1
|
Wang C, He Y, Tang J, Mao J, Liang X, Xu M, Zhang Z, Tian J, Jiang J, Li C, Zhou X. Chondroitin sulfate functionalized nanozymes inhibit the inflammation feedback loop for enhanced atherosclerosis therapy by regulating intercellular crosstalk. Int J Biol Macromol 2024; 282:136918. [PMID: 39471920 DOI: 10.1016/j.ijbiomac.2024.136918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/01/2024]
Abstract
In the inflammatory microenvironment of atherosclerotic plaques, metabolic dysregulation of superoxide anion (O2-) and hydrogen peroxide (H2O2) leads to the activation of feedback mechanisms involving IL-1β, TNF-α, and MCP-1, which triggers inflammatory cascades between macrophages and vascular smooth muscle cells (VSMCs) in atherosclerosis (AS). To address this, a chondroitin sulfate (CS)-functionalized dual-targeted engineered nanozyme, CS-Lip/PB@Rap, was developed by encapsulating mesoporous Prussian blue nanoparticles (PBs) loaded with rapamycin (Rap) within CS-modified liposomes. CS functionalization endowed CS-Lip/PB@Rap with a specific targeting ability for CD44 receptors, thus enabling targeted delivery to inflammatory macrophages and VSMCs. Moreover, its enhanced multiple enzyme-like activities effectively modulated the imbalance of oxidative stress. The underlying mechanism of crosstalk regulation by these engineered nanozymes may inhibit the NF-κB pathway by restoring normal metabolism of O2- and H2O2, thereby blocking the TNF-α, IL-1β, and MCP-1 feedback loops between macrophages and VSMCs. This process reduced the production of inflammatory macrophages and inhibited the VSMC transformation from a contractile phenotype to a synthetic phenotype, preventing the formation of fibrous caps. Furthermore, the elimination of oxidative stress could decrease the production of oxygenized low-density lipoprotein (ox-LDL), which inhibited the formation of foam cells and alleviated the atherogenic progression.
Collapse
Affiliation(s)
- Chenglong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jingying Mao
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ji Tian
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Basic Medicine Research Innovation Center for Cardiometabolic Disease, Ministry of Education, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Duan M, Jie J, Li C, Bai X, Hua S, Tang M, Li D. Echinatin alleviates sepsis severity through modulation of the NF-κB and MEK/ERK signaling pathways. Biomed Pharmacother 2024; 179:117359. [PMID: 39236479 DOI: 10.1016/j.biopha.2024.117359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
Sepsis, a frequently fatal condition, emerges from an exaggerated inflammatory response to infection, resulting in multi-organ dysfunction and alarmingly high mortality rates. Despite the urgent need for effective treatments, current therapeutic options remain limited to antibiotics, with no other efficacious alternatives available. Echinatin (Ecn), a potent bioactive compound extracted from the roots and rhizomes of licorice, has gained significant attention for its broad pharmacological properties, particularly its ability to combat oxidative stress. Recent research highlights the crucial role that oxidative stress plays in the onset and progression of sepsis further emphasizing the potential therapeutic value of Ecn in this context. In this study, we explored the protective effects of Ecn in a murine model of sepsis induced by cecal ligation and puncture (CLP). Ecn demonstrated a significant reduction in the levels of inflammatory cytokines and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Network pharmacology analysis identified 41 targets and top 15 pathways involved in the Ecn-mediated signaling network, revealing that Ecn might exert its effects through key targets including the NF-κB and MAPK signaling pathways. Molecular docking studies suggested a strong affinity between Ecn and MEK, with kinetic simulations and binding energy calculations confirming a stable interaction. Mechanistically, Ecn treatment inhibited NF-κB and the MEK/ERK signaling pathway, as evidenced by decreased phosphorylation of IκBα and nuclear p65, along with reduced phosphorylation of MEK and ERK in both LPS-stimulated RAW 264.7 macrophages and septic mice. Furthermore, the administration of MEK signaling agonists reversed the anti-inflammatory effects of Ecn, indicating the involvement of this signaling pathway in Ecn's protective mechanism. Notably, our investigation revealed that Ecn did not affect bacterial proliferation either in vivo or in vitro, underscoring its specific immunomodulatory effects rather than direct antimicrobial activity. In summation, our findings underscored the potential of Ecn as an innovative therapeutic remedy for sepsis-induced injury, particularly through the regulation of the NF-κB and MEK/ERK signaling pathway. This exploration unveiled a promising therapeutic approach for treating sepsis, supplementing existing interventions and addressing their constraints.
Collapse
Affiliation(s)
- Meina Duan
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Jing Jie
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Chunxiuli Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoxue Bai
- Department of General Practice, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shucheng Hua
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China.
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
3
|
Zuo X, Ding X, Zhang Y, Kang YJ. Reversal of atherosclerosis by restoration of vascular copper homeostasis. Exp Biol Med (Maywood) 2024; 249:10185. [PMID: 38978540 PMCID: PMC11228934 DOI: 10.3389/ebm.2024.10185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
Atherosclerosis has traditionally been considered as a disorder characterized by the accumulation of cholesterol and thrombotic materials within the arterial wall. However, it is now understood to be a complex inflammatory disease involving multiple factors. Central to the pathogenesis of atherosclerosis are the interactions among monocytes, macrophages, and neutrophils, which play pivotal roles in the initiation, progression, and destabilization of atherosclerotic lesions. Recent advances in our understanding of atherosclerosis pathogenesis, coupled with results obtained from experimental interventions, lead us to propose the hypothesis that atherosclerosis may be reversible. This paper outlines the evolution of this hypothesis and presents corroborating evidence that supports the potential for atherosclerosis regression through the restoration of vascular copper homeostasis. We posit that these insights may pave the way for innovative therapeutic approaches aimed at the reversal of atherosclerosis.
Collapse
Affiliation(s)
- Xiao Zuo
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Xueqin Ding
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaya Zhang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Y James Kang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Qian G, Adeyanju O, Cai D, Tucker TA, Idell S, Chen SY, Guo X. DOCK2 Promotes Atherosclerosis by Mediating the Endothelial Cell Inflammatory Response. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:599-611. [PMID: 37838011 PMCID: PMC10988758 DOI: 10.1016/j.ajpath.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/16/2023]
Abstract
The pathology of atherosclerosis, a leading cause of mortality in patients with cardiovascular disease, involves inflammatory phenotypic changes in vascular endothelial cells. This study explored the role of the dedicator of cytokinesis (DOCK)-2 protein in atherosclerosis. Mice with deficiencies in low-density lipoprotein receptor and Dock2 (Ldlr-/-Dock2-/-) and controls (Ldlr-/-) were fed a high-fat diet (HFD) to induce atherosclerosis. In controls, Dock2 was increased in atherosclerotic lesions, with increased intercellular adhesion molecule (Icam)-1 and vascular cell adhesion molecule (Vcam)-1, after HFD for 4 weeks. Ldlr-/-Dock2-/- mice exhibited significantly decreased oil red O staining in both aortic roots and aortas compared to that in controls after HFD for 12 weeks. In control mice and in humans, Dock2 was highly expressed in the ECs of atherosclerotic lesions. Dock2 deficiency was associated with attenuation of Icam-1, Vcam-1, and monocyte chemoattractant protein (Mcp)-1 in the aortic roots of mice fed HFD. Findings in human vascular ECs in vitro suggested that DOCK2 was required in TNF-α-mediated expression of ICAM-1/VCAM-1/MCP-1. DOCK2 knockdown was associated with attenuated NF-κB phosphorylation with TNF-α, partially accounting for DOCK2-mediated vascular inflammation. With DOCK2 knockdown in human vascular ECs, TNF-α-mediated VCAM-1 promoter activity was inhibited. The findings from this study suggest the novel concept that DOCK2 promotes the pathogenesis of atherosclerosis by modulating inflammation in vascular ECs.
Collapse
Affiliation(s)
- Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Oluwaseun Adeyanju
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Dunpeng Cai
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, Missouri
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Shi-You Chen
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, Missouri; The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri; Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.
| | - Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.
| |
Collapse
|
5
|
Lu M, Pan J, Hu Y, Ding L, Li Y, Cui X, Zhang M, Zhang Z, Li C. Advances in the study of vascular related protective effect of garlic (Allium sativum) extract and compounds. J Nutr Biochem 2024; 124:109531. [PMID: 37984733 DOI: 10.1016/j.jnutbio.2023.109531] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 11/22/2023]
Abstract
Garlic (Allium sativum) is a functional food containing multiple bioactive compounds that find widespread applications in culinary and medicinal practices. It consists of multiple chemical components, including allicin and alliin. This article offers a comprehensive review of the protective effects of garlic extracts and their active constituents on the vascular system. In vitro and in vivo experiments have shown that garlic extracts and their active ingredients possess various bioactive properties. These substances demonstrate beneficial effects on blood vessels by demonstrating anti-inflammatory and antioxidant activities, inhibiting lipid accumulation and migration, preventing lipid peroxidation, promoting angiogenesis, reducing platelet aggregation, enhancing endothelial function, and inhibiting endothelial cell apoptosis. In clinical studies, garlic and its extracts have demonstrated their efficacy in managing vascular system diseases, including atherosclerosis, diabetes, and high cholesterol levels. In summary, these studies highlight the potential therapeutic roles and underlying mechanisms of garlic and its constituents in managing conditions like diabetes, atherosclerosis, ischemic diseases, and other vascular disorders.
Collapse
Affiliation(s)
- Mengkai Lu
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinyuan Pan
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liang Ding
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhiyuan Zhang
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chao Li
- Innovation Research Institute of traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
6
|
Garcia VP, Fandl HK, Hijmans JG, Berry AR, Cardenas HL, Stockelman KA, DeSouza NM, Treuth JW, Greiner JJ, Park AJ, Stauffer BL, DeSouza CA. Effects of circulating endothelial microvesicles isolated from adults with obesity on endothelial cell inflammation, apoptosis, and nitric oxide production. Am J Physiol Endocrinol Metab 2024; 326:E38-E49. [PMID: 37991453 PMCID: PMC11193534 DOI: 10.1152/ajpendo.00139.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/27/2023] [Accepted: 11/14/2023] [Indexed: 11/23/2023]
Abstract
Circulating endothelial cell-derived microvesicles (EMVs) have been shown to be elevated with obesity and associated with endothelial dysfunction; however, their direct effect on endothelial cells is unknown. The experimental aim of this study was to determine the effect of EMVs isolated from adults with obesity on endothelial cell inflammation, apoptosis, and nitric oxide (NO) production. EMVs (CD144+ microvesicles) were identified, enumerated, and isolated from plasma by flow cytometry from 24 sedentary adults: 12 normal-weight adults [8 M/4 F; age: 55 ± 6 yr; body mass index (BMI): 24.3 ± 0.7 kg/m2; EMV: 144 ± 53 EMVs/µL] and 12 adults with obesity (6 M/6 F; 59 ± 7 yr; BMI: 31.0 ± 1.1 kg/m2; EMV: 245 ± 89 EMVs/µL). Human umbilical vein endothelial cells were cultured and treated with EMVs from either normal-weight adults or adults with obesity. EMVs from obese adults induced significantly higher release of interleukin (IL)-6 (108.2 ± 7.7 vs. 90.9 ± 10.0 pg/mL) and IL-8 (75.4 ± 9.8 vs. 59.5 ± 11.5 pg/mL) from endothelial cells vs. EMVs from normal-weight adults, concordant with greater intracellular expression of phosphorylated NF-κB p65 (Ser536; active NF-κB) [145.0 ± 34.1 vs. 114.5 ± 30.4 arbitrary units (AU)]. Expression of phosphorylated p38-MAPK (15.4 ± 5.7 vs. 9.2 ± 2.5 AU) and active caspase-3 (168.2 ± 65.5 vs. 107.8 ± 40.5 AU), markers of cell apoptosis, was higher in cells treated with obesity-related EMVs. Phosphorylated endothelial nitric oxide synthase (eNOS) (Ser1177) expression (23.5 ± 7.2 vs. 34.7 ± 9.7 AU) and NO production (6.9 ± 1.4 vs. 8.7 ± 0.7 µmol/L) were significantly lower in the cells treated with EMVs from obese adults. These data indicate that circulating EMVs from adults with obesity promote a proinflammatory, proapoptotic, and NO-compromised endothelial phenotype. Circulating EMVs are a potential mediator of obesity-related endothelial dysfunction.NEW & NOTEWORTHY In the present study, we determined the effect of circulating endothelial cell-derived microvesicles (EMVs) isolated from adults with obesity on endothelial cell inflammation, apoptosis, and nitric oxide (NO) production in vitro. Circulating EMVs harvested from adults with obesity promoted a proinflammatory, proapoptotic, and NO-compromised endothelial phenotype. Elevated circulating EMVs in adults with obesity, independent of other cardiometabolic risk factors, are a potential novel systemic mediator of obesity-related endothelial dysfunction and vascular risk.
Collapse
Affiliation(s)
- Vinicius P Garcia
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Hannah K Fandl
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Jamie G Hijmans
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Auburn R Berry
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Hannah L Cardenas
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Kelly A Stockelman
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Noah M DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - J William Treuth
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
| | - Andrew J Park
- Rocky Mountain Regional Spinal Injury System, Craig Hospital, Englewood, Colorado, United States
- Department of Medicine, University of Colorado Anschutz Medical Center, Denver, Colorado, United States
| | - Brian L Stauffer
- Department of Medicine, University of Colorado Anschutz Medical Center, Denver, Colorado, United States
- Denver Health Medical Center, Denver, Colorado, United States
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado, United States
- Department of Medicine, University of Colorado Anschutz Medical Center, Denver, Colorado, United States
| |
Collapse
|
7
|
Terasawa M, Zang L, Hiramoto K, Shimada Y, Mitsunaka M, Uchida R, Nishiura K, Matsuda K, Nishimura N, Suzuki K. Oral Administration of Rhamnan Sulfate from Monostroma nitidum Suppresses Atherosclerosis in ApoE-Deficient Mice Fed a High-Fat Diet. Cells 2023; 12:2666. [PMID: 37998401 PMCID: PMC10670814 DOI: 10.3390/cells12222666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
Oral administration of rhamnan sulfate (RS), derived from the seaweed Monostroma nitidum, markedly suppresses inflammatory damage in the vascular endothelium and organs of lipopolysaccharide-treated mice. This study aimed to analyze whether orally administered RS inhibits the development of atherosclerosis, a chronic inflammation of the arteries. ApoE-deficient female mice were fed a normal or high-fat diet (HFD) with or without RS for 12 weeks. Immunohistochemical and mRNA analyses of atherosclerosis-related genes were performed. The effect of RS on the migration of RAW264.7 cells was also examined in vitro. RS administration suppressed the increase in blood total cholesterol and triglyceride levels. In the aorta of HFD-fed mice, RS reduced vascular smooth muscle cell proliferation, macrophage accumulation, and elevation of VCAM-1 and inhibited the reduction of Robo4. Increased mRNA levels of Vcam1, Mmp9, and Srebp1 in atherosclerotic areas of HFD-fed mice were also suppressed with RS. Moreover, RS directly inhibited the migration of RAW264.7 cells in vitro. Thus, in HFD-fed ApoE-deficient mice, oral administration of RS ameliorated abnormal lipid metabolism and reduced vascular endothelial inflammation and hyperpermeability, macrophage infiltration and accumulation, and smooth muscle cell proliferation in the arteries leading to atherosclerosis. These results suggest that RS is an effective functional food for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Masahiro Terasawa
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Liqing Zang
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Yasuhito Shimada
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Mari Mitsunaka
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Ryota Uchida
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Kaoru Nishiura
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Koichi Matsuda
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Norihiro Nishimura
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Koji Suzuki
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| |
Collapse
|
8
|
Allbritton-King JD, García-Cardeña G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023; 11:1278166. [PMID: 37965580 PMCID: PMC10642230 DOI: 10.3389/fcell.2023.1278166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The vascular endothelium is a multifunctional cellular system which directly influences blood components and cells within the vessel wall in a given tissue. Importantly, this cellular interface undergoes critical phenotypic changes in response to various biochemical and hemodynamic stimuli, driving several developmental and pathophysiological processes. Multiple studies have indicated a central role of the endothelium in the initiation, progression, and clinical outcomes of cardiac disease. In this review we synthesize the current understanding of endothelial function and dysfunction as mediators of the cardiomyocyte phenotype in the setting of distinct cardiac pathologies; outline existing in vivo and in vitro models where key features of endothelial cell dysfunction can be recapitulated; and discuss future directions for development of endothelium-targeted therapeutics for cardiac diseases with limited existing treatment options.
Collapse
Affiliation(s)
- Jules D. Allbritton-King
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Guillermo García-Cardeña
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
9
|
Pickett JR, Wu Y, Zacchi LF, Ta HT. Targeting endothelial vascular cell adhesion molecule-1 in atherosclerosis: drug discovery and development of vascular cell adhesion molecule-1-directed novel therapeutics. Cardiovasc Res 2023; 119:2278-2293. [PMID: 37595265 PMCID: PMC10597632 DOI: 10.1093/cvr/cvad130] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/14/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) has been well established as a critical contributor to atherosclerosis and consequently as an attractive therapeutic target for anti-atherosclerotic drug candidates. Many publications have demonstrated that disrupting the VCAM-1 function blocks monocyte infiltration into the sub-endothelial space, which effectively prevents macrophage maturation and foam cell transformation necessary for atherosclerotic lesion formation. Currently, most VCAM-1-inhibiting drug candidates in pre-clinical and clinical testing do not directly target VCAM-1 itself but rather down-regulate its expression by inhibiting upstream cytokines and transcriptional regulators. However, the pleiotropic nature of these regulators within innate immunity means that optimizing dosage to a level that suppresses pathological activity while preserving normal physiological function is extremely challenging and oftentimes infeasible. In recent years, highly specific pharmacological strategies that selectively inhibit VCAM-1 function have emerged, particularly peptide- and antibody-based novel therapeutics. Studies in such VCAM-1-directed therapies so far remain scarce and are limited by the constraints of current experimental atherosclerosis models in accurately representing the complex pathophysiology of the disease. This has prompted the need for a comprehensive review that recounts the evolution of VCAM-1-directed pharmaceuticals and addresses the current challenges in novel anti-atherosclerotic drug development.
Collapse
Affiliation(s)
- Jessica R Pickett
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| | - Yuao Wu
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
| | - Lucia F Zacchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), University of Queensland, St. Lucia, QLD 4072, Australia
- School of Chemistry and Molecular Biosciences, the University of Queensland, St. Lucia, QLD 4072, Australia
| | - Hang T Ta
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, West Creek Road, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Kessels Road, Nathan, QLD 4111, Australia
| |
Collapse
|
10
|
Hussain MS, Afzal O, Gupta G, Altamimi ASA, Almalki WH, Alzarea SI, Kazmi I, Kukreti N, Gupta S, Sulakhiya K, Singh SK, Dua K. Probing the links: Long non-coding RNAs and NF-κB signalling in atherosclerosis. Pathol Res Pract 2023; 249:154773. [PMID: 37647827 DOI: 10.1016/j.prp.2023.154773] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease that involves the accumulation of lipids and immune cells in the arterial wall. NF-kB signaling is a key regulator of inflammation and is known to play a critical role in atherosclerosis. Recent studies have shown that lncRNAs can regulate NF-kB and contribute to the development and progression of atherosclerosis. Preliminary findings reveal significant alterations in the expression of specific lncRNAs in atherosclerotic lesions compared to healthy arterial tissue. Experimental evidence suggests that these dysregulated lncRNAs can influence the NF-kB pathway. By unravelling the crosstalk between lncRNAs and NF-kB signaling, this review aims to enhance our understanding of the molecular mechanisms underlying atherosclerosis. Identifying novel therapeutic targets and diagnostic markers may lead to developing interventions and management strategies for this prevalent cardiovascular disease. This review summarizes the current knowledge on the role of lncRNAs in NF-kB signaling in atherosclerosis and highlights their potential as therapeutic targets for this disease.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, 302017 Jaipur, Rajasthan, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura 302017, Jaipur, India; Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Kunjbihari Sulakhiya
- Neuro Pharmacology Research Laboratory (NPRL), Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
11
|
Murphy JM, Jeong K, Tran DTK, Cioffi DL, Campbell PM, Kim JH, Jo H, Ahn EYE, Lim STS. Nuclear FAK in endothelium: An intrinsic inhibitor of NF-κB activation in atherosclerosis. Atherosclerosis 2023; 379:117189. [PMID: 37527611 PMCID: PMC10530536 DOI: 10.1016/j.atherosclerosis.2023.117189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND AIMS Hyperlipidemia leads to the accumulation of oxidized low-density lipoprotein (oxLDL) within the vessel wall where it causes chronic inflammation in endothelial cells (ECs) and drives atherosclerotic lesions. Although focal adhesion kinase (FAK) is critical in proinflammatory NF-κB activation in ECs, it is unknown if hyperlipidemia alters FAK-mediated NF-κB activity in vivo to affect atherosclerosis progression. METHODS We investigated changes in EC FAK and NF-κB activation using Apoe-/- mice fed a western diet (WD). Both pharmacological FAK inhibition and EC-specific FAK inhibited mouse models were utilized. FAK and NF-κB localization and activity were also analyzed in human atherosclerotic samples. RESULTS ECs of hyperlipidemic mice clearly showed much higher levels of FAK activation in the cytoplasm, which was associated with increased NF-κB activation compared to normal diet (ND) group. On the contrary, FAK is mostly localized in the nucleus and inactive in ECs under healthy conditions with a low NF-κB activity. Both pharmacological and EC-specific genetic FAK inhibition in WD fed Apoe-/- mice exhibited a significant decrease in FAK activity and cytoplasmic localization, NF-κB activation, macrophage recruitment, and atherosclerotic lesions compared to the vehicle or FAK wild-type groups. Analyses of human atherosclerotic specimens revealed a positive correlation between increased active cytoplasmic FAK within ECs and NF-κB activation in the lesions. CONCLUSIONS Hyperlipidemic conditions activate NF-κB pathway by increasing EC FAK activity and cytoplasmic localization in mice and human atherosclerotic samples. As FAK inhibition can efficiently reduce vascular inflammation and atherosclerotic lesions in mice by reversing EC FAK localization and NF-κB activation, these findings support a potential use for FAK inhibitors in treating atherosclerosis.
Collapse
Affiliation(s)
- James M Murphy
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kyuho Jeong
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Duyen Thi Kieu Tran
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL, 36688, USA
| | - Pamela Moore Campbell
- Department of Pathology, University of South Alabama College of Medicine, Mobile, AL, 36617, USA
| | - Jin H Kim
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, 36688, USA
| | - Hanjoong Jo
- Department of Bioengineering, Emory University and Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Eun-Young Erin Ahn
- Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ssang-Taek Steve Lim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
12
|
Worapongsatitaya P, Chaikijurajai T, Ponvilawan B, Ungprasert P. Hidradenitis Suppurativa and Risk of Coronary Artery Disease: A Systematic Review and Meta-Analysis. Indian J Dermatol 2023; 68:359-365. [PMID: 37822371 PMCID: PMC10564198 DOI: 10.4103/ijd.ijd_245_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Background Patients with hidradenitis suppurativa (HS) may have a higher risk of coronary artery disease (CAD) due to the excessive inflammatory burden. However, data on this association is still relatively limited. Aims To investigate the association between HS and risk of prevalent and incident CAD by combining result from all available studies using systematic review and meta-analysis technique. Materials and Methods Potentially eligible studies were identified from Medline and EMBASE databases from inception to November 2021 using search strategy that comprised of terms for 'hidradenitis suppurativa' (HS) and 'coronary artery disease' (CAD). Eligible study must be cohort study that consisted of one cohort of patients with HS and another cohort of individuals without HS. The study must report incidence or prevalence of CAD in both groups. The retrieved point estimates with standard errors from each study were summarized into pooled result using random-effect model and generic inverse variance method. Meta-analyses of the prevalent and incident CAD were conducted separately. Results A total of 876 articles were identified. After two rounds of independent review by three investigators, seven cohort studies (four incident studies and three prevalent studies) met the eligibility criteria and were analysed in the meta-analyses. The meta-analysis found a significantly elevated risk of both incident and prevalent CAD in patients with HS compared to individuals without psoriasis with the pooled risk ratio of 1.38 (95% CI, 1.21-1.58; I2 83%) and 1.70 (95% CI, 1.13-2.57; I2 89%), respectively. Limitations Limited accuracy of diagnosis of HS and CSD as most included studies relied on diagnostic codes and high between-study statistical heterogeneity. Conclusions The current systematic review and meta-analysis found a significantly increased risk of both prevalent and incident CAD among patients with HS.
Collapse
Affiliation(s)
- Pitchaya Worapongsatitaya
- From the Internal Medicine Residency Program, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thanat Chaikijurajai
- From the Internal Medicine Residency Program, University of Minnesota Medical School, Minneapolis, MN, USA
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Bangkok, Thailand
| | - Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Patompong Ungprasert
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
13
|
Yuan H, Liu Y, Zhang J, Dong JF, Zhao Z. Transcription factors in megakaryocytes and platelets. Front Immunol 2023; 14:1140501. [PMID: 36969155 PMCID: PMC10034027 DOI: 10.3389/fimmu.2023.1140501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
Transcription factors bind promoter or regulatory sequences of a gene to regulate its rate of transcription. However, they are also detected in anucleated platelets. The transcription factors RUNX1, GATA1, STAT3, NFκB, and PPAR have been widely reported to play key roles in the pathophysiology of platelet hyper-reactivity, thrombosis, and atherosclerosis. These non-transcriptional activities are independent of gene transcription or protein synthesis but their underlying mechanisms of action remain poorly defined. Genetic and acquired defects in these transcription factors are associated with the production of platelet microvesicles that are known to initiate and propagate coagulation and to promote thrombosis. In this review, we summarize recent developments in the study of transcription factors in platelet generation, reactivity, and production of microvesicles, with a focus on non-transcriptional activities of selected transcription factors.
Collapse
Affiliation(s)
- Hengjie Yuan
- Tianjin Institute of Neurology, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- BloodWorks Research Institute, Seattle, WA, United States
| | - Yafan Liu
- Tianjin Institute of Neurology, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Tianjin Institute of Neurology, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-fei Dong
- BloodWorks Research Institute, Seattle, WA, United States
- Division of Hematology, Department of Medicine, University of Washington, School of Medicine, Seattle, WA, United States
- *Correspondence: Zilong Zhao, ; Jing-fei Dong,
| | - Zilong Zhao
- Tianjin Institute of Neurology, Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- BloodWorks Research Institute, Seattle, WA, United States
- *Correspondence: Zilong Zhao, ; Jing-fei Dong,
| |
Collapse
|
14
|
Zhang B, Tian M, Zhu J, Zhu A. Global research trends in atherosclerosis-related NF-κB: a bibliometric analysis from 2000 to 2021 and suggestions for future research. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:57. [PMID: 36819535 PMCID: PMC9929801 DOI: 10.21037/atm-22-6145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
Background Atherosclerosis (AS) is closely related to stroke and cardiovascular diseases. Nuclear factor kappa-B (NF-κB) is the master regulator of inflammation, and thus, modulating the transcription of NF-κB can improve AS. Methods In this study, we conducted a bibliometric analysis to identify the frontiers, hotspots, and features of global research output on NF-κB in AS from 2000 to 2021. Papers published from 2000 to 2021 and the recorded information were retrieved from the Science Citation Index-Expanded of the Web of Science Core Collection. Bibliometric analysis and visualization were performed using VOSviewer and CiteSpace, including an analysis of the general distribution of annual output, highly productive countries, active journals, active institutions and authors, keywords, and co-cited references. Results A total of 5,439 original articles and reviews were retrieved and analyzed, and the results indicated that the annual number of publications on NF-κB in AS has been increasing in waves over the past 22 years. The majority of papers were published in China, while the USA had the highest number of citations and H-index. The most productive affiliation and journal were the University of California System and Arteriosclerosis Thrombosis and Vascular Biology, respectively. The papers of Chiu JJ. received the highest number of citations globally in 2011. The keywords, "nlrp3 inflammasome" and "microRNA", have recently attracted considerable attention, and very frequently occurring keywords included "NF kappa B", "atherosclerosis", "expression", "activation", "endogenous cell", and "oxidative stress". New keywords in 2021 included "muscle", "attenuates atherosclerosis", "mesenchymal transition", "metabolic disorder", and "palmitic acid". Conclusions AS and inflammation have become research hotspots lately. Over the past decade, most studies have focused on basic research, and pathways associated with the regulatory role of NF-κB in AS have become a particular focus in recent studies. Moreover, our study revealed that NF-κB plays a remarkable role in AS and may be a therapeutic target.
Collapse
Affiliation(s)
- Bingyue Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Mengyao Tian
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China
| | - Jinling Zhu
- Zhejiang Engineering Research Center for “Preventive Treatment” Smart Health of Traditional Chinese Medicine, Hangzhou, China
| | - Aisong Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China;,Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China;,Zhejiang Engineering Research Center for “Preventive Treatment” Smart Health of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
15
|
Zhang Z, Qin S, Wang R, Fang Z, Wang Y, Li F. Circ_0003575 knockdown alleviates ox-LDL-induced human aortic endothelial cell dysfunction in atherosclerosis by miR-637/TRAF6 axis. Clin Hemorheol Microcirc 2023; 85:173-187. [PMID: 37599528 DOI: 10.3233/ch-231858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) are involved in the progression of atherosclerosis (AS). The present study aimed to determine the functions and mechanism of circ_0003575 in AS. METHODS Oxidized low-density lipoprotein (ox-LDL) was used to induce human aortic endothelial cells (HAECs) to establish an AS cell model. Cell Counting Kit-8 (CCK-8) assay and 5'-ethynyl-2'-deoxyuridine (EdU) assay were conducted to assess cell proliferation. Flow cytometry analysis was utilized to quantify cell apoptosis. Tube formation assay was performed to analyze angiogenesis ability. Enzyme linked immunosorbent assay (ELISA) was used to examine the concentrations of inflammatory factors. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blot were manipulated for the expression of circ_0003575, microRNA-637 (miR-637) and TNF receptor associated factor 6 (TRAF6). Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were adopted to estimate the downstream targets of circ_0003575. RESULTS Ox-LDL treatment repressed the proliferation and angiogenesis and promoted the apoptosis and inflammation in HAECs. Circ_0003575 knockdown ameliorated ox-LDL-induced injury of HAECs. Circ_0003575 interacted with mi-R-637, which directly targeted TRAF6. Inhibition of miR-637 reversed the impacts of circ_0003575 knockdown on HAEC injury. Moreover, miR-637 overexpression promoted cell proliferation and angiogenesis and inhibited cell apoptosis and inflammation by targeting TRAF6 in ox-LDL-treated HAECs. Further, circ_0003575 silencing inhibited the activation of NF-κB pathway. CONCLUSION Circ_0003575 knockdown alleviated ox-LDL-induced HAEC damage by regulating miR-637/TRAF6 and NF-κB pathways.
Collapse
Affiliation(s)
- Zhanshuai Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| | - Shaoqiang Qin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| | - Rui Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| | - Zhiqin Fang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| | - Yaling Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| | - Fangjiang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou City, Hebei, China
| |
Collapse
|
16
|
Li W, Jin K, Luo J, Xu W, Wu Y, Zhou J, Wang Y, Xu R, Jiao L, Wang T, Yang G. NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front Cardiovasc Med 2022; 9:988266. [PMID: 36204587 PMCID: PMC9530249 DOI: 10.3389/fcvm.2022.988266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis (AS) is a common cardiovascular disease with complex pathogenesis, in which multiple pathways and their interweaving regulatory mechanism remain unclear. The primary transcription factor NF-κB plays a critical role in AS via modulating the expression of a series of inflammatory mediators under various stimuli such as cytokines, microbial antigens, and intracellular stresses. Endoplasmic reticulum (ER) stress, caused by the disrupted synthesis and secretion of protein, links inflammation, metabolic signals, and other cellular processes via the unfolded protein response (UPR). Both NF-κB and ER stress share the intersection regarding their molecular regulation and function and are regarded as critical individual contributors to AS. In this review, we summarize the multiple interactions between NF-κB and ER stress activation, including the UPR, NLRP3 inflammasome, and reactive oxygen species (ROS) generation, which have been ignored in the pathogenesis of AS. Given the multiple links between NF-κB and ER stress, we speculate that the integrated network contributes to the understanding of molecular mechanisms of AS. This review aims to provide an insight into these interactions and their underlying roles in the progression of AS, highlighting potential pharmacological targets against the atherosclerotic inflammatory process.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liqun Jiao,
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Tao Wang,
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- Tao Wang,
| |
Collapse
|
17
|
Meka Farid Babu, A.R. S, Benerji G.V.. 3-Nitrotyrosine (NT) levels in serum and its association with insulin resistance in patients with type 2 diabetes mellitus: Biomarker role of NT in the assessment of oxidative stress mediated impending vascular complications in nephropathy. Biomedicine (Taipei) 2022. [DOI: 10.51248/.v42i4.1433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Introduction and Aim: 3-Nitrotyrosine (NT) has been recognized as a marker of oxidative stress in diabetes mellitus. NT has also been studied in diverse metabolic conditions. The aim of our study was oriented towards the role of NT as a predictor of oxidative stress mediated impending nephropathy in diabetes mellitus and that with reference to albuminuria.
Materials and Methods: A total of 150 type 2 diabetics in the age group 35 - 50 years were enrolled as three groups, comprising 50 each, based on albuminuria. 50 healthy age and gender matched subjects constituted the control group. Serum NT and Insulin were assessed by ELISA. HbA1c was quantitated by immunoturbidimetric method and microalbumin was assessed by turbilatex method. Routine biochemistry was enabled through ERBA EM-200 fully automated analyzer. Stringent quality control was affected. The study was begun following approval accorded by the competent committees.
Results: NT levels were positively correlated with albumin-creatinine ratio and insulin resistance. NT could be used as a predictor of impending vascular complications in diabetic nephropathy.
Conclusion: NT levels could act as a predictor of oxidative stress mediated diabetic nephropathy in the light of albuminuria.
Collapse
|
18
|
van den Hoek AM, Özsezen S, Caspers MPM, van Koppen A, Hanemaaijer R, Verschuren L. Unraveling the Transcriptional Dynamics of NASH Pathogenesis Affecting Atherosclerosis. Int J Mol Sci 2022; 23:8229. [PMID: 35897797 PMCID: PMC9331250 DOI: 10.3390/ijms23158229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/07/2022] [Accepted: 07/22/2022] [Indexed: 12/10/2022] Open
Abstract
The prevalence of non-alcoholic steatohepatitis (NASH) is rapidly increasing and associated with cardiovascular disease (CVD), the major cause of mortality in NASH patients. Although sharing common risk factors, the mechanisms by which NASH may directly contribute to the development to CVD remain poorly understood. The aim of this study is to gain insight into key molecular processes of NASH that drive atherosclerosis development. Thereto, a time-course study was performed in Ldlr-/-.Leiden mice fed a high-fat diet to induce NASH and atherosclerosis. The effects on NASH and atherosclerosis were assessed and transcriptome analysis was performed. Ldlr-/-.Leiden mice developed obesity, hyperlipidemia and insulin resistance, with steatosis and hepatic inflammation preceding atherosclerosis development. Transcriptome analysis revealed a time-dependent increase in pathways related to NASH and fibrosis followed by an increase in pro-atherogenic processes in the aorta. Gene regulatory network analysis identified specific liver regulators related to lipid metabolism (SC5D, LCAT and HMGCR), inflammation (IL1A) and fibrosis (PDGF, COL3A1), linked to a set of aorta target genes related to vascular inflammation (TNFA) and atherosclerosis signaling (CCL2 and FDFT1). The present study reveals pathogenic liver processes that precede atherosclerosis development and identifies hepatic key regulators driving the atherogenic pathways and regulators in the aorta.
Collapse
Affiliation(s)
- Anita M. van den Hoek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (A.v.K.); (R.H.)
| | - Serdar Özsezen
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (S.Ö.); (M.P.M.C.); (L.V.)
| | - Martien P. M. Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (S.Ö.); (M.P.M.C.); (L.V.)
| | - Arianne van Koppen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (A.v.K.); (R.H.)
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, The Netherlands; (A.v.K.); (R.H.)
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (S.Ö.); (M.P.M.C.); (L.V.)
| |
Collapse
|
19
|
Huang Z, Guo S, Fu C, Zhou W, Stalin A, Zhang J, Liu X, Jia S, Wu C, Lu S, Li B, Wu Z, Tan Y, Fan X, Cheng G, Mou Y, Wu J. Identification of molecular mechanisms underlying the therapeutic effects of Xintong granule in coronary artery disease by a network pharmacology and molecular docking approach. Medicine (Baltimore) 2022; 101:e29829. [PMID: 35801781 PMCID: PMC9259182 DOI: 10.1097/md.0000000000029829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Coronary artery disease (CAD) is a cardiovascular disease characterized by atherosclerosis, angiogenesis, thrombogenesis, inflammation, etc. Xintong granule (XTG) is considered a practical therapeutic strategy in China for CAD. Although its therapeutic role in CAD has been reported, the molecular mechanisms of XTG in CAD have not yet been explored. A network pharmacology approach including drug-likeness (DL) evaluation, oral bioavailability (OB) prediction, protein-protein interaction (PPI) network construction and analysis, and Gene Ontology term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses was used to predict the active ingredients, potential targets, and molecular mechanisms of XTG associated with the treatment of CAD. Molecular docking analysis was performed to investigate the interactions between the active compounds and the underlying targets. Fifty-one active ingredients of XTG and 294 CAD-related targets were screened for analysis. Gene Ontology enrichment analysis showed that the therapeutic targets of XTG in CAD are mainly involved in blood circulation and vascular regulation. KEGG pathway analysis indicated that XTG intervenes in CAD mainly through the regulation of fluid shear stress and atherosclerosis, the AGE-RAGE signaling pathway in diabetic complications, and the relaxin signaling pathway. Molecular docking analysis showed that each key active ingredient (quercetin, luteolin, kaempferol, stigmasterol, resveratrol, fisetin, gamma-sitosterol, and beta-sitosterol) of XTG can bind to the core targets of CAD (AKT1, JUN, RELA, MAPK8, NFKB1, EDN1, and NOS3). The present study revealed the CAD treatment-related active ingredients, underlying targets, and potential molecular mechanisms of XTG acting by regulating fluid shear stress and atherosclerosis, AGE-RAGE signaling pathway in diabetic complications, and relaxin signaling pathway.
Collapse
Affiliation(s)
- Zhihong Huang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Guo
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei Zhou
- China-Japan Friendship Hospital, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingyuan Zhang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinkui Liu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Jia
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Bingbing Li
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhishan Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guoliang Cheng
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong Lunan Pharmaceutical Group Co. Ltd., Linyi, China
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yanfang Mou
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Shandong Lunan Pharmaceutical Group Co. Ltd., Linyi, China
| | - Jiarui Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Jiarui Wu (e-mail: )
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The purpose of our review was to evaluate current standards in clinical practice in determining overall cardiac risk in female patients with chronic rheumatologic diseases. We hoped to not only summarize known cardiac manifestations of various chronic rheumatologic diseases but also determine the effectiveness of new risk scores in determining cardiac risk in this patient population. RECENT FINDINGS Chronic rheumatologic diseases have been associated with various cardiac manifestations for some time, with initial studies involving risk of coronary artery disease (CAD) in rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). However, recent studies have shown numerous other cardiac manifestations associated with these and other chronic rheumatologic diseases. Risk scores have been used for several decades to help determine overall cardiac risk in the general population, but these risk scores have notoriously underestimated the risk of cardiac disease in woman and in patients with chronic rheumatologic diseases. These diseases, often with a female predominance, can impact long-term mortality and have devastating consequences if not monitored and treated appropriately. Thus, new risk scores have been developed over the last several years to help improve detection and awareness of cardiac disease in these patients. Novel modified risk scores have found some success at improving the detection of cardiac disease in patients with chronic rheumatologic diseases. Further studies looking at these risk scores need to determine the accuracy of these scores and where they fall short. With the advent of advanced imaging technologies, future risk scores may involve certain imaging-based markers to help guide accurate risk determination.
Collapse
Affiliation(s)
- Tyler Schmidt
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Rekha Mankad
- Department of Cardiovascular Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
21
|
Zhang ML, Zhang MN, Wang WL, Chen H, Wang X, Li X, Li L, Tong F. 17β-estradiol inhibits vascular proliferation and inflammation by reducing NF-κB expression induced by increased crosstalk between KLF5 and ERα. Biochem Biophys Res Commun 2022; 625:181-187. [DOI: 10.1016/j.bbrc.2022.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022]
|
22
|
WU H, ZHANG Z, WANG Y, ZHANG T, QI S, TANG Y, GAO X. Investigation into the Properties of L-5-Methyltetrahydrofolate and Seal Oil as a Potential Atherosclerosis Intervention in Rats. J Nutr Sci Vitaminol (Tokyo) 2022; 68:87-96. [DOI: 10.3177/jnsv.68.87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Hong WU
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Zhengduo ZHANG
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Yuxin WANG
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Tianran ZHANG
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Shaojun QI
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Yanjin TANG
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| | - Xibao GAO
- Department of Physical and Chemical Inspection, School of Public Health, Cheeloo College of Medicine, Shandong University
| |
Collapse
|
23
|
Hong S, Niu M, Meng D, Li A, Dong Q, Zhang J, Tian X, Lu S, Wang Y. High-density lipoprotein reduces microglia activation and protects against experimental autoimmune encephalomyelitis in mice. Int Immunopharmacol 2022; 105:108566. [DOI: 10.1016/j.intimp.2022.108566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/05/2022]
|
24
|
A Review of the Associations Between Obstructive Sleep Apnea and Gestational Diabetes Mellitus and Possible Mechanisms of Disease. Reprod Sci 2022; 30:81-92. [PMID: 35257355 PMCID: PMC9810675 DOI: 10.1007/s43032-022-00904-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 02/25/2022] [Indexed: 01/07/2023]
Abstract
Obstructive sleep apnea (OSA) usually leads to the occurrence of diabetes. Gestational diabetes mellitus (GDM) is a common gestational complication associated with adverse maternal and fetal outcomes. Increasing studies suggest that women with OSA during pregnancy may be at a significantly greater risk of developing GDM. It is crucial to explore the association between OSA and GDM and the mechanisms underlying this association. In this review, we presented a comprehensive literature review of the following: the association between OSA and GDM, the possible mechanisms of this association, and the effects of continuous positive airway pressure (CPAP) on OSA with GDM. The results showed that most authors suggested that there was an association between OSA and GDM. The intermittent hypoxemia (IH) and reduction of slow-wave sleep (SWS) may be the key to this association. IH induces the products of oxidative stress and inflammation as well as dysregulation of the hypothalamic-pituitary-adrenal, which lead to diabetes. In addition, SWS reduction in OSA enhances the inflammation by increasing the inflammatory cytokines, increases the sympathetic activation, and causes changes in leptin level, which result in the development of GDM. Additionally, whether CPAP is beneficial to GDM remains still unclear.
Collapse
|
25
|
Baicalin Alleviates Thrombin-Induced Inflammation in Vascular Smooth Muscle Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5799308. [PMID: 35097121 PMCID: PMC8799346 DOI: 10.1155/2022/5799308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/27/2021] [Indexed: 11/26/2022]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of the arterial intima. As AS represents the most common type of vascular disease, it affects millions of individuals and is a source of high morbidity and mortality rates worldwide. Overwhelming evidence indicates that AS-related inflammation is mediated by proinflammatory cytokines, chemokines, adhesion molecules and inflammatory signaling pathways, with each of these factors being shown to play critical roles during the entire progression of AS. While a number of drugs have been approved for use in the treatment of AS, their benefits are modest, which underscores the urgency for the development of new drug therapies. In part, these deficits in effective drugs can be attributable to the lack of a clear understanding of the molecular mechanisms of AS. In this study, we investigate the capacity for thrombin to trigger inflammation and induce cell proliferation in vascular smooth muscle cells (VSMCs). We then assessed the effects of baicalin and its potential mechanisms on VSMC inflammation as induced by thrombin. Baicalin, which is a natural bioactive compound of S. baicalensis Georgi (SBG), exerted a protective effect against thrombin-induced VSMC inflammation as resulting from the upregulation of PAR-1. This protection as exerted by baicalin appears to reside in its capacity to produce an inhibitory effect on the thrombin-induced activation of the ERK1/2 pathway. These findings suggest that baicalin may be a promising candidate for the treatment of atherosclerosis.
Collapse
|
26
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Hou J, Deng Q, Deng X, Zhong W, Liu S, Zhong Z. MicroRNA-146a-5p alleviates lipopolysaccharide-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production via the regulation of TRAF6 and IRAK1 in human umbilical vein endothelial cells (HUVECs). ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1433. [PMID: 34733985 PMCID: PMC8506750 DOI: 10.21037/atm-21-3903] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 01/02/2023]
Abstract
Background Microribonucleic acids (miRNAs) have an evident role in regulating endothelial inflammation and dysfunction, which characterizes the early stages of atherosclerosis. The NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome has been reported to contribute to the endothelial inflammatory response that promotes atherosclerosis development and progression. This study sought to investigate the effects of miR-146a-5p on lipopolysaccharide (LPS)-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production in human umbilical vein endothelial cells (HUVECs). Methods HUVECs were transfected with a miR-146a-5p mimic, small-interfering RNA (siRNA) (si-TRAF6, and si-IRAK1), and were then stimulated with LPS for 24 h. The messenger (mRNA) and the protein levels of p-NF-κB/NF-κB, NLRP3, Caspase-1, pro-inflammatory cytokine [interleukin (IL)-6, IL-1β and tumor necrosis factor alpha (TNF-α)] in the HUVECs were analyzed by quantitative real-time polymerase chain reactions (PCRs) and western blot assays, respectively. The secretion of IL-6 from the cells was detected by enzyme-linked immunoassay (ELISA). Bioinformatic and dual-luciferase reporter assays were performed to identify the targets of miR-146a-5p. Results LPS promoted pro-inflammatory cytokine expression in a dose-dependent manner and significantly increased the expression levels of p-NF-κB/NF-κB p65, NLRP3, and Caspase-1. After transfection with a miR-146a-5p mimic, or si-TRAF6 or si-IRAK1, we observed that the mRNA and protein levels of NF-κB/p-NF-κB, NLRP3, Caspase-1, and pro-inflammatory cytokine in the HUVECs were all down-regulated, and the secretion of IL-6 from cells declined significantly. After transfection with a miR-146-5p mimic, the expression of TRAF6 and IRAK1 in HUVECs were both down-regulated. Dual-luciferase reporter assays confirmed that miR-146-5p directly targets the 3'-untranslated region (3'-UTR) of TRAF6 and IRAK1 to regulate their expression. Conclusions As a modulator of TRAF6 and IRAK1, miR-146a-5p negatively regulated LPS-induced NF-κB activation and the NLRP3 inflammasome signaling pathway in HUVECs. Thus, miRNA-146a-5p may serve as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Jingyuan Hou
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China
| | - Qiaoting Deng
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Xunwei Deng
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Wei Zhong
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
| | - Sudong Liu
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Zhixiong Zhong
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
| |
Collapse
|
28
|
Nyambuya TM, Dludla PV, Mxinwa V, Nkambule BB. The pleotropic effects of fluvastatin on complement-mediated T-cell activation in hypercholesterolemia. Biomed Pharmacother 2021; 143:112224. [PMID: 34649351 DOI: 10.1016/j.biopha.2021.112224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
T-cells orchestrate the inflammatory responses in atherosclerosis, and their function is modified by the lipoprotein milieu and complement activity. We investigated the effects of fluvastatin on the expression of complement decay-accelerating factor (DAF/CD55) antigen, and the levels of transcription factors in circulating T-cells in hypercholesterolemia. The hypercholesterolemic state was associated with the upregulation of DAF expression on circulating T-cells and increased levels nuclear factor kappa B (NF-kB) and interferon regulatory factor 4 (IRF4). Notably, the elevated levels of DAF and NF-kB expression persisted following treatment with fluvastatin. Therefore, the pleiotropic effects of fluvastatin are partially ascribed to its ability to mediate T-cell activation and regulate complement activity. Consequently, enhanced therapeutic interventions that targets complement-induced T-cell activation may be important in mitigating the development of atherosclerosis and major cardiovascular events in individuals with hypercholesterolemia.
Collapse
Affiliation(s)
- Tawanda Maurice Nyambuya
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek, Namibia.
| | - Phiwayinkosi Vusi Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa.
| | - Vuyolwethu Mxinwa
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - Bongani Brian Nkambule
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
29
|
Pillai SC, Borah A, Le MNT, Kawano H, Hasegawa K, Kumar DS. Co-Delivery of Curcumin and Bioperine via PLGA Nanoparticles to Prevent Atherosclerotic Foam Cell Formation. Pharmaceutics 2021; 13:pharmaceutics13091420. [PMID: 34575496 PMCID: PMC8468720 DOI: 10.3390/pharmaceutics13091420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/20/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cholesterol-rich arterial plaques characterize atherosclerosis, a significant cause of heart disease. Nutraceuticals have received attention over the years, demonstrating potential benefits towards treating and preventing cardiovascular diseases (CVD), including atherosclerosis. Curcumin, a potent polyphenol present in Curcuma longa, has shown remarkable anti-atherosclerotic activity via anti-inflammatory and anti-oxidative properties. The bioavailability and low water solubility of curcumin limit its clinical translational purposes. These issues can be circumvented effectively by nano-drug delivery systems that can target atherosclerotic plaque sites. In this work, we chose to use curcumin and a natural bioenhancer called Bioperine (derived from Piper nigrum) inside a polymeric nano-drug delivery system for targeting atherosclerotic plaque sites. We selected two different ratios of curcumin:Bioperine to study its comparative effect on the inhibition of oxidized low-density lipoprotein (Ox-LDL)-induced foam cell formation. Our studies demonstrated that Cur-Bio PLGA NPs (both ratios) maintained the cell viability in THP-1 monocyte-derived macrophages above 80% at all periods. The 1:0.2:10 ratio of Cur-Bio PLGA NPs at a concentration of 250 μg/mL illustrated an enhanced reduction in the relative cholesterol content in the THP-1-derived foam cells compared to the 1:1:10 ratio. Confocal microscopy analysis also revealed a reduction in macrophage-mediated foam cell formation when administered with both the ratios of Cur-Bio PLGA NPs. Relative fold change in the mRNA expression of the genes involved in the inflammatory pathways in the atherosclerotic process downregulated NF-κB, CCL2/MCP-1, CD-36, and STAT-3 activity while upregulating the SCAR-B1 expression when treated with the Cur-Bio PLGA NPs. This study thus highlights the importance of natural-based compounds towards the therapeutic intervention against atherosclerotic activity when administered as preventive medicine.
Collapse
Affiliation(s)
- Sindhu C. Pillai
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary Science, Toyo University, 2100, Kujirai, Saitama, Kawagoe 350-8585, Japan; (S.C.P.); (A.B.)
| | - Ankita Borah
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary Science, Toyo University, 2100, Kujirai, Saitama, Kawagoe 350-8585, Japan; (S.C.P.); (A.B.)
| | - Minh Nguyen Tuyet Le
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8302, Japan; (M.N.T.L.); (H.K.); (K.H.)
| | - Hiroaki Kawano
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8302, Japan; (M.N.T.L.); (H.K.); (K.H.)
| | - Kouichi Hasegawa
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8302, Japan; (M.N.T.L.); (H.K.); (K.H.)
| | - D. Sakthi Kumar
- Bio-Nano Electronics Research Centre, Graduate School of Interdisciplinary Science, Toyo University, 2100, Kujirai, Saitama, Kawagoe 350-8585, Japan; (S.C.P.); (A.B.)
- Correspondence: ; Tel.: +81-(0)492-39-1636/1375 or +81-(0)90-9964-7605; Fax: +81-(0)366-77-1140
| |
Collapse
|
30
|
Sumransub N, El Jurdi N, Chiraphapphaiboon W, Maakaron JE. Putting function back in dysfunction: Endothelial diseases and current therapies in hematopoietic stem cell transplantation and cellular therapies. Blood Rev 2021; 51:100883. [PMID: 34429234 DOI: 10.1016/j.blre.2021.100883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/16/2021] [Accepted: 08/12/2021] [Indexed: 01/28/2023]
Abstract
Endothelial dysfunction is characterized by altered vascular permeability and prothrombotic, pro-inflammatory phenotypes. Endothelial dysfunction results in end-organ damage and has been associated with diverse disease pathologies. Complications observed after hematopoietic stem cell transplantation (HCT) and chimeric antigen receptor-T cell (CAR-T) therapy for hematologic and neoplastic disorders share overlapping clinical manifestations and there is increasing evidence linking these complications to endothelial dysfunction. Despite advances in supportive care and treatments, end-organ toxicity remains the leading cause of mortality. A new strategy to mitigate endothelial dysfunction could lead to improvement of clinical outcomes for patients. Statins have demonstrated pleiotropic effects of immunomodulatory and endothelial protection by various molecular mechanisms. Recent applications in immune-mediated diseases such as autoimmune disorders, chronic inflammatory conditions, and graft-versus-host disease (GVHD) have shown promising results. In this review, we cover the mechanisms underlying endothelial dysfunction in GVHD and CAR-T cell-related toxicities. We summarize the current knowledge about statins and other agents used as endothelial protectants. We propose further studies using statins for prophylaxis and prevention of end-organ damage related to extensive endothelial dysfunction in HCT and CAR-T.
Collapse
Affiliation(s)
- Nuttavut Sumransub
- Department of Medicine, University of Minnesota, 420 Delaware St. SE MMC 480, Minneapolis, MN 55455, United States of America
| | - Najla El Jurdi
- Department of Medicine, University of Minnesota, 420 Delaware St. SE MMC 480, Minneapolis, MN 55455, United States of America
| | - Wannasiri Chiraphapphaiboon
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd, Bangkok-Noi, Bangkok 10700, Thailand
| | - Joseph E Maakaron
- Department of Medicine, University of Minnesota, 420 Delaware St. SE MMC 480, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
31
|
Role of Histone Deacetylases in Monocyte Function in Health and Chronic Inflammatory Diseases. Rev Physiol Biochem Pharmacol 2021; 180:1-47. [PMID: 33974124 DOI: 10.1007/112_2021_59] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Histone deacetylases (HDACs) are a family of 18 members that participate in the epigenetic regulation of gene expression. In addition to histones, some HDACs also deacetylate transcription factors and specific cytoplasmic proteins.Monocytes, as part of the innate immune system, maintain tissue homeostasis and help fight infections and cancer. In these cells, HDACs are involved in multiple processes including proliferation, migration, differentiation, inflammatory response, infections, and tumorigenesis. Here, a systematic description of the role that most HDACs play in these functions is reviewed. Specifically, some HDACs induce a pro-inflammatory response and play major roles in host defense. Conversely, other HDACs reprogram monocytes and macrophages towards an immunosuppressive phenotype. The right balance between both types helps monocytes to respond correctly to the different physiological/pathological stimuli. However, aberrant expressions or activities of specific HDACs are associated with autoimmune diseases along with other chronic inflammatory diseases, infections, or cancer.This paper critically reviews the interesting and extensive knowledge regarding the role of some HDACs in these pathologies. It also shows that as yet, very little progress has been made toward the goal of finding effective HDAC-targeted therapies. However, given their obvious potential, we conclude that it is worth the effort to develop monocyte-specific drugs that selectively target HDAC subtypes with the aim of finding effective treatments for diseases in which our innate immune system is involved.
Collapse
|
32
|
Cai X, Sha F, Zhao C, Zheng Z, Zhao S, Zhu Z, Zhu H, Chen J, Chen Y. Synthesis and anti-inflammatory activity of novel steroidal chalcones with 3β-pregnenolone ester derivatives in RAW 264.7 cells in vitro. Steroids 2021; 171:108830. [PMID: 33836205 DOI: 10.1016/j.steroids.2021.108830] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/05/2021] [Accepted: 03/28/2021] [Indexed: 02/05/2023]
Abstract
To identify new potential anti-inflammatory agents, we herein report the synthesis of novel steroidal chalcones with 3β-pregnenolone esters of cinnamic acid derivatives using pregnenolone as the starting material. The structures of the newly synthesised compounds were confirmed by 1H NMR, 13C NMR, HRMS and infrared imaging. All the derivatives were examined to determine their in vitro anti-inflammatory profiles against LPS-induced inflammation in RAW 264.7 cells; the derivates were evaluated by the quantification of the pro-inflammatory mediator nitric oxide (NO) in the cell culture supernatant based on the Griess reaction, which measures nitrite levels, followed by an in vitro cytotoxicity study. Among these novel derivatives, compound 11e [3β-3-phenyl acrylate-pregn-5-en-17β-yl-3' -(p-fluoro)-phenylprop-2'-en-1'-one] was identified as the most potent anti-inflammatory agent, which showed significant anti-inflammatory activity by inhibiting the LPS-induced pro-inflammatory mediator NO in a dose-dependent manner without any cytotoxicity. Moreover, compound 11e markedly inhibited the expression of pro-inflammatory cytokines, including inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), tumour necrosis factor-α (TNF-α), and cyclooxygenase-2 (COX-2), in LPS-induced RAW 264.7 cells. Further studies confirmed that compound 11e significantly suppressed the transcriptional activity of NF-κB in activated RAW 264.7 cells. Molecular docking study revealed the strong binding affinity of compound 11e to the active site of the pro-inflammatory proteins, which confirmed that compound 11e acted as an anti-inflammatory mediator. These results indicated that steroidal chalcones with 3β-pregnenolone esters of cinnamic acid derivatives might be considered for further research in the design of anti-inflammatory drugs, and compound 11e might be a promising therapeutic anti-inflammatory drug candidate.
Collapse
Affiliation(s)
- Xiaorui Cai
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Fei Sha
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Chuanyi Zhao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhiwei Zheng
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Shulin Zhao
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Zhiwei Zhu
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Huide Zhu
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Jiaoling Chen
- Department of Pharmacy, The Affiliated Tumor Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yicun Chen
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
33
|
Poles J, Karhu E, McGill M, McDaniel HR, Lewis JE. The effects of twenty-four nutrients and phytonutrients on immune system function and inflammation: A narrative review. J Clin Transl Res 2021; 7:333-376. [PMID: 34239993 PMCID: PMC8259612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/10/2021] [Accepted: 04/27/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND AND AIM Recently, optimal immune function has become a primary focus of worldwide attention not only in the prevention of chronic disease but also as one strategy to reduce the severity of acute illness. Inflammation, a process largely controlled by the immune system, has long been studied and recognized for its role in chronic disease. Optimizing immune function or managing inflammation using individual nutrients and phytonutrients is not well understood by the average person. Thus, this narrative literature review summarizes many of the more recent findings about how certain nutrients and phytonutrients affect immune function and inflammation, and how they may best be utilized considering the growing worldwide interest in this topic. METHODS A comprehensive literature search of PubMed was performed to find clinical trials in humans that assessed the effect of nutrients and phytonutrients on immune function and inflammation, in individuals with acute and chronic health conditions, published in English between 2000 and 2020. Two independent reviewers evaluated the articles for their inclusion. RESULTS Eighty-seven articles were summarized in this narrative review. In total 24 nutrients and phytonutrients were included in the study, that is, acetyl-L-carnitine, Aloe vera polysaccharides, beta-glucans, bilberry, black seed oil, coenzyme Q10, curcumin (turmeric), frankincense, garlic, ginger, hydrolyzed rice bran, isoflavones, lipoic acid, mistletoe, N-acetyl cysteine, omega-3 fatty acids, resveratrol, selenium, shiitake mushroom and its derivatives, Vitamin B12, Vitamin C, Vitamin D3 (cholecalciferol), Vitamin E (d-alpha- and gamma-tocopherol), and zinc. Some of the noteworthy immune function and anti-inflammatory responses to these interventions included modulation of nuclear factor-Kappa B, tumor necrosis factor-a, interferon-g, interleukin-6, and CD4+ T cells, among others. These findings are not completely consistent or ubiquitous across all patient populations or health status. CONCLUSIONS Based on this review, many nutrients and phytonutrients are capable of significantly modulating immune function and reducing inflammation, according to multiple biomarkers in clinical trials in different populations of adults with varying health statuses. Thus, dietary supplementation may serve as an adjunct to conventional pharmaceutical or medical therapies, but evaluation of risks and benefits for each person and health status is necessary. Additional larger studies are also needed to investigate the safety and efficacy of nutritional compounds in various health conditions, with emphases on potential drug-supplement interactions and clinical endpoints. RELEVANCE FOR PATIENTS As demonstrated in the reviewed clinical trials, patients of various health challenges with a wide range of severity may benefit from select nutrients and phytonutrients to improve their immune function and reduce inflammation.
Collapse
Affiliation(s)
- Jillian Poles
- Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elisa Karhu
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Megan McGill
- Department of Internal Medicine, Mount Sinai Medical Center Miami Beach, FL, USA
| | | | - John E. Lewis
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
34
|
Ge J, Guo K, Zhang C, Talukder M, Lv MW, Li JY, Li JL. Comparison of nanoparticle-selenium, selenium-enriched yeast and sodium selenite on the alleviation of cadmium-induced inflammation via NF-kB/IκB pathway in heart. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 773:145442. [PMID: 33940727 DOI: 10.1016/j.scitotenv.2021.145442] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/09/2021] [Accepted: 01/22/2021] [Indexed: 06/12/2023]
Abstract
Cadmium (Cd) has been confirmed as an environmental contaminant, which potential threats health impacts to humans and animals. Selenium (Se) as a beneficial element that alleviates the negative effects of Cd toxicity. Se mainly exists in two forms in food nutrients including organic Se usually as (Se-enriched yeast (SeY)) and inorganic Se (sodium selenite (SSe)). Nanoparticle of Se (Nano-Se), a new form Se, which is synthesized by the bioreduction of Se species, which attracted significant attention recently. However, compared the superiority alleviation effects of Nano-Se, SeY or SSe on Cd-induced toxicity and related mechanisms are still poorly understood. The purpose of this study was to compare the superiority antagonism effects of Nano-Se, SeY and SSe on Cd-induced inflammation response via NF-kB/IκB pathway in the heart. The present study demonstrated that exposed to Cd obviously increased the accumulation of Cd, disruption of ion homeostasis and depressed the ratios of K+/Na+ and Mg2+/Ca2+ via ion chromatography mass spectrometry (ICP-MS) detecting the heart specimens. In the results of histological and ultrastructure observation, typical inflammatory infiltrate characteristics and mitochondria and nuclear structure alterations in the hearts of Cd group were confirmed. Cd treatment enhanced the inducible nitric oxide synthase (iNOS) activities and NOS isoforms expression via NF-kB/IκB pathway to promote inflammation response. However, the combined treatment of Cd-exposed animals with Nano-Se was more effective than SeY and SSe in reversing Cd-induced histopathological changes and iNOS activities increased, reducing Cd accumulation and antagonizing Cd-triggered inflammation response via NF-kB/IκB pathway in chicken hearts. Overall, Se applications, especially Nano-Se, can be most efficiently used for relieving cardiotoxicity by exposed to Cd compared to other Se compound.
Collapse
Affiliation(s)
- Jing Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Kai Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Chifeng Animal Health Supervision Institute, Chifeng County 024000, PR China
| | - Cong Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, 450046 Zhengzhou, Henan, PR China
| | - Milton Talukder
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
| | - Mei-Wei Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Yang Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
35
|
Alfaidi M, Scott ML, Orr AW. Sinner or Saint?: Nck Adaptor Proteins in Vascular Biology. Front Cell Dev Biol 2021; 9:688388. [PMID: 34124074 PMCID: PMC8187788 DOI: 10.3389/fcell.2021.688388] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 12/28/2022] Open
Abstract
The Nck family of modular adaptor proteins, including Nck1 and Nck2, link phosphotyrosine signaling to changes in cytoskeletal dynamics and gene expression that critically modulate cellular phenotype. The Nck SH2 domain interacts with phosphotyrosine at dynamic signaling hubs, such as activated growth factor receptors and sites of cell adhesion. The Nck SH3 domains interact with signaling effectors containing proline-rich regions that mediate their activation by upstream kinases. In vascular biology, Nck1 and Nck2 play redundant roles in vascular development and postnatal angiogenesis. However, recent studies suggest that Nck1 and Nck2 differentially regulate cell phenotype in the adult vasculature. Domain-specific interactions likely mediate these isoform-selective effects, and these isolated domains may serve as therapeutic targets to limit specific protein-protein interactions. In this review, we highlight the function of the Nck adaptor proteins, the known differences in domain-selective interactions, and discuss the role of individual Nck isoforms in vascular remodeling and function.
Collapse
Affiliation(s)
- Mabruka Alfaidi
- Department of Pathology and Translational Pathobiology, Louisiana State University Health - Shreveport, Shreveport, LA, United States
| | - Matthew L Scott
- Department of Pathology and Translational Pathobiology, Louisiana State University Health - Shreveport, Shreveport, LA, United States
| | - Anthony Wayne Orr
- Department of Pathology and Translational Pathobiology, Louisiana State University Health - Shreveport, Shreveport, LA, United States.,Department of Cell Biology and Anatomy, LSU Health - Shreveport, Shreveport, LA, United States.,Department of Molecular & Cellular Physiology, LSU Health - Shreveport, Shreveport, LA, United States
| |
Collapse
|
36
|
Li X, Liu L, Li T, Liu M, Wang Y, Ma H, Mu N, Wang H. SIRT6 in Senescence and Aging-Related Cardiovascular Diseases. Front Cell Dev Biol 2021; 9:641315. [PMID: 33855020 PMCID: PMC8039379 DOI: 10.3389/fcell.2021.641315] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/11/2021] [Indexed: 12/14/2022] Open
Abstract
SIRT6 belongs to the nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases and has established diverse roles in aging, metabolism and disease. Its function is similar to the Silent Information Regulator 2 (SIR2), which prolongs lifespan and regulates genomic stability, telomere integrity, transcription, and DNA repair. It has been demonstrated that increasing the sirtuin level through genetic manipulation extends the lifespan of yeast, nematodes and flies. Deficiency of SIRT6 induces chronic inflammation, autophagy disorder and telomere instability. Also, these cellular processes can lead to the occurrence and progression of cardiovascular diseases (CVDs), such as atherosclerosis, hypertrophic cardiomyopathy and heart failure. Herein, we discuss the implications of SIRT6 regulates multiple cellular processes in cell senescence and aging-related CVDs, and we summarize clinical application of SIRT6 agonists and possible therapeutic interventions in aging-related CVDs.
Collapse
Affiliation(s)
- Xiaokang Li
- Department of Cardiology, China Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lin Liu
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian Li
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Haiyan Wang
- Department of Cardiology, China Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
37
|
Brewster LM, Bain AR, Garcia VP, Fandl HK, Stone R, DeSouza NM, Greiner JJ, Tymko MM, Vizcardo-Galindo GA, Figueroa-Mujica RJ, Villafuerte FC, Ainslie PN, DeSouza CA. Global REACH 2018: dysfunctional extracellular microvesicles in Andean highlander males with excessive erythrocytosis. Am J Physiol Heart Circ Physiol 2021; 320:H1851-H1861. [PMID: 33710927 DOI: 10.1152/ajpheart.00016.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
High altitude-related excessive erythrocytosis (EE) is associated with increased cardiovascular risk. The experimental aim of this study was to determine the effects of microvesicles isolated from Andean highlanders with EE on endothelial cell inflammation, oxidative stress, apoptosis, and nitric oxide (NO) production. Twenty-six male residents of Cerro de Pasco, Peru (4,340 m), were studied: 12 highlanders without EE (age: 40 ± 4 yr; BMI: 26.4 ± 1.7; Hb: 17.4 ± 0.5 g/dL, Spo2: 86.9 ± 1.0%) and 14 highlanders with EE (43 ± 4 yr; 26.2 ± 0.9; 24.4 ± 0.4 g/dL; 79.7 ± 1.6%). Microvesicles were isolated, enumerated, and collected from plasma by flow cytometry. Human umbilical vein endothelial cells were cultured and treated with microvesicles from highlanders without and with EE. Microvesicles from highlanders with EE induced significantly higher release of interleukin (IL)-6 (89.8 ± 2.7 vs. 77.1 ± 1.9 pg/mL) and IL-8 (62.0 ± 2.7 vs. 53.3 ± 2.2 pg/mL) compared with microvesicles from healthy highlanders. Although intracellular expression of total NF-κB p65 (65.3 ± 6.0 vs. 74.9 ± 7.8.9 AU) was not significantly affected in cells treated with microvesicles from highlanders without versus with EE, microvesicles from highlanders with EE resulted in an ∼25% higher (P < 0.05) expression of p-NF-κB p65 (173.6 ± 14.3 vs. 132.8 ± 12.2 AU). Cell reactive oxygen species production was significantly higher (76.4.7 ± 5.4 vs. 56.7 ± 1.7% of control) and endothelial nitric oxide synthase (p-eNOS) activation (231.3 ± 15.5 vs. 286.6 ± 23.0 AU) and NO production (8.3 ± 0.6 vs. 10.7 ± 0.7 μM/L) were significantly lower in cells treated with microvesicles from highlanders with versus without EE. Cell apoptotic susceptibility was not significantly affected by EE-related microvesicles. Circulating microvesicles from Andean highlanders with EE increased endothelial cell inflammation and oxidative stress and reduced NO production.NEW & NOTEWORTHY In this study, we determined the effects of microvesicles isolated from Andean highlanders with excessive erythrocytosis (EE) on endothelial cell inflammation, oxidative stress, apoptosis, and NO production. Microvesicles from highlanders with EE induced a dysfunctional response from endothelial cells characterized by increased cytokine release and expression of active nuclear factor-κB and reduced nitric oxide production. Andean highlanders with EE exhibit dysfunctional circulating extracellular microvesicles that induce a proinflammatory, proatherogenic endothelial phenotype.
Collapse
Affiliation(s)
- L Madden Brewster
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - Anthony R Bain
- Department of Kinesiology, University of Windsor, Windsor, Ontario, Canada
| | - Vinicius P Garcia
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - Hannah K Fandl
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | - Rachel Stone
- Department of Kinesiology, University of Windsor, Windsor, Ontario, Canada
| | - Noah M DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado.,Faculty of Health and Social Development, Centre for Heart, Lung and Vascular Health, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Jared J Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| | | | | | | | | | - Philip N Ainslie
- Faculty of Health and Social Development, Centre for Heart, Lung and Vascular Health, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Christopher A DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, Boulder, Colorado
| |
Collapse
|
38
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
39
|
Insulin Receptor Substrate p53 Ameliorates High-Glucose-Induced Activation of NF- κB and Impaired Mobility of HUVECs. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3210586. [PMID: 33506012 PMCID: PMC7806382 DOI: 10.1155/2021/3210586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/20/2020] [Accepted: 12/24/2020] [Indexed: 01/14/2023]
Abstract
Diabetes-related macrovascular and microvascular complications lead to poor prognosis. Insulin receptor substrate p53 (IRSp53) is known to act as a substrate for the insulin receptor tyrosine kinase, but its role in endothelial dysfunction remains unclear. Human umbilical vein endothelial cells (HUVECs) treated with D-glucose at different concentrations and a streptozocin-induced rat diabetes mellitus (DM) model were used to investigate the effects of hyperglycemia on the expression levels of IRSp53 and galectin-3 (gal-3) and the inflammatory state and mobility of HUVECs. Thereafter, IRSp53-overexpressing HUVECs and IRSp53-knockdown HUVECs were established using IRSp53-overexpressing lentivirus or IRSp53-siRNA to explore the role of IRSp53 in the HUVEC inflammatory state and HUVEC mobility. D-glucose at high concentration (HG) and hyperglycemia were found to induce downregulation of IRSp53 and upregulation of gal-3 in vitro and in vivo. Treatment with HG resulted in activation of NF-κB in HUVECs and impaired HUVEC mobility. Insulin restored HG-induced changes in the expression levels of IRSp53 and gal-3 in HUVECs and protected the cells from NF-κB activation and impaired mobility. Overexpression of IRSp53 inhibited the activation of NF-κB in HUVECs and strengthened HUVEC migration. Knockdown of IRSp53 facilitated the activation of NF-κB in HUVECs and decreased HUVEC migration. However, neither overexpression nor knockdown of IRSp53 altered the effects of insulin on HG-induced detrimental changes in HUVECs. HG and hyperglycemia resulted in downregulation of IRSp53 in vitro and in vivo. IRSp53 is concluded to inhibit the activation of NF-κB in HUVECs and to strengthen HUVEC migration.
Collapse
|
40
|
Karunakaran D, Nguyen MA, Geoffrion M, Vreeken D, Lister Z, Cheng HS, Otte N, Essebier P, Wyatt H, Kandiah JW, Jung R, Alenghat FJ, Mompeon A, Lee R, Pan C, Gordon E, Rasheed A, Lusis AJ, Liu P, Matic LP, Hedin U, Fish JE, Guo L, Kolodgie F, Virmani R, van Gils JM, Rayner KJ. RIPK1 Expression Associates With Inflammation in Early Atherosclerosis in Humans and Can Be Therapeutically Silenced to Reduce NF-κB Activation and Atherogenesis in Mice. Circulation 2020; 143:163-177. [PMID: 33222501 DOI: 10.1161/circulationaha.118.038379] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic activation of the innate immune system drives inflammation and contributes directly to atherosclerosis. We previously showed that macrophages in the atherogenic plaque undergo RIPK3 (receptor-interacting serine/threonine-protein kinase 3)-MLKL (mixed lineage kinase domain-like protein)-dependent programmed necroptosis in response to sterile ligands such as oxidized low-density lipoprotein and damage-associated molecular patterns and that necroptosis is active in advanced atherosclerotic plaques. Upstream of the RIPK3-MLKL necroptotic machinery lies RIPK1 (receptor-interacting serine/threonine-protein kinase 1), which acts as a master switch that controls whether the cell undergoes NF-κB (nuclear factor κ-light-chain-enhancer of activated B cells)-dependent inflammation, caspase-dependent apoptosis, or necroptosis in response to extracellular stimuli. We therefore set out to investigate the role of RIPK1 in the development of atherosclerosis, which is driven largely by NF-κB-dependent inflammation at early stages. We hypothesize that, unlike RIPK3 and MLKL, RIPK1 primarily drives NF-κB-dependent inflammation in early atherogenic lesions, and knocking down RIPK1 will reduce inflammatory cell activation and protect against the progression of atherosclerosis. METHODS We examined expression of RIPK1 protein and mRNA in both human and mouse atherosclerotic lesions, and used loss-of-function approaches in vitro in macrophages and endothelial cells to measure inflammatory responses. We administered weekly injections of RIPK1 antisense oligonucleotides to Apoe-/- mice fed a cholesterol-rich (Western) diet for 8 weeks. RESULTS We find that RIPK1 expression is abundant in early-stage atherosclerotic lesions in both humans and mice. Treatment with RIPK1 antisense oligonucleotides led to a reduction in aortic sinus and en face lesion areas (47.2% or 58.8% decrease relative to control, P<0.01) and plasma inflammatory cytokines (IL-1α [interleukin 1α], IL-17A [interleukin 17A], P<0.05) in comparison with controls. RIPK1 knockdown in macrophages decreased inflammatory genes (NF-κB, TNFα [tumor necrosis factor α], IL-1α) and in vivo lipopolysaccharide- and atherogenic diet-induced NF-κB activation. In endothelial cells, knockdown of RIPK1 prevented NF-κB translocation to the nucleus in response to TNFα, where accordingly there was a reduction in gene expression of IL1B, E-selectin, and monocyte attachment. CONCLUSIONS We identify RIPK1 as a central driver of inflammation in atherosclerosis by its ability to activate the NF-κB pathway and promote inflammatory cytokine release. Given the high levels of RIPK1 expression in human atherosclerotic lesions, our study suggests RIPK1 as a future therapeutic target to reduce residual inflammation in patients at high risk of coronary artery disease.
Collapse
Affiliation(s)
- Denuja Karunakaran
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Dianne Vreeken
- Leiden University Medical Center, The Netherlands (D.V., J.M.v.G.)
| | - Zachary Lister
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Henry S Cheng
- Toronto General Research Hospital Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (H.S.C.)
| | - Nicola Otte
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Patricia Essebier
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Joshua W Kandiah
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Jung
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Francis J Alenghat
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Ana Mompeon
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Richard Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Carlsbad, CA (R.L.)
| | - Calvin Pan
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Emma Gordon
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (D.K., N.O., P.E., E.G.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Aldons J Lusis
- David Geffen School of Medicine, University of California Los Angeles (C.P., A.J.L.)
| | - Peter Liu
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.)
| | - Ljubica Perisic Matic
- Vascular Surgery Division, Department of Molecular Medicine and Surgery, Karolinska Institute, Sweden (L.P.M.)
| | | | - Jason E Fish
- Cardiology, Department of Medicine, University of Chicago, IL (F.J.A., J.E.F.)
| | - Liang Guo
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Frank Kolodgie
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | - Renu Virmani
- CVPath Institute Inc., Gaithersburg, MD (L.G., F.K., R.V.)
| | | | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (D.K., M.-A.N., M.G., Z.L., H.W., J.W.K., R.J., A.M., A.R., P.L., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ontario, Canada (M.-A.N., K.J.R.)
| |
Collapse
|
41
|
Al-Yafeai Z, Pearson BH, Peretik JM, Cockerham ED, Reeves KA, Bhattarai U, Wang D, Petrich BG, Orr AW. Integrin affinity modulation critically regulates atherogenic endothelial activation in vitro and in vivo. Matrix Biol 2020; 96:87-103. [PMID: 33157226 DOI: 10.1016/j.matbio.2020.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/30/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022]
Abstract
While vital to platelet and leukocyte adhesion, the role of integrin affinity modulation in adherent cells remains controversial. In endothelial cells, atheroprone hemodynamics and oxidized lipoproteins drive an increase in the high affinity conformation of α5β1 integrins in endothelial cells in vitro, and α5β1 integrin inhibitors reduce proinflammatory endothelial activation to these stimuli in vitro and in vivo. However, the importance of α5β1 integrin affinity modulation to endothelial phenotype remains unknown. We now show that endothelial cells (talin1 L325R) unable to induce high affinity integrins initially adhere and spread but show significant defects in nascent adhesion formation. In contrast, overall focal adhesion number, area, and composition in stably adherent cells are similar between talin1 wildtype and talin1 L325R endothelial cells. However, talin1 L325R endothelial cells fail to induce high affinity α5β1 integrins, fibronectin deposition, and proinflammatory responses to atheroprone hemodynamics and oxidized lipoproteins. Inducing the high affinity conformation of α5β1 integrins in talin1 L325R endothelial cells suggest that NF-κB activation and maximal fibronectin deposition require both integrin activation and other integrin-independent signaling. In endothelial-specific talin1 L325R mice, atheroprone hemodynamics fail to promote inflammation and macrophage recruitment, demonstrating a vital role for integrin activation in regulating endothelial phenotype.
Collapse
Affiliation(s)
- Zaki Al-Yafeai
- Departments of Molecular and Cellular Physiology, LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Brenna H Pearson
- Departments of Molecular and Cellular Physiology, LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Jonette M Peretik
- Pathology and Translational Pathobiology,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Elizabeth D Cockerham
- Pathology and Translational Pathobiology,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Kaylea A Reeves
- Pathology and Translational Pathobiology,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Umesh Bhattarai
- Departments of Molecular and Cellular Physiology, LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Dongdong Wang
- Pathology and Translational Pathobiology,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States
| | - Brian G Petrich
- Department of Pediatrics, Shreveport, LA, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - A Wayne Orr
- Departments of Molecular and Cellular Physiology, LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States.; Cell Biology and Anatomy,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States.; Pathology and Translational Pathobiology,LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States.; Department of Pathology and Translational Pathobiology, 1501 Kings Hwy, Biomedical Research Institute, Rm. 6-21, LSU Health Sciences Center - Shreveport, Shreveport, LA 71130, United States.
| |
Collapse
|
42
|
Shafi O. Switching of vascular cells towards atherogenesis, and other factors contributing to atherosclerosis: a systematic review. Thromb J 2020; 18:28. [PMID: 33132762 PMCID: PMC7592591 DOI: 10.1186/s12959-020-00240-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Onset, development and progression of atherosclerosis are complex multistep processes. Many aspects of atherogenesis are not yet properly known. This study investigates the changes in vasculature that contribute to switching of vascular cells towards atherogenesis, focusing mainly on ageing. Methods Databases including PubMed, MEDLINE and Google Scholar were searched for published articles without any date restrictions, involving atherogenesis, vascular homeostasis, aging, gene expression, signaling pathways, angiogenesis, vascular development, vascular cell differentiation and maintenance, vascular stem cells, endothelial and vascular smooth muscle cells. Results Atherogenesis is a complex multistep process that unfolds in a sequence. It is caused by alterations in: epigenetics and genetics, signaling pathways, cell circuitry, genome stability, heterotypic interactions between multiple cell types and pathologic alterations in vascular microenvironment. Such alterations involve pathological changes in: Shh, Wnt, NOTCH signaling pathways, TGF beta, VEGF, FGF, IGF 1, HGF, AKT/PI3K/ mTOR pathways, EGF, FOXO, CREB, PTEN, several apoptotic pathways, ET - 1, NF-κB, TNF alpha, angiopoietin, EGFR, Bcl - 2, NGF, BDNF, neurotrophins, growth factors, several signaling proteins, MAPK, IFN, TFs, NOs, serum cholesterol, LDL, ephrin, its receptor pathway, HoxA5, Klf3, Klf4, BMPs, TGFs and others.This disruption in vascular homeostasis at cellular, genetic and epigenetic level is involved in switching of the vascular cells towards atherogenesis. All these factors working in pathologic manner, contribute to the development and progression of atherosclerosis. Conclusion The development of atherosclerosis involves the switching of gene expression towards pro-atherogenic genes. This happens because of pathologic alterations in vascular homeostasis. When pathologic alterations in epigenetics, genetics, regulatory genes, microenvironment and vascular cell biology accumulate beyond a specific threshold, then the disease begins to express itself phenotypically. The process of biological ageing is one of the most significant factors in this aspect as it is also involved in the decline in homeostasis, maintenance and integrity.The process of atherogenesis unfolds sequentially (step by step) in an interconnected loop of pathologic changes in vascular biology. Such changes are involved in 'switching' of vascular cells towards atherosclerosis.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
43
|
Guanxinshutong Alleviates Atherosclerosis by Suppressing Oxidative Stress and Proinflammation in ApoE -/- Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1219371. [PMID: 33014098 PMCID: PMC7519182 DOI: 10.1155/2020/1219371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 01/01/2023]
Abstract
Atherosclerosis (AS) is a chronic progressive disease related to dyslipidemia, inflammation, and oxidative stress. Guanxinshutong capsule (GXST), a traditional Chinese medicine, has been widely used in treating coronary atherosclerotic heart disease, while its mechanism actions on AS are still not to be well addressed. Our present study is aimed to examine the effect of GXST on AS and elucidate the multitarget mechanisms of GXST on AS. Network pharmacology analysis was employed to screen the multitarget mechanisms of GXST on AS. ApoE−/− mice were used to validate these effects. Circulating lipid profile and oxidative stress-related factors were measured by the Elisa kit. Furthermore, the aortic trunk and aortic root were excised for oil red O staining, histopathological and immunohistochemical analysis. We first discovered that GXST was clued to exert synergistically antiatherosclerosis properties including lipid-lowering, anti-inflammation, and antioxidation through the computational prediction based on a network pharmacology simulation. Next, the validation experiments in atherosclerosis mice provided evidence that GXST significantly reduced atherosclerotic lesions, increased collagen deposition, and attenuated LV remodeling to some extent. Mechanistically, GXST modulated lipid profile, downregulated the level of inflammatory cytokines and NF-κBp65. GXST also reduced the activity of oxidative parameter MDA and upregulated the activities of antioxidant enzymes (SOD and GSH) compared with the AS model group. In conclusion, GXST intervention might attenuate atherosclerosis by mechanisms involving reducing lipid deposition, modulating oxidative stress and inflammatory responses, but a larger controlled trial is necessary for confirmation.
Collapse
|
44
|
Azizoğlu M, Ayaz L, Bayrak G, Yılmaz BC, Birbiçer H, Doruk N. Evaluation of miRNAs Related with Nuclear Factor Kappa B Pathway in Lipopolysaccharide Induced Acute Respiratory Distress Syndrome. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:130-139. [PMID: 32934950 PMCID: PMC7489110 DOI: 10.22088/ijmcm.bums.9.2.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/15/2020] [Indexed: 01/28/2023]
Abstract
This study aimed to determine the expression of nuclear factor kappa B (NF-κB) pathway related miRNAs in experimental acute respiratory distress syndrome (ARDS) induced by lipopolysaccharide (LPS) in rats, and to elucidate the underlying molecular mechanism. Twenty four sprague dawley rats were randomly divided into two groups; LPS (n = 12) and control (n = 12). Experimental ARDS was induced by intraperitoneal injection of E. coli LPS in LPS group. Intraperitoneal saline was administered in control group. Serum and lung samples were collected from both groups. Immunohistochemistry staining was performed for interleukin 1β (IL-1β), tumor necrosis factor α (TNF-α), CD 68, and caspase-3 in lung samples. Intensity of staining was scored as strong, moderate, weak, and no for evaluation of IL-1β and TNF-α. In addition, caspase-3 and CD68-positive stained cells were counted in sections. Expressions of 9 miRNAs were determined by quantitative real-time PCR in serum samples. IL-1β and TNF-α staining scores were significantly higher in the LPS group in comparison with the control group (P = 0.04 and P = 0.02, respectively). In addition, caspase-3 and CD68-positive stained cells were significantly higher in the LPS group (P = 0.02). Expressions of seven miRNAs were significantly changed in the LPS group in comparison with the control group. While six miRNAs (miR-7a-5p, miR-7b, miR-9a-5p, miR-21-5p, miR-29a-3p, and miR-138-5p) were up regulated, only miR-124-3p was down regulated. This study suggests that these miRNAs may have a role in the pathogenesis of ARDS related to NF-κB. However, this relationship needs to be examined in new studies by evaluation of pathways and target genes.
Collapse
Affiliation(s)
- Mustafa Azizoğlu
- Mersin University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Mersin, Turkey
| | - Lokman Ayaz
- Trakya University, Faculty of Pharmacy, Department of Biochemistry, Edirne, Turkey
| | - Gülsen Bayrak
- Mersin University, Faculty of Medicine, Department of Histology & Embriyology, Mersin, Turkey
| | - Banu Coşkun Yılmaz
- Mersin University, Faculty of Medicine, Department of Histology & Embriyology, Mersin, Turkey
| | - Handan Birbiçer
- Mersin University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Mersin, Turkey
| | - Nurcan Doruk
- Mersin University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Mersin, Turkey
| |
Collapse
|
45
|
Shi C, Deng J, Chiu M, Chen YX, O'Brien ER. Heat shock protein 27 immune complex altered signaling and transport (ICAST): Novel mechanisms of attenuating inflammation. FASEB J 2020; 34:14287-14301. [PMID: 32888229 DOI: 10.1096/fj.202001389rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 01/04/2023]
Abstract
Blood levels of heat shock protein (HSP27) and natural IgG auto-antibodies to HSP27 (AAbs) are higher in healthy controls compared to cardiovascular disease patients. Vaccination of mice with recombinant HSP25 (rHSP25, murine ortholog of human rHSP27) increased AAb levels, attenuated atherogenesis and reduced plaque inflammation and cholesterol content. We sought to determine if the HSP27 immune complex (IC) altered MΦ inflammation signaling (Toll Like Receptor 4; TLR4), and scavenger receptors involved in cholesterol uptake (SR-AI, CD-36). Combining a validated polyclonal IgG anti-HSP27 antibody (PAb) with rHSP27 enhanced binding to THP-1 MΦ cell membranes and activation of NF-κB signaling via TLR4, competing away LPS and effecting an anti-inflammatory cytokine profile. Similarly, adding the PAb with rHSP27 enhanced binding to SR-AI and CD-36, as well as lowered oxLDL binding in HEK293 cells separately transfected with SR-AI and CD-36, or THP-1 MΦ. Finally, the PAb enhanced the uptake and internalization of rHSP27 in THP-1 MΦ. Thus, the HSP27 IC potentiates HSP27 cell membrane signaling with receptors involved in modulating inflammation and cholesterol uptake, as well as HSP27 internalization. Going forward, we are focusing on the development of HSP27 Immune Complex Altered Signaling and Transport (ICAST) as a means of modulating inflammation.
Collapse
Affiliation(s)
- Chunhua Shi
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Jingti Deng
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Michael Chiu
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Yong-Xiang Chen
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Edward R O'Brien
- Division of Cardiology, Department of Cardiac Sciences, University of Calgary Cumming School of Medicine, Libin Cardiovascular Institute, Calgary, AB, Canada
| |
Collapse
|
46
|
Zhang E, Huang J, Wang K, Yu Q, Zhu C, Ren H. Pterostilbene Protects Against Lipopolysaccharide/D-Galactosamine-Induced Acute Liver Failure by Upregulating the Nrf2 Pathway and Inhibiting NF- κB, MAPK, and NLRP3 Inflammasome Activation. J Med Food 2020; 23:952-960. [PMID: 32701014 DOI: 10.1089/jmf.2019.4647] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The purpose of this study was to evaluate the protective effect of pterostilbene (Psb) against lipopolysaccharide and D-galactosamine (L/D)-induced acute liver failure (ALF) in mice and its potential mechanisms. Histology of liver was detected by H&E staining. Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels in serum and malondialdehyde (MDA), myeloperoxidase (MPO), glutathione (GSH), and superoxide dismutase (SOD) contents in liver were examined using detection kits. The levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) secretion were detected by ELISA. Meanwhile, MAPK, NF-κB, NLRP3 inflammasome, and Nrf2 were assessed by western blotting. Our findings showed that pretreatment with Psb protected against L/D-induced ALF by lowering the lethality, improving liver histology, reducing ALT, AST, IL-6, IL-1β, TNF-α, MDA, and MPO levels, and boosting liver GSH content and SOD activity. Moreover, Psb pretreatment effectively suppressed inflammation by decreasing NLRP3 inflammasome, MAPK, and NF-κB pathway activations. Moreover, Psb pretreatment efficiently enhanced the expression of several antioxidant enzymes, mainly depending on Nrf2 activation. This was the first study to demonstrate that Psb protects against L/D-induced ALF by inactivating MAPK, NF-κb, and NLRP3 inflammasome and upregulating the Nrf2 signaling pathway, indicating a potential therapeutic application for ALF treatment.
Collapse
Affiliation(s)
- Erli Zhang
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jingbo Huang
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun, China
| | - Kun Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Qinlei Yu
- Jilin Provincial Animal Disease Control Center, Changchun, China
| | - Chao Zhu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Hua Ren
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
47
|
Sehnert B, Burkhardt H, Dübel S, Voll RE. Cell-Type Targeted NF-kappaB Inhibition for the Treatment of Inflammatory Diseases. Cells 2020; 9:E1627. [PMID: 32640727 PMCID: PMC7407293 DOI: 10.3390/cells9071627] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022] Open
Abstract
Deregulated NF-k activation is not only involved in cancer but also contributes to the pathogenesis of chronic inflammatory diseases like rheumatoid arthritis (RA) and multiple sclerosis (MS). Ideally, therapeutic NF-KappaB inhibition should only take place in those cell types that are involved in disease pathogenesis to maintain physiological cell functions in all other cells. In contrast, unselective NF-kappaB inhibition in all cells results in multiple adverse effects, a major hindrance in drug development. Hitherto, various substances exist to inhibit different steps of NF-kappaB signaling. However, powerful tools for cell-type specific NF-kappaB inhibition are not yet established. Here, we review the role of NF-kappaB in inflammatory diseases, current strategies for drug delivery and NF-kappaB inhibition and point out the "sneaking ligand" approach. Sneaking ligand fusion proteins (SLFPs) are recombinant proteins with modular architecture consisting of three domains. The prototype SLC1 binds specifically to the activated endothelium and blocks canonical NF-kappaB activation. In vivo, SLC1 attenuated clinical and histological signs of experimental arthritides. The SLFP architecture allows an easy exchange of binding and effector domains and represents an attractive approach to study disease-relevant biological targets in a broad range of diseases. In vivo, SLFP treatment might increase therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Bettina Sehnert
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| | - Harald Burkhardt
- Division of Rheumatology, University Hospital Frankfurt, Goethe University, and Branch for Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany;
| | - Stefan Dübel
- Institute of Biochemistry and Biotechnology, Technical University Braunschweig, 38106 Braunschweig, Germany;
| | - Reinhard E. Voll
- Department of Rheumatology and Clinical Immunology, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79098 Freiburg, Germany
| |
Collapse
|
48
|
Sertedaki E, Veroutis D, Zagouri F, Galyfos G, Filis K, Papalambros A, Aggeli K, Tsioli P, Charalambous G, Zografos G, Sigala F. Carotid Disease and Ageing: A Literature Review on the Pathogenesis of Vascular Senescence in Older Subjects. Curr Gerontol Geriatr Res 2020; 2020:8601762. [PMID: 32582337 PMCID: PMC7306882 DOI: 10.1155/2020/8601762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/11/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023] Open
Abstract
Aging is a natural process that affects all systems of the human organism, leading to its inability to adapt to environmental changes. Advancing age has been correlated with various pathological conditions, especially cardiovascular and cerebrovascular diseases. Carotid artery (CA) is mainly affected by age-induced functional and morphological alterations causing atheromatous disease. The evolvement of biomedical sciences has allowed the elucidation of many aspects of this condition. Symptomatic carotid disease (CD) derives from critical luminar stenosis or eruption of an atheromatous plaque due to structural modifications of the vessels, such as carotid intima-media thickening. At a histologic level, the aforementioned changes are mediated by elastin fragmentation, collagen deposition, immune cell infiltration, and accumulation of cytokines and vasoconstrictors. Underlying mechanisms include chronic inflammation and oxidative stress, dysregulation of cellular homeostatic systems, and senescence. Thus, there is an imbalance in components of the vessel wall, which fails to counteract exterior stress stimuli. Consequently, arterial relaxation is impaired and atherosclerotic lesions progress. This is a review of current evidence regarding the relationship of aging with vascular senescence and CD. A deeper understanding of these mechanisms can contribute to the production of efficient prevention methods and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Eleni Sertedaki
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Veroutis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Flora Zagouri
- Clinical Therapeutics Department, Alexandra General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - George Galyfos
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Konstadinos Filis
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Papalambros
- First Department of Surgery, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Konstantina Aggeli
- First Department of Cardiology, University of Athens Medical School, Hippocration Hospital, Athens, Greece
| | - Panagiota Tsioli
- First Department of Pathology, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - George Charalambous
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - George Zografos
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| | - Fragiska Sigala
- First Department of Propaedeutic Surgery, Hippocration General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
49
|
Johns EC, Denison FC, Reynolds RM. Sleep disordered breathing in pregnancy: A review of the pathophysiology of adverse pregnancy outcomes. Acta Physiol (Oxf) 2020; 229:e13458. [PMID: 32087033 DOI: 10.1111/apha.13458] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022]
Abstract
Sleep disordered breathing (SDB) is a common obesity-related co-morbidity with strong associations to cardiometabolic disease. The risk of SDB is increased during pregnancy, particularly among obese pregnant women. Accumulating evidence suggests that an association exists between maternal SDB and the development of adverse pregnancy outcomes, particularly gestational diabetes and hypertensive disorders of pregnancy. Intermittent hypoxia, a central characteristic of SDB, has been shown in animal and clinical studies to dysregulate several biological pathways. This includes the promotion of oxidative stress, increased inflammation, activation of the hypothalamic-pituitary-adrenal axis, increased sympathetic activity and impaired glucose and insulin metabolism. This review considers how, during pregnancy, these pathophysiological processes are plausible mechanisms through which SDB may contribute to an increased risk of adverse outcomes, for the mother and perhaps also the offspring. However, a lack of robust evidence specific to the pregnant population, including limited evaluation of the placental function in affected pregnancies, limits our ability to draw definite conclusions on mechanisms contributing to adverse pregnancy outcomes and, indeed, the strength of association between SDB and certain pregnancy complications.
Collapse
Affiliation(s)
- Emma C. Johns
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health University of Edinburgh, Queen's Medical Research Institute Edinburgh United Kingdom
| | - Fiona C. Denison
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health University of Edinburgh, Queen's Medical Research Institute Edinburgh United Kingdom
| | - Rebecca M. Reynolds
- Tommy's Centre for Maternal and Fetal Health, MRC Centre for Reproductive Health University of Edinburgh, Queen's Medical Research Institute Edinburgh United Kingdom
- BHF/University Centre for Cardiovascular Science University of Edinburgh, Queen's Medical Research Institute Edinburgh United Kingdom
| |
Collapse
|
50
|
Omentin-1 Ameliorated Free Fatty Acid-Induced Impairment in Proliferation, Migration, and Inflammatory States of HUVECs. Cardiol Res Pract 2020; 2020:3054379. [PMID: 32300482 PMCID: PMC7140148 DOI: 10.1155/2020/3054379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/20/2020] [Accepted: 02/28/2020] [Indexed: 02/05/2023] Open
Abstract
Objectives Endothelial cell injury is a critical pathological change during the development of atherosclerosis. Here, we explored the effect of omentin-1 on free fatty acid- (FFA-) induced endothelial cell injury. Methods An FFA-induced endothelial cell injury model was established to investigate the role of omentin-1 in this process. Cell proliferation was analyzed with the Cell Counting Kit assay and flow cytometry. Scratch and transwell assays were used to evaluate cell migration. Factors secreted by endothelial cells after injury were detected by western blotting, reverse-transcription quantitative polymerase chain reaction, and cellular fluorescence assay. Results Omentin-1 rescued the FFA-induced impaired proliferation and migration capabilities of human umbilical vein endothelial cells (HUVECs). It decreased the number of THP-1 cells attached to HUVECs in response to injury and inhibited the FFA-induced proinflammatory state of HUVECs. Conclusion Omentin-1 could partly ameliorate FFA-induced endothelial cell injury.
Collapse
|