1
|
Mao J, Wu C, Zheng L, Li Y, Yang R, Yuan P, Jiang J, Li C, Zhou X. Advances in stimulus-responsive nanomedicine for treatment and diagnosis of atherosclerosis. Colloids Surf B Biointerfaces 2024; 245:114298. [PMID: 39378703 DOI: 10.1016/j.colsurfb.2024.114298] [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: 08/20/2024] [Revised: 09/22/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
Atherosclerosis (AS), an inflammatory cardiovascular disease driven by lipid deposition, presents global prevalence with high mortality. Effective anti-inflammatory or lipid removal is a promising strategy. However, current conventional drug delivery methods may face challenges in targeting disease sites and are deficient in the treatment of AS because of the nonspecific tissue distribution and uncontrollable release of the drug. In contrast, stimulus-responsive nanodrug delivery systems (NDDSs) can respond to stimulation and achieve controlled drug release rates at specific disease sites owing to the abnormal pathological microenvironment in plaques with low pH, excessive reactive oxygen species (ROS) and enzymes, and high shear stress. As a consequence, the efficacy of treatment is improved, and adverse reactions are reduced. On the other hand, NDDSs can combine exogenous stimulus responses (photothermal, ultrasound, etc.) to precisely control their function in time and space. This review for the first time focuses on the application of stimulus-responsive NDDSs in the treatment and diagnosis of AS in the last five years. In addition, its pivotal challenges and prospects are emphasized, aiming to facilitate its application for AS.
Collapse
Affiliation(s)
- Jingying Mao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China
| | - Chengxi Wu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Lixin Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yaoyao Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ronghao Yang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ping Yuan
- Department of Neurology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Jiang
- Department of Thyroid and Vascular 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.
| | - Xiangyu Zhou
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Luzhou, Sichuan 646000, China; Department of Thyroid and Vascular Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Jia X, Bai X, Yin Z, Zheng Q, Zhao Y, Lu Y, Shu Y, Wang Y, Zhang Y, Jin S. Siglec-5 as a novel receptor mediates endothelial cells oxLDL transcytosis to promote atherosclerosis. Transl Res 2024; 274:49-66. [PMID: 39341359 DOI: 10.1016/j.trsl.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Excessive subendothelial retention of oxidized low-density lipoprotein (oxLDL) and subsequent oxLDL engulfment by macrophages leads to the formation of foam cells and the development of atherosclerosis. Our previous study showed that the plasma level of sialic acid-binding immunoglobulin-like lectin 5 (Siglec-5) was a novel biomarker for the prognosis of atherosclerosis in diabetic patients. However, the role and underlying mechanisms of Siglec-5 in atherosclerosis have not been elucidated. METHODS The interaction between oxLDL and Siglec-5 was detected by fluorescence colocalization and coimmunoprecipitation. The effect of oxLDL on Siglec-5 expression was detected in endothelial cells and macrophages, and the effect of Siglec-5 on oxLDL transcytosis and uptake was investigated. Siglec-5 was overexpressed in mice using recombinant adeno-associated virus vector serotype 9 (rAAV9-Siglec-5) to evaluate the effect of Siglec-5 on oxLDL uptake and atherogenesis in vivo. In addition, the effects of Siglec-5 antibodies and soluble Siglec-5 proteins on oxLDL transcytosis and uptake and their role in atherogenesis were investigated in vivo and in vitro. RESULTS We found that oxLDL interacted with Siglec-5 and that oxLDL stimulated the expression of Siglec-5. Siglec-5 promotes the transcytosis and uptake of oxLDL, while both anti-Siglec-5 antibodies and soluble Siglec-5 protein attenuated oxLDL transcytosis and uptake. Interestingly, overexpression of Siglec-5 by recombinant adeno-associated viral vector serotype 9 (rAAV9-Siglec-5) promoted the retention of oxLDL in the aorta of C57BL/6 mice. Moreover, overexpression of Siglec-5 significantly accelerated the formation of atherosclerotic lesions in Apoe-/- mice. Moreover, both anti-Siglec-5 antibodies and soluble Siglec-5 protein significantly alleviated the retention of oxLDL in the aorta of rAAV9-Siglec-5-transfected C57BL/6 mice and the formation of atherosclerotic plaques in rAAV9-Siglec-5-transfected Apoe-/- mice. CONCLUSION Our results suggested that Siglec-5 was a novel receptor that mediated oxLDL transcytosis and promoted the formation of foam cells. Interventions that inhibit the interaction between oxLDL and Siglec-5, including anti-Siglec-5 antibody or soluble Siglec-5 protein treatment, may provide novel therapeutic strategies in treating atherosclerosis.
Collapse
Affiliation(s)
- Xiong Jia
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China; Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Xiangli Bai
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China; Department of Laboratory Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Zhiqiang Yin
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yin Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yajing Lu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yifei Zhang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan 430077, China.
| |
Collapse
|
3
|
Wang D, Ling J, Tan R, Wang H, Qu Y, Li X, Lin J, Zhang Q, Hu Q, Liu Z, Lu Z, Lin Y, Sun L, Wang D, Zhou M, Shi Z, Gao W, Ye H, Lin X. CD169 + classical monocyte as an important participant in Graves' ophthalmopathy through CXCL12-CXCR4 axis. iScience 2024; 27:109213. [PMID: 38439953 PMCID: PMC10910260 DOI: 10.1016/j.isci.2024.109213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/11/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
Patients with Graves' disease (GD) can develop Graves' ophthalmopathy (GO), but the underlying pathological mechanisms driving this development remain unclear. In our study, which included patients with GD and GO, we utilized single-cell RNA sequencing (scRNA-seq) and multiplatform analyses to investigate CD169+ classical monocytes, which secrete proinflammatory cytokines and are expanded through activated interferon signaling. We found that CD169+ clas_mono was clinically significant in predicting GO progression and prognosis, and differentiated into CD169+ macrophages that promote inflammation, adipogenesis, and fibrosis. Our murine model of early-stage GO showed that CD169+ classical monocytes accumulated in orbital tissue via the Cxcl12-Cxcr4 axis. Further studies are needed to investigate whether targeting circulating monocytes and the Cxcl12-Cxcr4 axis could alleviate GO progression.
Collapse
Affiliation(s)
- Dongliang Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jie Ling
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - RongQiang Tan
- The First People’s Hospital of Zhaoqing, Zhaoqing 526000, China
| | - Huishi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yixin Qu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xingyi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jinshan Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Qikai Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Qiuling Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhaojing Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yuheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Li Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Dingqiao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Ming Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhuoxing Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Wuyou Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Huijing Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
4
|
Wang SQ, Xiang J, Zhang GQ, Fu LY, Xu YN, Chen Y, Tao L, Hu XX, Shen XC. Essential oil from Fructus Alpinia zerumbet ameliorates atherosclerosis by activating PPARγ-LXRα-ABCA1/G1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155227. [PMID: 38128398 DOI: 10.1016/j.phymed.2023.155227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/11/2023] [Accepted: 11/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Atherosclerosis (AS) is a progressive chronic disease. Currently, cardiovascular diseases (CVDs) caused by AS is responsible for the global increased mortality. Yanshanjiang as miao herb in Guizhou of China is the dried and ripe fruit of Fructus Alpinia zerumbet. Accumulated evidences have confirmed that Yanshanjiang could ameliorate CVDs, including AS. Nevertheless, its effect and mechanism on AS are still largely unknown. PURPOSE To investigate the role of essential oil from Fructus Alpinia zerumbet (EOFAZ) on AS, and the potential mechanism. METHODS A high-fat diet (HFD) ApoE-/- mice model of AS and a oxLDL-induced model of macrophage-derived foam cells (MFCs) were reproduced to investigate the pharmacological properties of EOFAZ on AS in vivo and foam cell formation in vitro, respectively. The underlying mechanisms of EOFAZ were investigated using Network pharmacology and molecular docking. EOFAZ effect on PPARγ protein stability was measured using a cellular thermal shift assay (CETSA). Pharmacological agonists and inhibitors and gene interventions were employed for clarifying EOFAZ's potential mechanism. RESULTS EOFAZ attenuated AS progression in HFD ApoE-/- mice. This attenuation was manifested by the reduced aortic intima plaque development, increased collagen content in aortic plaques, notable improvement in lipid profiles, and decreased levels of inflammatory factors. Moreover, EOFAZ inhibited the formation of MFCs by enhancing cholesterol efflux through activiting the PPARγ-LXRα-ABCA1/G1 pathway. Interestingly, the pharmacological knockdown of PPARγ impaired the beneficial effects of EOFAZ on MFCs. Additionally, our results indicated that EOFAZ reduced the ubiquitination degradation of PPARγ, and the chemical composition of EOFAZ directly bound to the PPARγ protein, thereby increasing its stability. Finally, PPARγ knockdown mitigated the protective effects of EOFAZ on AS in HFD ApoE-/- mice. CONCLUSION These findings represent the first confirmation of EOFAZ's in vivo anti-atherosclerotic effects in ApoE-/- mice. Mechanistically, its chemical constituents can directly bind to PPARγ protein, enhancing its stability, while reducing PPARγ ubiquitination degradation, thereby inhibiting foam cell formation via activation of the PPARγ-LXRα-ABCA1/G1 pathway. Simultaneously, EOFAZ could ameliorates blood lipid metabolism and inflammatory microenvironment, thus synergistically exerting its anti-atherosclerotic effects.
Collapse
Affiliation(s)
- Sheng-Quan Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Jun Xiang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Guang-Qiong Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling-Yun Fu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yi-Ni Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Xiao-Xia Hu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, China.
| | - Xiang-Chun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
5
|
Poznyak A, Kashirskikh D, Postnov A, Popov M, Sukhorukov V, Orekhov A. Sialic acid as the potential link between lipid metabolism and inflammation in the pathogenesis of atherosclerosis. Braz J Med Biol Res 2023; 56:e12972. [PMID: 38088673 PMCID: PMC10712282 DOI: 10.1590/1414-431x2023e12972] [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: 07/22/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
In the modern world, cardiovascular diseases have a special place among the most common causes of death. Naturally, this widespread problem cannot escape the attention of scientists and researchers. One of the main conditions preceding the development of fatal cardiovascular diseases is atherosclerosis. Despite extensive research into its pathogenesis and possible prevention and treatment strategies, many gaps remain in our understanding of this disease. For example, the concept of multiple low-density lipoprotein modifications was recently stated, in which desialylation is of special importance. Apart from this, sialic acids are known to be important contributors to processes such as endothelial dysfunction and inflammation, which in turn are major components of atherogenesis. In this review, we have collected information on sialic acid metabolism, analyzed various aspects of its implication in atherosclerosis at different stages, and provided an overview of the role of particular groups of enzymes responsible for sialic acid metabolism in the context of atherosclerosis.
Collapse
Affiliation(s)
- A.V. Poznyak
- Institute for Atherosclerosis Research, Moscow, Russia
| | | | - A.Y. Postnov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - M.A. Popov
- Department of Cardiac Surgery, Moscow Regional Research and Clinical Institute (MONIKI), Moscow, Russia
| | - V.N. Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| | - A.N. Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific Institution, Petrovsky National Research Centre of Surgery (FSBSI “Petrovsky NRCS”), Moscow, Russia
| |
Collapse
|
6
|
Prenzler S, Rudrawar S, Waespy M, Kelm S, Anoopkumar-Dukie S, Haselhorst T. The role of sialic acid-binding immunoglobulin-like-lectin-1 (siglec-1) in immunology and infectious disease. Int Rev Immunol 2023; 42:113-138. [PMID: 34494938 DOI: 10.1080/08830185.2021.1931171] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Siglec-1, also known as Sialoadhesin (Sn) and CD169 is highly conserved among vertebrates and with 17 immunoglobulin-like domains is Siglec-1 the largest member of the Siglec family. Expression of Siglec-1 is found primarily on dendritic cells (DCs), macrophages and interferon induced monocyte. The structure of Siglec-1 is unique among siglecs and its function as a receptor is also different compared to other receptors in this class as it contains the most extracellular domains out of all the siglecs. However, the ability of Siglec-1 to internalize antigens and to pass them on to lymphocytes by allowing dendritic cells and macrophages to act as antigen presenting cells, is the main reason that has granted Siglec-1's key role in multiple human disease states including atherosclerosis, coronary artery disease, autoimmune diseases, cell-cell signaling, immunology, and more importantly bacterial and viral infections. Enveloped viruses for example have been shown to manipulate Siglec-1 to increase their virulence by binding to sialic acids present on the virus glycoproteins allowing them to spread or evade immune response. Siglec-1 mediates dissemination of HIV-1 in activated tissues enhancing viral spread via infection of DC/T-cell synapses. Overall, the ability of Siglec-1 to bind a variety of target cells within the immune system such as erythrocytes, B-cells, CD8+ granulocytes and NK cells, highlights that Siglec-1 is a unique player in these essential processes.
Collapse
Affiliation(s)
- Shane Prenzler
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Santosh Rudrawar
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Mario Waespy
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Sørge Kelm
- Centre for Biomolecular Interactions Bremen, Department of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Shailendra Anoopkumar-Dukie
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
| | - Thomas Haselhorst
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
7
|
Zhang J, Zhang L. Bioinformatics approach to identify the influences of SARS-COV2 infections on atherosclerosis. Front Cardiovasc Med 2022; 9:907665. [PMID: 36061537 PMCID: PMC9433720 DOI: 10.3389/fcvm.2022.907665] [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: 03/30/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been a global pandemic since early 2020. Understanding the relationship between various systemic disease and COVID-19 through disease ontology (DO) analysis, an approach based on disease similarity studies, has found that COVID-19 is most strongly associated with atherosclerosis. The study provides new insights for the common pathogenesis of COVID-19 and atherosclerosis by looking for common transcriptional features. Two datasets (GSE152418 and GSE100927) were downloaded from GEO database to search for common differentially expressed genes (DEGs) and shared pathways. A total of 34 DEGs were identified. Among them, ten hub genes with high degrees of connectivity were picked out, namely C1QA, C1QB, C1QC, CD163, SIGLEC1, APOE, MS4A4A, VSIG4, CCR1 and STAB1. This study suggests the critical role played by Complement and coagulation cascades in COVID-19 and atherosclerosis. Our findings underscore the importance of C1q in the pathogenesis of COVID-19 and atherosclerosis. Activation of the complement system can lead to endothelial dysfunction. The DEGs identified in this study provide new biomarkers and potential therapeutic targets for the prevention of atherosclerosis.
Collapse
|
8
|
Zhou Y, Wang S, Liang X, Heger Z, Xu M, Lu Q, Yu M, Adam V, Li N. Turning Hot into Cold: Immune Microenvironment Reshaping for Atherosclerosis Attenuation Based on pH-Responsive shSiglec-1 Delivery System. ACS NANO 2022; 16:10517-10533. [PMID: 35762565 DOI: 10.1021/acsnano.2c01778] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Current atherosclerosis treatment is based on a combination of cholesterol-lowering medication and low-fat diets; however, the clinical effect is unsatisfactory. It has been shown that the level of immune cell infiltration and pro-inflammatory factors in the atherosclerotic immune microenvironment (AIM) play important roles in the development and progression of atherosclerosis. Therefore, we hypothesized that reshaping "hot AIM" into "cold AIM" could attenuate atherosclerosis. For this purpose, we designed a pH-responsive and charge-reversible nanosystem, referred to as Au-PEI/shSiglec-1/PEI-acetylsalicylic acid (ASPA NPs) to effectively deliver shSiglec-1, which blocked the interactions between macrophages with CD8+ T/NKT cells, thus inhibiting immune cell infiltration. Further, we demonstrated that acetylsalicylic acid (ASA), detached from the pH-responsive PEI-ASA polymer, and inhibited lipid accumulation in macrophage, thereby decreasing the lipid antigen presentation. Additionally, reduced macrophage-produced inflammatory factors by ASA and low CD8+ T/NKT cell infiltration levels synergistically inhibit Th17 cell differentiation, thus further dramatically attenuating inflammation in AIM by decreasing the IL-17A production. Eventually, ASPA NPs efficiently reshaped AIM by inhibiting immune cell infiltration, lipid antigen presentation, and pro-inflammation, which provided a feasible therapeutic strategy for atherosclerosis immunotherapy.
Collapse
Affiliation(s)
- Yue Zhou
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Siyu Wang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Qiang Lu
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Meng Yu
- School of Pharmaceutical Science Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-61300 Brno, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, CZ-61200 Brno, Czech Republic
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
9
|
Hsu YW, Hsu FF, Chiang MT, Tsai DL, Li FA, Angata T, Crocker PR, Chau LY. Siglec-E retards atherosclerosis by inhibiting CD36-mediated foam cell formation. J Biomed Sci 2021; 28:5. [PMID: 33397354 PMCID: PMC7784283 DOI: 10.1186/s12929-020-00698-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/21/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The accumulation of lipid-laden macrophages, foam cells, within sub-endothelial intima is a key feature of early atherosclerosis. Siglec-E, a mouse orthologue of human Siglec-9, is a sialic acid binding lectin predominantly expressed on the surface of myeloid cells to transduce inhibitory signal via recruitment of SH2-domain containing protein tyrosine phosphatase SHP-1/2 upon binding to its sialoglycan ligands. Whether Siglec-E expression on macrophages impacts foam cell formation and atherosclerosis remains to be established. METHODS ApoE-deficient (apoE-/-) and apoE/Siglec-E-double deficient (apoE-/-/Siglec-E-/-) mice were placed on high fat diet for 3 months and their lipid profiles and severities of atherosclerosis were assessed. Modified low-density lipoprotein (LDL) uptake and foam cell formation in wild type (WT) and Siglec-E-/-- peritoneal macrophages were examined in vitro. Potential Siglec-E-interacting proteins were identified by proximity labeling in conjunction with proteomic analysis and confirmed by coimmunoprecipitation experiment. Impacts of Siglec-E expression and cell surface sialic acid status on oxidized LDL uptake and signaling involved were examined by biochemical assays. RESULTS Here we show that genetic deletion of Siglec-E accelerated atherosclerosis without affecting lipid profile in apoE-/- mice. Siglec-E deficiency promotes foam cell formation by enhancing acetylated and oxidized LDL uptake without affecting cholesterol efflux in macrophages in vitro. By performing proximity labeling and proteomic analysis, we identified scavenger receptor CD36 as a cell surface protein interacting with Siglec-E. Further experiments performed in HEK293T cells transiently overexpressing Siglec-E and CD36 and peritoneal macrophages demonstrated that depletion of cell surface sialic acids by treatment with sialyltransferase inhibitor or sialidase did not affect interaction between Siglec-E and CD36 but retarded Siglec-E-mediated inhibition on oxidized LDL uptake. Subsequent experiments revealed that oxidized LDL induced transient Siglec-E tyrosine phosphorylation and recruitment of SHP-1 phosphatase in macrophages. VAV, a downstream effector implicated in CD36-mediated oxidized LDL uptake, was shown to interact with SHP-1 following oxidized LDL treatment. Moreover, oxidized LDL-induced VAV phosphorylation was substantially lower in WT macrophages comparing to Siglec-E-/- counterparts. CONCLUSIONS These data support the protective role of Siglec-E in atherosclerosis. Mechanistically, Siglec-E interacts with CD36 to suppress downstream VAV signaling involved in modified LDL uptake.
Collapse
Affiliation(s)
- Yaw-Wen Hsu
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Fu-Fei Hsu
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Ming-Tsai Chiang
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Dong-Lin Tsai
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Fu-An Li
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan
| | - Takashi Angata
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan
| | - Paul R Crocker
- Division of Cell Signaling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Lee-Young Chau
- Institute of Biomedical Sciences, Academia Sinica, No.128, Sec.II, Academy Road, Taipei, 115, Taiwan.
| |
Collapse
|
10
|
Hu Y, Zhang T, Chen J, Cheng W, Chen J, Zheng Z, Lin J, Zhu G, Zhang Y, Bai X, Wang Y, Song B, Wang Q, Qin L, Zhang P. Downregulation of Hypoxia-Inducible Factor-1α by RNA Interference Alleviates the Development of Collagen-Induced Arthritis in Rats. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1330-1342. [PMID: 32160704 PMCID: PMC7038004 DOI: 10.1016/j.omtn.2020.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis (RA) is the most common type of autoimmune arthritis. Hypoxia-inducible factor-1α (HIF-1α) as a transcription factor in response to hypoxia suggests that it could be a potential therapeutic target for the treatment of RA. In this study, we assessed whether the HIF pathway blockade attenuates the manifestations of RA in the collagen-induced arthritis (CIA) rat model. We constructed a short hairpin RNA (shRNA) lentiviral expression vector targeting HIF-1α (pLVX-shRNA-HIF-1α) and to achieve HIF-1α RNA interference. Quantitative RT-PCR, immunofluorescence staining, and western blot were used to detect the expressions of HIF-1α, vascular endothelial growth factor (VEGF), phsopho (p)-p65, and p-IКBɑ mRNA and protein, respectively. Micro-computed tomography was used to investigate joint morphology at different time points after CIA induction. Moreover, enzyme-linked immunosorbent assay (ELISA) was used to monitor the expression of inflammatory cytokines. In vitro analyses revealed that pLVX-shRNA-HIF-1α effectively inhibited the expression of HIF-1α and VEGF and led to the activation of p-65 and p-IКBɑ, as well as decreased proinflammatory cytokine expression in cell culture. Inhibition of HIF-1α in rats decreased signs of a systemic inflammatory condition, together with decreased pathological changes of RA. Moreover, downregulation of HIF-1α expression markedly reduced the synovitis and angiogenesis. In conclusion, we have shown that pharmacological inhibition of HIF-1 may improve the clinical manifestations of RA.
Collapse
Affiliation(s)
- Yiping Hu
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Tiantian Zhang
- Department of Rheumatology, People's Hospital of Bao'an District, Shenzhen, Guangdong 518128, China
| | - Jingqin Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - WenXiang Cheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Jianhai Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Zhengtan Zheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Jietao Lin
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Yong Zhang
- Shenzhen Pingle Orthopaedic Hospital, Guangdong 518000, China
| | - Xueling Bai
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Bing Song
- School of Dentistry, Cardiff University, Cardiff, Heath Park, CF23 6AL Wales, UK
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China.
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China; University of Chinese Academy of Sciences, Shenzhen Hospital, Shenzhen 518000, China.
| |
Collapse
|
11
|
Zhang C, Chen J, Liu Y, Xu D. Sialic acid metabolism as a potential therapeutic target of atherosclerosis. Lipids Health Dis 2019; 18:173. [PMID: 31521172 PMCID: PMC6745061 DOI: 10.1186/s12944-019-1113-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 08/30/2019] [Indexed: 01/08/2023] Open
Abstract
Sialic acid (Sia), the acylated derivative of the nine-carbon sugar neuraminic acid, is a terminal component of the oligosaccharide chains of many glycoproteins and glycolipids. In light of its important biological and pathological functions, the relationship between Sia and coronary artery disease (CAD) has been drawing great attentions recently. Large-scale epidemiological surveys have uncovered a positive correlation between plasma total Sia and CAD risk. Further research demonstrated that N-Acetyl-Neuraminic Acid, acting as a signaling molecule, triggered myocardial injury via activation of Rho/ROCK-JNK/ERK signaling pathway both in vitro and in vivo. Moreover, there were some evidences showing that the aberrant sialylation of low-density lipoprotein, low-density lipoprotein receptor and blood cells was involved in the pathological process of atherosclerosis. Significantly, the Sia regulates immune response by binding to sialic acid-binding immunoglobulin-like lectin (Siglecs). The Sia-Siglecs axis is involved in the immune inflammation of atherosclerosis. The generation of Sia and sialylation of glycoconjugate both depend on many enzymes, such as sialidase, sialyltransferase and trans-sialidase. Abnormal activation or level of these enzymes associated with atherosclerosis, and inhibitors of them might be new CAD treatments. In this review, we focus on summarizing current understanding of Sia metabolism and of its relevance to atherosclerosis.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.,Department of Health Management Center, Hunan Provincial People's Hospital, 61 Jiefang West Road, Changsha, 410005, Hunan, China
| | - Jingyuan Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Yuhao Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Danyan Xu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
12
|
Qiu Q, Li C, Song Y, Shi T, Luo X, Zhang H, Hu L, Yan X, Zheng H, Liu M, Liu M, Liu M, Yang S, Liu X, Chen G, Deng Y. Targeted delivery of ibrutinib to tumor-associated macrophages by sialic acid-stearic acid conjugate modified nanocomplexes for cancer immunotherapy. Acta Biomater 2019; 92:184-195. [PMID: 31108259 DOI: 10.1016/j.actbio.2019.05.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022]
Abstract
Ibrutinib (IBR), an irreversible Bruton's tyrosine kinase (BTK) inhibitor, is expected to be a potent therapeutic modality, given that BTK is overexpressed in tumor-associated macrophages (TAMs) and participates in promoting tumor progression, angiogenesis, and immunosuppression. However, rapid clearance in vivo and low tumor accumulation have rendered effective uptake of IBR by TAMs challenge. Herein, we designed and synthesized a sialic acid (SA)-stearic acid conjugate modified on the surface of nanocomplexes to encapsulate IBR (SA/IBR/EPG) for targeted immunotherapy. Amphiphilic egg phosphatidylglycerol (EPG) structure and strong IBR-EPG interactions render these nanocomplexes high IBR loading capacity, prolonged blood circulation, and optimal particle sizes (∼30 nm), which can effectively deliver IBR to the tumor, followed by subsequent internalization of IBR by TAMs through SA-mediated active targeting. In vitro and in vivo tests showed that the prepared SA/IBR/EPG nanocomplexes could preferentially accumulate in TAMs and exert potent antitumor activity. Immunofluorescence staining analysis further confirmed that SA/IBR/EPG remarkably inhibited angiogenesis and tumorigenic cytokines released by TAM and eventually suppressed tumor progression, without eliciting any unwanted effect. Thus, SA-decorated IBR nanocomplexes present a promising strategy for cancer immunotherapy. STATEMENT OF SIGNIFICANCE: Ibrutinib (IBR), an irreversible Bruton's tyrosine kinase (BTK) inhibitor, is expected to be a potent therapeutic modality, given that BTK is overexpressed in tumor-associated macrophages (TAMs) and participates in promoting tumor progression, angiogenesis, and immunosuppression. However, rapid clearance in vivo and low tumor accumulation have rendered effective uptake of IBR by TAMs challenge. Herein, we designed and synthesized a sialic acid (SA)-stearic acid conjugate modified on the surface of nanocomplexes to encapsulate IBR (SA/IBR/EPG) for targeted delivery of IBR to TAMs. The developed SA/IBR/EPG nanocomplexes exhibited high efficiency in targeting TAMs and inhibiting BTK activation, consequently inhibiting Th2 tumorigenic cytokine release, reducing angiogenesis, and suppressing tumor growth. These results implied that the SA/IBR/EPG nanocomplex could be a promising strategy for TAM-targeting immunotherapy with minimal systemic side effects.
Collapse
|
13
|
Li JY, Yang XY, Wang XF, Jia X, Wang ZJ, Deng AP, Bai XL, Zhu L, Li BH, Feng ZB, Li Y, Wang L, Jin S. Siglec-5 is a novel marker of critical limb ischemia in patients with diabetes. Sci Rep 2017; 7:11272. [PMID: 28900239 PMCID: PMC5595823 DOI: 10.1038/s41598-017-11820-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/30/2017] [Indexed: 11/09/2022] Open
Abstract
Critical Limb Ischemia (CLI) is common but uncommonly diagnosed. Improved recognition and early diagnostic markers for CLI are needed. Therefore, the aim of our study was to identify plasma biomarkers of CLI in patients with type 2 diabetes mellitus (T2DM). In this study, antibody-coated glass slide arrays were used to determine the plasma levels of 274 human cytokines in four matched cases of diabetes with and without CLI. Potential biomarkers were confirmed in an independent cohort by ELISA. After adjusting for confounding risk factors, only plasma level of Siglec-5 remained significantly associated with an increased odds ratio (OR) for diabetes with CLI by binary logistic regression analysis. Receiver operating characteristic (ROC) curve analysis revealed the optimal cut-off points for Siglec-5 was 153.1 ng/ml. After entering Siglec-5, the AUC was 0.99, which was higher than that of confounding risk factors only (AUC = 0.97, P < 0.05). Siglec-5 was expressed in plaques, but not in healthy artery wall in T2DM patients. Elevated plasma Siglec-5 was independently associated with CLI in T2DM. Plasma Siglec-5 levels are implicated as an early diagnostic marker of CLI in T2DM patients and it may become a target for the prevention or treatment of CLI in diabetes.
Collapse
Affiliation(s)
- Ju-Yi Li
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of Pharmacy, The central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiao-Yan Yang
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiu-Fang Wang
- Department of Pain, The central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiong Jia
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhong-Jing Wang
- Department of Endocrinology, The central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ai-Ping Deng
- Department of Pharmacy, The central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang-Li Bai
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Zhu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bing-Hui Li
- Department of Wound Repair, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zi-Bo Feng
- Department of Wound Repair, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ye Li
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Wang
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Jin
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China. .,Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Tam C, Wong JH, Cheung RCF, Zuo T, Ng TB. Therapeutic potentials of short interfering RNAs. Appl Microbiol Biotechnol 2017; 101:7091-7111. [PMID: 28791440 DOI: 10.1007/s00253-017-8433-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/19/2017] [Indexed: 01/10/2023]
Abstract
Short interfering RNA (siRNA) is one of the members of the family of RNA interference (RNAi). Coupled with the RNA-induced silencing complex (RISC), siRNA is able to trigger the cleavage of target RNAs which serve as a defensive system against pathogens. Meanwhile, siRNA in gene silencing opens a new avenue for the treatment of various diseases. SiRNA can effectively inhibit viral infection and replication and suppress tumorigenesis and various inflammation-associated diseases and cardiovascular diseases by inactivation of viral genes and downregulation of oncogene expression. Recently, endogenous siRNAs (endo-siRNAs) were discovered in the reproductive cells of animals which may be associated with regulation of cell division. Structural modification of siRNA enhances the delivery, specificity and efficacy and bioavailability to the target cells. There are at least five categories of siRNA delivery systems including viral vectors, lipid-based nanoparticles, peptide-based nanoparticles, polymer-based nanoparticles and inorganic small molecules like metal ions, silica and carbon. Sufficient preclinical and clinical studies supported that siRNA may be a potential medicine for targeted therapy of various diseases in the near future.
Collapse
Affiliation(s)
- Chit Tam
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China.
| | - Jack Ho Wong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China
| | - Randy Chi Fai Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China
| | - Tao Zuo
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong, China
| | - Tzi Bun Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Sha Tin, New Territories, Hong Kong, China.
| |
Collapse
|