1
|
Liu S, Zhang H, Wu M, Zhou Z, Xiao Y, Wan Q, Lan Z, Rong C. Association between the triglyceride-glucose index and carotid artery plaque burden in patients with primary hypertension: A cross-sectional study. Clin Exp Hypertens 2024; 46:2383232. [PMID: 39045803 DOI: 10.1080/10641963.2024.2383232] [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: 05/30/2024] [Revised: 06/29/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Studies have shown an association between the triglyceride-glucose (TyG) index and carotid artery plaque (CAP). However, the relationship between the TyG index and plaque burden in individuals with primary hypertension remains uncertain. Our study specifically aimed to explore this relationship among primary hypertension patients. METHODS This study involved 5,153 hospitalized patients diagnosed with primary hypertension who were undergoing treatment at the Affiliated Hospital of Jiangxi University of Chinese Medicine. We utilized multivariate logistic regression, penalized spline regression, and generalized additive models to assess the association between the TyG index and CAP burden. RESULTS There were 2,400 patients with primary hypertension in all. The multivariate study, which took into account all covariables, showed a positive correlation between the TyG index and CAP (OR: 1.25, 95% CI: 1.04-1.5). When the TyG index was evaluated as quartiles, the risk of CAP in the Q3 and Q4 levels of the TyG index were 1.4 (95% CI: 1.03-1.91) and 1.54 (95% CI: 1.11-2.14) times greater than in the Q1 level after adjusting for all covariables (P for trend < .05). Regardless of whether the TyG index was used as a continuous variable or a categorical variable, it has no significant association with the risk of single plaque after adjusting for all confounders (p ≥ .05). The TyG index was found to be substantially correlated with the presence of multiple plaques when analyzed as a continuous variable (OR: 1.32, 95% CI: 1.09-1.59, p = .004). When the TyG index was evaluated as quartiles, the adjusted OR in Q3 and Q4 were 1.49 (95% CI: 1.06-2.1) and 1.67 (95% CI: 1.16-2.41), respectively, with Q1 as reference (P for trend = .005). The relationship between the TyG index and the presence of multiple plaques is also consistent in all subgroups. CONCLUSION The TyG index is positively associated with the presence of multiple plaques in patients with primary hypertension, whereas no association is found between the TyG index and the presence of a single carotid plaque.
Collapse
Affiliation(s)
- Shanshan Liu
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Hao Zhang
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Miao Wu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Zhixian Zhou
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Yao Xiao
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Qiang Wan
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Zhihui Lan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Chao Rong
- Department of Cardiology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Chen S, Zhang W, Tang C, Rong X, Liu Y, Luo Y, Xu L, Xu Z, Wang J, Wang Y, Du Q, Liu B, Zhang Y, Liu J, Guo D. Macrophage membrane-functionalized manganese dioxide nanomedicine for synergistic treatment of atherosclerosis by mitigating inflammatory storms and promoting cholesterol efflux. J Nanobiotechnology 2024; 22:664. [PMID: 39465387 PMCID: PMC11514794 DOI: 10.1186/s12951-024-02939-x] [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: 07/11/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024] Open
Abstract
Atherosclerosis (AS) poses a significant threat to human life and health. However, conventional antiatherogenic medications exhibit insufficient targeting precision and restricted therapeutic effectiveness. Moreover, during the progression of AS, macrophages undergo polarization toward the proinflammatory M1 phenotype and generate reactive oxygen species (ROS) to accelerate the occurrence of inflammatory storms, and ingest excess lipids to form foam cells by inhibiting cholesterol efflux. In our study, we developed a macrophage membrane-functionalized hollow mesoporous manganese dioxide nanomedicine (Col@HMnO2-MM). This nanomedicine has the ability to evade immune cell phagocytosis, enables prolonged circulation within the body, targets the inflammatory site of AS for effective drug release, and alleviates the inflammatory storm at the AS site by eliminating ROS. Furthermore, Col@HMnO2-MM has the ability to generate oxygen autonomously by breaking down surplus hydrogen peroxide generated at the inflammatory AS site, thereby reducing the hypoxic microenvironment of the plaque by downregulating hypoxia-inducible factor (HIF-1α), which in turn enhances cholesterol efflux to inhibit foam cell formation. In an APOE-/- mouse model, Col@HMnO2-MM significantly reduced inflammatory factor levels, lipid storage, and plaque formation without significant long-term toxicity. In summary, this synergistic treatment significantly improved the effectiveness of nanomedicine and may offer a novel strategy for precise AS therapy.
Collapse
Affiliation(s)
- Sijin Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wenli Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Chun Tang
- Department of Spine Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xiyue Rong
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yun Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ying Luo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lian Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhongsheng Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Junrui Wang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yi Wang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qianying Du
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yu Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Jia Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Dajing Guo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
3
|
Tang C, Shen Y, Xing Y, Wu Y, Zhang M, Zhang H, Zhao S, Zhou Z, Sun Y, Mo X, Wang W. 3D-Printed Stents Loaded with Panax notoginseng Saponin for Promoting Re-endothelialization and Reducing Local Inflammation in the Carotid Artery of Rabbits. ACS Biomater Sci Eng 2024; 10:6483-6497. [PMID: 39141849 DOI: 10.1021/acsbiomaterials.4c00925] [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: 08/16/2024]
Abstract
Endovascular treatment (EVT) using stents has become the primary option for severe cerebrovascular stenosis. However, considerable challenges remain to be addressed, such as in-stent restenosis (ISR) and late thrombosis. Many modified stents have been developed to inhibit the hyperproliferation of vascular smooth muscle cells (SMCs) and protect vascular endothelial cells (VECs), thereby reducing such complications. Some modified stents, such as those infused with rapamycin, have improved in preventing acute thrombosis. However, ISR and late thrombosis, which are long-term complications, remain unavoidable. Panax notoginseng saponin (PNS), a traditional Chinese medicine consisting of various compounds, is beneficial in promoting the proliferation and migration of VECs and inhibiting the proliferation of SMCs. Herein, a 3D-printed polycaprolactone (PCL) stent loaded with PNS (PNS-PCL stent) was developed based on a previous study. In vitro studies confirmed that PNS promotes the migration and proliferation of VECs, which were damaged, by increasing the expression levels of microRNA-126, p-AKT, and endothelial nitric oxide synthase. In vivo, the PNS-PCL stents maintained the patency of the carotid artery in rabbits for up to three months, outperforming the PCL stents. The PNS-PCL stents may present a new solution for the EVT of cerebrovascular atherosclerotic stenosis in the future.
Collapse
Affiliation(s)
- Chaojie Tang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Yihong Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Yazhi Xing
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Yufan Wu
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
- Department of Otolaryngology Head and Neck Surgery, Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Mianmian Zhang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - He Zhang
- Department of Gastroenterology, Guang'anmen Hospital, China Academy of Traditional Chinese Medical Sciences, Beijing 100053, PR China
| | - Shuo Zhao
- Joint International Research Laboratory of Resource Chemistry, Ministry of Education, Shanghai Normal University, Shanghai 200234, PR China
| | - Zhiguo Zhou
- Joint International Research Laboratory of Resource Chemistry, Ministry of Education, Shanghai Normal University, Shanghai 200234, PR China
| | - Yongning Sun
- Department Cardiovascular Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, PR China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Wu Wang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
- Department of Radiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| |
Collapse
|
4
|
Geng S, Lu R, Zhang Y, Wu Y, Xie L, Caldwell BA, Pradhan K, Yi Z, Hou J, Xu F, Chen X, Li L. Monocytes Reprogrammed by 4-PBA Potently Contribute to the Resolution of Inflammation and Atherosclerosis. Circ Res 2024; 135:856-872. [PMID: 39224974 PMCID: PMC11424066 DOI: 10.1161/circresaha.124.325023] [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: 06/04/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis. METHODS Systems analyses integrating single-cell RNA sequencing and complementary immunologic approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming were assessed by integrated biochemical and genetic approaches. The intercellular propagation of homeostasis resolution was evaluated by coculture assays with donor monocytes trained by 4-PBA and recipient naive monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high-fat diet-fed ApoE-/- mouse model with IP 4-PBA administration. Furthermore, the selective efficacy of 4-PBA-trained monocytes was examined by IV transfusion of ex vivo trained monocytes by 4-PBA into recipient high-fat diet-fed ApoE-/- mice. RESULTS In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 (intercellular adhesion molecule 1) via reducing peroxisome stress and attenuating SYK (spleen tyrosine kinase)-mTOR (mammalian target of rapamycin) signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ (peroxisome proliferator-activated receptor γ) neddylation mediated by TOLLIP (toll-interacting protein). 4-PBA-trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA-trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo. CONCLUSIONS Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ran Lu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Yajun Wu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ling Xie
- Department of Biochemistry and Molecular Biology, University of North Carolina at Chappell Hill, NC (L.X., X.C.)
| | - Blake A Caldwell
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Kisha Pradhan
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ziyue Yi
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Jacqueline Hou
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Feng Xu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Xian Chen
- Department of Biochemistry and Molecular Biology, University of North Carolina at Chappell Hill, NC (L.X., X.C.)
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| |
Collapse
|
5
|
Ahmad A, Kim SJ, Jeong YJ, Khan MS, Park J, Lee DW, Lee C, Choi YJ, Yi HG. Coaxial bioprinting of a stentable and endothelialized human coronary artery-sized in vitro model. J Mater Chem B 2024; 12:8633-8646. [PMID: 39119756 DOI: 10.1039/d4tb00601a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Atherosclerosis accounts for two-thirds of deaths attributed to cardiovascular diseases, which continue to be the leading cause of mortality. Current clinical management strategies for atherosclerosis, such as angioplasty with stenting, face numerous challenges, including restenosis and late thrombosis. Smart stents, integrated with sensors that can monitor cardiovascular health in real-time, are being developed to overcome these limitations. This development necessitates rigorous preclinical trials on either animal models or in vitro models. Despite efforts being made, a suitable human-scale in vitro model compatible with a cardiovascular stent has remained elusive. To address this need, this study utilizes an in-bath bioprinting method to create a human-scale, freestanding in vitro model compatible with cardiovascular stents. Using a coaxial nozzle, a tubular structure of human coronary artery (HCA) size is bioprinted with a collagen-based bioink, ensuring good biocompatibility and suitable rheological properties for printing. We precisely replicated the dimensions of the HCA, including its internal diameter and wall thickness, and simulated the vascular barrier functionality. To simplify post-processing, a pumpless perfusion bioreactor is fabricated to culture a HCA-sized model, eliminating the need for a peristaltic pump and enabling scalability for high-throughput production. This model is expected to accelerate stent development in the future.
Collapse
Affiliation(s)
- Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Republic of Korea
| | - Seon-Jin Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea
| | - Yun-Jin Jeong
- Department of Automatic System, Chosun College of Science & Technology, Gwangju, 61453, Republic of Korea
| | - Muhammad Soban Khan
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Dong-Weon Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Changho Lee
- Department of Artificial Intelligence Convergence, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, 58128, Republic of Korea
| | - Yeong-Jin Choi
- Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon, 51508, Republic of Korea.
- Advanced Materials Engineering, Korea National University of Science and Technology (UST), Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Republic of Korea
| |
Collapse
|
6
|
Huang H, Liao D, He B, Zhou G, Cui Y. Effects of Citrus Flavanone Hesperidin Extracts or Purified Hesperidin Consumption on Risk Factors for Cardiovascular Disease: Evidence From an Updated Meta-analysis of Randomized Controlled Trials. Curr Dev Nutr 2024; 8:102055. [PMID: 39279783 PMCID: PMC11399677 DOI: 10.1016/j.cdnut.2023.102055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/21/2023] [Accepted: 12/04/2023] [Indexed: 09/18/2024] Open
Abstract
Background Cardiovascular disease (CVD) is a serious public health problem worldwide. The role of citrus flavanone hesperidin consumption on cardiovascular disease risk factors (CVDRFs) has been examined in many clinical trials, but conflicting results have been found. Objectives This study aimed to systematically evaluate the effects of hesperidin extracts or purified hesperidin on CVDRFs in humans with an updated meta-analysis of randomized controlled trials. Methods According to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 guidelines, we systematically screened and searched electronic databases from their establishment to March 2023. Reference lists and previous reviews were also searched. Intervention trials assessing hesperidin consumption on CVD outcomes were included for pooling. To assess the quality of the included articles, the tool of Cochrane risk-of-bias tool was applied. We synthesized the effect sizes with 95% CIs and weighted mean difference (WMD). The I 2 index was used to evaluate the between-study heterogeneity. To explore the heterogeneity source, we used meta-regression and subgroup analysis. Publication bias and sensitivity analysis were also performed. We used the Grading of Recommendations Assessment, Development, and Evaluation approach to evaluate the evidence quality. Results We included 12 trials with 589 participants. We found evident effects of hesperidin on low-density lipoprotein cholesterol (WMD: -0.22 mmol/L; 95% CI: -0.33, -0.11 mmol/L), total cholesterol (WMD: -0.20 mmol/L; 95% CI: -0.31, -0.08 mmol/L), fasting blood glucose (WMD: -0.15 mg/dL; 95% CI: -0.29, -0.02 mg/dL), quantitative insulin-sensitivity check index (WMD 0.06, 95% CI 0.01 to 0.10), intercellular adhesion molecule 1 (WMD: -13.60 ng/mL; 95% CI: -23.72, -3.48 ng/mL), vascular cell adhesion molecule 1 (WMD: -15.60 ng/mL; 95% CI: -30.13, -1.06 ng/mL), and C-reactive protein (WMD: -0.56 mg/L; 95% CI: -1.11, -0.01 mg/L), whereas no effects were found for other CVDRFs. Conclusions Our current findings demonstrate that hesperidin might be advantageous in improving numerous CVDRFs in humans, such as blood lipid concentrations, blood glucose control, and management of inflammatory indicators.
Collapse
Affiliation(s)
- Haohai Huang
- Clinical Translational Medical Center, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
- Department of Clinical Pharmacy, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Dan Liao
- Department of Gynaecology, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bin He
- Clinical Translational Medical Center, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Guanghui Zhou
- Department of Rehabilitation Medicine, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yejia Cui
- Department of Clinical Laboratory, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
7
|
Singh SK, Parihar S, Jain S, Ho JAA, Vankayala R. Light-responsive functional nanomaterials as pioneering therapeutics: a paradigm shift to combat age-related disorders. J Mater Chem B 2024; 12:8212-8234. [PMID: 39058026 DOI: 10.1039/d4tb00578c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Aging, marked by dysregulated cellular systems, gives rise to a spectrum of age-related disorders, including neurodegeneration, atherosclerosis, immunosenescence, and musculoskeletal issues. These conditions contribute significantly to the global disease burden, posing challenges to health span and economic resources. Current therapeutic approaches, although diverse in mechanism, often fall short in targeting the underlying cellular pathologies. They fail to address the issues compounded by altered pharmacokinetics in the elderly. Nanotechnology emerges as a transformative solution, offering tissue-specific targeted therapies through nanoparticles. Functional nanomaterials (FNMs) respond to internal or external stimuli, with light-responsive nanomaterials gaining prominence. Harnessing the benefits of deep tissue penetration and ease of manipulation particularly in the near-infrared spectrum, light-responsive FNMs present innovative strategies for age-related comorbidities. This review comprehensively summarizes the potential of light-responsive FNM-based approaches for targeting cellular environments in age-related disorders, and also emphasizes the advantages over traditional treatment modalities. Specifically, it focuses on the development of various classes of light-responsive functional nanomaterials including plasmonic nanomaterials, nanomaterials as carriers, upconversion nanomaterials, 2D nanomaterials, transition metal oxide and dichalcogenide nanomaterials and carbon-based nanomaterials against age related diseases. We foresee that such advanced developments in the field of nanotechnology could provide a new hope for clinical diagnosis and treatment of age-related disorders.
Collapse
Affiliation(s)
- Shubham Kumar Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Karwar 342030, India.
| | - Shivay Parihar
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Karwar 342030, India.
| | - Sanskar Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Karwar 342030, India.
| | - Ja-An Annie Ho
- Bioanalytical Chemistry and Nanobiomedicine Laboratory, Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
- Center for Emerging Materials and Advanced Devices, National Taiwan University, Taipei 10617, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
| | - Raviraj Vankayala
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Karwar 342030, India.
- Interdisciplinary Research Platform, Smart Healthcare, Indian Institute of Technology Jodhpur, Karwar 342030, India
| |
Collapse
|
8
|
Mo F, Wang C, Li S, Li Z, Xiao C, Zhang Y, Hu C, Wang E, Lin P, Yuan T, Zuo Z, Fu W, Chen X, Ren L, Wang L. A Dual-Targeting, Multi-Faceted Biocompatible Nanodrug Optimizes the Microenvironment to Ameliorate Abdominal Aortic Aneurysm. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405761. [PMID: 38923441 DOI: 10.1002/adma.202405761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Abdominal aortic aneurysm (AAA) is a highly lethal cardiovascular disease that currently lacks effective pharmacological treatment given the complex pathophysiology of the disease. Here, single-cell RNA-sequencing data from patients with AAA and a mouse model are analyzed, which reveals pivotal pathological changes, including the M1-like polarization of macrophages and the loss of contractile function in smooth muscle cells (SMCs). Both cell types express the integrin αvβ3, allowing for their dual targeting with a single rationally designed molecule. To this end, a biocompatible nanodrug, which is termed EVMS@R-HNC, that consists of the multifunctional drug everolimus (EVMS) encapsulated by the hepatitis B virus core protein modifies to contain the RGD sequence to specifically bind to integrin αvβ3 is designed. Both in vitro and in vivo results show that EVMS@R-HNC can target macrophages as well as SMCs. Upon binding of the nanodrug, the EVMS is released intracellularly where it exhibits multiple functions, including inhibiting M1 macrophage polarization, thereby suppressing the self-propagating inflammatory cascade and immune microenvironment imbalance, while preserving the normal contractile function of SMCs. Collectively, these results suggest that EVMS@R-HNC presents a highly promising therapeutic approach for the management of AAA.
Collapse
Affiliation(s)
- Fandi Mo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Chufan Wang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Shiyi Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Zheyun Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Cheng Xiao
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Yuchong Zhang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Chengkai Hu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Peng Lin
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Tong Yuan
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Ziang Zuo
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Lei Ren
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen, 361005, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Vascular Surgery Institute of Fudan University, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Vascular Surgery (Xiamen), Zhongshan hospital, Fudan University, Xiamen, 361015, China
| |
Collapse
|
9
|
Kounatidis D, Tentolouris N, Vallianou NG, Mourouzis I, Karampela I, Stratigou T, Rebelos E, Kouveletsou M, Stamatopoulos V, Tsaroucha E, Dalamaga M. The Pleiotropic Effects of Lipid-Modifying Interventions: Exploring Traditional and Emerging Hypolipidemic Therapies. Metabolites 2024; 14:388. [PMID: 39057711 PMCID: PMC11278853 DOI: 10.3390/metabo14070388] [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: 06/18/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerotic cardiovascular disease poses a significant global health issue, with dyslipidemia standing out as a major risk factor. In recent decades, lipid-lowering therapies have evolved significantly, with statins emerging as the cornerstone treatment. These interventions play a crucial role in both primary and secondary prevention by effectively reducing cardiovascular risk through lipid profile enhancements. Beyond their primary lipid-lowering effects, extensive research indicates that these therapies exhibit pleiotropic actions, offering additional health benefits. These include anti-inflammatory properties, improvements in vascular health and glucose metabolism, and potential implications in cancer management. While statins and ezetimibe have been extensively studied, newer lipid-lowering agents also demonstrate similar pleiotropic effects, even in the absence of direct cardiovascular benefits. This narrative review explores the diverse pleiotropic properties of lipid-modifying therapies, emphasizing their non-lipid effects that contribute to reducing cardiovascular burden and exploring emerging benefits for non-cardiovascular conditions. Mechanistic insights into these actions are discussed alongside their potential therapeutic implications.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | | | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
10
|
Huang Z, Zhou Z, Ma Y, Hu YM. Mito-Tempo alleviates ox-LDL-provoked foam cell formation by regulating Nrf2/NLRP3 signaling. Biosci Biotechnol Biochem 2024; 88:759-767. [PMID: 38719485 DOI: 10.1093/bbb/zbae058] [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: 11/30/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024]
Abstract
Our previous studies have demonstrated that Mito-Tempol (also known as 4-hydroxy-Tempo), a mitochondrial reactive oxygen species scavenger, alleviates oxidized low-density lipoprotein (ox-LDL)-triggered foam cell formation. Given the effect of oxidative stress on activating the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome, which promotes foam cell formation, we aimed to explore whether Mito-Tempo inhibits ox-LDL-triggered foam cell formation by regulating NLRP3 inflammasome. The results revealed that Mito-Tempo re-activated Nrf2 and alleviated macrophage foam cell formation induced by ox-LDL, whereas the effects were reversed by ML385 (a specific Nrf2 inhibitor). Mito-Tempo restored the expression and nuclear translocation of Nrf2 by decreasing ox-LDL-induced ubiquitination. Furthermore, Mito-Tempo suppressed ox-LDL-triggered NLRP3 inflammasome activation and subsequent pyroptosis, whereas the changes were blocked by ML385. Mito-Tempo decreased lipoprotein uptake by inhibiting CD36 expression and suppressed foam cell formation by regulating the NLRP3 inflammasome. Taken together, Mito-Tempo exhibits potent anti-atherosclerotic effects by regulating Nrf2/NLRP3 signaling.
Collapse
Affiliation(s)
- Zhenyu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhaoli Zhou
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Scientific Research Center, Shanghai University of Medicine & Health Science, Shanghai, China
- Department of Pharmacology, School of Pharmacy, Shanghai University of Medicine & Health Science, Shanghai, China
| | - Ying Ma
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao-Min Hu
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
11
|
Xu F, Chen H, Zhou C, Zang T, Wang R, Shen S, Li C, Yu Y, Pei Z, Shen L, Qian J, Ge J. Targeting deubiquitinase OTUB1 protects vascular smooth muscle cells in atherosclerosis by modulating PDGFRβ. Front Med 2024; 18:465-483. [PMID: 38644399 DOI: 10.1007/s11684-024-1056-8] [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: 07/20/2023] [Accepted: 12/04/2023] [Indexed: 04/23/2024]
Abstract
Atherosclerosis is a chronic artery disease that causes various types of cardiovascular dysfunction. Vascular smooth muscle cells (VSMCs), the main components of atherosclerotic plaque, switch from contractile to synthetic phenotypes during atherogenesis. Ubiquitylation is crucial in regulating VSMC phenotypes in atherosclerosis, and it can be reversely regulated by deubiquitinases. However, the specific effects of deubiquitinases on atherosclerosis have not been thoroughly elucidated. In this study, RNAi screening in human aortic smooth muscle cells was performed to explore the effects of OTU family deubiquitinases, which revealed that silencing OTUB1 inhibited PDGF-BB-stimulated VSMC phenotype switch. Further in vivo studies using Apoe-/- mice revealed that knockdown of OTUB1 in VSMCs alleviated atherosclerosis plaque burden in the advanced stage and led to a stable plaque phenotype. Moreover, VSMC proliferation and migration upon PDGF-BB stimulation could be inhibited by silencing OTUB1 in vitro. Unbiased RNA-sequencing data indicated that knocking down OTUB1 influenced VSMC differentiation, adhesion, and proliferation. Mass spectrometry of ubiquitinated protein confirmed that proteins related to cell growth and migration were differentially ubiquitylated. Mechanistically, we found that OTUB1 recognized the K707 residue ubiquitylation of PDGFRβ with its catalytic triad, thereby reducing the K48-linked ubiquitylation of PDGFRβ. Inhibiting OTUB1 in VSMCs could promote PDGFRβ degradation via the ubiquitin-proteasome pathway, so it was beneficial in preventing VSMCs' phenotype switch. These findings revealed that knocking down OTUB1 ameliorated VSMCs' phenotype switch and atherosclerosis progression, indicating that OTUB1 could be a valuable translational therapeutic target in the future.
Collapse
Affiliation(s)
- Fei Xu
- Department of Cardiology and Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Han Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Changyi Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Tongtong Zang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Rui Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Shutong Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Yue Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Zhiqiang Pei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| |
Collapse
|
12
|
Wei Q, Xiao Y, Du L, Li Y. Advances in Nanoparticles in the Prevention and Treatment of Myocardial Infarction. Molecules 2024; 29:2415. [PMID: 38893291 PMCID: PMC11173599 DOI: 10.3390/molecules29112415] [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: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) is one of the most prevalent types of cardiovascular disease. During MI, myocardial cells become ischemic and necrotic due to inadequate blood perfusion, leading to irreversible damage to the heart. Despite the development of therapeutic strategies for the prevention and treatment of MI, their effects are still unsatisfactory. Nanoparticles represent a new strategy for the pre-treatment and treatment of MI, and novel multifunctional nanoparticles with preventive and therapeutic capabilities hold promise for the prevention and treatment of this disease. This review summarizes the common types and properties of nanoparticles, and focuses on the research progress of nanoparticles for the prevention and treatment of MI.
Collapse
Affiliation(s)
| | | | | | - Ya Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Q.W.); (Y.X.); (L.D.)
| |
Collapse
|
13
|
Sun Z, Yun Z, Lin J, Sun X, Wang Q, Duan J, Li C, Zhang X, Xu S, Wang Z, Xiong X, Yao K. Comprehensive mendelian randomization analysis of plasma proteomics to identify new therapeutic targets for the treatment of coronary heart disease and myocardial infarction. J Transl Med 2024; 22:404. [PMID: 38689297 PMCID: PMC11061979 DOI: 10.1186/s12967-024-05178-8] [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: 02/25/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Ischemic heart disease is one of the leading causes of mortality worldwide, and thus calls for development of more effective therapeutic strategies. This study aimed to identify potential therapeutic targets for coronary heart disease (CHD) and myocardial infarction (MI) by investigating the causal relationship between plasma proteins and these conditions. METHODS A two-sample Mendelian randomization (MR) study was performed to evaluate more than 1600 plasma proteins for their causal associations with CHD and MI. The MR findings were further confirmed through Bayesian colocalization, Summary-data-based Mendelian Randomization (SMR), and Transcriptome-Wide Association Studies (TWAS) analyses. Further analyses, including enrichment analysis, single-cell analysis, MR analysis of cardiovascular risk factors, phenome-wide Mendelian Randomization (Phe-MR), and protein-protein interaction (PPI) network construction were conducted to verify the roles of selected causal proteins. RESULTS Thirteen proteins were causally associated with CHD, seven of which were also causal for MI. Among them, FES and PCSK9 were causal proteins for both diseases as determined by several analytical methods. PCSK9 was a risk factor of CHD (OR = 1.25, 95% CI: 1.13-1.38, P = 7.47E-06) and MI (OR = 1.36, 95% CI: 1.21-1.54, P = 2.30E-07), whereas FES was protective against CHD (OR = 0.68, 95% CI: 0.59-0.79, P = 6.40E-07) and MI (OR = 0.65, 95% CI: 0.54-0.77, P = 5.38E-07). Further validation through enrichment and single-cell analysis confirmed the causal effects of these proteins. Moreover, MR analysis of cardiovascular risk factors, Phe-MR, and PPI network provided insights into the potential drug development based on the proteins. CONCLUSIONS This study investigated the causal pathways associated with CHD and MI, highlighting the protective and risk roles of FES and PCSK9, respectively. FES. Specifically, the results showed that these proteins are promising therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Ziyi Sun
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Zhangjun Yun
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 10070, China
| | - Jianguo Lin
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, 10070, China
| | - Xiaoning Sun
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, 10070, China
| | - Qingqing Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Jinlong Duan
- Department of Andrology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Cheng Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, 10040, China
| | - Xiaoxiao Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, 10070, China
| | - Siyu Xu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Zeqi Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 10070, China
| | - Xingjiang Xiong
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| | - Kuiwu Yao
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, 10040, China.
| |
Collapse
|
14
|
Zhang S, Zhang Q, Lu Y, Chen J, Liu J, Li Z, Xie Z. Roles of Integrin in Cardiovascular Diseases: From Basic Research to Clinical Implications. Int J Mol Sci 2024; 25:4096. [PMID: 38612904 PMCID: PMC11012347 DOI: 10.3390/ijms25074096] [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: 02/23/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cardiovascular diseases (CVDs) pose a significant global health threat due to their complex pathogenesis and high incidence, imposing a substantial burden on global healthcare systems. Integrins, a group of heterodimers consisting of α and β subunits that are located on the cell membrane, have emerged as key players in mediating the occurrence and progression of CVDs by regulating the physiological activities of endothelial cells, vascular smooth muscle cells, platelets, fibroblasts, cardiomyocytes, and various immune cells. The crucial role of integrins in the progression of CVDs has valuable implications for targeted therapies. In this context, the development and application of various integrin antibodies and antagonists have been explored for antiplatelet therapy and anti-inflammatory-mediated tissue damage. Additionally, the rise of nanomedicine has enhanced the specificity and bioavailability of precision therapy targeting integrins. Nevertheless, the complexity of the pathogenesis of CVDs presents tremendous challenges for monoclonal targeted treatment. This paper reviews the mechanisms of integrins in the development of atherosclerosis, cardiac fibrosis, hypertension, and arrhythmias, which may pave the way for future innovations in the diagnosis and treatment of CVDs.
Collapse
Affiliation(s)
- Shuo Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Qingfang Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Yutong Lu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China; (S.Z.); (Q.Z.); (Y.L.); (J.C.); (J.L.); (Z.L.)
| |
Collapse
|
15
|
Volpatti LR, de Matos SN, Borjas G, Reda J, Watkins EA, Zhou Z, Nguyen M, Solanki A, Fang Y, Hubbell JA. LDL-Binding IL-10 Reduces Vascular Inflammation in Atherosclerotic Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.582839. [PMID: 38496521 PMCID: PMC10942346 DOI: 10.1101/2024.03.04.582839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease associated with the accumulation of low-density lipoprotein (LDL) in arterial walls. Higher levels of the anti-inflammatory cytokine IL-10 in serum are correlated with reduced plaque burden. However, cytokine therapies have not translated well to the clinic, partially due to their rapid clearance and pleiotropic nature. Here, we engineered IL-10 to overcome these challenges by hitchhiking on LDL to atherosclerotic plaques. Specifically, we constructed fusion proteins in which one domain is IL-10 and the other is an antibody fragment (Fab) that binds to protein epitopes of LDL. In murine models of atherosclerosis, we show that systemically administered Fab-IL-10 constructs bind circulating LDL and traffic to atherosclerotic plaques. One such construct, 2D03-IL-10, significantly reduces aortic immune cell infiltration to levels comparable to healthy mice, whereas non-targeted IL-10 has no therapeutic effect. Mechanistically, we demonstrate that 2D03-IL-10 preferentially associates with foamy macrophages and reduces pro-inflammatory activation markers. This platform technology can be applied to a variety of therapeutics and shows promise as a potential targeted anti-inflammatory therapy in atherosclerosis.
Collapse
Affiliation(s)
- Lisa R. Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, United States
| | - Salvador Norton de Matos
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Medical Scientist Training Program, Pritzker School of Medicine, University of Chicago, IL, 60637, United States
| | - Gustavo Borjas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Joseph Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Zhengjie Zhou
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Yun Fang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, USA
- Committee on Molecular Medicine, University of Chicago, Chicago, IL, 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Committee on Immunology, University of Chicago, Chicago, IL 60637, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
16
|
He Y, Cai Y, Wei D, Cao L, He Q, Zhang Y. Elucidating the mechanisms of formononetin in modulating atherosclerotic plaque formation in ApoE-/- mice. BMC Cardiovasc Disord 2024; 24:121. [PMID: 38388385 PMCID: PMC10882812 DOI: 10.1186/s12872-024-03774-6] [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: 11/28/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Atherosclerosis(AS) poses a pressing challenge in contemporary medicine. Formononetin (FMN) plays a crucial role in its prevention and treatment. However, the detailed impact of FMN on the stability of atherosclerotic plaques and its underlying mechanisms remain to be elucidated. METHODS An intervention consisting of FMN was given along with a high-fat food regimen in the ApoE-/- mouse model. The investigation included the evaluation of the degree of atherosclerotic lesion, the main components of the plaque, lipid profiles, particular markers indicating M1/M2 macrophage phenotypes, the quantities of factors related to inflammation, the infiltration of macrophages, and the identification of markers linked to the α7nAChR/JAK2/STAT3 axis effect molecules. RESULTS The evaluation of aortic morphology in ApoE-/-mice revealed that FMN significantly improved the plaque area, fibrous cap protrusion, lipid deposition, and structural alterations on the aortic surface, among other markers of atherosclerosis,and there is concentration dependence. Furthermore, the lipid content of mouse serum was assessed, and the results showed that the low-, medium-, and high-dosage FMN groups had significantly lower levels of LDL-C, ox-LDL, TC, and TG. The results of immunohistochemical staining indicated that the low-, medium-, and high-dose FMN therapy groups had enhanced CD206 expression and decreased expression of CD68 and iNOS. According to RT-qPCR data, FMN intervention has the potential to suppress the expression of iNOS, COX-2, miR-155-5p, IL-6, and IL-1β mRNA, while promoting the expression of IL-10, SHIP1, and Arg-1 mRNA levels. However, the degree of inhibition varied among dosage groups. Western blot investigation of JAK/STAT signaling pathway proteins and cholinergic α7nAChR protein showed that p-JAK2 and p-STAT3 protein expression was suppressed at all dosages, whereas α7nAChR protein expression was enhanced. CONCLUSIONS According to the aforementioned findings, FMN can reduce inflammation and atherosclerosis by influencing macrophage polarization, blocking the JAK/STAT signaling pathway, and increasing α7nAChR expression.
Collapse
Affiliation(s)
- Ying He
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Youde Cai
- Jinyang Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550081, China
| | - Dingling Wei
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Liping Cao
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China
| | - Qiansong He
- First Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550001, China.
| | - Yazhou Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
17
|
Zhang Y, Qi Y, Jia Z, Li Y, Wu L, Zhou Q, Xu F. Effects and mechanisms of Zhizi Chuanxiong herb pair against atherosclerosis: an integration of network pharmacology, molecular docking, and experimental validation. Chin Med 2024; 19:8. [PMID: 38212797 PMCID: PMC10782628 DOI: 10.1186/s13020-023-00874-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/25/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND The Zhizi Chuanxiong herb pair (ZCHP) can delay pathological progression of atherosclerosis (AS); however, its pharmacological mechanism remains unclear because of its complex components. The purpose of current study is to systematically investigate the anti-AS mechanism of ZCHP. METHODS The databases of TCMSP, STITCH, SwissTargetPrediction, BATMAN-TCM, and ETCM were searched to predict the potential targets of ZCHP components. Disease targets associated with AS was retrieved from the GEO database. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway analyses were executed using DAVID 6.8. Molecular docking method was employed to evaluate the core target binding to blood components, and animal experiments were performed to test action mechanism. RESULTS A ZCHP-components-targets-AS network was constructed by using Cytoscape, included 11 main components and 52 candidate targets. Crucial genes were shown in the protein-protein interaction network, including TNF, IL-1β, IGF1, MMP9, COL1A1, CCR5, HMOX1, PTGS1, SELE, and SYK. KEGG enrichment illustrated that the NF-κB, Fc epsilon RI, and TNF signaling pathways were important for AS treatment. These results were validated by molecular docking. In ApoE-/- mice, ZCHP significantly reduced intima-media thickness, pulse wave velocity, plaque area, and serum lipid levels while increasing the difference between the end-diastolic and end-systolic diameters. Furthermore, ZCHP significantly decreased the mRNA and protein levels of TNF-α and IL-1β, suppressed NF-κB activation, and inhibited the M1 macrophage polarization marker CD86 in ApoE-/- mice. CONCLUSION This study combining network pharmacology, molecular biology, and animal experiments showed that ZCHP can alleviate AS by suppressing the TNF/NF-κB axis and M1 macrophage polarization.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yifei Qi
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zijun Jia
- Comprehensive Care of China-Japan Friendship Hospital, Beijing, China
| | - Yiming Li
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Integrated Traditional and Western Medicine, Peking University, Beijing, China
| | - Liqi Wu
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingbing Zhou
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Fengqin Xu
- Institute of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Zhan M, Ma M, Mo X, Zhang Y, Li T, Yang Y, Dong L. Dracocephalum moldavica L.: An updated comprehensive review of its botany, traditional uses, phytochemistry, pharmacology, and application aspects. Fitoterapia 2024; 172:105732. [PMID: 37952760 DOI: 10.1016/j.fitote.2023.105732] [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: 08/26/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
Dracocephalum moldavica, known as Xiang-qing-lan (in Chinese), is a traditional folk medicine, which was commonly used by Mongolian and Xinjiang Uyghurs area. Dracocephalum moldavica has the effects of purging liver fire, clearing stomach heat, hemostasis. It is used for treating insufficient heart and blood, weakened brain function, weak feeling and spirit disease etc. This review aimed to summarize the botany, traditional uses, phytochemistry, pharmacology and application of Dracocephalum moldavica, which expected to provide theoretical support for future utilization and highlight the further investigation of this vital plant. In addition to the essential oil, approximately 154 compounds have been isolated and identified from aerial parts of the Dracocephalum moldavica, including flavonoids, terpenoids, lignans, phenylpropanoids, phenols, glycosides, polysaccharide and other compounds. Extensive pharmacological activities of the extracts or compounds of Dracocephalum moldavica in vivo and in vitro were confirmed including cardiovascular protection, antioxidative, antimicrobial, antifungal, anti-complementary and chronic mountain sickness. Moreover, Dracocephalum moldavica is used in a wide range of applications in food, biological pesticides and cosmetics. In the future, Dracocephalum moldavica needs further study, such as paying more attention to quality control, toxicity, pharmacological mechanism and pharmacokinetics.
Collapse
Affiliation(s)
- Meng Zhan
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Miao Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Xiyan Mo
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yuanyuan Zhang
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Tingting Li
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education (Ningxia Medical University), Yinchuan 750004, China
| | - Yong Yang
- Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan 750004, China
| | - Lin Dong
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; Ningxia Collaborative Innovation Center of Regional Characteristic Traditional Chinese Medicine, Yinchuan 750004, China; Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education (Ningxia Medical University), Yinchuan 750004, China.
| |
Collapse
|
19
|
Wong M, Dai Y, Ge J. Pan-vascular disease: what we have done in the past and what we can do in the future? CARDIOLOGY PLUS 2024; 9:1-5. [PMID: 38584611 PMCID: PMC10994062 DOI: 10.1097/cp9.0000000000000078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 04/09/2024] Open
Affiliation(s)
- Mingjen Wong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
20
|
Namous H, Strillacci MG, Braz CU, Shanmuganayagam D, Krueger C, Peppas A, Soffregen WC, Reed J, Granada JF, Khatib H. ITGB2 is a central hub-gene associated with inflammation and early fibro-atheroma development in a swine model of atherosclerosis. ATHEROSCLEROSIS PLUS 2023; 54:30-41. [PMID: 38116576 PMCID: PMC10728570 DOI: 10.1016/j.athplu.2023.11.001] [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: 05/12/2023] [Revised: 09/14/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023]
Abstract
Background and aim The complex dynamic interplay between different biological pathways involved in atherosclerosis development has rendered the identification of specific therapeutic targets a challenging quest. We aimed to identify specific genes and mechanistic pathways associated with the early development of fibro-atheromas in a swine model of atherosclerosis. Methods The Wisconsin Miniature Swine™ model of Familial Hypercholesterolemia (WMS-FH, n = 11) and genetically related WMS controls (WMS-N, n = 11) were used. The infrarenal aorta was harvested from both groups for histopathologic and transcriptomic profiling at 12 months. Bioinformatic analysis was performed to identify hub genes and pathways central to disease pathophysiology. The expression of ITGB2, the top ranked hub gene, was manipulated in cell culture and the expression of interconnected genes was tested. Results Fibro-atheromatous lesions were documented in all WMS-FH aortic tissues and displayed internal elastic lamina (IEL) disruption, significant reduction of myofibroblast presence and disorganized collagen deposition. No fibro-atheromas were observed in the control group. A total of 266 differentially expressed genes (DEGs) were upregulated in WMS-FH aortic tissues, while 29 genes were downregulated. Top identified hub genes included ITGB2, C1QA, LCP2, SPI1, CSF1R, C5AR1, CTSS, MPEG1, C1QC, and CSF2RB. Overexpression of ITGB2 resulted in elevated expression of other interconnected genes expressed in porcine endothelial cells. Conclusion In a swine translational model of atherosclerosis, transcriptomic analysis identified ITGB2 as a central hub gene associated inflammation and early fibroatheroma development making it a potential therapeutic target at this stage of disease.
Collapse
Affiliation(s)
- Hadjer Namous
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | | | - Camila Urbano Braz
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | | | - Christian Krueger
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | - Athanasios Peppas
- Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - William C. Soffregen
- Northstar Preclinical and Pathology Services, LLC and Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - Jess Reed
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| | - Juan F. Granada
- Skirball Center for Innovation, Cardiovascular Research Foundation, New York, NY, USA
| | - Hasan Khatib
- Department of Animal and Dairy Sciences – University of Wisconsin Madison, WI, USA
| |
Collapse
|
21
|
Tscheuschner L, Tzafriri AR. Cardiovascular Tissue Engineering Models for Atherosclerosis Treatment Development. Bioengineering (Basel) 2023; 10:1373. [PMID: 38135964 PMCID: PMC10740643 DOI: 10.3390/bioengineering10121373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
In the early years of tissue engineering, scientists focused on the generation of healthy-like tissues and organs to replace diseased tissue areas with the aim of filling the gap between organ demands and actual organ donations. Over time, the realization has set in that there is an additional large unmet need for suitable disease models to study their progression and to test and refine different treatment approaches. Increasingly, researchers have turned to tissue engineering to address this need for controllable translational disease models. We review existing and potential uses of tissue-engineered disease models in cardiovascular research and suggest guidelines for generating adequate disease models, aimed both at studying disease progression mechanisms and supporting the development of dedicated drug-delivery therapies. This involves the discussion of different requirements for disease models to test drugs, nanoparticles, and drug-eluting devices. In addition to realistic cellular composition, the different mechanical and structural properties that are needed to simulate pathological reality are addressed.
Collapse
Affiliation(s)
- Linnea Tscheuschner
- Department of Vascular Surgery, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Abraham R. Tzafriri
- Department of Research and Innovation, CBSET Inc., Lexington, MA 02421, USA;
| |
Collapse
|
22
|
Altabas V, Marinković Radošević J, Špoljarec L, Uremović S, Bulum T. The Impact of Modern Anti-Diabetic Treatment on Endothelial Progenitor Cells. Biomedicines 2023; 11:3051. [PMID: 38002051 PMCID: PMC10669792 DOI: 10.3390/biomedicines11113051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes is one of the leading chronic diseases globally with a significant impact on mortality. This condition is associated with chronic microvascular and macrovascular complications caused by vascular damage. Recently, endothelial progenitor cells (EPCs) raised interest due to their regenerative properties. EPCs are mononuclear cells that are derived from different tissues. Circulating EPCs contribute to regenerating the vessel's intima and restoring vascular function. The ability of EPCs to repair vascular damage depends on their number and functionality. Diabetic patients have a decreased circulating EPC count and impaired EPC function. This may at least partially explain the increased risk of diabetic complications, including the increased cardiovascular risk in these patients. Recent studies have confirmed that many currently available drugs with proven cardiovascular benefits have beneficial effects on EPC count and function. Among these drugs are also medications used to treat different types of diabetes. This manuscript aims to critically review currently available evidence about the ways anti-diabetic treatment affects EPC biology and to provide a broader context considering cardiovascular complications. The therapies that will be discussed include lifestyle adjustments, metformin, sulphonylureas, gut glucosidase inhibitors, thiazolidinediones, dipeptidyl peptidase 4 inhibitors, glucagon-like peptide 1 receptor analogs, sodium-glucose transporter 2 inhibitors, and insulin.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Jelena Marinković Radošević
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | - Lucija Špoljarec
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
| | | | - Tomislav Bulum
- Department of Endocrinology, Diabetes and Metabolic Diseases, Sestre Milosrdnice University Clinical Hospital, 10000 Zagreb, Croatia
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 10000 Zagreb, Croatia
| |
Collapse
|
23
|
Geng S, Lu R, Zhang Y, Wu Y, Xie L, Caldwell B, Pradhan K, Yi Z, Hou J, Xu F, Chen X, Li L. Monocytes reprogrammed by 4-PBA potently contribute to the resolution of inflammation and atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563200. [PMID: 37961551 PMCID: PMC10634693 DOI: 10.1101/2023.10.19.563200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis. Methods Systems analyses integrating single-cell RNA-sequencing and complementary immunological approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming was assessed by integrated biochemical and genetic approaches. The inter-cellular propagation of homeostasis resolution was evaluated by co-culture assays with donor monocytes trained by 4-PBA and recipient naïve monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high fat diet-fed ApoE -/- mouse model with i.p. 4-PBA administration. Furthermore, the selective efficacy of 4-PBA trained monocytes were examined by i.v. transfusion of ex vivo trained monocytes by 4-PBA into recipient high fat diet-fed ApoE -/- mice. Results In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 via reducing peroxisome stress and attenuating SYK-mTOR signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ neddylation mediated by TOLLIP. 4-PBA trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo . Conclusion Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision-therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.
Collapse
|
24
|
Aleman Oliva C, Aleman Espino E, Demory Beckler M, Kesselman MM. Radiation-Induced Peripheral Artery Disease in a 63-Year-Old Patient. Cureus 2023; 15:e47372. [PMID: 38022038 PMCID: PMC10657155 DOI: 10.7759/cureus.47372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Tobacco use, hypertension, diabetes, and hypercholesterolemia are known risk factors for peripheral artery disease (PAD). However, additional causes of PAD, such as radiation therapy, should be considered for the prevention and diagnosis of this disease. The patient described in this report had 36 radiation therapies directly to the pelvis and bladder area due to bladder cancer. The presence of severe PAD on this patient's right external iliac artery, the same area where he received radiation therapy, raises the question of whether radiation therapy contributed to the development of PAD. In addition, his history of anal intraepithelial neoplasia, obstructive uropathy, and chronic kidney disease further demonstrated that he possibly suffered extensive tissue damage due to radiation to the pelvis. This case report explores the current diagnosis guidelines and treatment options for patients with radiation-induced PAD. Through this case study, we aim to bring awareness to this lesser-known cause of PAD among medical providers and promote research for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Claudia Aleman Oliva
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Erik Aleman Espino
- Osteopathic Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Marc M Kesselman
- Rheumatology, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
25
|
Chen X, Xia Y, Shen S, Wang C, Zan R, Yu H, Yang S, Zheng X, Yang J, Suo T, Gu Y, Zhang X. Research on the Current Application Status of Magnesium Metal Stents in Human Luminal Cavities. J Funct Biomater 2023; 14:462. [PMID: 37754876 PMCID: PMC10532415 DOI: 10.3390/jfb14090462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023] Open
Abstract
The human body comprises various tubular structures that have essential functions in different bodily systems. These structures are responsible for transporting food, liquids, waste, and other substances throughout the body. However, factors such as inflammation, tumors, stones, infections, or the accumulation of substances can lead to the narrowing or blockage of these tubular structures, which can impair the normal function of the corresponding organs or tissues. To address luminal obstructions, stenting is a commonly used treatment. However, to minimize complications associated with the long-term implantation of permanent stents, there is an increasing demand for biodegradable stents (BDS). Magnesium (Mg) metal is an exceptional choice for creating BDS due to its degradability, good mechanical properties, and biocompatibility. Currently, the Magmaris® coronary stents and UNITY-BTM biliary stent have obtained Conformité Européene (CE) certification. Moreover, there are several other types of stents undergoing research and development as well as clinical trials. In this review, we discuss the required degradation cycle and the specific properties (anti-inflammatory effect, antibacterial effect, etc.) of BDS in different lumen areas based on the biocompatibility and degradability of currently available magnesium-based scaffolds. We also offer potential insights into the future development of BDS.
Collapse
Affiliation(s)
- Xiang Chen
- School of Medicine, Anhui University of Science and Technology, Huainan 232000, China;
| | - Yan Xia
- School of Stomatology, Anhui Medical College, Hefei 230601, China;
| | - Sheng Shen
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.S.); (R.Z.); (T.S.)
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China;
| | - Chunyan Wang
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China;
- Department of General Surgery, Shanghai Xuhui Central Hospital, Shanghai 200031, China
| | - Rui Zan
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.S.); (R.Z.); (T.S.)
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China;
| | - Han Yu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; (H.Y.); (S.Y.)
| | - Shi Yang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; (H.Y.); (S.Y.)
| | - Xiaohong Zheng
- Department of Hepatopancreatobiliary Surgery, Huainan Xinhua Hospital Affiliated to Anhui University of Science and Technology, Huainan 232000, China; (X.Z.); (J.Y.)
| | - Jiankang Yang
- Department of Hepatopancreatobiliary Surgery, Huainan Xinhua Hospital Affiliated to Anhui University of Science and Technology, Huainan 232000, China; (X.Z.); (J.Y.)
| | - Tao Suo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (S.S.); (R.Z.); (T.S.)
- Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, Shanghai 200032, China;
| | - Yaqi Gu
- School of Medicine, Anhui University of Science and Technology, Huainan 232000, China;
- Department of Hepatopancreatobiliary Surgery, Huainan Xinhua Hospital Affiliated to Anhui University of Science and Technology, Huainan 232000, China; (X.Z.); (J.Y.)
| | - Xiaonong Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; (H.Y.); (S.Y.)
| |
Collapse
|
26
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
27
|
González‐López P, Álvarez‐Villarreal M, Ruiz‐Simón R, López‐Pastor AR, de Ceniga MV, Esparza L, Martín‐Ventura JL, Escribano Ó, Gómez‐Hernández A. Role of miR-15a-5p and miR-199a-3p in the inflammatory pathway regulated by NF-κB in experimental and human atherosclerosis. Clin Transl Med 2023; 13:e1363. [PMID: 37605307 PMCID: PMC10442475 DOI: 10.1002/ctm2.1363] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) prevalence has significantly increased in the last decade and atherosclerosis development is the main trigger. MicroRNAs (miRNAs) are non-coding RNAs that negatively regulate gene expression of their target and their levels are frequently altered in CVDs. METHODS By RT-qPCR, we analysed miR-9-5p, miR-15a-5p, miR-16-5p and miR-199a-3p levels in aorta from apolipoprotein knockout (ApoE-/- ) mice, an experimental model of hyperlipidemia-induced atherosclerosis, and in human aortic and carotid atherosclerotic samples. By in silico studies, Western blot analysis and immunofluorescence studies, we detected the targets of the altered miRNAs. RESULTS Our results show that miR-15a-5p and miR-199a-3p are significantly decreased in carotid and aortic samples from patients and mice with atherosclerosis. In addition, we found an increased expression in targets of both miRNAs that participate in the inflammatory pathway of nuclear factor kappa B (NF-κB), such as IKKα, IKKβ and p65. In human vein endothelial cells (HUVECs) and vascular smooth muscle cells (VSMCs), the overexpression of miR-15a-5p or miR-199a-3p decreased IKKα, IKKβ and p65 protein levels as well as NF-κB activation. On the other hand, miR-15a-5p and miR-199a-3p overexpression reduced ox-LDL uptake and the inflammation regulated by NF-κB in VSMCs. Moreover, although miR-15a-5p and miR-199a-3p were significantly increased in exosomes from patients with advanced carotid atherosclerosis, only in the ROC analyses for miR-15a-5p, the area under the curve was 0.8951 with a p value of .0028. CONCLUSIONS Our results suggest that the decrease of miR-199a-3p and miR-15a-5p in vascular samples from human and experimental atherosclerosis could be involved in the NF-κB activation pathway, as well as in ox-LDL uptake by VSMCs, contributing to inflammation and progression atherosclerosis. Finally, miR-15a-5p could be used as a novel diagnostic biomarker for advanced atherosclerosis.
Collapse
Affiliation(s)
- Paula González‐López
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Marta Álvarez‐Villarreal
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Rubén Ruiz‐Simón
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Andrea R. López‐Pastor
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Melina Vega de Ceniga
- Department of Angiology and Vascular SurgeryHospital of Galdakao‐UsansoloGaldakaoBizkaiaSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoBizkaiaSpain
| | - Leticia Esparza
- Department of Angiology and Vascular SurgeryHospital of Galdakao‐UsansoloGaldakaoBizkaiaSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoBizkaiaSpain
| | | | - Óscar Escribano
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| | - Almudena Gómez‐Hernández
- Hepatic and Vascular Diseases Laboratory. Biochemistry and Molecular Biology Department, School of PharmacyComplutense University of MadridMadridSpain
| |
Collapse
|
28
|
Puricelli C, Gigliotti CL, Stoppa I, Sacchetti S, Pantham D, Scomparin A, Rolla R, Pizzimenti S, Dianzani U, Boggio E, Sutti S. Use of Poly Lactic-co-glycolic Acid Nano and Micro Particles in the Delivery of Drugs Modulating Different Phases of Inflammation. Pharmaceutics 2023; 15:1772. [PMID: 37376219 DOI: 10.3390/pharmaceutics15061772] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation contributes to the pathogenesis of many diseases, including apparently unrelated conditions such as metabolic disorders, cardiovascular diseases, neurodegenerative diseases, osteoporosis, and tumors, but the use of conventional anti-inflammatory drugs to treat these diseases is generally not very effective given their adverse effects. In addition, some alternative anti-inflammatory medications, such as many natural compounds, have scarce solubility and stability, which are associated with low bioavailability. Therefore, encapsulation within nanoparticles (NPs) may represent an effective strategy to enhance the pharmacological properties of these bioactive molecules, and poly lactic-co-glycolic acid (PLGA) NPs have been widely used because of their high biocompatibility and biodegradability and possibility to finely tune erosion time, hydrophilic/hydrophobic nature, and mechanical properties by acting on the polymer's composition and preparation technique. Many studies have been focused on the use of PLGA-NPs to deliver immunosuppressive treatments for autoimmune and allergic diseases or to elicit protective immune responses, such as in vaccination and cancer immunotherapy. By contrast, this review is focused on the use of PLGA NPs in preclinical in vivo models of other diseases in which a key role is played by chronic inflammation or unbalance between the protective and reparative phases of inflammation, with a particular focus on intestinal bowel disease; cardiovascular, neurodegenerative, osteoarticular, and ocular diseases; and wound healing.
Collapse
Affiliation(s)
- Chiara Puricelli
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Ian Stoppa
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Sara Sacchetti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Deepika Pantham
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
29
|
Chen M, Fang CY, Guo JC, Pang LM, Zhou Y, Hong Y, Yang LF, Zhang J, Zhang T, Zhou BF, Hu GQ. Predictive value of atherogenic index of plasma and atherogenic index of plasma combined with low-density lipoprotein cholesterol for the risk of acute myocardial infarction. Front Cardiovasc Med 2023; 10:1117362. [PMID: 37304956 PMCID: PMC10250605 DOI: 10.3389/fcvm.2023.1117362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Background and aims Acute myocardial infarction (AMI) is a prevalent medical condition associated with significant morbidity and mortality rates. The principal underlying factor leading to myocardial infarction is atherosclerosis, with dyslipidemia being a key risk factor. Nonetheless, relying solely on a single lipid level is insufficient for accurately predicting the onset and progression of AMI. The present investigation aims to assess established clinical indicators in China, to identify practical, precise, and effective tools for predicting AMI. Methods The study enrolled 267 patients diagnosed with acute myocardial infarction as the experimental group, while the control group consisted of 73 hospitalized patients with normal coronary angiography. The investigators collected general clinical data and relevant laboratory test results and computed the Atherogenic Index of Plasma (AIP) for each participant. Using acute myocardial infarction status as the dependent variable and controlling for confounding factors such as smoking history, fasting plasma glucose (FPG), low-density lipoprotein cholesterol (LDL-C), blood pressure at admission, and diabetes history, the researchers conducted multivariate logistic regression analysis with AIP as an independent variable. Receiver operating characteristic (ROC) curves were employed to determine the predictive value of AIP and AIP combined with LDL-C for acute myocardial infarction. Result The results of the multivariate logistic regression analysis indicated that the AIP was an independent predictor of acute myocardial infarction. The optimal cut-off value for AIP to predict AMI was -0.06142, with a sensitivity of 81.3%, a specificity of 65.8%, and an area under the curve (AUC) of 0.801 (95% confidence interval [CI]: 0.743-0.859, P < 0.001). When AIP was combined with LDL-C, the best cut-off value for predicting acute myocardial infarction was 0.756107, with a sensitivity of 79%, a specificity of 74%, and an AUC of 0.819 (95% CI: 0.759-0.879, P < 0.001). Conclusions The AIP is considered an autonomous determinant of risk for AMI. Utilizing the AIP index alone, as well as in conjunction with LDL-C, can serve as effective predictors of AMI.
Collapse
Affiliation(s)
- Min Chen
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Cao-yang Fang
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jiong-chao Guo
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, China
| | - La-mei Pang
- Department of Endocrinology, Hefei BOE Hospital, Hefei, China
| | - Yuan Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, China
| | - Yu Hong
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, China
| | - Lin-fei Yang
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Jing Zhang
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Ting Zhang
- Department of Cardiology, The Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Bing-feng Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, China
| | - Guang-quan Hu
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Yang HH, Wang X, Li S, Liu Y, Akbar R, Fan GC. Lipocalin family proteins and their diverse roles in cardiovascular disease. Pharmacol Ther 2023; 244:108385. [PMID: 36966973 PMCID: PMC10079643 DOI: 10.1016/j.pharmthera.2023.108385] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
The lipocalin (LCN) family members, a group of small extracellular proteins with 160-180 amino acids in length, can be detected in all kingdoms of life from bacteria to human beings. They are characterized by low similarity of amino acid sequence but highly conserved tertiary structures with an eight-stranded antiparallel β-barrel which forms a cup-shaped ligand binding pocket. In addition to bind small hydrophobic ligands (i.e., fatty acids, odorants, retinoids, and steroids) and transport them to specific cells, lipocalins (LCNs) can interact with specific cell membrane receptors to activate their downstream signaling pathways, and with soluble macromolecules to form the complex. Consequently, LCNs exhibit great functional diversity. Accumulating evidence has demonstrated that LCN family proteins exert multiple layers of function in the regulation of many physiological processes and human diseases (i.e., cancers, immune disorders, metabolic disease, neurological/psychiatric disorders, and cardiovascular disease). In this review, we firstly introduce the structural and sequence properties of LCNs. Next, six LCNs including apolipoprotein D (ApoD), ApoM, lipocalin 2 (LCN2), LCN10, retinol-binding protein 4 (RBP4), and Lipocalin-type prostaglandin D synthase (L-PGDS) which have been characterized so far are highlighted for their diagnostic/prognostic values and their potential effects on coronary artery disease and myocardial infarction injury. The roles of these 6 LCNs in cardiac hypertrophy, heart failure, diabetes-induced cardiac disorder, and septic cardiomyopathy are also summarized. Finally, their therapeutic potential for cardiovascular disease is discussed in each section.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Siru Li
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yueying Liu
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
31
|
Yang Y, Shao M, Cheng W, Yao J, Ma L, Wang Y, Wang W. A Pharmacological Review of Tanshinones, Naturally Occurring Monomers from Salvia miltiorrhiza for the Treatment of Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3801908. [PMID: 36793978 PMCID: PMC9925269 DOI: 10.1155/2023/3801908] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/23/2022] [Accepted: 11/25/2022] [Indexed: 02/09/2023]
Abstract
Cardiovascular diseases (CVDs) are a set of heart and blood vessel disorders that include coronary heart disease (CHD), rheumatic heart disease, and other conditions. Traditional Chinese Medicine (TCM) has definite effects on CVDs due to its multitarget and multicomponent properties, which are gradually gaining national attention. Tanshinones, the major active chemical compounds extracted from Salvia miltiorrhiza, exhibit beneficial improvement on multiple diseases, especially CVDs. At the level of biological activities, they play significant roles, including anti-inflammation, anti-oxidation, anti-apoptosis and anti-necroptosis, anti-hypertrophy, vasodilation, angiogenesis, combat against proliferation and migration of smooth muscle cells (SMCs), as well as anti-myocardial fibrosis and ventricular remodeling, which are all effective strategies in preventing and treating CVDs. Additionally, at the cellular level, Tanshinones produce marked effects on cardiomyocytes, macrophages, endothelia, SMCs, and fibroblasts in myocardia. In this review, we have summarized a brief overview of the chemical structures and pharmacological effects of Tanshinones as a CVD treatment to expound on different pharmacological properties in various cell types in myocardia.
Collapse
Affiliation(s)
- Ye Yang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
| | - Mingyan Shao
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenkun Cheng
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Junkai Yao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
| | - Lin Ma
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
- Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
32
|
Wu Q, Lv Q, Liu X, Ye X, Cao L, Wang M, Li J, Yang Y, Li L, Wang S. Natural compounds from botanical drugs targeting mTOR signaling pathway as promising therapeutics for atherosclerosis: A review. Front Pharmacol 2023; 14:1083875. [PMID: 36744254 PMCID: PMC9894899 DOI: 10.3389/fphar.2023.1083875] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease that is a major cause of cardiovascular diseases (CVDs), including coronary artery disease, hypertension, myocardial infarction, and heart failure. Hence, the mechanisms of AS are still being explored. A growing compendium of evidence supports that the activity of the mechanistic/mammalian target of rapamycin (mTOR) is highly correlated with the risk of AS. The mTOR signaling pathway contributes to AS progression by regulating autophagy, cell senescence, immune response, and lipid metabolism. Various botanical drugs and their functional compounds have been found to exert anti- AS effects by modulating the activity of the mTOR signaling pathway. In this review, we summarize the pathogenesis of AS based on the mTOR signaling pathway from the aspects of immune response, autophagy, cell senescence, and lipid metabolism, and comb the recent advances in natural compounds from botanical drugs to inhibit the mTOR signaling pathway and delay AS development. This review will provide a new perspective on the mechanisms and precision treatments of AS.
Collapse
Affiliation(s)
- Qian Wu
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Qianyu Lv
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao’an Liu
- Capital University of Medical, Beijing, China
| | - Xuejiao Ye
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Linlin Cao
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Manshi Wang
- Beijing Xicheng District Guangwai Hospital, Beijing, China
| | - Junjia Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Yingtian Yang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Lanlan Li
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Wang
- Guang’anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
33
|
Pan K, Xu C, Chen C, Chen S, Zhang Y, Ding X, Xu X, Lv Q. Soluble interleukin-2 receptor combined with interleukin-8 is a powerful predictor of future adverse cardiovascular events in patients with acute myocardial infarction. Front Cardiovasc Med 2023; 10:1110742. [PMID: 37139133 PMCID: PMC10150071 DOI: 10.3389/fcvm.2023.1110742] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/21/2023] [Indexed: 05/05/2023] Open
Abstract
Background Little is known about the role of interleukin (IL) in patients with acute myocardial infarction (MI), especially soluble IL-2 receptor (sIL-2R) and IL-8. We aim to evaluate, in MI patients, the predictive value of serum sIL-2R and IL-8 for future major adverse cardiovascular events (MACEs), and compare them with current biomarkers reflecting myocardial inflammation and injury. Methods This was a prospective, single-center cohort study. We measured serum concentrations of IL-1β, sIL-2R, IL-6, IL-8 and IL-10. Levels of current biomarkers for predicting MACEs were measured, including high-sensitivity C reactive protein, cardiac troponin T and N-terminal pro-brain natriuretic peptide. Clinical events were collected during 1-year and a median of 2.2 years (long-term) follow-up. Results Twenty-four patients (13.8%, 24/173) experienced MACEs during 1-year follow-up and 40 patients (23.1%, 40/173) during long-term follow-up. Of the five interleukins studied, only sIL-2R and IL-8 were independently associated with endpoints during 1-year or long-term follow-up. Patients with high sIL-2R or IL-8 levels (higher than the cutoff value) had a significantly higher risk of MACEs during 1-year (sIL-2R: HR 7.7, 3.3-18.0, p < 0.001; IL-8: HR 4.8, 2.1-10.7, p < 0.001) and long-term (sIL-2R: HR 7.7, 3.3-18.0, p < 0.001; IL-8: HR 4.8, 2.1-10.7, p < 0.001) follow-up. Receiver operator characteristic curve analysis regarding predictive accuracy for MACEs during 1-year follow-up showed that the area under the curve for sIL-2R, IL-8, sIL-2R combined with IL-8 was 0.66 (0.54-0.79, p = 0.011), 0.69 (0.56-0.82, p < 0.001) and 0.720 (0.59-0.85, p < 0.001), whose predictive value were superior to that of current biomarkers. The addition of sIL-2R combined with IL-8 to the existing prediction model resulted in a significant improvement in predictive power (p = 0.029), prompting a 20.8% increase in the proportion of correct classifications. Conclusions High serum sIL-2R combined with IL-8 levels was significantly associated with MACEs during follow-up in patients with MI, suggesting that sIL-2R combined with IL-8 may be a helpful biomarker for identifying the increased risk of new cardiovascular events. IL-2 and IL-8 would be promising therapeutic targets for anti-inflammatory therapy.
Collapse
Affiliation(s)
- Kunming Pan
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Chenqi Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
| | - Can Chen
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuqing Chen
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqian Zhang
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Kidney Disease and Dialysis, Shanghai, China
- Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Correspondence: Qianzhou Lv Xialian Xu
| | - Qianzhou Lv
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
- Correspondence: Qianzhou Lv Xialian Xu
| |
Collapse
|