analysis. RESULTS This study will provide a synthesis of current evidence of IL-17/IL-17R inhibitors on atherosclerosis in PSO and PSA. CONCLUSION The conclusion of our study will provide updated evidence to judge whether IL-17/IL-17R inhibitors is an effective solution to atherosclerosis as comorbidity of PSO and PSA. PROSPERP REGISTRATION NUMBER CRD42020209897.
Collapse
Affiliation(s)
- Ningyuan Liu
- Beijing University of Chinese Medicine
- Department of Dermatology and Venereology, China-Japan Friendship Hospital
| | - Danni Su
- Department of Dermatology and Venereology, China-Japan Friendship Hospital
- Peking University Health Science Center, Beijing
| | - Keshuai Liu
- Beijing University of Chinese Medicine
- Department of Dermatology and Venereology, China-Japan Friendship Hospital
| | - Binbin Liu
- Beijing University of Chinese Medicine
- Department of Dermatology and Venereology, China-Japan Friendship Hospital
| | - Shibo Wang
- Hangzhou Third Hospital, Hangzhou, China
| | - Xiaoyan Zhang
- Department of Dermatology and Venereology, China-Japan Friendship Hospital
- Peking University Health Science Center, Beijing
| |
Collapse
|
416
|
Deng X, Liang X, Yang H, Huang Z, Huang X, Liang C, Kuang Y, Qin Y, Lin F, Luo Z. Nicotinamide mononucleotide (NMN) protects bEnd.3 cells against H 2 O 2 -induced damage via NAMPT and the NF-κB p65 signalling pathway. FEBS Open Bio 2021; 11:866-879. [PMID: 33340447 PMCID: PMC7931234 DOI: 10.1002/2211-5463.13067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 01/14/2023] Open
Abstract
An increasing number of studies have shown that nicotinamide mononucleotide (NMN) can inhibit not only ageing but also oxidative stress and inflammatory reactions by improving energy metabolism. However, the role of NMN in regulating the anti‐apoptotic, antioxidative stress and inflammatory responses of brain microvascular endothelial cells is still unknown. Therefore, here we studied the effects of NMN on H2O2‐induced oxidative damage of bEnd.3 cells. In this study, we found that NMN could inhibit the NF‐κBp65 inflammatory signalling pathway and increase the expression of the enzymes NAMPT, VEGF and eNOS, alleviating H2O2‐induced apoptosis in bEnd.3 cells. Taken together, these results suggest that NMN reduces H2O2‐induced oxidative stress and apoptosis and improves cell functions by inhibiting the NF‐κBp65 inflammatory pathway and increasing NAMPT expression.
Collapse
Affiliation(s)
- Xiujun Deng
- Department of Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinghuan Liang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Haiyan Yang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenxing Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xuemei Huang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chunfeng Liang
- Department of Blood transfusion, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaqi Kuang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingfen Qin
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Faquan Lin
- Department of Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zuojie Luo
- Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
417
|
Zhao Y, Chen W, Liu Y, Li H, Chi J, Chang Q, Shen L, Yan R, Li J, Yin X, Fu Y. Promoting plaque stability by gene silencing of monocyte chemotactic protein-3 or overexpression of tissue factor pathway inhibitor in ApoE-/- mice. J Drug Target 2021; 29:669-675. [PMID: 33472448 DOI: 10.1080/1061186x.2021.1878363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chemokines may promote the formation and instability of atherosclerotic plaque, which is the most common cause of acute coronary syndrome. The aim of this study was to clarify the function of monocyte chemotactic protein-3 (MCP-3) in the stability of atherosclerotic plaque, to determine the role of tissue factor pathway inhibitor (TFPI) on the development and stability of atherosclerotic plaques, and to further elucidate the anti-atherosclerotic mechanism of TFPI with the emphasis on chemokine MCP-3. We constructed an adenovirus-mediated shRNA against mouse MCP-3 (Ad-MCP-3-shRNA) and an adenovirus-containing TFPI (Ad-TFPI), and tranferred them in a model of vulnerable plaque in ApoE-/- mice respectively. Here, we reported that MCP-3-shRNA and TFPI could both reduce the plaque area and decrease the content of lipids and macrophages, on the contrary, the fibrous cap thickness and content of collagen and smooth muscle cells were increased. In addition, the expression of MCP-3 and CC chemokine receptor 2 (CCR2) was decreased by TFPI transfer. These data provide the first in vivo evidence that MCP-3 is a major contributor to the unstability of atherosclerotic plaque and TFPI may exert its anti-atherosclerotic effects and promote stabilisation of plaque at least partly through inhibiting MCP-3/CCR2 pathway, which may be a new therapeutic method for atherosclerosis.
Collapse
Affiliation(s)
- Yong Zhao
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenjia Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Li
- Department of Cardiology, The Fifth Hospital in Harbin, Harbin, China
| | - Jinyu Chi
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qing Chang
- Department of Cardiology, Heilongjiang Provincial Hospital, Harbin, China
| | - Li Shen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Runan Yan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiashu Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhua Yin
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Fu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
418
|
Xu Y, Tian H, Qiao G, Zheng W. Telocytes in the atherosclerotic carotid artery: Immunofluorescence and TEM evidence. Acta Histochem 2021; 123:151681. [PMID: 33493960 DOI: 10.1016/j.acthis.2021.151681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/15/2022]
Abstract
Telocytes, which possess distinct body shapes and long telopodes, are allocated in the vascular wall. As a fundamental cell type, telocytes construct a three-dimensional network to form a support structure for the artery. This study aims to characterize the morphology and ultrastructure of telocytes in atherosclerotic arteries. ApoE gene-deficient mice were selected as the atherosclerosis animal model and fed a high-fat diet for at least 12 weeks, and immunofluorescence assays and transmission electron microscopy techniques were used to observe changes in telocytes in atherosclerotic arteries. By immunofluorescence staining, CD34, CD117 and PDGFR-α were positive compared with negative CD28/vimentin in telocytes in the atherosclerotic carotid artery, and they were distributed in the tunica intima and tunica adventitia. Under transmission electron microscopy, the bodies of telocytes became larger, while telopodes became shorter compared with their normal condition, and a mass of lipidosomes was present during the progression of atherosclerosis. These results demonstrate that immunofluorescence with TEM is the critical method for identifying TCs and that steatosis of TCs is a reason for atherosclerotic artery dysfunction.
Collapse
|
419
|
Yang W, Lee JH, Lee JW, Kim Y, Kim YS, You HS, Kang HT. Increased Omega-3 Fatty Acid Intake Is Inversely Associated with Subclinical Inflammation in Healthy Elderly Men, Based on the 2015-2018 Korean National Health and Nutrition Examination Survey. Nutrients 2021; 13:nu13020338. [PMID: 33498799 PMCID: PMC7911968 DOI: 10.3390/nu13020338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Subclinical inflammation as a risk factor of cardiovascular diseases was clinically measured using C-reactive protein (CRP) level. (2) Methods: This study was cross-sectionally designed based the 2015–2018 Korean National Health and Nutrition Examination Survey (KNHANES). The ratio of daily omega-3 fatty acids to energy intake (ω3FA ratio) was classified into four quartile groups (Q1, <0.3%; Q2, 0.3%–<0.6%; Q3, 0.6%–<1.0%; and Q4, ≥1.0% in both sexes). Logistic regression analysis was conducted to investigate the association between the ω3FA ratio and subclinical inflammation defined as CRP levels ≥3 mg/dL. (3) Results: The ω3FA ratio in subjects without and with subclinical inflammation was 0.8% and 0.7% in men (p-value = 0.001), and 0.8% and 0.8% in women (p-value = 0.491), respectively. The prevalence of subclinical inflammation in males decreased with increasing quartile of ω3FA ratio (12.9%, 9.6%, 7.4%, and 7.7%, p-value = 0.033), while female prevalence was not significant among quartile groups. Compared to Q1, odds ratios (95% confidence intervals) for subclinical inflammation of Q2, Q3, and Q4 were 0.740 (0.465–1.177), 0.564 (0.341–0.930), and 0.549 (0.317–0.953) in males, and 1.066 (0.653–1.741), 1.105 (0.600–1.718), and 0.934 (0.556–1.571) in females after full adjustment. (4) Conclusion: The ω3FA ratio is associated with subclinical inflammation in men.
Collapse
Affiliation(s)
- Woojung Yang
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
| | - Jong Hun Lee
- Department of Food Science and Biotechnology, Gachon University, Seongnam 13120, Korea;
| | - Jae-woo Lee
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
| | - Yonghwan Kim
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
| | - Ye-Seul Kim
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
| | - Hyo-Sun You
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
| | - Hee-Taik Kang
- Department of Family Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea; (W.Y.); (J.-w.L.); (Y.K.); (Y.-S.K.); (H.-S.Y.)
- Department of Family Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Korea
- Correspondence: ; Tel.: +82-43-269-6301
| |
Collapse
|
420
|
Chang R, Mamun A, Dominic A, Le NT. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front Physiol 2021; 11:605908. [PMID: 33519510 PMCID: PMC7844210 DOI: 10.3389/fphys.2020.605908] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells have emerged as key players in SARS-CoV-2 infection and COVID-19 inflammatory pathologies. Dysfunctional endothelial cells can promote chronic inflammation and disease processes like thrombosis, atherosclerosis, and lung injury. In endothelial cells, mitochondria regulate these inflammatory pathways via redox signaling, which is primarily achieved through mitochondrial reactive oxygen species (mtROS). Excess mtROS causes oxidative stress that can initiate and exacerbate senescence, a state that promotes inflammation and chronic endothelial dysfunction. Oxidative stress can also activate feedback loops that perpetuate mitochondrial dysfunction, mtROS overproduction, and inflammation. In this review, we provide an overview of phenotypes mediated by mtROS in endothelial cells - such as mitochondrial dysfunction, inflammation, and senescence - as well as how these chronic states may be initiated by SARS-CoV-2 infection of endothelial cells. We also propose that SARS-CoV-2 activates mtROS-mediated feedback loops that cause long-term changes in host redox status and endothelial function, promoting cardiovascular disease and lung injury after recovery from COVID-19. Finally, we discuss the implications of these proposed pathways on long-term vascular health and potential treatments to address these chronic conditions.
Collapse
Affiliation(s)
- Ryan Chang
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Abrar Mamun
- Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Abishai Dominic
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
421
|
Tapoi L, Benchea L, Siriopol D, Kanbay M, Covic A. Colchicine Use in Acute Coronary Syndromes: An Update. Angiology 2021; 72:503-510. [PMID: 33412929 DOI: 10.1177/0003319720985644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Coronary artery disease is the leading cause of death worldwide, and its main pathological substrate is represented by atherosclerosis. Inflammation is a major promoter of the atherosclerotic process and is involved in both the initiation and progression of atherosclerosis, as well as in the occurrence of fatal complications. Until the present moment, Colchicine Cardiovascular Outcomes Trial is the largest trial to demonstrate a major benefit of low-dose colchicine on major adverse cardiac events in patients with recent myocardial infarction (MI), but the mechanisms behind this relation are not completely known. The purpose of this review is to emphasize the possible pathways through which colchicine improves the clinical outcomes in the acute setting of acute coronary syndromes by referring to the results of the studies published in the past 5 years. Aside from its stated systemic anti-inflammatory effect, colchicine could be a valuable addition to the therapeutic approach of acute MI by reducing the infarct size, stabilizing the coronary plaque, as well as reducing platelet aggregation. Moreover, colchicine may improve endothelial function, reduce the transcoronary release of cytokines, and prevent a rise in inflammatory markers after percutaneous coronary intervention, thus diminishing the residual inflammatory risk.
Collapse
Affiliation(s)
- Laura Tapoi
- Cardiovascular Diseases Institute "Prof. Dr. George I.M. Georgescu," Iasi, Romania
| | - Laura Benchea
- Cardiovascular Diseases Institute "Prof. Dr. George I.M. Georgescu," Iasi, Romania
| | - Dimitrie Siriopol
- 162290"Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.,Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C.I. Parhon" University Hospital, Iasi, Romania
| | - Mehmet Kanbay
- Department of Medicine, 52979Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Adrian Covic
- 162290"Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.,Nephrology Department, Dialysis and Renal Transplant Center, "Dr. C.I. Parhon" University Hospital, Iasi, Romania
| |
Collapse
|
422
|
Gallic acid: Pharmacological activities and molecular mechanisms involved in inflammation-related diseases. Biomed Pharmacother 2021; 133:110985. [DOI: 10.1016/j.biopha.2020.110985] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022] Open
|
423
|
Endothelial Dysfunction as a Primary Consequence of SARS-CoV-2 Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1321:33-43. [PMID: 33656711 DOI: 10.1007/978-3-030-59261-5_3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A number of different viral species are known to have effects on the endothelium. These include dengue, Ebola, Marburg, Lassa fever, yellow fever and influenza viruses, cytomegalovirus and coronaviruses. There are currently seven human endemic coronaviruses, all of which cause respiratory diseases and bind to receptors found within the endothelium. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes the coronavirus disease 2019 (COVID-19) is highly infectious. Like its predecessor, SARS-CoV, it binds to angiotensin-converting enzyme-2 (ACE-2), which is expressed in many cell types, particularly in the lung, including endothelial cells. The initiation of a cytokine storm by the virus along with infection of endothelial cells leads to apoptosis and structural and functional changes that attenuate vascular integrity in many organs including the lungs, heart, liver and kidney. Endothelial damage also enhances the coagulation pathway leading to thrombus formation in major vessels and capillaries. Infection with SARS-CoV-2 has an adverse outcome for individuals with particular comorbid diseases, e.g. hypertension, obesity, type 2 diabetes and cardiovascular disease. It is possible that this is due to the presence of pre-existing endothelial dysfunction and systemic inflammation in subjects with these diseases. Therapies for COVID-19 that target the endothelium, the inflammatory response and the coagulation pathway are currently under trial.
Collapse
|
424
|
Wang W, Zheng Y, Li M, Lin S, Lin H. Recent Advances in Studies on the Role of Neuroendocrine Disorders in Obstructive Sleep Apnea-Hypopnea Syndrome-Related Atherosclerosis. Nat Sci Sleep 2021; 13:1331-1345. [PMID: 34349578 PMCID: PMC8326525 DOI: 10.2147/nss.s315375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is a common cause of death worldwide, and atherosclerosis (AS) and obstructive sleep apnea-hypopnea syndrome (OSAHS) critically contribute to the initiation and progression of cardiovascular diseases. OSAHS promotes endothelial injury, vascular smooth muscle cell (VSMC) proliferation, abnormal lipid metabolism, and elevated arterial blood pressure. However, the exact OSAHS mechanism that causes AS remains unclear. The nervous system is widely distributed in the central and peripheral regions. It regulates appetite, energy metabolism, inflammation, oxidative stress, insulin resistance, and vasoconstriction by releasing regulatory factors and participates in the occurrence and development of AS. Studies showed that OSAHS can cause changes in neurophysiological plasticity and affect modulator release, suggesting that neuroendocrine dysfunction may be related to the OSAHS mechanism causing AS. In this article, we review the possible mechanisms of neuroendocrine disorders in the pathogenesis of OSAHS-induced AS and provide a new basis for further research on the development of corresponding effective intervention strategies.
Collapse
Affiliation(s)
- Wanda Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Yanli Zheng
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Meimei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| | - Shu Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Huili Lin
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, People's Republic of China
| |
Collapse
|
425
|
Zhai Y, Yang J, Zhang J, Yang J, Li Q, Zheng T. Src-family Protein Tyrosine Kinases: A promising target for treating Cardiovascular Diseases. Int J Med Sci 2021; 18:1216-1224. [PMID: 33526983 PMCID: PMC7847615 DOI: 10.7150/ijms.49241] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
The Src-family protein tyrosine kinases (SFKs), a subfamily of non-receptor tyrosine kinases, are ubiquitously expressed in various cell types. Numerous studies have suggested that SFKs are related to signal transduction in major cardiac physiological and pathological processes, it is the activity of SFKs that is connected with the maintenance of cardiovascular homeostasis. Upon stimulation of various injury factors or stress, the phosphorylation state of SFKs is changed, which has been found to modulate different cardiac pathological conditions, such as hypertension, coronary heart disease, ischemic heart disease, myocardial ischemia-reperfusion injury, arrhythmia and cardiomyopathy via regulating cell growth, differentiation, movement and function, electrophysiologic signals. This review summarizes the basic information about SFKs, updates its role in the different processes underlying the development of multiple cardiovascular diseases (CVDs), and highlights their potential role as disease biomarkers and therapeutic targets, which would help understand the pathophysiology of CVDs and promote the further potential clinical adhibition.
Collapse
Affiliation(s)
- Yuhong Zhai
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China.,Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
| | - Jing Zhang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China.,Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
| | - Qi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China.,Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Tao Zheng
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China.,Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China.,Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| |
Collapse
|
426
|
Geng P, Xu X, Gao Z. Sinomenine Suppress the Vitamin D3 and High Fat Induced Atherosclerosis in Rats via Suppress of Oxidative Stress and Inflammation. J Oleo Sci 2021; 70:1815-1828. [PMID: 34866111 DOI: 10.5650/jos.ess21255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a cardiovascular disease that arise due to dysfunction of lipid deposition and metabolism. AS is causes the mortality and morbidity worldwide. Sinomenine isolated from the Sinomenium acutum is used extensively against the various cardiac diseases in China. However, the anti-atherosclerosis effect of sinomenine still not explore. In this study, we explore the cardioprotective and anti-atherosclerosis effect of sinomenine against Vitamin D3 and High fat induced atherosclerosis in rats. Sprague Dawley (SD) rats were used in this study. The rats were received the vitamin D (60000) and High fat diet to induce the atherosclerosis and divided into groups and received the oral administration of sinomenine (2.5, 5 and 10 mg/kg) and simvastatin (5 mg/kg). Body weight, organ weight and biochemical parameters were estimated. The mRNA expression of MyD88, TLR4, NF-κB and IκB were estimated. Sinomenine treated rats significantly (p<0.001) suppressed the body weight and modulated the organ weight (hepatic, renal and heart). Sinomenine significantly (p<0.001) decreased the level of triacylglycerols (TG), low density lipoprotein cholesterol (LDL-c), total cholesterol (TC), very low-density lipoprotein cholesterol (VLDL-c) and augmented the level of high-density lipoprotein cholesterol (HDL-c). Sinomenine treatment also reduced the level of atherogenic index (TC/HDL-c and LDL-c/HDL-c). Sinomenine treatment decrease the ratio of HMG CoA/Mevalonate and level of collagen and total protein. Sinomenine significantly (p<0.001) altered the level of heart parameters, antioxidant parameters and inflammatory cytokines. Sinomenine significantly (p<0.001) reduced the expression of MyD88, TLR4, NF-κB and IκB. Taken together, sinomenine exhibited the protective effect against the atherosclerosis via alteration of TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Pengbo Geng
- Department of Cardiology, Xi'an XD Group Hospital
| | - Xiaohui Xu
- Department of Cardiology, Xi'an XD Group Hospital
| | - Zhao Gao
- Department of Cardiology of Heart Disease Hospital, XI'AN International Medical Center Hospital
| |
Collapse
|
427
|
Harp KO, Botchway F, Dei-Adomakoh Y, Wilson MD, Hood JL, Adjei AA, Stiles JK, Driss A. Hemoglobin Genotypes Modulate Inflammatory Response to Plasmodium Infection. Front Immunol 2020; 11:593546. [PMID: 33424841 PMCID: PMC7786007 DOI: 10.3389/fimmu.2020.593546] [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: 09/02/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
In 2018, 228 million cases and 405,000 malaria-associated deaths were reported worldwide with a majority being in Africa. A wide range of factors, including parasitemia, host immunity, inflammatory responses to infection, and host hemoglobin genotype, mediate the severity of malaria. Among the hemoglobinopathies, hemoglobin S (HbS) is caused by a single amino acid substitution of Glutamic Acid replaced by Valine at the sixth position of the beta-globin chain (E6V). Hemoglobin C (HbC) on the other hand, involves a single amino acid substitution of Glutamic Acid by a Lysine (E6K), which has received the most attention. These substitutions alter the stability of Hb leading to wide-ranging hematological disorders. The homozygous state of hemoglobin S (HbSS) results in sickle cell anemia (SCA) whereas the heterozygous state (HbAS) results in sickle cell trait (SCT). Both mutations are reported to mediate the reduction in the severity and fatality of Plasmodium falciparum malaria. The mechanism underlying this protection is poorly understood. Since both malaria and sickle cell disease (SCD) are associated with the destruction of erythrocytes and widespread systemic inflammation, identifying which inflammatory factor(s) mediate susceptibility of individuals with different hemoglobin genotypes to Plasmodium infection could result in the discovery of new predictive markers and interventions against malaria or SCD severity. We hypothesized that hemoglobin genotypes modulate the inflammatory response to Plasmodium infection. We conducted a cross-sectional study in Ghana, West Africa, between 2014 and 2019 to ascertain the relationships between blood inflammatory cytokines, Plasmodium infection, and hemoglobin genotype. A total of 923 volunteers were enrolled in the study. A total of 74, age and sex-matched subjects were identified with various genotypes including HbAS, HbAC, HbSS, HbSC, HbCC, or HbAA. Complete blood counts and serum inflammatory cytokine expression levels were assessed. The results indicate that differential expression of CXCL10, TNF-α, CCL2, IL-8, and IL-6 were tightly linked to hemoglobin genotype and severity of Plasmodium infection and that these cytokine levels may be predictive for susceptibility to severe malaria or SCD severity.
Collapse
Affiliation(s)
- Keri Oxendine Harp
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Felix Botchway
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Accra, Ghana
| | | | - Michael D Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Joshua L Hood
- Department of Pharmacology and Toxicology & the James Graham Brown Cancer Center & the Hepatobiology and Toxicology COBRE, University of Louisville, School of Medicine, Louisville, KY, United States
| | - Andrew A Adjei
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Accra, Ghana
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Adel Driss
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
428
|
Wang Y, Ruan Y, Wu S. ET-1 regulates the human umbilical vein endothelial cell cycle by adjusting the ERβ/FOXN1 signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1499. [PMID: 33313244 PMCID: PMC7729364 DOI: 10.21037/atm-20-6560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Atherosclerosis (AS) is a chronic and progressive disease primarily induced by inflammation of the arterial blood vessel wall. Investigating the function and molecular regulation mechanisms of ET-1, ERβ, and FOXN1 in disease models will provide new targets and means for clinical treatment. Methods The effects of ET-1 on oxidative stress in HUVEC were verified through quantitative polymerase chain reaction (qPCR), western blot, flow cytometry, as well as dual luciferase reporter gene and biochemical assays. Results Compared with the ET-1+ negative control (NC) group, the ERβ messenger ribonucleic acid (mRNA) expression level was significantly reduced, and the FOXN1 mRNA expression level increased markedly in the ET-1 + ERβ small interfering ribonucleic acid (siRNA) group. Meanwhile, the FOXN1 mRNA expression level was significantly reduced in the ET-1 + FOXN1 siRNA group. FOXN1 promoter luciferase reporter gene activity was notably enhanced in the ERβ siRNA group compared with the siRNA control group. Compared with the ET-1 + NC group, the levels of reaction oxygen species (ROS) in the ET-1 + ERβ siRNA group increased considerably, the superoxide dismutase (SOD) level was significantly reduced, and the G0/G1 phase cell ratio was reduced. In addition, the protein expression of ERβ and cyclin B1 (CCNB1) was markedly reduced, whereas the protein expression of cyclin A2 (CCNA2), cyclin D1 (CCND1), and cyclin E1 (CCNE1) increased substantially. The opposite result was observed in the ET-1 + FOXN1 siRNA group. Conclusions ET-1 can contribute to the expression of ERβ and FOXN1. ERβ can inhibit the expression of FOXN1 by regulating promoter activity. The ET-1/ERβ/FOXN1 signaling pathway is involved in the regulation of oxidative stress and cycle progression in HUVEC. This study provides a new mechanism for the regulation of umbilical vein endothelial cells. The ET-1/ERβ/FOXN1 signaling pathway may provide novel therapeutic targets and strategies for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuyan Wang
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunjun Ruan
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Saizhu Wu
- Department of Gerontology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
429
|
Zhang R, Ji Z, Yao Y, Zuo W, Yang M, Qu Y, Su Y, Ma G, Li Y. Identification of hub genes in unstable atherosclerotic plaque by conjoint analysis of bioinformatics. Life Sci 2020; 262:118517. [PMID: 33011223 DOI: 10.1016/j.lfs.2020.118517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 02/09/2023]
Abstract
AIMS Unstable atherosclerotic plaque is the main pathological basis of acute coronary syndrome, which is the leading cause of death and disability worldwide. Therefore, we combined multiple bioinformatics tools to identify key genes related to unstable plaque. MAIN METHODS GSE94605 contained 7 plasma sample pools of 175 healthy and 6 sample pools of 150 unstable angina pectoris (UAP) patients, and detected with miRNA array while GSE60993 collected peripheral blood from 7 normal and 9 UAP, and detected with mRNA array. GSE120521 collected carotid plaques from 4 patients and dissected in stable and unstable regions, then detected with RNA-seq. Differentially expressed miRNAs (DEMs) and genes (DEGs) in UAP were re-analyzed. Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG) and Protein-protein interaction (PPI) network were applied on top 10 up-regulated or down-regulated DEMs targets, and whole DEGs. MiRNAs-mRNAs network was constructed with these DEMs and DEGs, and the expression profile of genes within the network was finally validated in GSE120521. KEY FINDINGS Totally, 263 up-regulated and 201 down-regulated DEMs were identified in GSE94605, and 78 up-regulated and 29 down-regulated DEGs were identified in GSE60993. Subsequently, a miRNAs-mRNAs network was constructed with 6 up-regulated miRNAs targeted to 12 down-regulated genes, and 4 down-regulated miRNAs targeted to 8 up-regulated genes. Finally, MORF4L2, RAB3IL1 and MMP9 within the network were considered as hub genes in unstable plaque progression after being validated in GSE120521. SIGNIFICANCE These 3 genes may provide new targets for diagnosis and therapy of unstable atherosclerotic plaque.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Wenjie Zuo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Mingming Yang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Yangyang Qu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Yamin Su
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China.
| | - Yongjun Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, PR China.
| |
Collapse
|
430
|
Zhang L, Yuan JQ, Song FC, Zhu MD, Li Q, Liu SH, Zhao K, Zhao C. Ameliorative effects of the traditional Chinese medicine formula Qing-Mai-Yin on arteriosclerosis obliterans in a rabbit model. PHARMACEUTICAL BIOLOGY 2020; 58:785-795. [PMID: 33073642 PMCID: PMC7592894 DOI: 10.1080/13880209.2020.1803368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
CONTEXT Qing-Mai-Yin (QMY) is a clinically used herbal formula for treating arteriosclerosis obliterans (ASO). OBJECTIVE To evaluate the chemical constituents and effects of QMY on ASO rabbit model. MATERIALS AND METHODS Forty-eight New Zealand rabbits were divided into six groups (n = 8): normal (normal rabbits treated with 0.5% CMC-Na), vehicle (ASO rabbits treated with 0.5% CMC-Na), positive (simvastatin, 1.53 mg/kg), and QMY treatment (300, 600, and 1200 mg/kg). ASO rabbit model was prepared by high fatty feeding, roundly shortening artery, and bovine serum albumin immune injury. QMY (300, 600 and 1200 mg/kg) was orally administered for 8 weeks. The effects and possible mechanisms of QMY on ASO rabbits were evaluated by pathological examination, biochemical assays, and immunohistochemical assays. The compositions of QMY were analysed using HPLC-Q-TOF-MS/MS analysis. RESULTS Compared to the vehicle rabbit, QMY treatment suppressed plaque formation and intima thickness in aorta, and decreased intima thickness, whereas increased lumen area of femoral artery. Additionally, QMY treatment decreased TC, TG and LDL, decreased CRP and ET, and increased NO and 6-K-PGF1α in serum. Furthermore, the potential mechanisms studied revealed that QMY treatment could suppress expression of TNF-α, IL-6, ICAM-1 and NF-κB in endothelial tissues, and increase IκB. In addition, HPLC analysis showed QMY had abundant anthraquinones, stilbenes, and flavonoids. CONCLUSION QMY has ameliorative effects on ASO rabbit, and the potential mechanisms are correlated to reducing inflammation and down-regulating NF-κB. Our study provides a scientific basis for the future application and investigation of QMY.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Jia-Qin Yuan
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Fu-Chen Song
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Mei-Dong Zhu
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Qi Li
- Yueyang Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Sheng-Hua Liu
- Yueyang Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Kai Zhao
- Department of Traditional of Chinese Medicine, General Hospital of Ningxia Medical University, Yinchuan, PR China
- CONTACT Kai Zhao Department of Traditional of Chinese Medicine, General Hospital of Ningxia Medical University, 804 Shengli Road, Yinchuan, Ningxia750004, PR China
| | - Cheng Zhao
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
- Cheng Zhao Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai200437, PR China
| |
Collapse
|
431
|
Guo W, Li XN, Li J, Lu J, Wu J, Zhu WF, Qin P, Xu NZ, Zhang Q. Increased plasma miR-146a levels are associated with subclinical atherosclerosis in newly diagnosed type 2 diabetes mellitus. J Diabetes Complications 2020; 34:107725. [PMID: 32981813 DOI: 10.1016/j.jdiacomp.2020.107725] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/02/2020] [Accepted: 08/23/2020] [Indexed: 11/29/2022]
Abstract
AIMS The association between circulating miR-146a and subclinical atherosclerosis in type 2 diabetes mellitus (T2DM) remains poorly understood. This study aimed to investigate the correlation between plasma miR-146a levels and subclinical atherosclerosis as measured by the carotid intima-media thickness (CIMT) and brachial-ankle pulse wave velocity (baPWV) in patients with newly diagnosed T2DM. METHODS We studied 100 patients with newly diagnosed T2DM. Subclinical atherosclerosis was defined as a thickened CIMT (≥1.0 mm) and high baPWV defined as a value greater than the 75th percentile. Plasma miR-146a levels and metabolic parameters were measured. RESULTS Patients with thickened CIMT had higher plasma miR-146a levels than those without thickened CIMT (3.36 ± 1.32 vs 1.38 ± 1.11, P < 0.001). Patients in the high baPWV group had higher plasma miR-146a levels than those in the normal baPWV group (3.43 ± 1.32 vs 1.98 ± 1.48, P < 0.001). Both CIMT (β = 0.569, P < 0.001) and baPWV (β = 0.274, P = 0.001) positively correlated with plasma miR-146a levels after adjustment for confounding factors by multiple stepwise regression. On binary logistic regression, plasma miR-146a level was an independent risk factor for thickened CIMT (OR = 3.890, 95% CI 1.415-7.698, P = 0.008) and high baPWV (OR = 1.954, 95% CI 1.256-3.040, P = 0.002) after adjustment for established cardiovascular risk factors. The area under the receiver operating characteristics curve (AUROC) of plasma miR-146a level for predicting thickened CIMT was 0.795 (95%CI 0.708-0.883, P < 0.001) and for predicting high baPWV was 0.773 (95%CI 0.679-0.867, P < 0.001). CONCLUSION Plasma miR-146a levels correlate with CIMT and baPWV and could act as a biomarker for early diagnosis and as a therapeutic target for atherosclerosis in T2DM.
Collapse
Affiliation(s)
- Wen Guo
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Xiao-Na Li
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jie Li
- Department of Endocrinology, Nanjing Central Hospital, Nanjing 210018, China
| | - Jing Lu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Juan Wu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wen-Fang Zhu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Pei Qin
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| | - Nian-Zhen Xu
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Qun Zhang
- Department of Health Promotion Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
432
|
Zhang L, Yuan JQ, Song FC, Zhu MD, Li Q, Liu SH, Zhao K, Zhao C. Ameliorative effects of the traditional Chinese medicine formula Qing-Mai-Yin on arteriosclerosis obliterans in a rabbit model. PHARMACEUTICAL BIOLOGY 2020; 58:785-795. [PMID: 33073642 DOI: 10.1080/13880209.2020.1803368if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
CONTEXT Qing-Mai-Yin (QMY) is a clinically used herbal formula for treating arteriosclerosis obliterans (ASO). OBJECTIVE To evaluate the chemical constituents and effects of QMY on ASO rabbit model. MATERIALS AND METHODS Forty-eight New Zealand rabbits were divided into six groups (n = 8): normal (normal rabbits treated with 0.5% CMC-Na), vehicle (ASO rabbits treated with 0.5% CMC-Na), positive (simvastatin, 1.53 mg/kg), and QMY treatment (300, 600, and 1200 mg/kg). ASO rabbit model was prepared by high fatty feeding, roundly shortening artery, and bovine serum albumin immune injury. QMY (300, 600 and 1200 mg/kg) was orally administered for 8 weeks. The effects and possible mechanisms of QMY on ASO rabbits were evaluated by pathological examination, biochemical assays, and immunohistochemical assays. The compositions of QMY were analysed using HPLC-Q-TOF-MS/MS analysis. RESULTS Compared to the vehicle rabbit, QMY treatment suppressed plaque formation and intima thickness in aorta, and decreased intima thickness, whereas increased lumen area of femoral artery. Additionally, QMY treatment decreased TC, TG and LDL, decreased CRP and ET, and increased NO and 6-K-PGF1α in serum. Furthermore, the potential mechanisms studied revealed that QMY treatment could suppress expression of TNF-α, IL-6, ICAM-1 and NF-κB in endothelial tissues, and increase IκB. In addition, HPLC analysis showed QMY had abundant anthraquinones, stilbenes, and flavonoids. CONCLUSION QMY has ameliorative effects on ASO rabbit, and the potential mechanisms are correlated to reducing inflammation and down-regulating NF-κB. Our study provides a scientific basis for the future application and investigation of QMY.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Jia-Qin Yuan
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Fu-Chen Song
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Mei-Dong Zhu
- Department of Vascular Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Qi Li
- Yueyang Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Sheng-Hua Liu
- Yueyang Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Kai Zhao
- Department of Traditional of Chinese Medicine, General Hospital of Ningxia Medical University, Yinchuan, PR China
| | - Cheng Zhao
- Department of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
433
|
Biochanin A Mitigates Atherosclerosis by Inhibiting Lipid Accumulation and Inflammatory Response. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8965047. [PMID: 33959213 PMCID: PMC8074550 DOI: 10.1155/2020/8965047] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/13/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022]
Abstract
Biochanin A (BCA), a dietary isoflavone extracted from red clover and cabbage, has been shown to antagonize hypertension and myocardial ischemia/reperfusion injury. However, very little is known about its role in atherogenesis. The aim of this study was to observe the effects of BCA on atherosclerosis and explore the underlying mechanisms. Our results showed that administration of BCA promoted reverse cholesterol transport (RCT), improved plasma lipid profile, and decreased serum proinflammatory cytokine levels and atherosclerotic lesion area in apoE-/- mice fed a Western diet. In THP-1 macrophage-derived foam cells, treatment with BCA upregulated ATP-binding cassette (ABC) transporter A1 (ABCA1) and ABCG1 expression and facilitated subsequent cholesterol efflux and diminished intracellular cholesterol contents by activating the peroxisome proliferator-activated receptor γ (PPARγ)/liver X receptor α (LXRα) and PPARγ/heme oxygenase 1 (HO-1) pathways. BCA also activated these two signaling pathways to inhibit the secretion of proinflammatory cytokines. Taken together, these findings suggest that BCA is protective against atherosclerosis by inhibiting lipid accumulation and inflammatory response through the PPARγ/LXRα and PPARγ/HO-1 pathways. BCA may be an attractive drug for the prevention and treatment of atherosclerotic cardiovascular disease.
Collapse
|
434
|
Wu M, Li X, Guo Q, Li J, Xu G, Li G, Wang J, Zhang X. Magnetic mesoporous silica nanoparticles-aided dual MR/NIRF imaging to identify macrophage enrichment in atherosclerotic plaques. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 32:102330. [PMID: 33171287 DOI: 10.1016/j.nano.2020.102330] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/08/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023]
Abstract
Active foamy macrophage enrichment drives atherosclerotic plaque initiation and evolution, and is the prominent target for precisely identifying vulnerable plaque. Precise imaging of high-risk plaque allows promotion of treatment and prevention of vascular pathema. However, current iron oxide (IO) nanoparticles-based magnetic resonance (MR) imaging of plaque is often limited by insufficient perfusion and nonspecific accumulation of peri-aortic lymph nodes. Besides that, intrinsic defects of MR also impede its use for accurately identifying plaque details. Herein, by conjugating with PP1 peptide, a novel magnetic mesoporous silica nanoparticle (PIMI) loaded with near-infrared fluorescence (NIRF) dye (IR820) was fabricated to specifically target and quantify macrophage enrichment of atherosclerotic plaque in ApoE-/- mice using dual MR/NIRF imaging. Biocompatibility experiments ulteriorly confirmed the high safety of PIMI nanoparticles in vivo, which lays the foundation of next-generation contrast agent for recognizing macrophage-rich plaque in the near future.
Collapse
Affiliation(s)
- Menglin Wu
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Xue Li
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China.
| | - Qi Guo
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Jiang Li
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Guoping Xu
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Guilai Li
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Jiahui Wang
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China
| | - Xuening Zhang
- Department of Radiology, Second Hospital of Tianjin Medical University, Tianjin, PR China.
| |
Collapse
|
435
|
Shu H, Peng Y, Hang W, Nie J, Zhou N, Wang DW. The role of CD36 in cardiovascular disease. Cardiovasc Res 2020; 118:115-129. [PMID: 33210138 PMCID: PMC8752351 DOI: 10.1093/cvr/cvaa319] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
CD36, also known as the scavenger receptor B2, is a multifunctional receptor widely expressed in various organs. CD36 plays a crucial role in the uptake of long-chain fatty acids, the main metabolic substrate in myocardial tissue. The maturation and transportation of CD36 is regulated by post-translational modifications, including phosphorylation, ubiquitination, glycosylation, and palmitoylation. CD36 is decreased in pathological cardiac hypertrophy caused by ischaemia-reperfusion and pressure overload, and increased in diabetic cardiomyopathy and atherosclerosis. Deficiency of CD36 alleviates diabetic cardiomyopathy and atherosclerosis, while overexpression of CD36 eliminates ischaemia-reperfusion damage, together suggesting that CD36 is closely associated with the progression of cardiovascular diseases and may be a new therapeutic target. This review summarizes the regulation and post-translational modifications of CD36 and evaluates its role in cardiovascular diseases and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jiali Nie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.,Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
436
|
Xin L, Gao J, Lin H, Qu Y, Shang C, Wang Y, Lu Y, Cui X. Regulatory Mechanisms of Baicalin in Cardiovascular Diseases: A Review. Front Pharmacol 2020; 11:583200. [PMID: 33224035 PMCID: PMC7667240 DOI: 10.3389/fphar.2020.583200] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular diseases (CVDs) is the leading cause of high morbidity and mortality worldwide, which emphasizes the urgent necessity to develop new pharmacotherapies. In eastern countries, traditional Chinese medicine Scutellaria baicalensis Georgi has been used clinically for thousands of years. Baicalin is one of the main active ingredients extracted from Chinese herbal medicine S. baicalensis. Emerging evidence has established that baicalin improves chronic inflammation, immune imbalance, disturbances in lipid metabolism, apoptosis and oxidative stress. Thereby it offers beneficial roles against the initiation and progression of CVDs such as atherosclerosis, hypertension, myocardial infarction and reperfusion, and heart failure. In this review, we summarize the pharmacological features and relevant mechanisms by which baicalin regulates CVDs in the hope to reveal its application for CVDs prevention and/or therapy.
Collapse
Affiliation(s)
- Laiyun Xin
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialiang Gao
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongchen Lin
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Qu
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chang Shang
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuling Wang
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingdong Lu
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiangning Cui
- Department of Cardiology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
437
|
韩 晓, 吴 文, 刘 小, 祝 烨. [Study on visfatin-induced inflammation and necroptosis via LOX-1 in human umbilical vein endothelial cells]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2020; 37:834-841. [PMID: 33140607 PMCID: PMC10320536 DOI: 10.7507/1001-5515.202003067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 02/05/2023]
Abstract
The aim of the study is to identify the effects and underlying mechanisms of visfatin on inflammation and necroptosis in vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) were stimulated with visfatin or pretreated with Polyinosinic acid (LOX-1 inhibitor). By using the Western blot, RT-PCR, immunocytochemistry, enzyme-linked immunosorbent assay (ELISA), MTT and flow cytometry technique, the occurrence of inflammation and necroptosis in HUVECs were evaluated. Our results showed that 100 ng/mL visfatin significantly increased the mRNA and protein expression of monocyte chemotactic protein 1 (MCP-1) and LOX-1 after 24 hours' treatment in HUVECs. However, pretreatment with Polyinosinic acid could significantly reduce the expression of MCP-1 compared with visfatin group. Additionally, 100 ng/mL visfatin could induce the production of necrotic features and increase the mRNA expression of BMF (one of the markers of necroptosis), while pretreating with Polyinosinic acid markedly downregulated the mRNA expression of BMF gene and promoted the cell proliferation. These results indicate that visfatin might induce inflammation and necroptosis via LOX-1 in HUVECs, suggesting that visfatin plays a central role in the development of atherosclerosis.
Collapse
Affiliation(s)
- 晓宇 韩
- 成都市第二人民医院 老年医学科(成都 610017)Geriatrics Department, Chengdu Second People’s Hospital, Chengdu 610017, P.R.China
- 四川大学华西医院 心血管疾病研究室(成都 610041)Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - 文超 吴
- 成都市第二人民医院 老年医学科(成都 610017)Geriatrics Department, Chengdu Second People’s Hospital, Chengdu 610017, P.R.China
| | - 小菁 刘
- 成都市第二人民医院 老年医学科(成都 610017)Geriatrics Department, Chengdu Second People’s Hospital, Chengdu 610017, P.R.China
- 四川大学华西医院 心血管疾病研究室(成都 610041)Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, P.R.China
| | - 烨 祝
- 成都市第二人民医院 老年医学科(成都 610017)Geriatrics Department, Chengdu Second People’s Hospital, Chengdu 610017, P.R.China
| |
Collapse
|
438
|
Meng Q, Pu L, Luo X, Wang B, Li F, Liu B. Regulatory Roles of Related Long Non-coding RNAs in the Process of Atherosclerosis. Front Physiol 2020; 11:564604. [PMID: 33192561 PMCID: PMC7604474 DOI: 10.3389/fphys.2020.564604] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis (AS) is the main cause of coronary heart disease, cerebral infarction, and peripheral vascular disease, which comprise serious hazards to human health. Atherosclerosis is characterized by the deposition of lipids on the interior walls of blood vessels, causing an inflammatory response of immune cells, endothelial cells, and smooth muscle cells, and a proliferation cascade reaction. Despite years of research, the underlying pathogenesis of AS is not fully defined. Recent advances in our understanding of the molecular mechanisms by which non-coding RNA influences the initiation and progression of AS have shown that long non-coding RNAs (lncRNAs) regulate important stages in the atherosclerotic process. In this review, we summarize current knowledge of lncRNAs, which influence the development of AS. We review the regulatory processes of lncRNAs on core stages of atherosclerotic progression, including lipid metabolism, inflammation, vascular cell proliferation, apoptosis, adhesion and migration, and angiogenesis. A growing body of evidence suggests that lncRNAs have great potential as new therapeutic targets for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Luya Pu
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Xizi Luo
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Baisen Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China.,The Key Laboratory for Bionics Engineering, Ministry of Education, Jilin University, Changchun, China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China.,Key Laboratory for Health Biomedical Materials of Jilin Province, Jilin University, Changchun, China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
439
|
Wang F, Liang S, Hu J, Xu Y. Aryl hydrocarbon receptor connects dysregulated immune cells to atherosclerosis. Immunol Lett 2020; 228:55-63. [PMID: 33053378 DOI: 10.1016/j.imlet.2020.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 11/20/2022]
Abstract
As a chronic inflammatory disease with autoimmune components, atherosclerosis is the major cause of cardiovascular morbidity and mortality. Recent studies have revealed that the development of atherosclerosis is strongly linked to the functional activities of aryl hydrocarbon receptor (AHR), a chemical sensor that is also important for the development, maintenance, and function of a variety of immune cells. In this review, we focus on the impact of AHR signaling on the different cell types that are closely related to the atherogenesis, including T cells, B cells, dendritic cells, macrophages, foam cells, and hematopoietic stem cells in the arterial walls, and summarize the latest development on the interplay between this environmental sensor and immune cells in the context of atherosclerosis. Hopefully, elucidation of the role of AHR in atherosclerosis will facilitate the understanding of case variation in disease prevalence and may aid in the development of novel therapies.
Collapse
Affiliation(s)
- Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China
| | - Shuangchao Liang
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Jiqiong Hu
- Department of Vascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, School of Life Science, Anhui Normal University, Wuhu, 241000, China.
| |
Collapse
|
440
|
Kostyunin A, Mukhamadiyarov R, Glushkova T, Bogdanov L, Shishkova D, Osyaev N, Ovcharenko E, Kutikhin A. Ultrastructural Pathology of Atherosclerosis, Calcific Aortic Valve Disease, and Bioprosthetic Heart Valve Degeneration: Commonalities and Differences. Int J Mol Sci 2020; 21:E7434. [PMID: 33050133 PMCID: PMC7587971 DOI: 10.3390/ijms21207434] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 01/24/2023] Open
Abstract
Atherosclerosis, calcific aortic valve disease (CAVD), and bioprosthetic heart valve degeneration (alternatively termed structural valve deterioration, SVD) represent three diseases affecting distinct components of the circulatory system and their substitutes, yet sharing multiple risk factors and commonly leading to the extraskeletal calcification. Whereas the histopathology of the mentioned disorders is well-described, their ultrastructural pathology is largely obscure due to the lack of appropriate investigation techniques. Employing an original method for sample preparation and the electron microscopy visualisation of calcified cardiovascular tissues, here we revisited the ultrastructural features of lipid retention, macrophage infiltration, intraplaque/intraleaflet haemorrhage, and calcification which are common or unique for the indicated types of cardiovascular disease. Atherosclerotic plaques were notable for the massive accumulation of lipids in the extracellular matrix (ECM), abundant macrophage content, and pronounced neovascularisation associated with blood leakage and calcium deposition. In contrast, CAVD and SVD generally did not require vasculo- or angiogenesis to occur, instead relying on fatigue-induced ECM degradation and the concurrent migration of immune cells. Unlike native tissues, bioprosthetic heart valves contained numerous specialised macrophages and were not capable of the regeneration that underscores ECM integrity as a pivotal factor for SVD prevention. While atherosclerosis, CAVD, and SVD show similar pathogenesis patterns, these disorders demonstrate considerable ultrastructural differences.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, 650002 Kemerovo, Russia; (A.K.); (R.M.); (T.G.); (L.B.); (D.S.); (N.O.); (E.O.)
| |
Collapse
|
441
|
Yang Y, Lai C, Yan F, Wang J. Clinical Significance of MRI Contrast Enhancement of the Oculomotor Nerve in Ischemic Isolated Oculomotor Nerve Palsy. J Clin Neurol 2020; 16:653-658. [PMID: 33029972 PMCID: PMC7541971 DOI: 10.3988/jcn.2020.16.4.653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose Contrast enhancement of the oculomotor nerve in MRI was recently noticed in patients with clinical ischemic isolated oculomotor nerve palsy (iIONP). The opinions about whether this is a sign of inflammation and whether or not to administer steroids vary between doctors. The study aimed to determine the associations between this enhancement and vascular-disease risk factors (VRFs) and inflammatory factors in iIONP patients. Methods The study recruited patients who had experienced iIONP during the previous 2 years. They were divided into groups A and B based on whether or not they exhibited an enhanced oculomotor nerve in MRI of the cavernous sinus using thin-section, fat-suppressed, and contrast-enhanced sequences. VRFs, inflammatory factors, and improvement scores were compared between the two groups. Results Most (71.1%) of the 45 included iIONP patients had enhanced oculomotor nerves in MRI. VRFs, periorbital pain, elevated C-reactive protein and erythrocyte sedimentation rate, the neutrophil-to-lymphocyte ratio, and the platelet-to-lymphocyte ratio were not significantly associated with the enhancement. Four of the five patients in group A exhibited an elevated cerebrospinal fluid (CSF) IgG synthesis rate. The improvement score of eight patients who received 80 mg of methylprednisolone in addition to the routine therapy was not significantly different from the scores of the other patients (p=0.485). Conclusions More than half of the iIONP patients had an enhanced oculomotor nerve in MRI. A few of them also had elevated CSF IgG synthesis rate, but no further evidence for inflammation was found. The administration of steroids seemed to have no benefit other than increasing the blood glucose level.
Collapse
Affiliation(s)
- Yan Yang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chuntao Lai
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Fei Yan
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiawei Wang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
442
|
Tan L, Song X, Ren Y, Wang M, Guo C, Guo D, Gu Y, Li Y, Cao Z, Deng Y. Anti-inflammatory effects of cordycepin: A review. Phytother Res 2020; 35:1284-1297. [PMID: 33090621 DOI: 10.1002/ptr.6890] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 09/13/2020] [Indexed: 01/08/2023]
Abstract
Cordycepin is the major bioactive component extracted from Cordyceps militaris. In recent years, cordycepin has received increasing attention owing to its multiple pharmacological activities. This study reviews recent researches on the anti-inflammatory effects and the related activities of cordycepin. The results from our review indicate that cordycepin exerts protective effects against inflammatory injury for many diseases including acute lung injury (ALI), asthma, rheumatoid arthritis, Parkinson's disease (PD), hepatitis, atherosclerosis, and atopic dermatitis. Cordycepin regulates the NF-κB, RIP2/Caspase-1, Akt/GSK-3β/p70S6K, TGF-β/Smads, and Nrf2/HO-1 signaling pathways among others. Several studies focusing on cordycepin derivatives were reviewed and found to down metabolic velocity of cordycepin and increase its bioavailability. Moreover, cordycepin enhanced immunity, inhibited the proliferation of viral RNA, and suppressed cytokine storms, thereby suggesting its potential to treat COVID-19 and other viral infections. From the collected and reviewed information, this article provides the theoretical basis for the clinical applications of cordycepin and discusses the path for future studies focusing on expanding the medicinal use of cordycepin. Taken together, cordycepin and its analogs show great potential as the next new class of anti-inflammatory agents.
Collapse
Affiliation(s)
- Lu Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Xiaominting Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yali Ren
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Miao Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Chuanjie Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Dale Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yucheng Gu
- Syngenta Jealott's Hill International Research Centre, Berkshire, UK
| | - Yuzhi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Zhixing Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yun Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| |
Collapse
|
443
|
Butt AK, Cave B, Maturana M, Towers WF, Khouzam RN. The Role of Colchicine in Coronary Artery Disease. Curr Probl Cardiol 2020; 46:100690. [PMID: 32994053 DOI: 10.1016/j.cpcardiol.2020.100690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022]
Abstract
There is increasing experimental and clinical evidence that inflammation appears to play an important role in atherosclerosis and coronary artery disease. Treatment of coronary artery disease currently involves management of cardiovascular risk factors, lipid-lowering strategies and antiplatelet medications. Inflammation seems to be central to the pathogenesis of atherosclerotic plaque development, instability, and rupture seen in coronary artery disease. Colchicine, a well-known and relatively inexpensive drug, has unique anti-inflammatory properties, which is generating considerable interest in its potential role in reducing cardiovascular morbidity and potentially mortality. This review discusses the mechanism of action of colchicine in preventing and treating atherosclerosis as well as the literature from recent clinical studies supporting its use in coronary artery disease.
Collapse
|
444
|
Wang H, Xie Y, Salvador AM, Zhang Z, Chen K, Li G, Xiao J. Exosomes: Multifaceted Messengers in Atherosclerosis. Curr Atheroscler Rep 2020; 22:57. [PMID: 32772195 DOI: 10.1007/s11883-020-00871-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Atherosclerosis (AS) is a chronic inflammatory disease that contributes to the development of coronary artery disease, which has become a leading health burden worldwide. Though several strategies such as pharmacological treatment, exercise intervention, and surgery have been used in clinical practice, there is still no effective strategy to cure AS. Exosomes are extensively studied both as diagnostic markers as well as for therapeutic purposes due to their role in pathological processes related to AS. To elucidate the role of exosomes in AS and thus provide a new insight into AS therapy, we review recent advances concerning exosome targets and their function in mediating intercellular communication in AS, and expect to provide a reference for novel effective strategies to cure AS. RECENT FINDINGS Exosomes exert important roles in the diagnosis, development, and potential therapy of AS. For AS development, (1) activation of CD-137 in endothelial cells represses exosomal-TET2 production, causing a phenotypic switch of vascular smooth muscle cells (VSMC) and promoting plaque formation; (2) exosomal-MALTA1 derived from endothelial cells causes neutrophil extracellular traps (NETs) and M2 macrophage polarization, which aggravates AS; and (3) exosomal-miR-21-3p derived from macrophages inhibits PTEN expression and further promotes VSMC migration/proliferation, leading to AS development. For AS diagnosis, plasma exosomal-miR30e and miR-92a are considered to be potential diagnostic markers. For AS therapy, adipose mesenchymal stem cell-derived exosomes protect endothelial cells from AS aggravation, via inhibiting miR-342-5p. Exosome-mediated cross-talk between different cells within the vasculature exerts crucial roles in regulating endothelial function, proliferation and differentiation of vascular smooth muscle cells, and platelet activation as well as macrophage activation, collectively leading to the development and progression of AS. Exosomes can potentially be used as diagnostic biomarkers and constitute as a new therapeutic strategy for AS.
Collapse
Affiliation(s)
- Hongyun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuling Xie
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Ane M Salvador
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02214, USA
| | - Zhongrong Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China
| | - Kaichuan Chen
- Department of Ophthalmology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02214, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, 333 Nan Chen Road, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
445
|
Mostafa MN, Osama M. The implications of neutrophil extracellular traps in the pathophysiology of atherosclerosis and atherothrombosis. Exp Biol Med (Maywood) 2020; 245:1376-1384. [PMID: 32727216 DOI: 10.1177/1535370220945989] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
IMPACT STATEMENT Fatal consequences of atherosclerosis and atherothrombosis give research in this field great importance. This review provides recent information about the implications of neutrophils in the pathophysiology of atherosclerosis and atherothrombosis via formation and release of neutrophil extracellular traps (NETs), thereby enhancing our understanding on how the process of atherosclerosis develops and how its consequences occur. Information provided in this review suggests NETs as a new therapeutic target and a rich point for research. This review gives answers to questions about the mechanisms of atherosclerosis and atherothrombosis progression through studying the implications of NETs in these processes.
Collapse
Affiliation(s)
| | - Mahmoud Osama
- Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
446
|
Vinciguerra M, Romiti S, Fattouch K, De Bellis A, Greco E. Atherosclerosis as Pathogenetic Substrate for Sars-Cov2 Cytokine Storm. J Clin Med 2020; 9:E2095. [PMID: 32635302 PMCID: PMC7408959 DOI: 10.3390/jcm9072095] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (Sars-CoV-2) outbreak is a public health emergency affecting different regions around the world. The lungs are often damaged due to the presence of Sars-CoV-2 binding receptor ACE2 on epithelial alveolar cells. Severity of infection varies from complete absence of symptomatology to more aggressive symptoms, characterized by sudden acute respiratory distress syndrome (ARDS), multiorgan failure, and sepsis, requiring treatment in intensive care unit (ICU). It is not still clear why the immune system is not able to efficiently suppress viral replication in a small percentage of patients. It has been documented as pathological conditions affecting the cardiovascular system, strongly associated to atherosclerotic progression, such as heart failure (HF), coronary heart disease (CHD), hypertension (HTN) and diabetes mellitus (DM), could serve as predictive factors for severity and susceptibility during Sars-CoV-2 infection. Atherosclerotic progression, as a chronic inflammation process, is characterized by immune system dysregulation leading to pro-inflammatory patterns, including interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), and IL-1β. Reviewing immune system and inflammation profiles in atherosclerosis and laboratory results reported in severe COVID-19 infections, we hypothesized a pathogenetic correlation. Atherosclerosis may be an ideal pathogenetic substrate for high viral replication ability, leading to adverse outcomes, as reported in patients with cardiovascular factors. The level of atherosclerotic progression may affect a different degree of severe infection; in a vicious circle, feeding itself, Sars-CoV-2 may exacerbate atherosclerotic evolution due to excessive and aberrant plasmatic concentration of cytokines.
Collapse
Affiliation(s)
- Mattia Vinciguerra
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (M.V.); (S.R.); (E.G.)
| | - Silvia Romiti
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (M.V.); (S.R.); (E.G.)
| | - Khalil Fattouch
- Department of Cardiovascular Surgery, GVM Care and Research, Maria Eleonora Hospital, 90135 Palermo, Italy
| | - Antonio De Bellis
- Department of Cardiology and Cardiac Surgery, Casa di Cura “S. Michele”, Maddaloni, 81024 Caserta, Italy;
| | - Ernesto Greco
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (M.V.); (S.R.); (E.G.)
| |
Collapse
|
447
|
Wang JL, Cai F, Liu XH, Li LM, He X, Hu XM, Kang CM, Bai HL, Zhang RY, Wu CM, Wu LM, Wang J, Zheng L, Ping BH, Hu YW, Wang Q. Lipopolysaccharide Promotes Inflammatory Response via Enhancing IFIT1 Expression in Human Umbilical Vein Endothelial Cells. DNA Cell Biol 2020; 39:1274-1281. [PMID: 32551893 DOI: 10.1089/dna.2020.5454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Jia-Li Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Linyi People's Hospital of Shandong Province, Linyi, China
| | - Fen Cai
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, China
| | - Xue-Hui Liu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Li-Min Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin He
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiu-Mei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun-Min Kang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huan-Lan Bai
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ru-Yi Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chang-Meng Wu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Li-Mei Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Jia Wang
- Rizhao People's Hospital of Shandong Province, Rizhao, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bao-Hong Ping
- Department of Hui Qiao, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
448
|
Wu B, Jiang M, Liu X, Huang C, Gu Z, Cao Y. Evaluation of toxicity of halloysite nanotubes and multi-walled carbon nanotubes to endothelial cells in vitro and blood vessels in vivo. Nanotoxicology 2020; 14:1017-1038. [PMID: 32574508 DOI: 10.1080/17435390.2020.1780642] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanomaterials (NMs) with tubular structures, such as halloysite nanotubes (HNTs), have potential applications in biomedicine. Although the biocompatibility of HNTs has been investigated before, the toxicity of HNTs to blood vessels is rarely systemically evaluated. Herein, we compared the toxicity of HNTs and multi-walled carbon nanotubes (MWCNTs) to human umbilical vein endothelial cells (HUVECs) in vitro and blood vessels of mice in vivo. HUVECs internalized HNTs and MWCNTs, but the uptake of HNTs was not obviously changed by clathrin inhibitor. Exposure to NMs decreased cellular viability, activated apoptotic proteins and up-regulated adhesion molecules, including soluble vascular cell adhesion molecule 1 (sVCAM-1) and VCAM-1. As the mechanisms, NMs decreased NO levels, eNOS mRNA and eNOS/p-eNOS proteins. Meanwhile, NMs promoted intracellular ROS and autophagy dysfunction, shown as decreased protein levels of LC3, beclin-1 and ATG5. The eNOS regulator Kruppel-like factor 4 (KLF4) was inhibited, but another eNOS regulator KLF4 was surprisingly up-regulated. Under in vivo conditions, ICR mice intravenously injected with NMs (50 μg/mouse, once a day for 5 days) showed an increased percentage of neutrophils, monocytes and basophils. Meanwhile, autophagy dysfunction, eNOS uncoupling, activation of apoptotic proteins and alteration of KLF proteins were also observed in mouse aortas. All of the toxic effects were more pronounced for MWCNTs in comparison with HNTs based on the same mass concentrations. Our results may provide novel insights about the toxicity of NMs with tubular structures to blood vessels. Considering the toxicological data reported here, HNTs are probably safer nanocarriers compared with MWCNTs.
Collapse
Affiliation(s)
- Bihan Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P.R. China
| | - Mengdie Jiang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P.R. China
| | - Xuewu Liu
- Hunan Laboratory Animal Center, Hunan Drug Safety Evaluation Center, Liuyang, P.R. China
| | - Chaobo Huang
- College of Chemical Engineering, Nanjing Forestry University (NFU), Nanjing, P.R. China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, P.R. China
| |
Collapse
|
449
|
Hu B, Xu L, Li Y, Bai X, Xing M, Cao Q, Liang H, Song S, Ji A. A peptide inhibitor of macrophage migration in atherosclerosis purified from the leech Whitmania pigra. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112723. [PMID: 32119950 DOI: 10.1016/j.jep.2020.112723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/25/2019] [Accepted: 02/25/2020] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis has become a worldwide public health problem that seriously threatens human health. Leech is traditional Chinese medicine that can be utilized to treat cardiovascular disease. Based on the anti-atherosclerosis activity of leech hydrolysate, we separated and purified the leech peptide capable of inhibiting macrophage migration and studied the pathways of the anti-migration leech peptide. MATERIALS AND METHODS The leech peptide capable of inhibiting macrophage migration that measured by cell migration assays from the leech Whitmania pigra was separated and purified by Q Sepharose FF strong alkaline anion exchange column chromatography, Superdex 30, Superdex peptide and G10 gel column chromatography. And the purity, molecular weight of the leech peptide was determined by high-performance liquid chromatography and high-resolution mass spectrometry. The pathways of anti-migration to macrophages of the leech peptide were studied by inhibitors, Western blotting and RT-PCR. RESULTS We obtained a purified leech peptide with a sequence of EAGSAKELEGDPVAG from the leech Whitmania pigra. We also showed that the anti-migration to macrophages of the leech peptide was blocked by c-Jun N-terminal kinase (JNK) inhibitor and p38 mitogen-activated protein kinase (p38 MAPK) inhibitor. Moreover, the result of RT-PCR and Western blotting revealed that the leech peptide induced an increase in JNK, p38 phosphorylation and the transcription of mitogen-activated protein kinase kinase kinase 4 (MEKK4) and apoptosis signal-regulating kinase 2 (ASK2). These data indicated that the anti-migration to macrophages of the leech peptide occurred through JNK and p38 MAPK pathways. In addition, the results demonstrated that the leech peptide had no significant effect on the immunological activity of macrophages including phagocytic ability, lysozyme activity, and levels of expression of inflammatory factors. CONCLUSION A sequence peptide was obtained from the hydrolysate of leech Whitmania pigra that inhibits macrophage migration.
Collapse
Affiliation(s)
- Bo Hu
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Lixu Xu
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Ying Li
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Xu Bai
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Maochen Xing
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Qi Cao
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Hao Liang
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China
| | - Shuliang Song
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China.
| | - Aiguo Ji
- Marine College, Shandong University, Weihai, 264209, PR China; Weihai International Biotechnology Research and Development Centre, Shandong University Weihai, 264209, PR China; School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
450
|
An L, Ma J, Yang X, Liang Y, Wang H, Tuerhong M, Lall N, Abudukeremu M, Zhang Y, Lee D, Xu J, Wu X, Guo Y. Caseahomopene A, a ring-expanded homotriterpenoid from Casearia kurzii showing anti-inflammatory activities in vitro and in vivo. Bioorg Chem 2020; 98:103758. [DOI: 10.1016/j.bioorg.2020.103758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/01/2020] [Accepted: 03/13/2020] [Indexed: 12/27/2022]
|