1
|
Zhou X, Chen X, Zhang L, Yuan J, Lin H, Zhu M, Xu X, Dong G, Fu J, Wu W. Mannose-Binding Lectin Reduces Oxidized Low-Density Lipoprotein Induced Vascular Endothelial Cells Injury by Inhibiting LOX1-ox-LDL Binding and Modulating Autophagy. Biomedicines 2023; 11:1743. [PMID: 37371838 DOI: 10.3390/biomedicines11061743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Objective: To investigate the role of mannose-binding lectin (MBL) in modulating autophagy and protecting endothelial cells (ECs) from oxidized low-density lipoprotein (ox-LDL)-induced injury. Methods: Serum MBL concentration and carotid intima-media thickness (cIMT) were measured in 94 obese and 105 healthy children. ECs were transfected with MBL over-expression plasmid, LOX1 was knocked-down to explore the protective role of MBL in ox-LDL induced ECs injury. Dendritic cells (DCs) were co-cultured with ECs, and inflammatory factors, DC maturation, and autophagy was assessed. WT and ApoE-/- mice were fed with a high fat diet (HFD) with or without MBL-adenovirus injection for 16 weeks and aortic vascular endothelial tissue was isolated, then atherosclerotic plaque, cell injury and autophagy were analyzed. Results: Serum MBL concentration in obese children was lower than healthy controls and was negatively correlated with cIMT. The uptake of ox-LDL was decreased in LOX1 knock-down ECs. MBL over-expression in vitro inhibited LOX1-ox-LDL binding. Both LOX1 knock-down and MBL over-expression can ameliorate EC autophagy and cell injury. MBL over-expression in vivo alleviated atherosclerotic plaque formation, influenced DC maturation and down-regulated IL-6, IL-12, and TNF-a levels. Conclusions: MBL exerts a protective role in ox-LDL-induced EC injury by modulating DC maturation and EC autophagy via inhibiting LOX1-ox-LDL binding.
Collapse
Affiliation(s)
- Xuelian Zhou
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Xuefeng Chen
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Li Zhang
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Jinna Yuan
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Hu Lin
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Mingqiang Zhu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Xiaoqin Xu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Guanping Dong
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| | - Wei Wu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou 310052, China
| |
Collapse
|
2
|
Chavda V, Chaurasia B, Garg K, Deora H, Umana GE, Palmisciano P, Scalia G, Lu B. Molecular mechanisms of oxidative stress in stroke and cancer. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2021.100029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
3
|
Li C, He K, Yin M, Zhang Q, Lin J, Niu Y, Wang Q, Xu Q, Jiang N, Zhao G. LOX-1 Regulates Neutrophil Apoptosis and Fungal Load in A. Fumigatus Keratitis. Curr Eye Res 2021; 46:1800-1811. [PMID: 34264144 DOI: 10.1080/02713683.2021.1948063] [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] [Indexed: 10/20/2022]
Abstract
PURPOSE To determine whether LOX-1 regulates neutrophil apoptosis and fungal load in A. fumigatus keratitis. METHODS Fas, FasL, CASP3, CASP8, CASP9 and BCL2 were tested in normal and infected corneas of C57BL/6 mice. Mice corneas were infected with A. fumigatus with or without pretreatment of LOX-1 neutralizing antibody or inhibitor (Poly I). Clinical score was recored and HE staining was tested. Fungal load in mice corneas was observed by plate counting. Poly morphonuclear neutrophilic leukocytes (PMNs) were stimulated with 75% ethanol-killed A. fumigatus with or without pretreatment of LOX-1 neutralizing antibody or Poly I. PCR, western blot and immunostaining tested expression of Fas, FasL, CASP3, CASP8, CASP9, BCL2 and cleaved caspase-3. PMNs infiltration and TUNEL-positive cells were assessed by immunofluorescent staining. Flow cytometry assay tested the percentage of apoptosis neutrophils. RESULTS Fas, Fas ligand, caspase-8, caspase-9 and caspase-3 mRNA levels were significantly higher in C57BL/6 mice corneas infected with A. fumigatus than normal corneas. Poly I treatment alleviated the severity and decreased clinical score at 3, 5 and 7 days post infecrion (p.i.). HE staining showed less infiltration in corneal tissue after LOX-1 inhibition. Plate counting experiment showed that number of viable fungus in corneas of Poly I treated group was significantly less than control group. LOX-1 neutralizing antibody or Poly I treatment significantly decreased neutrophil infiltration, the quantity of TUNEL-positive cells, the expression of Fas, Fas ligand, caspase-8, caspase-9, caspase-3, cleaved caspase-3 and the percentage of apoptosis neutrophils compared with control corneas. LOX-1 neutralizing antibody treatment significantly decreased Fas, FasL, CASP3, CASP8, CASP9 and cleaved caspase-3 expression in neutrophils. CONCLUSION LOX-1 inhibition decrease neutrophil apoptosis and fungal load in A. fumigatus keratitis.
Collapse
Affiliation(s)
- Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kun He
- Department of Ophthalmology, Zhejiang Quhua Hospital Quzhou, Zhejiang, China
| | - Min Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiuqiu Zhang
- Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yawen Niu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Nan Jiang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
4
|
Yang L, Geng T, Yang G, Ma J, Wang L, Ketkar H, Yang D, Lin T, Hwang J, Zhu S, Wang Y, Dai J, You F, Cheng G, Vella AT, Flavell RA, Fikrig E, Wang P. Macrophage scavenger receptor 1 controls Chikungunya virus infection through autophagy in mice. Commun Biol 2020; 3:556. [PMID: 33033362 PMCID: PMC7545163 DOI: 10.1038/s42003-020-01285-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophage scavenger receptor 1 (MSR1) mediates the endocytosis of modified low-density lipoproteins and plays an important antiviral role. However, the molecular mechanism underlying MSR1 antiviral actions remains elusive. We report that MSR1 activates autophagy to restrict infection of Chikungunya virus (CHIKV), an arthritogenic alphavirus that causes acute and chronic crippling arthralgia. Msr1 expression was rapidly upregulated after CHIKV infection in mice. Msr1 knockout mice had elevated viral loads and increased susceptibility to CHIKV arthritis along with a normal type I IFN response. Induction of LC3 lipidation by CHIKV, a marker of autophagy, was reduced in Msr1-/- cells. Mechanistically, MSR1 interacted with ATG12 through its cytoplasmic tail and this interaction was enhanced by CHIKV nsP1 protein. MSR1 repressed CHIKV replication through ATG5-ATG12-ATG16L1 and this was dependent on the FIP200-and-WIPI2-binding domain, but not the WD40 domain of ATG16L1. Our results elucidate an antiviral role for MSR1 involving the autophagic function of ATG5-ATG12-ATG16L1.
Collapse
Affiliation(s)
- Long Yang
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA
| | - Tingting Geng
- grid.208078.50000000419370394Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Guang Yang
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA ,grid.258164.c0000 0004 1790 3548Department of Parasitology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jinzhu Ma
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA
| | - Leilei Wang
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA
| | - Harshada Ketkar
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA
| | - Duomeng Yang
- grid.208078.50000000419370394Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Tao Lin
- grid.208078.50000000419370394Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Jesse Hwang
- grid.47100.320000000419368710Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Shu Zhu
- grid.47100.320000000419368710Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520 USA ,grid.59053.3a0000000121679639Present Address: Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027 China
| | - Yanlin Wang
- grid.208078.50000000419370394Department of Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Jianfeng Dai
- grid.263761.70000 0001 0198 0694Institutes of Biology and Medical Sciences, Soochow University, Jiangsu, China
| | - Fuping You
- grid.11135.370000 0001 2256 9319School of Basic Medical Sciences, Peking University, Beijing, China
| | - Gong Cheng
- grid.12527.330000 0001 0662 3178Department of Basic Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Anthony T. Vella
- grid.208078.50000000419370394Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Richard. A. Flavell
- grid.47100.320000000419368710Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520 USA ,grid.413575.10000 0001 2167 1581Howard Hughes Medical Institute, Chevy Chase, MD USA
| | - Erol Fikrig
- grid.47100.320000000419368710Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 06520 USA ,grid.413575.10000 0001 2167 1581Howard Hughes Medical Institute, Chevy Chase, MD USA
| | - Penghua Wang
- grid.260917.b0000 0001 0728 151XDepartment of Microbiology & Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595 USA ,grid.208078.50000000419370394Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| |
Collapse
|
5
|
Abstract
BACKGROUND Atherosclerotic lesions develop preferentially at certain sites in the human arterial system, such as the inner wall of curved segments and the outer wall of bifurcations. Local wall shear stress (WSS) and concentration of low density lipoprotein (LDL) have been identified as two important factors contributing to these lesions. OBJECTIVE To determine if a connection exists between arterial curvature and the formation of atherosclerosis. METHODS A set of 3-D vessel models with different bend angles was constructed. By comparing blood flow, WSS, and LDL aggregation, the influence of bend curvature on atherosclerotic lesions was assessed. RESULTS Upon increasing arterial bending, low WSS regions were formed at the outer wall of the junction between straight and curved segments, as well as the inner wall of curved segments. However, high LDL concentrations only appeared at the inner wall of the bend region. A connection between secondary flow and LDL concentration was observed; high LDL concentration regions had stronger secondary flow. Higher water infiltration velocity could enhance LDL aggregation, while blood non-Newtonian properties, by easing secondary flow, diminished its aggregation. CONCLUSIONS Under the same flow rate, a larger bend angle increased flow resistance, lowered WSS, and increased LDL surface concentrations, thus indicating an increased risk of atherosclerosis.
Collapse
Affiliation(s)
- Sen Lu
- Institute of Thermal Science of Technology, Shandong University, Jinan, China
| | - Shusheng Zhang
- Institute of Thermal Science of Technology, Shandong University, Jinan, China
| |
Collapse
|
6
|
Wang Y, Che J, Zhao H, Tang J, Shi G. Paeoniflorin attenuates oxidized low‐density lipoprotein‐induced apoptosis and adhesion molecule expression by autophagy enhancement in human umbilical vein endothelial cells. J Cell Biochem 2018; 120:9291-9299. [PMID: 30548681 DOI: 10.1002/jcb.28204] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 11/15/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Yi Wang
- Department of Cardiothoracic Surgery Huaihe Hospital of Henan University Kaifeng China
| | - Jianbo Che
- Department of Cardiothoracic Surgery Huaihe Hospital of Henan University Kaifeng China
| | - Hui Zhao
- Department of Cardiothoracic Surgery Huaihe Hospital of Henan University Kaifeng China
| | - Jianyu Tang
- Department of Cardiothoracic Surgery Huaihe Hospital of Henan University Kaifeng China
| | - Gongning Shi
- Department of Cardiothoracic Surgery Huaihe Hospital of Henan University Kaifeng China
| |
Collapse
|
7
|
Luo Y, Meng X, Zhou P, Lu S, Qin M, Xu X, Sun G, Sun X. Elatoside C protects against ox-LDL-induced HUVECs injury by FoxO1-mediated autophagy induction. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1654-1665. [DOI: 10.1016/j.bbadis.2017.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/31/2016] [Accepted: 01/23/2017] [Indexed: 12/27/2022]
|
8
|
Zhou H, Wang C. Cytoprotective Effects and Mechanisms of Δ-17 Fatty Acid Desaturase in Injured Human Umbilical Vein Endothelial Cells (HUVECs). Med Sci Monit 2017; 23:1627-1635. [PMID: 28373637 PMCID: PMC5388310 DOI: 10.12659/msm.903654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The beneficial effect of Δ-17 FAD is poorly understood. The aim of this study was to investigate the protective mechanism of fatty acids against atherosclerotic (AS) damage induced by oxidized low-density lipoprotein (ox-LDL) in human umbilical vein endothelial cells (HUVECs), and to identify the molecular mechanisms involved. Material/Methods The ox-LDL was used to induce lipotoxicity in HUVECs to establish a model of oxidative injury. HUVECs were transfected with Δ-17FAD lentivirus to induce cytoprotective effects. We evaluated the alterations in cell proliferation and apoptosis, and oxidative stress index, including levels of nitric oxide (NO), malonyldialdehyde (MDA), SOD enzyme, LDH, GSH-PX, and vascular endothelial growth factor (VEGF) expression. Results The ox-LDL-induced excessive cellular apoptosis of HUVECs was abrogated by upregulation of Δ-17 FAD. Importantly, Δ-17 FAD converted ω-3 polyunsaturated fatty acid ARA into ω-6 polyunsaturated fatty acid EPA. Further, Δ-17 FAD overexpression promoted the proliferation of HUVECS, and inhibited ox-LDL-induced lipid peroxidation of HUVECs. The levels of nitric oxide, GSH-PX, and SOD enzyme were increased, and the activity of MDA and LDH was suppressed by the upregulation of Δ-17 FAD. In addition, upregulation of Δ-17 FAD significantly increased VEGF expression. In vitro tube formation assay showed that Δ-17 FAD significantly promoted angiogenesis. Conclusions These results suggest that Δ-17 fatty acid desaturase plays a beneficial role in the prevention of ox-LDL-induced cellular damage.
Collapse
Affiliation(s)
- Haoyu Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China (mainland)
| | - Chengming Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
9
|
Yang J, Yu J, Li D, Yu S, Ke J, Wang L, Wang Y, Qiu Y, Gao X, Zhang J, Huang L. Store-operated calcium entry-activated autophagy protects EPC proliferation via the CAMKK2-MTOR pathway in ox-LDL exposure. Autophagy 2016; 13:82-98. [PMID: 27791458 DOI: 10.1080/15548627.2016.1245261] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Improving biological functions of endothelial progenitor cells (EPCs) is beneficial to maintaining endothelium homeostasis and promoting vascular re-endothelialization. Because macroautophagy/autophagy has been documented as a double-edged sword in cell functions, its effects on EPCs remain to be elucidated. This study was designed to explore the role and molecular mechanisms of store-operated calcium entry (SOCE)-activated autophagy in proliferation of EPCs under hypercholesterolemia. We employed oxidized low-density lipoprotein (ox-LDL) to mimic hypercholesterolemia in bone marrow-derived EPCs from rat. Ox-LDL dose-dependently activated autophagy flux, while inhibiting EPC proliferation. Importantly, inhibition of autophagy either by silencing Atg7 or by 3-methyladenine treatment, further aggravated proliferative inhibition by ox-LDL, suggesting the protective effects of autophagy against ox-LDL. Interestingly, ox-LDL increased STIM1 expression and intracellular Ca2+ concentration. Either Ca2+ chelators or deficiency in STIM1 attenuated ox-LDL-induced autophagy activation, confirming the involvement of SOCE in the process. Furthermore, CAMKK2 (calcium/calmodulin-dependent protein kinase kinase 2, β) activation and MTOR (mechanistic target of rapamycin [serine/threonine kinase]) deactivation were associated with autophagy modulation. Together, our results reveal a novel signaling pathway of SOCE-CAMKK2 in the regulation of autophagy and offer new insights into the important roles of autophagy in maintaining proliferation and promoting the survival capability of EPCs. This may be beneficial to improving EPC transplantation efficacy and enhancing vascular re-endothelialization in patients with hypercholesterolemia.
Collapse
Affiliation(s)
- Jie Yang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jie Yu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Dongdong Li
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Sanjiu Yu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jingbin Ke
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Lianyou Wang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Yanwei Wang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Youzhu Qiu
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Xubin Gao
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Jihang Zhang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Lan Huang
- a Institute of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| |
Collapse
|
10
|
He F, Xu X, Yuan S, Tan L, Gao L, Ma S, Zhang S, Ma Z, Jiang W, Liu F, Chen B, Zhang B, Pang J, Huang X, Weng J. Oxidized Low-density Lipoprotein (ox-LDL) Cholesterol Induces the Expression of miRNA-223 and L-type Calcium Channel Protein in Atrial Fibrillation. Sci Rep 2016; 6:30368. [PMID: 27488468 PMCID: PMC4973266 DOI: 10.1038/srep30368] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia causing high morbidity and mortality. While changing of the cellular calcium homeostasis plays a critical role in AF, the L-type calcium channel α1c protein has suggested as an important regulator of reentrant spiral dynamics and is a major component of AF-related electrical remodeling. Our computational modeling predicted that miRNA-223 may regulate the CACNA1C gene which encodes the cardiac L-type calcium channel α1c subunit. We found that oxidized low-density lipoprotein (ox-LDL) cholesterol significantly up-regulates both the expression of miRNA-223 and L-type calcium channel protein. In contrast, knockdown of miRNA-223 reduced L-type calcium channel protein expression, while genetic knockdown of endogenous miRNA-223 dampened AF vulnerability. Transfection of miRNA-223 by adenovirus-mediated expression enhanced L-type calcium currents and promoted AF in mice while co-injection of a CACNA1C-specific miR-mimic counteracted the effect. Taken together, ox-LDL, as a known factor in AF-associated remodeling, positively regulates miRNA-223 transcription and L-type calcium channel protein expression. Our results implicate a new molecular mechanism for AF in which miRNA-223 can be used as an biomarker of AF rheumatic heart disease.
Collapse
Affiliation(s)
- Fengping He
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Xin Xu
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Shuguo Yuan
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Liangqiu Tan
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Lingjun Gao
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Shaochun Ma
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Shebin Zhang
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Zhanzhong Ma
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Wei Jiang
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Fenglian Liu
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Baofeng Chen
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Beibei Zhang
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Jungang Pang
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Xiuyan Huang
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Jiaqiang Weng
- Department Institute of Cardiovascular Diseases, The Yuebei People’s Hospital, Medical College, Shantou University, Shantou, Guangdong, China
| |
Collapse
|
11
|
Geng Z, Xu F, Zhang Y. MiR-129-5p-mediated Beclin-1 suppression inhibits endothelial cell autophagy in atherosclerosis. Am J Transl Res 2016; 8:1886-1894. [PMID: 27186312 PMCID: PMC4859917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/06/2016] [Indexed: 06/05/2023]
Abstract
Endothelial cell injury and subsequent death play an essential role in the pathogenesis of atherosclerosis. Autophagy of endothelial cells antagonizes the development of atherosclerosis, whereas the underlying molecular mechanisms are unclear. MicroRNA-129-5p (miR-129-5p) is a well-defined tumor suppressorin some types of cancer, while it is unknown whether miR-129-5p may also play a role in the development of atherosclerosis. Here, we addressed this question in the current study. We examined the levels of endothelial cell autophagy in ApoE (-/-) mice suppled with high-fat diet (HFD), a mouse model for atherosclerosis (simplified as HFD mice). We analyzed the levels of Beclin-1 and the levels of miR-129-5p in the purified CD31+ endothelial cells from mouse aorta. Prediction of the binding between miR-129-5p and 3'-UTR of Beclin-1 mRNA was performed by bioinformatics analyses and confirmed by a dual luciferase reporter assay. The effects of miR-129-5p were further analyzed in an in vitro model using oxidized low-density lipoprotein (ox-LDL)-treated human aortic endothelial cells (HAECs). We found that HFD mice developed atherosclerosisin 12 weeks, while the control ApoE (-/-) mice that had received normal diet (simplified as CTL mice) did not. Compared to CTL mice, HFD mice had significantly lower levels of endothelial cell autophagy, resulting from decreases in Beclin-1 protein, but not mRNA. The decreases in Beclin-1 in endothelial cells were due to HFD-induced increases inmiR-129-5p, which suppressed the translation of Beclin-1 mRNA via 3'-UTR binding. These in vivo findings were reproduced in vitro on ox-LDL-treated HAECs. Together, these data suggest that upregulation of miR-129-5p by HFD may impair the protective effects of endothelial cell autophagy against development of atherosclerosis through suppressing protein translation of Beclin-1.
Collapse
Affiliation(s)
- Zhaohua Geng
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical UniversityChongqing 400037, China
| | - Fei Xu
- Oncology Radiotherapy Center of PLA 302 HospitalBeijing 100039, China
| | - Yiguan Zhang
- Josephine Ford Cancer Center, Department of Internal Medicine, Henry Ford Health SystemDetroit, MI 48202, USA
| |
Collapse
|
12
|
Lectin-like oxidized low-density lipoprotein receptor-1 regulates autophagy and Toll-like receptor 4 in the brain of hypertensive mice. J Hypertens 2016; 33:525-33; discussion 533. [PMID: 25380158 DOI: 10.1097/hjh.0000000000000411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) regulates blood pressure and is important for the development of inflammation, oxidative stress and autophagy. We posited that LOX-1 via NADPH oxidase activation may affect autophagy and Toll-like receptor (TLR)4 expression in the brains of hypertensive mice. METHODS To examine this postulate, wild-type mice were given continuous infusion of angiotensin II (50 ng/min) for 28 days. As expected, these mice developed significant increase in blood pressure. RESULTS Corpus callosum in the brains of these hypertensive mice revealed intense expression of NADPH oxidase (subunits P22phox and P47phox), activation of P38 MAPK and nuclear factor-kappaB (P65), autophagy-related proteins (beclin-1 and conversion of LC3-I to LC3-II), and TLR4 (and associated signaling molecules myeloid differentiation primary response gene (88) and TIR-domain-containing adapter-inducing interferon-β). These observations suggested activation of redox signals, autophagy and immune system. In parallel experiments, mice with LOX-1 deletion given similar infusion of angiotensin II showed much less expression of NADPH oxidase, activation of P38 MAPK and nuclear factor-kappaB, autophagy-related proteins and TLR4 [and myeloid differentiation primary response gene (88) and TIR-domain-containing adapter-inducing interferon-β]. Mice with LOX-1 deletion also showed a smaller rise in blood pressure than wild-type mice, both groups given similar infusion of angiotensin II. CONCLUSION These studies suggest immune activation in the brains of mice with angiotensin II-induced hypertension. Further, these observations imply the existence of a link between LOX-1, NADPH oxidase expression, development of autophagy and immune activation in hypertension.
Collapse
|
13
|
Resveratrol Enhances Autophagic Flux and Promotes Ox-LDL Degradation in HUVECs via Upregulation of SIRT1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7589813. [PMID: 27069532 PMCID: PMC4812467 DOI: 10.1155/2016/7589813] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/09/2016] [Accepted: 02/21/2016] [Indexed: 12/03/2022]
Abstract
Oxidized low-density lipoprotein- (Ox-LDL-) induced autophagy dysfunction in human vascular endothelial cells contributes to the development of atherosclerosis (AS). Resveratrol (RSV) protects against Ox-LDL-induced endothelium injury. The objective of this study was to determine the mechanisms underlying Ox-LDL-induced autophagy dysfunction and RSV-mediated protection in human umbilical vein endothelial cells (HUVECs). The results showed that Ox-LDL suppressed the expression of sirtuin 1 (SIRT1) and increased LC3-II and sequestosome 1 (p62) protein levels without altering p62 mRNA levels in HUVECs. Pretreatment with bafilomycin A1 (BafA1) to inhibit lysosomal degradation abrogated the Ox-LDL-induced increase in LC3-II protein level. Ox-LDL increased colocalization of GFP and RFP puncta in mRFP-GFP-tandem fluorescent LC3- (tf-LC3-) transfected cells. Moreover, Ox-LDL decreased the expression of mature cathepsin D and attenuated cathepsin D activity. Pretreatment with RSV increased the expression of SIRT1 and LC3-II and increased p62 protein degradation. RSV induced RFP-LC3 aggregation more than GFP-LC3 aggregation. RSV restored lysosomal function and promoted Ox-LDL degradation in HUVECs. All the protective effects of RSV were blocked after SIRT1 was knocked down. These findings demonstrated that RSV upregulated the expression of SIRT1, restored lysosomal function, enhanced Ox-LDL-induced impaired autophagic flux, and promoted Ox-LDL degradation through the autophagy-lysosome degradation pathway in HUVECs.
Collapse
|
14
|
|
15
|
Thakkar S, Wang X, Khaidakov M, Dai Y, Gokulan K, Mehta JL, Varughese KI. Structure-based Design Targeted at LOX-1, a Receptor for Oxidized Low-Density Lipoprotein. Sci Rep 2015; 5:16740. [PMID: 26578342 PMCID: PMC4649741 DOI: 10.1038/srep16740] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/19/2015] [Indexed: 01/16/2023] Open
Abstract
Atherosclerosis related cardiovascular diseases continue to be the primary cause of mortality in developed countries. The elevated level of low density lipoprotein (LDL) is generally considered to be the driver of atherosclerosis, but recent years have seen a shift in this perception in that the vascular plaque buildup is mainly caused by oxidized LDL (ox-LDL) rather than native-LDL. The scavenger receptor LOX-1 found in endothelial cells binds and internalizes ox-LDL which leads to the initiation of plaque formation in arteries. Using virtual screening techniques, we identified a few potential small molecule inhibitors of LOX-1 and tested their inhibitory potential using differential scanning fluorimetry and various cellular assays. Two of these molecules significantly reduced the uptake of ox-LDL by human endothelial cells, LOX-1 transcription and the activation of ERK1/2 and p38 MAPKs in human endothelial cells. In addition, these molecules suppressed ox-LDL-induced VCAM-1 expression and monocyte adhesion onto human endothelial cells demonstrating their therapeutic potential.
Collapse
Affiliation(s)
- Shraddha Thakkar
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Xianwei Wang
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Magomed Khaidakov
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Yao Dai
- Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kuppan Gokulan
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jawahar L Mehta
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Medicine, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kottayil I Varughese
- Department of Physiology and Biophysics College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
16
|
Ding Z, Liu S, Wang X, Deng X, Fan Y, Shahanawaz J, Shmookler Reis RJ, Varughese KI, Sawamura T, Mehta JL. Cross-talk between LOX-1 and PCSK9 in vascular tissues. Cardiovasc Res 2015; 107:556-67. [PMID: 26092101 DOI: 10.1093/cvr/cvv178] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022] Open
Abstract
AIMS Lectin-like ox-LDL receptor-1 (LOX-1) plays an important role in inflammatory diseases, such as atherosclerosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) modulates LDL receptor degradation and influences serum LDL levels. The present study was designed to investigate the possible interaction between PCSK9 and LOX-1. METHODS AND RESULTS In the first set of experiments, human vascular endothelial cells and smooth muscle cells were studied at baseline and after lipopolysaccharide (LPS) treatment (to create an inflammatory state). Both PCSK9 and LOX-1 were strongly induced by LPS treatment. To define the role of PCSK9 in LOX-1 expression, cells were transfected with siRNA against PCSK9, which resulted in reduced LOX-1 expression and function. On the other hand, cells exposed to recombinant hPCSK9 revealed enhanced LOX-1 expression (P < 0.05). To determine whether LOX-1 also regulates PCSK9, cultured cells in which LOX-1 was knocked down by siRNA expressed less PCSK9, whereas those transfected with hLOX-1 cDNA showed increased PCSK9 expression. The second set of experiments was carried out in wild-type (WT) and gene knockout (KO; LOX-1 and PCSK9) mice; LOX-1 KO mice showed much less PCSK9 (P < 0.05 vs. WT mice). PCSK9-KO mice showed much less LOX-1 (P < 0.05 vs. WT mice). Furthermore, we observed that mitochondrial reactive oxygen species (mtROS) plays an initiating role in the LOX-1/PCSK9 interaction, since mtROS induction enhanced and its inhibition reduced the expression of both PCSK9 and LOX-1. We also found that both LOX-1 and PCSK9 regulate adhesion molecules vascular cell adhesion molecule-1 expression. Finally, oxidized low-density lipoprotein and tumour necrosis factor-α, pro-inflammatory stimuli besides LPS, regulated PCSK9 expression that is mediated by the NF-κB signalling pathway. CONCLUSIONS These observations suggest that LOX-1 and PCSK9 positively influence each other's expression, especially during an inflammatory reaction. mtROS appear to be important initiators of PCSK9/LOX-1 expression.
Collapse
Affiliation(s)
- Zufeng Ding
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Shijie Liu
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xianwei Wang
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xiaoyan Deng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiwani Shahanawaz
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Robert J Shmookler Reis
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kattayi I Varughese
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tatsuya Sawamura
- Department of Physiology, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto 390-8621, Japan
| | - Jawahar L Mehta
- Central Arkansas Veterans Healthcare System and the University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
17
|
Hemodynamic shear stress modulates endothelial cell autophagy: Role of LOX-1. Int J Cardiol 2015; 184:86-95. [DOI: 10.1016/j.ijcard.2015.01.065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/23/2014] [Accepted: 01/25/2015] [Indexed: 11/21/2022]
|
18
|
Ding Z, Liu S, Wang X, Deng X, Fan Y, Sun C, Wang Y, Mehta JL. Hemodynamic shear stress via ROS modulates PCSK9 expression in human vascular endothelial and smooth muscle cells and along the mouse aorta. Antioxid Redox Signal 2015; 22:760-71. [PMID: 25490141 PMCID: PMC4361218 DOI: 10.1089/ars.2014.6054] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIMS To investigate a possible link between hemodynamic shear stress, reactive oxygen species (ROS) generation, and proprotein convertase subtilisin/kexin type 9 (PCSK9) expression. RESULTS Using a parallel-plate flow chamber, we observed that PCSK9 expression in vascular smooth muscle cells (SMCs) and endothelial cells (ECs) reached maximal value at low shear stress (3-6 dynes/cm(2)), and then began to decline with an increase in shear stress. PCSK9 expression increased when cells were treated with lipopolysaccharide. PCSK9 expression was always greater in SMCs than in ECs. ROS generation followed the same pattern as PCSK9 expression. Aortic branching and aorta-iliac bifurcation regions of mouse aorta that express low shear stress were also found to have greater PCSK9 expression (vs. other regions). To determine a relationship between ROS and PCSK9 expression, ECs and SMCs were treated with ROS inhibitors diphenylene-iodonium chloride and apocynin, and both markedly reduced PCSK9 expression. Relationship between PCSK9 and ROS was further studied in p47(phox) and gp91(phox) knockout mice; both mice strains revealed low PCSK9 levels in serum and mRNA levels in aorta-iliac bifurcation regions (vs. wild-type mice). Other studies showed that ROS and NF-κB activation plays a bridging role in PCSK9 expression via lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1). INNOVATION Low shear stress induces PCSK9 expression, which is mediated by NADPH oxidase-dependent ROS production. CONCLUSIONS This study provides evidence that low shear stress enhances PCSK9 expression in concert with ROS generation in vascular ECs and SMCs. ROS seem to regulate PCSK9 expression. We propose that PCSK9-ROS interaction may be important in the development of atherosclerosis in arterial channels with low shear stress.
Collapse
Affiliation(s)
- Zufeng Ding
- 1 Central Arkansas Veterans Healthcare System and the Departments of Medicine, and Physiology and Biophysics, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | | | | | | | | | | | | | | |
Collapse
|
19
|
ZHAO XI, LIU YOUJUN, DING JINLI, REN XIAOCHEN, BAI FAN, ZHANG MINGZI, MA LIANCAI, WANG WENXIN, XIE JINSHENG, QIAO AIKE. HEMODYNAMIC EFFECTS OF THE ANASTOMOSES IN THE MODIFIED BLALOCK–TAUSSIG SHUNT: A NUMERICAL STUDY USING A 0D/3D COUPLING METHOD. J MECH MED BIOL 2015. [DOI: 10.1142/s0219519415500177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The modified Blalock–Taussig (BT) shunt is a palliative surgery which can help the tetralogy of Fallot (TOF) patient increase the blood oxygen saturation by interposing a systemic-to-pulmonary artery shunt. Two typical anastomotic shapes are frequently used in clinical practice: the end-to-side (ETS) and the side-to-side (STS) anastomosis. This paper examines the hemodynamic influence of the anastomotic shape in the modified BT shunt. Three models with different anastomotic shapes were reconstructed. The ETS anastomoses were applied in the first model. For the innominate artery (IA) and the pulmonary artery (PA) in the second model, the ETS and the STS anastomosis were applied, respectively. Finally, the STS anastomoses were applied in the third model. The 0D/3D coupling method was used to perform a numerical simulation by coupling the three-dimensional (3D) artery model with a zero-dimensional (0D) lumped parameter model for the cardiovascular system. The simulation results showed that the perfusion into the left and right PA in Model 1 was unbalanced. Swirling flow appeared in the shunt in Model 3, but the shunt flow rate in Model 3 was lower. The ETS anastomosis at the PA may cause unbalanced blood perfusion into the left and right PA. Conversely, the STS anastomosis can make the blood perfusion more balanced. Otherwise, the STS anastomosis at the IA could generate a swirling flow in the shunt which may provide a better hemodynamic environment while decreasing the pulmonary perfusion.
Collapse
Affiliation(s)
- XI ZHAO
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - YOUJUN LIU
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - JINLI DING
- Department of Diagnostic Radiology, Beijing You An Hospital affiliated to Capital Medical University, No. 8, Xi Tou Tiao, You An Men, Fengtai District, Beijing 100069, P. R. China
| | - XIAOCHEN REN
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - FAN BAI
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - MINGZI ZHANG
- Graduate School of Engineering, Tohoku University, Ohta Laboratory, Institute of Fluid Science, 2-1-1 Katahira Aoba-ku Sendai Miyagi 980-8577, Japan
| | - LIANCAI MA
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - WENXIN WANG
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| | - JINSHENG XIE
- Beijing An Zhen Hospital affiliated to Capital Medical University, No. 2 Anzhen Road Chaoyang District, Beijing 100029, P. R. China
| | - AIKE QIAO
- College of Life Science and Bio-engineering, Beijing University of Technology, No. 100 Pingleyuan, Chaoyang District, Beijing 100124, P. R. China
| |
Collapse
|
20
|
Impact of hepatitis C seropositivity on the risk of coronary heart disease events. Am J Cardiol 2014; 114:1841-5. [PMID: 25438910 PMCID: PMC4372470 DOI: 10.1016/j.amjcard.2014.09.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/14/2014] [Accepted: 09/14/2014] [Indexed: 12/14/2022]
Abstract
Chronic infections have been shown to enhance atherogenicity. However, the association between chronic hepatitis C (HCV) and coronary heart disease (CHD) remains controversial. We examined the risk for CHD events in patients with HCV with an emphasis on the risk of CHD events with active infection. We conducted a retrospective cohort study using the Enterprise Data Warehouse at the University of Arkansas for Medical Sciences. HCV positive and negative patients were identified based on serology and incident CHD events were studied. Patient characteristics at entry were compared either by analysis of variance/F-test (continuous variables) or by a Chi-squared test (categorical variables). The joint effect of risk factors for incident CHD was evaluated using logistic regression. A total of 8,251 HCV antibody positive, 1,434 HCV RNA positive and 14,799 HCV negative patients were identified. HCV antibody and RNA positive patients had a higher incidence of hypertension, diabetes mellitus, obesity and chronic lung disease, but lower serum cholesterol levels compared to HCV negative patients (p< 0.001). HCV seropositive patients had a higher incidence of CHD events when compared to controls (4.9% vs. 3.2%, p<0.001). In the HCV cohort, patients with detectable HCV RNA had a significantly higher incidence of CHD events when compared to patients who were only HCV antibody positive with no detectable RNA (5.9% vs. 4.7%, p=0.04). In multivariate logistic regression analysis, both HCV antibody positivity (OR 1.32, 95% CI 1.09-1.60, p<0.001) and HCV RNA positivity (OR 1.59, 95% CI 1.13-2.26, p<0.001) were independent risk factors for incident CHD events. In conclusion, there is increased incidence of CHD events in HCV seropositive patients and the incidence is much higher in patients with detectable HCV RNA when compared to patients with remote infection who are only antibody positive. Lipid profile does not appear to be a good cardiovascular risk stratification tool in HVC patients.
Collapse
|
21
|
Chen Z, Martin M, Li Z, Shyy JYJ. Endothelial dysfunction: the role of sterol regulatory element-binding protein-induced NOD-like receptor family pyrin domain-containing protein 3 inflammasome in atherosclerosis. Curr Opin Lipidol 2014; 25:339-49. [PMID: 25188917 PMCID: PMC4339278 DOI: 10.1097/mol.0000000000000107] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Great effort has been devoted to elucidate the molecular mechanisms by which inflammasome in macrophages contributes to atherosclerosis. Inflammasome in vascular endothelial cells and its causal relationship with endothelial dysfunction in atherosclerosis are less understood. Here, we review the recent studies of inflammasome and its activation in endothelial cells, and highlight such endothelial inflammatory response in atherosclerosis. RECENT FINDINGS Inflammasomes are critical effectors in innate immunity, and their activation in macrophages and the arterial wall contributes to atherogenesis. Sterol regulatory element-binding protein 2, a master regulator in cholesterol biosynthesis, can be activated in a noncanonical manner, which leads to the activation of the NOD-like receptor family pyrin domain-containing protein inflammasome in macrophages and endothelial cells. Results from in-vitro and in-vivo models suggest that sterol regulatory element-binding protein 2 is a key molecule in aggravating proinflammatory responses in endothelial cells and promoting atherosclerosis. SUMMARY The SREBP-induced NOD-like receptor family pyrin domain-containing protein inflammasome and its instigation of innate immunity is an important contributor to atherosclerosis. Elucidating the underlying mechanisms will expand our understanding of endothelial dysfunction and its dynamic interaction with vascular inflammation. Furthermore, targeting SREBP-inflammasome pathways can be a therapeutic strategy for attenuating atherosclerosis.
Collapse
Affiliation(s)
- Zhen Chen
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Marcy Martin
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Biochemistry and Molecular Biology Graduate Program, University of California, Riverside, Riverside, CA 92521
| | - Zhao Li
- Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, PRC
| | - John Y-J. Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, PRC
| |
Collapse
|
22
|
Ding Z, Liu S, Wang X, Theus S, Fan Y, Deng X, Mehta JL. LOX-1 – dependent mitochondrial DNA damage and NLRP3 activation during systemic inflammation in mice. Biochem Biophys Res Commun 2014; 451:637-43. [DOI: 10.1016/j.bbrc.2014.08.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/08/2014] [Indexed: 01/05/2023]
|
23
|
Lectin-like ox-LDL receptor-1 (LOX-1)-Toll-like receptor 4 (TLR4) interaction and autophagy in CATH.a differentiated cells exposed to angiotensin II. Mol Neurobiol 2014; 51:623-32. [PMID: 24902807 DOI: 10.1007/s12035-014-8756-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/15/2014] [Indexed: 01/22/2023]
Abstract
Toll-like receptors (TLRs) play an essential role in innate immune response. Expression of TLRs has also been linked to autophagy. As the main receptor for oxidized low-density lipoprotein (ox-LDL) on the cell surface, lectin-like ox-LDL receptor-1 (LOX-1) is upregulated by proinflammatory cytokines and has been linked to the development of autophagy. However, the relationship between LOX-1, autophagy, and TLR4 in neurons has not been defined. Here, we show that Angiotensin II (Ang II) treatment of CATH.a differentiated neuronal cells resulted in the expression of TLR4 (and associated signals MyD88 and Toll/interleukin-1 receptor domain-containing adapter-inducing interferon (TRIF)), LOX-1 autophagy. LOX-1 knockdown (transfection with specific small interfering RNA (siRNA)) resulted in reduced expression of TLR4 (and associated signals MyD88 and TRIF) and P-P38 mitogen-activated protein kinase (MAPK) and autophagy. TLR4 knockdown with siRNA resulted in reduced LOX-1 expression and autophagy, indicating a positive feedback between LOX-1 and TLR4. Knockdown of TRIF as well as MyD88 or inhibition of P38 MAPK also inhibited the expression of LOX-1 and TLR4 and autophagy. Importantly, pretreatment with 3-methyladenine (autophagy inhibitor) enhanced while rapamycin (autophagy inducer) decreased the expression of LOX-1, TLR4, and P-P38 MAPK. These studies suggest the presence of a bidirectional link between LOX-1and TLR4 in cultured CATH.a differentiated cells exposed to Ang II with an important role for autophagy in this link.
Collapse
|
24
|
Ding Z, Liu S, Wang X, Dai Y, Khaidakov M, Romeo F, Mehta JL. LOX-1, oxidant stress, mtDNA damage, autophagy, and immune response in atherosclerosis. Can J Physiol Pharmacol 2014; 92:524-30. [PMID: 24959993 DOI: 10.1139/cjpp-2013-0420] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As a major receptor for oxidized low density lipoprotein (ox-LDL), lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is upregulated in many pathophysiological events, including endothelial cell dysfunction and smooth muscle cell growth, as well as monocyte migration and transformation into foam cells, which are present in atherosclerosis and myocardial ischemia. Excessive production of reactive oxygen species (ROS) increases LOX-1 expression, induces mitochondrial DNA damage, and activates autophagy. Damaged mitochondrial DNA that escapes from autophagy induces an inflammatory response. This paper reviews the potential link between LOX-1, mitochondrial DNA damage, autophagy, and immune response in atherosclerosis.
Collapse
Affiliation(s)
- Zufeng Ding
- a Central Arkansas Veterans Healthcare System and the Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | | | | | | | | | | | | |
Collapse
|